28.11.2012 Views

Contents - College of Medical and Dental Sciences - University of ...

Contents - College of Medical and Dental Sciences - University of ...

Contents - College of Medical and Dental Sciences - University of ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Workshop programme at a glance<br />

Scientific programme committee<br />

Organisers<br />

<strong>Contents</strong><br />

Confidentiality <strong>of</strong> results presented at this meeting<br />

Meeting venue <strong>and</strong> campus accommodation<br />

Meeting information including venues <strong>of</strong> meals<br />

Travel directions <strong>and</strong> maps<br />

Instructions for oral <strong>and</strong> poster presentations<br />

Details <strong>of</strong> excursion to Stratford Upon Avon<br />

Meeting agenda, including details <strong>of</strong> coach transport<br />

Detailed scientific meeting agenda<br />

Oral presentation abstracts<br />

Session 1 abstracts 1-7: Latency<br />

Session 2 abstracts 8-13: Immunology I<br />

Session 3 abstracts 14-21: Pathogenesis<br />

Session 4 abstracts 22-28: Virus-Cell Interactions I<br />

Session 5 abstracts 29-34: Gene Expression I<br />

Session 6 abstracts 35-41: Lytic Replication<br />

Session 7 abstracts 42-47: Immunology II<br />

Session 8 abstracts 48-55: Clinical & Epidemiology<br />

Session 10 abstracts 56-63: Virus-Cell Interactions II<br />

Session 11 abstracts 64-67: Gene Expression II<br />

Poster presentation abstracts P1-P18<br />

Delegate contact details<br />

Author index<br />

Page<br />

2<br />

3<br />

3<br />

4<br />

5<br />

6<br />

7<br />

10<br />

10<br />

11<br />

12<br />

19<br />

20<br />

28<br />

35<br />

44<br />

52<br />

59<br />

67<br />

74<br />

83<br />

92<br />

97<br />

116<br />

128<br />

1


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Workshop programme at a glance 1<br />

Tuesday<br />

Time<br />

July 22 nd<br />

Wednesday<br />

July 23 rd<br />

Thursday<br />

July 24 th<br />

Friday<br />

July 25th<br />

7:00-8:00 Breakfast<br />

Session 1<br />

Session 4<br />

Session 6<br />

8:45-10:30<br />

Latency<br />

Virus-Cell<br />

Interactions I<br />

Lytic Replication<br />

(abstracts 1-7) (abstracts 22-28) (abstracts 35-41)<br />

10:30-11:00 Tea/C<strong>of</strong>fee<br />

Session 2<br />

Session 5<br />

Session 7<br />

11:00-12:30<br />

Immunology I Gene Expression I Immunology II<br />

Saturday<br />

July 26 th<br />

Session 10<br />

Virus-Cell<br />

Interactions II<br />

(abstracts 56-63)<br />

Session 11<br />

Gene Expression II<br />

(abstracts 8-13) (abstracts 29-34) (abstracts 42-47) (abstracts 64-67)<br />

12:30-1:30 Lunch Workshop ends<br />

Session 3<br />

Session 8<br />

1:30-3:30<br />

Pathogenesis<br />

Clinical &<br />

Excursion to Epidemiology<br />

(abstracts 14-21) Stratford Upon (abstracts 48-55)<br />

3:30-4:00<br />

Poster session<br />

Avon<br />

Tea/C<strong>of</strong>fee<br />

5:30-8:30<br />

3:30-6:00<br />

Sponsored by<br />

Session 9<br />

4:00-5:30 Registration (abstracts P1-P18) Geneflow<br />

Round Table<br />

& welcome<br />

Sponsored by<br />

reception buffet<br />

Agilent<br />

Barber Institute Technologies<br />

Evening<br />

8:30<br />

8:00<br />

7:30<br />

Indian meal<br />

BBQ<br />

Conference<br />

Shimla Pinks Shackleton House<br />

dinner<br />

Sponsored by Edgbaston Cricket<br />

Miltenyi Biotec<br />

Ground<br />

Sponsored by BD<br />

Biosciences<br />

1<br />

Oral sessions 1-11 to be held in the Leonard Deacon lecture theatre, Wolfson Centre, The <strong>Medical</strong> School, <strong>University</strong> <strong>of</strong> Birmingham<br />

Poster session to be held on the Ground Floor <strong>of</strong> the Wolfson Centre<br />

2


Scientific Programme Committee<br />

David Blackbourn, PhD<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Juergen Haas, MD, PhD<br />

<strong>University</strong> <strong>of</strong> Edinburgh<br />

Paul Kellam, PhD<br />

<strong>University</strong> <strong>College</strong> London<br />

Frank Niepel, MD<br />

<strong>University</strong> <strong>of</strong> Erlangen<br />

Thomas Schulz, MD, PhD<br />

Hannover <strong>Medical</strong> School<br />

Scott Wong, PhD<br />

Oregon Health &Science <strong>University</strong>/Vaccine<br />

& Gene Therapy Institute<br />

Organisers<br />

David Blackbourn<br />

Co-chair, Organising Committee<br />

Cancer Research UK Institute for Cancer<br />

Studies<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Vincent Drive<br />

Edgbaston<br />

Birmingham<br />

B15 2TT<br />

United Kingdom<br />

Tel. +44 (0) 121 415 8804<br />

d.j.blackbourn@bham.ac.uk<br />

Conference Coordinators<br />

& Organising Committee<br />

Pr<strong>of</strong>essional Development Centre<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Birmingham<br />

B15 2TT<br />

United Kingdom<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Ethel Cesarman, MD, PhD<br />

Weill Cornell <strong>Medical</strong> <strong>College</strong>,<br />

Andrew Hislop, PhD<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

John Nicholas, PhD<br />

Johns Hopkins <strong>University</strong><br />

Rolf Renne, PhD<br />

<strong>University</strong> <strong>of</strong> Florida<br />

Adrian Whitehouse, PhD<br />

<strong>University</strong> <strong>of</strong> Leeds<br />

Paul Kellam<br />

Co-chair, Organising Committee<br />

Centre for Virology (Bloomsbury)<br />

<strong>University</strong> <strong>College</strong> London<br />

46 Clevel<strong>and</strong> Street<br />

London<br />

W1T 4JF<br />

United Kingdom<br />

Tel. +44 (0) 20 7679 9559<br />

p.kellam@ucl.ac.uk<br />

3


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Confidentiality <strong>of</strong> results presented at this meeting<br />

To continue in the sprit <strong>of</strong> the highly successful Workshops in the past, we hope that this 11 th<br />

International Workshop on KSHV <strong>and</strong> Related Agents will provide ample opportunity for informal<br />

discussions <strong>and</strong> exchange <strong>of</strong> ideas, data <strong>and</strong> reagents. To promote such interactions, <strong>and</strong> to<br />

encourage participants to present unpublished results, please treat all scientific presentations <strong>and</strong><br />

abstracts in this book as confidential unpublished research. Abstracts may not be cited. If you want to<br />

cite results, please do so as a “personal communication”, only with agreement from the respective<br />

authors. Results presented at this meeting may not be used as the basis <strong>of</strong> research without the<br />

explicit permission <strong>of</strong> the corresponding author. The KSHV field has already benefited from many<br />

collaborative ventures <strong>and</strong> a relatively free flow <strong>of</strong> reagents. Let’s encourage presentation <strong>of</strong> recent<br />

data <strong>and</strong> establish the KSHV workshops as an exciting series <strong>of</strong> meetings that are worth attending<br />

<strong>and</strong> cannot be replaced by scientific journals.<br />

4


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Welcome to the 11 th International Workshop on KSHV <strong>and</strong> Related Agents at<br />

the <strong>University</strong> <strong>of</strong> Birmingham, <strong>Medical</strong> School, Birmingham, Engl<strong>and</strong>, 22-26<br />

July 2008.<br />

Meeting Venue<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

<strong>Medical</strong> School<br />

Vincent Drive<br />

Edgbaston<br />

Birmingham<br />

B15 2TT<br />

Email – med-pdcbookings@bham.ac.uk<br />

Tel – +44 (0)121 414 8608<br />

Scientific sessions will be held in the Wolfson Centre <strong>of</strong> the <strong>Medical</strong> School:<br />

• The oral sessions will be held in the Leonard Deacon lecture theatre, Wolfson Centre<br />

• The poster session will be held on the Ground Floor <strong>of</strong> the Wolfson Centre<br />

Campus Accommodation<br />

Shackleton<br />

The Vale<br />

Edgbaston Park Road<br />

Birmingham<br />

B15 3SZ<br />

Check in from 2pm on 22 nd July<br />

Internet access available in all rooms, please ask at Shackleton reception<br />

24 hour reception<br />

Breakfast will be served from 7am; coaches will then transfer delegates to the conference please be at<br />

reception for 8am.<br />

5


Name Badges<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Please wear your name badge at all times during the conference.<br />

Meals<br />

The following meals are included in your registration:<br />

Welcome Reception<br />

Tuesday, 5.30pm – 8.30pm – Barber Institute, <strong>University</strong> <strong>of</strong> Birmingham, Edgbaston, Birmingham, B15<br />

2TS, Tel 0121 414 7333.<br />

Lunch<br />

Wednesday<br />

Thursday<br />

Friday<br />

Evening Meals<br />

Wednesday, 8pm – 11pm – Shimla Pinks, 214 Broad St, Birmingham, B15 1AY,<br />

Tel 0121 633 0366<br />

Thursday, 8pm – BBQ at Rio’s Bar, The Vale, Shackleton, Edgbaston Park Road, Edgbaston,<br />

Birmingham, B15 3SZ. Generously sponsored by Miltenyi Biotec.<br />

Friday, 7pm -11pm – Conference dinner at Edgbaston Cricket Ground, County Ground<br />

Edgbaston Rd, Birmingham, B5 7QU, Tel 0870 062 1902. Generously sponsored by BD Biosciences.<br />

Transport<br />

For those staying at Shackleton coaches have been arranged to transport delegates between<br />

accommodation <strong>and</strong> restaurant locations. Please wait for the coach at the times stated on the<br />

agenda.<br />

Please note coaches have been arranged for those delegates staying at Shackleton only.<br />

Delegates staying elsewhere must arrange own transport.<br />

Taxis - TOA – 0121 427 8888<br />

6


Travel Directions <strong>and</strong> Maps<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

7


By motorway<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Approaching from the north west or south east along the M6:<br />

• Leave at Junction 6 (signposted Birmingham Central) to join the A38(M)<br />

• At the end <strong>of</strong> the motorway, keep to the right, go over a flyover, then through some underpasses<br />

to join the A38 Bristol Road<br />

• The <strong>University</strong> is on your right, two <strong>and</strong> a half miles from the city centre<br />

Approaching from the M42 north:<br />

• Leave at Junction 8 to join the M6 northbound <strong>and</strong> follow the instructions above<br />

Approaching from the south west:<br />

• Leave the M5 at Junction 4 signposted Birmingham (SW) to join the A38<br />

• The <strong>University</strong> is approximately eight miles from the motorway<br />

Approaching from the M40:<br />

• It is easier to turn south on the M42 <strong>and</strong> leave at Junction 1, heading north on the A38 Bristol<br />

Road<br />

• The <strong>University</strong> is approximately eight miles from the motorway<br />

8


By rail<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Most cross-country services to Birmingham arrive at New Street Station. Up to six trains an hour<br />

depart for the <strong>University</strong> on the cross-city line (ten minutes to <strong>University</strong> station, final destination<br />

Longbridge or Redditch). The centre <strong>of</strong> the main campus is a five-minute walk from <strong>University</strong> Station.<br />

By coach<br />

There are frequent express coach services to Birmingham from London, Heathrow <strong>and</strong> Gatwick<br />

Airports, <strong>and</strong> many UK cities. The long-distance coach station is in Digbeth in the city centre.<br />

By bus<br />

Numbers 61, 62 <strong>and</strong> 63 travel to the <strong>University</strong>’s Edgbaston <strong>and</strong> Selly Oak campuses, while the 21<br />

<strong>and</strong> 44 serve the <strong>Medical</strong> School <strong>and</strong> Queen Elizabeth Hospital. The services all run frequently from<br />

the city centre. There is a travel information <strong>of</strong>fice outside New Street Station, where you can obtain<br />

bus timetables <strong>and</strong> departure point information. Maps can be found throughout the city centre<br />

indicating bus stop locations.<br />

By taxi<br />

There are taxi ranks at New Street Station <strong>and</strong> throughout the city centre. The journey to the<br />

<strong>University</strong> takes about ten minutes.<br />

By air<br />

Birmingham International Airport has direct flights from locations in the UK, as well as from the<br />

USA, Canada, Europe <strong>and</strong> the Middle East.<br />

The journey by taxi from the airport to the <strong>University</strong> takes approximately half an hour. Alternatively,<br />

Air-Rail Link provides a free, fast connection between the airport terminals <strong>and</strong> Birmingham<br />

International railway station. Air-Rail Link operates every two minutes (journey time 90 seconds).<br />

Birmingham International railway station has frequent services to New Street Station in the city centre<br />

(journey time around 15 minutes).<br />

If you are arriving at London, there is a frequent train service from London Euston railway station to<br />

New Street Station (journey time around 1 hour 30 minutes).<br />

• From Heathrow Airport. Take the Heathrow Express train to Paddington Station <strong>and</strong> then the<br />

Underground or a taxi to Euston Station. Alternatively, an Airbus runs from Heathrow Airport direct<br />

to Euston Station<br />

• From Gatwick Airport. Take the Airport Express train to Victoria Station <strong>and</strong> then the Underground<br />

or a taxi to Euston Station<br />

9


Oral Presentations<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Oral presentations are strictly limited to 12 minutes, allowing an additional 1 – 2 minutes for questions.<br />

Both PC <strong>and</strong> Macintosh computers are available for oral presentations in the Leonard Deacon lecture<br />

theatre, where the presentations will be made.<br />

Powerpoint presentations should be brought on USB stick <strong>and</strong> loaded by the times stated below.<br />

Identify the presentation with the abstract number, given previously, <strong>and</strong> with the last name <strong>of</strong><br />

the presenting author.<br />

To facilitate smooth running <strong>of</strong> the scientific sessions, the use <strong>of</strong> personal laptops for presentations is<br />

actively discouraged.<br />

Timetable for uploading presentations to the podium computers in the Leonard Deacon lecture<br />

theatre:<br />

Session Abstract numbers Upload by<br />

1 1-7 8:45 am, Wed July 23 rd<br />

2 8-13 11:00 am, Wed July 23 rd<br />

3 14-21 1:30 pm, Wed July 23 rd<br />

4 22-28 8:45 am, Thurs July 24 th<br />

5 29-34 11:00 am, Thurs July 24 th<br />

6 35-41 8:45 am, Fri July 25 th<br />

7 42-47 11:00 am, Fri July 25 th<br />

8 48-55 1:30 pm, Fri July 25 th<br />

10 56-63 8:45 am, Sat July 26 th<br />

11 64-67 11:00 am, Sat July 26 th<br />

Poster Viewing <strong>and</strong> Poster Session<br />

Generously sponsored by Agilent Technologies<br />

The poster board dimensions are 1m x 1m. Please identify in the poster title the abstract number,<br />

given previously.<br />

Posters can be placed on Wednesday 23 July from 8:30am. The poster boards are located on the<br />

Ground Floor in the Wolfson Centre. Posters should be removed by 5:00 pm Friday July 25 th .<br />

Poster <strong>and</strong> Drinks Reception, Wolfson Centre, Wednesday 23 July, 3:30 pm – 6:00 pm<br />

Excursion to Stratford Upon Avon<br />

Generously sponsored by Geneflow<br />

An excursion to Stratford Upon Avon has been arranged for Thursday 24 July, the coaches will depart<br />

from the <strong>Medical</strong> School following the morning session, at 12.30 sharp. A packed lunch will be<br />

provided, collect this on your way out. Please ensure you <strong>and</strong> your companions have your passes<br />

ready to board the coach.<br />

Upon arrival at Stratford you will be met by guides, the town walk will take in the three Shakespeare<br />

town houses, Shakespeare's Birthplace, Nahs's House <strong>and</strong> the site <strong>of</strong> the New Place <strong>and</strong> Hall's Cr<strong>of</strong>t,<br />

the Royal Shakespeare Theatres, Shakespeare's Grammar School <strong>and</strong> Holy Trinity Church, where<br />

William Shakespeare is buried. It is a very lively, entertaining <strong>and</strong> informative two hours.<br />

After the tour you will have the opportunity to explore the town for yourself, enjoying the views <strong>and</strong> an<br />

excellent variety <strong>of</strong> shops, from small individual retailers to major outlets, ideal for window shopping or<br />

browsing. Alternatively, perhaps you would prefer to relax <strong>and</strong> enjoy the views by the River Avon.<br />

Please ensure you are back at the collection point by 4.45pm, when the coaches will return to<br />

Shackleton.<br />

10


Agenda<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Date Time Event<br />

Tuesday 22nd 5.30 - 8.30pm<br />

Welcome Reception, Barber Institute, <strong>University</strong><br />

Campus<br />

5.30 – 8.30pm Shuttle coach between Shackleton & Barber Institute<br />

Wednesday 23rd 8.00am Coach pick up at Shackleton<br />

8.45 – 10.30am Session 1 – Latency (abstracts 1-7)<br />

10.30 – 11.00am Break<br />

11.00am - 12.30pm Session 2 – Immunology I (abstracts 8-13)<br />

12.30 - 1.30pm Lunch<br />

1.30 - 3.30pm Session 3 – Pathogenesis (abstracts 14-21)<br />

3.30 – 6.00pm Posters & Drinks Reception<br />

6.00pm Coach pick up at <strong>Medical</strong> School return to Shackleton<br />

8.00pm Coach pick up at Shackleton<br />

8.30 – 11.30pm Evening Meal - Shimla Pinks Restaurant<br />

11.30pm Coach pick up Shimla Pinks, Broad Street<br />

Thursday 24th 8.00am Coach pick up at Shackleton<br />

8.45 – 10.30am<br />

Session 4 – Virus-Cell Interactions<br />

(abstracts 22-28)<br />

10.30 - 11am Break<br />

11.00am - 12.30pm<br />

Session 5 – Gene Expression<br />

(abstracts 29-34)<br />

12.30 - 6pm Excursion - Stratford Upon Avon<br />

4.45pm Coach pick up Stratford Upon Avon<br />

8.00pm Evening Meal – BBQ, Shackleton<br />

Friday 25th 8.00am Coach pick up at Shackleton<br />

8.45 – 10.30am<br />

Session 6 – Lytic Replication<br />

(abstracts 35-41)<br />

10.30 – 11.00am Break<br />

11.00 - 12.30pm Session 7 – Immunology II (abstracts 42-47)<br />

12.30 - 1.30pm Lunch<br />

1.30 - 3.30pm Session 8 – Clinical & Epidemiology (abstracts 48-55)<br />

3.30 - 4.30pm Session 9 – Round Table<br />

5.30pm Coach pick up at <strong>Medical</strong> School return to Shackleton<br />

7.00pm Coach pick up at Shackleton<br />

7.30 – 11.00pm Evening Meal - Edgbaston Cricket Ground<br />

11.00pm Coach pick up Cricket Ground return to Shackleton<br />

Saturday 26th 7.30 - 8.30am Coach pick up at Shackleton<br />

8.45 – 10.30am Session 10 – Virus-Cell Interactions II (abstracts 56-63)<br />

10.30 - 11am Break<br />

11.00am - 12.30pm<br />

Session 11 – Gene Expression II<br />

(abstracts 64-67)<br />

12.30pm Conference Close<br />

11


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Detailed Scientific Meeting<br />

Agenda<br />

12


Wednesday, July 23 rd<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 1: Latency<br />

8:45 am – 10:30 am<br />

Chair: Rolf Renne<br />

Abstract # Title Authors<br />

1 DELETION OF LANA FROM RHESUS<br />

RHADINOVIRUS (RRV) GENERATES A<br />

HIGHLY LYTIC RECOMBINANT VIRUS<br />

2 DIVERGENCE OF THE NUCLEAR<br />

LOCALIZATION SIGNAL WITHIN THE<br />

ORF73 LATENCY-ASSOCIATED NUCLEAR<br />

ANTIGENS (LANA) OF KSHV AND THE<br />

MACAQUE RV1 AND RV2<br />

3<br />

RHADINOVIRUSES<br />

NUCLEOPHOSMIN IS A NOVEL<br />

REGULATOR OF KSHV REPLICATION VIA<br />

FUNCTIONAL INTERACTIONS WITH VIRAL<br />

CYCLIN AND LANA<br />

4 ROLE OF BET PROTEINS IN THE<br />

FUNCTION OF KSHV AND MHV68 LANA<br />

5 INTERACTION OF KSHV LANA-1 WITH<br />

USP7: IMPLICATIONS FOR P53 FUNCTION<br />

6 KSHV ENCODED LANA INTERACTS WITH<br />

THE NUCLEAR MITOTIC APPARATUS<br />

PROTEIN TO REGULATE GENOME<br />

7<br />

MAINTENANCE AND SEGREGATION<br />

MULTIPLE DOMAINS CONTRIBUTE TO THE<br />

ASSOCIATION OF LANA WITH HOST<br />

CHROMATIN COMPONENTS<br />

Session 2: Immunology I<br />

11:00 am – 12:30 pm<br />

Chair: Andrew Hislop<br />

Abstract # Title Authors<br />

8 INDUCTION OF CYTOKINES BY HHV-8 IN<br />

MYELOID DENDRITIC CELLS AND B CELLS<br />

9 KSHV K5 COUNTERACTS TETHERIN, A<br />

NOVEL COMPONENT OF INNATE<br />

ANTIVIRAL IMMUNITY, TO FACILITATE<br />

VIRUS RELEASE<br />

10 MOLECULAR MECHANISM OF<br />

11<br />

BST2/TETHERIN DOWNREGULATION BY<br />

KSHV-K5<br />

UPREGULATION OF THE TLR3 PATHWAY<br />

BY KSHV DURING PRIMARY INFECTION<br />

OF MONOCYTES<br />

12 KSHV EVADES INNATE IMMUNITY BY<br />

SUPPRESSION OF TLR4 EXPRESSION<br />

13 THE KSHV LYTIC PROTEIN VOX2 AND ITS<br />

CELLULAR COUNTERPART, CD200,<br />

INHIBIT EPITOPE-SPECIFIC T CELL<br />

RESPONSES<br />

Kwun Wah Wen, Chelsey Hilscher, Dirk P.<br />

Dittmer <strong>and</strong> Blossom Damania<br />

Kellie L. Burnside <strong>and</strong> Timothy M. Rose<br />

Grzegorz Sarek, Annika Järviluoma, Henna<br />

Syrjäkari, Sari Tojk<strong>and</strong>er, Salla Vartia,<br />

Marikki Laiho, <strong>and</strong> Päivi M. Ojala<br />

Magdalena Weidner-Glunde, Matthias<br />

Ottinger, Daniel Pliquet, Ronald Frank <strong>and</strong><br />

Thomas F. Schulz<br />

W. Albrecht, E. Gellermann, A. Viejo-<br />

Borbolla, T. F. Schulz<br />

Huaxin Si, Subhash C. Verma, Michael<br />

Lampson, Qiliang Cai, Erle S. Robertson<br />

Ryo Nasu, Satoko Matsumura, Naoko Tanese<br />

<strong>and</strong> Angus Wilson<br />

Emilee Knowlton, Giovanna Rappocciolo,<br />

Paolo Piazza, Heather Hensler, Frank J.<br />

Jenkins, Mariel Jais, Charles R. Rinaldo<br />

Edward Tsao, Sam Wilson, Claire Pardieu,<br />

Ben Webb, Imogen Lai, Stuart Neil, Greg<br />

Towers, <strong>and</strong> Paul Kellam<br />

M<strong>and</strong>ana Mansouri, Janet Douglas, Kasinath<br />

Viswanathan, Jean Gustin, Ashlee Moses <strong>and</strong><br />

Klaus Früh<br />

John West, Sean Gregory <strong>and</strong> Blossom<br />

Damania<br />

Dimitris Lagos, Richard James Vart, Fiona<br />

Gratrix, Samantha Jane Westrop, Ping-Pui<br />

Wong, Nesrina Imami, Mark Bower, Frances<br />

Gotch <strong>and</strong> Chris Bosh<strong>of</strong>f<br />

Rachel Colman, Karen Misstear, Heather<br />

Long, Omar Quereshi, David Sansom,<br />

Andrew Hislop, David J. Blackbourn<br />

13


Wednesday, July 23 rd<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 3: Pathogenesis<br />

1:30 am – 3:30 pm<br />

Chair: David Blackbourn<br />

Abstract # Title Authors<br />

14 GENERATION OF RHESUS RHADINOVIRUS<br />

LACKING EXPRESSION OF VGPCR AND<br />

VCD200 FOR THE ASSESSMENT OF THEIR<br />

ROLES IN DISEASE DEVELOPMENT IN A<br />

RHESUS MACAQUE MODEL OF KSHV<br />

15<br />

INFECTION<br />

GENERATION OF VFLIP TRANSGENIC<br />

MICE: A MODEL TO STUDY KSHV-<br />

ASSOCIATED LYMPHOMAGENESIS<br />

16 DEVELOPMENT OF ANIMAL MODELS OF<br />

KSHV INFECTION AND AIDS-ASSOCIATED<br />

DISEASE BASED ON MACAQUE<br />

RHADINOVIRUSES<br />

17 β ARRESTINS REGULATE KSHV GPCR<br />

ACTIVITY AND ARE MODULATED BY<br />

CANNABINOIDS<br />

18 ACTIVATION OF NF-⎢B BY KSHV K15<br />

19<br />

INVOLVES RECRUITMENT OF NIK (NF-ΚB<br />

INDUCING KINASE)<br />

KAPOSI'S SARCOMA ASSOCIATED HERPES<br />

VIRUS (KSHV/HHV-8) INDUCES<br />

ANGIOGENIN DURING INFECTION OF<br />

HUMAN DERMAL MICROVASCULAR<br />

ENDOTHELIAL (HMVEC-D) CELLS THAT IS<br />

CRITICAL FOR ANTI-APOPTOSIS, CELL<br />

20<br />

PROLIFERATION AND ANGIOGENESIS<br />

ROLE OF K15 IN KAPOSI’S SARCOMA<br />

HERPESVIRUS INDUCED ENDOTHELIAL<br />

CELL MIGRATION<br />

21 DOWNREGULATION OF GALECTIN-3 IN<br />

KSHV INFECTED DMVEC CELLS AND<br />

KAPOSI’S SARCOMA: IMPLICATIONS FOR<br />

TUMORIGENESIS<br />

Ryan D. Estep, Elisa Cardenas <strong>and</strong> Scott W.<br />

Wong<br />

Gianna Ballon, Amy Chadburn, Yi-Fang Liu,<br />

Yoshiteru Sasaki, Klaus Rajewsky, Ethel<br />

Cesarman<br />

A. Gregory Bruce, Jonathan T. Ryan,<br />

Courtney Gravett <strong>and</strong> Timothy M. Rose<br />

Xuefeng Zhang, Yehoshua Maor, Jerome E.<br />

Groopman<br />

Anika Hävemeier, Macel Pietrek <strong>and</strong> Thomas<br />

F. Schulz<br />

Sathish Sadagopan, Neelam-Sharma Walia,<br />

Mohanan Valiya Veettil, Virginie Bottero, Rita<br />

Levine <strong>and</strong> Bala Ch<strong>and</strong>ran<br />

Bernd Hillenbr<strong>and</strong>, Antje Bürger, Irina<br />

Fischer, Khaled Alkharsah <strong>and</strong> Thomas F.<br />

Schulz<br />

Donald J. Alcendor Wen Qui Zhu Prashant<br />

Desai <strong>and</strong> Gary S. Hayward<br />

14


Thursday, July 24 th<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 4: Virus-Cell Interactions I<br />

8:45 am – 10:30 am<br />

Chair: Scott Wong<br />

Abstract # Title Authors<br />

22 ANIMAL MODELS OF KAPOSI'S SARCOMA<br />

REVEAL A ROLE OF KSHV VGPCR AND<br />

RAC1 IN ANGIOGENESIS AND GENETIC<br />

INSTABILITY<br />

23 INSIGHTS INTO KSHV ACTIVATION OF<br />

THE IKK SIGNALOSOME: CRYSTAL<br />

24<br />

STRUCTURE OF A VFLIP-IKKγ COMPLEX<br />

THE VIRAL INHIBITOR OF APOPTOSIS<br />

VFLIP/K13 PROTECTS ENDOTHELIAL<br />

CELLS AGAINST SUPEROXIDE - INDUCED<br />

CELL DEATH<br />

25 ROLE OF DEFECTIVE OCT-2 AND OCA-B<br />

EXPRESSION IN IMMUNOGLOBULIN<br />

PRODUCTION AND KSHV LYTIC<br />

REACTIVATION IN PRIMARY EFFUSION<br />

LYMPHOMA<br />

26 KAPOSI’S SARCOMA ASSOCIATED<br />

HERPESVIRUS (KSHV) INDUCES<br />

TUMORIGENIC CELLULAR MIRNAS IN<br />

LATENTLY INFECTED ENDOTHELIAL CELLS<br />

27 KSHV AND CELLULAR MIRNA<br />

EXPRESSION DURING LATENCY AND<br />

LYTIC REACTIVATION IN PEL CELLS<br />

28 KAPOSI’S SARCOMA ASSOCIATED<br />

HERPESVIRUS (KSHV/HHV-8) UTILIZES<br />

MACROPINOCYTIC PATHWAY TO ENTER<br />

HUMAN DERMAL MICROVASCULAR<br />

ENDOTHELIAL (HMVEC-D) AND HUMAN<br />

UMBILICAL VEIN ENDOTHELIAL (HUVEC)<br />

CELLS<br />

Session 5: Gene Expression I<br />

11:00 am – 12:30 pm<br />

Chair: Adrian Whitehouse<br />

Abstract # Title Authors<br />

29 SCREENING FOR XINJIANG KS<br />

ASSOCIATED GENES AND THE STUDY ON<br />

THEIR MECHANISMS<br />

30 HOST CELL AND VIRAL MICRORNA<br />

(MIRNA) EXPRESSION IN B CELL<br />

31<br />

LYMPHOMAS<br />

CELLULAR AND VIRAL GENE EXPRESSION<br />

PROFILING<br />

MONOCYTES<br />

IN KSHV-INFECTED<br />

32 PROFILING OF CELLULAR AND VIRAL<br />

33<br />

MICRORNAS IN KAPOSI SARCOMA AND<br />

VIRAL-ASSOCIATED LYMPHOMA<br />

INEFFICIENT CODON USAGE IN V-FLIP<br />

MRNA LEADS TO TRANSCRIPT<br />

34<br />

INSTABILITY<br />

ROLE OF MURINE GAMMAHERPESVIRUS-<br />

68 ORF37 IN MEDIATING INHIBITION OF<br />

HOST GENE EXPRESSION<br />

Lucas Cavallin, Qi Ma, Rui Zhang, E.<br />

Margarita Duran, Pascal J. Goldschmidt-<br />

Clermont <strong>and</strong> Enrique A. Mesri<br />

Claire Bagnéris, Alex<strong>and</strong>er V Ageichik, Nora<br />

Cronin, Bonnie Wallace, Mary Collins, Chris<br />

Bosh<strong>of</strong>f, Gabriel Waksman <strong>and</strong> Tracey Barrett<br />

Michael Stürzl, Gaby S<strong>and</strong>er, Nathalie Gonin-<br />

Laurent, Kristina Weinländer, Elisabeth<br />

Naschberger, Ramona Jochmann, Khaled R.<br />

Alkharsah, Thomas F. Schulz, Margot Thome,<br />

Frank Neipel <strong>and</strong> Mathias Thurau<br />

Daniel DiBartolo, Elizabeth Hyjek, Shannon<br />

Keller, Ilaria Guasparri, Ren Sun, Amy<br />

Chadburn, Daniel M Knowles, <strong>and</strong> Ethel<br />

Cesarman<br />

Rebecca L. Skalsky Wendell Miley, Rachel<br />

Bagni, Soo-Jin Han Jianhong Hu, Chang Hee<br />

Kim, Rolf Renne <strong>and</strong> Denise Whitby<br />

Soo-Jin Han Jianhong Hu, Karlie Plaisance,<br />

Wendell Miley, Rachel Bagni, Chang Hee Kim,<br />

Denise Whitby <strong>and</strong> Rolf Renne<br />

Hari Raghu, Neelam Sharma Walia, Mohanan<br />

Valiya Veettil, Sathish Sadagopan <strong>and</strong> Bala<br />

Ch<strong>and</strong>ran<br />

Lei YANG, Yan ZENG, Dong-Mei LI,Ling-ling<br />

XIAN,Xiao –Fei ZHOU,Xiao-Hua TAN,Jin<br />

HUANG,Feng LI,Jian-Xin XIE,Hui<br />

ZHANG,Sheng Wang,Xian-Dao LUO<br />

Eve M Coulter, Dan Frampton, Ed Tsao, Paul<br />

Kellam<br />

Sean Gregory, Ling Wang, John West,<br />

Chelsey Hilscher, Dirk P. Dittmer <strong>and</strong><br />

Blossom Damania<br />

Andrea J. O’Hara, Bruce J. Dezube, William<br />

Harrington Jr., Blossom Damania, <strong>and</strong> Dirk P.<br />

Dittmer<br />

Priya Bellare, Andrew T Dufresne <strong>and</strong> Don<br />

Ganem<br />

L. Roaden, V. Sheridan, B. Lane, R. Sun, B.<br />

Dutia <strong>and</strong> B. Ebrahimi<br />

15


Friday July 25 th<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 6: Lytic Replication<br />

8:45 am – 10:30 am<br />

Chair: Thomas Schulz<br />

Abstract # Title Authors<br />

35 FUNCTIONAL CHARACTERIZATION OF Yan Wang, Charles Hollow <strong>and</strong> Yan Yuan<br />

KAPOSI'S SARCOMA–ASSOCIATED<br />

HERPESVIRUS<br />

MUTAGENESIS<br />

ORF K8 BY BAC-BASED<br />

36 APPLICATION OF ACTIVE KINOME<br />

COLLECTION FOR IDENTIFICATION OF A<br />

NOVEL KINASE FAMILY INVOLVED IN<br />

37<br />

REACTIVATION OF KSHV<br />

NOVEL FUNCTIONS OF K-RTA AND K-BZIP<br />

AS SUMO-TARGETING LIGASES AND<br />

38<br />

EPIGENETIC REGULATORS<br />

X-BOX BINDING PROTEIN 1 DOES NOT<br />

INDUCE EBV LYTIC REPLICATION IN<br />

39<br />

PRIMARY EFFUSION LYMPHOMA<br />

RECRUITMENT OF THE COMPLETE HTREX<br />

COMPLEX IS REQUIRED FOR KSHV<br />

INTRONLESS MRNA NUCLEAR EXPORT<br />

40<br />

AND VIRUS REPLICATION<br />

DOMAIN STRUCTURE AND FUNCTION OF<br />

KSHV ORF57 PROTEIN IN PROTEIN-<br />

PROTEIN<br />

INTERACTIONS<br />

AND PROTEIN-RNA<br />

41 KSHV ORF57 ENHANCES TRANSLATION<br />

OF VIRAL INTRONLESS MRNAS<br />

Fang Cheng, Markku Varjosalo, Anne<br />

Lehtonen, Magdalena Weidner-Glunde, Päivi<br />

J. Koskinen, Thomas Schulz, Jussi Taipale,<br />

<strong>and</strong> Päivi M. Ojala<br />

Hsing-Jien Kung, Pei-Ching Chang, Latricia<br />

Fitzgerald, Tom Ellison, Paul Luciw <strong>and</strong> Yoshi<br />

Izumiya<br />

Imogen Yi-Chun Lai <strong>and</strong> Paul Kellam<br />

James R. Boyne <strong>and</strong> Adrian Whitehouse<br />

Vladimir Majerciak <strong>and</strong> Zhi-Ming Zheng<br />

James R. Boyne, Adam Taylor <strong>and</strong> Adrian<br />

Whitehouse<br />

Session 7: Immunology II<br />

11:00 am – 12:30 pm<br />

Chair: Paul Kellam<br />

Abstract # Title Authors<br />

42 MODULATION BY KSHV OF LIGANDS Alexis Madrid <strong>and</strong> Don Ganem<br />

THAT MEDIATE NK CELL RECOGNITION<br />

43 SCREENING FOR NATURALLY OCCURRING<br />

ANTI-KCP (KSHV COMPLEMENT CONTROL<br />

PROTEIN; ORF4) ANTIBODIES IN KSHV-<br />

INFECTED PATIENTS AND GENERATION<br />

OF SPECIFIC MONOCLONAL ANTIBODIES<br />

FOR PATHOGENESIS STUDIES<br />

44 A KSHV VIRAL HOMOLOGUE OF CELLULAR<br />

CD200 (VOX2) INDIRECTLY SUPPRESSES<br />

GRANULOCYTE OXIDATIVE ACTIVITY IN<br />

HUMAN BLOOD<br />

45 CD8+ T CELL RECOGNITION OF THE KSHV<br />

LATENT PROTEIN LANA1<br />

46 CYTOKINE RESPONSES BY CD8 T<br />

LYMPHOCYTES TO KSHV/HHV8 LYTIC<br />

47<br />

AND LATENCY PROTEINS<br />

INFECTION OF LYMPHOID CELLS BY KSHV<br />

IN CULTURED PRIMARY HUMAN<br />

TONSILLAR LYMPHOID CELLS EX VIVO<br />

Anna M. Blom, Marcin Okroj, Linda Mark,<br />

Zoltan Korodi, Rosamaria Tedeschi, Joakim<br />

Dillner <strong>and</strong> O. Brad Spiller<br />

Karen Misstear, Rachel Colman, Hema<br />

Chahal, Janet Lord, David Blackbourn<br />

Shereen Sabbah & Andrew D Hislop<br />

Lauren Lepone, Giovanna Rappocciolo,<br />

Emilee Knowlton, Paolo Piazza, Mariel Jais,<br />

Frank J. Jenkins <strong>and</strong> Charles R. Rinaldo<br />

Jinjong Myoung <strong>and</strong> Don Ganem<br />

16


Friday July 25 th<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 8: Clinical & Epidemiology<br />

1:30 pm – 3:30 pm<br />

Chair: Ethel Cesarman<br />

Abstract # Title Authors<br />

48 PASSIVE TRANSFER OF HHV-8<br />

ANTIBODIES FROM BLOOD DONORS TO<br />

TRANSFUSION RECIPIENTS AND<br />

POSSIBLE<br />

INFECTION<br />

PROTECTION FROM HHV-8<br />

49 HEIGHTENED REDOX STATUS OF<br />

50<br />

PRIMARY EFFUSION LYMPHOMA CELLS<br />

CAN BE EXPLOITED THERAPEUTICALLY BY<br />

DECREASING RESISTANCE TO OXIDATIVE<br />

STRESS<br />

RAPAMYCIN IS EFFECTIVE AGAINST PTEN<br />

POSITIVE<br />

MALIGNANCIES<br />

AIDS-DEFINING<br />

51 GEOGRAPHIC VARIATION OF THE<br />

PREVALENCE OF KAPOSI’S SARCOMA-<br />

ASSOCIATED HERPESVIRUS AND RISK<br />

FACTORS FOR TRANSMISSION IN WOMEN<br />

FROM 8 COUNTRIES IN FOUR<br />

52<br />

CONTINENTS<br />

SERUM EPIDEMIOLOGICAL STUDIES ON<br />

KAPOSI'S SARCOMA IN XINJIANG<br />

53 KAPOSI’S SARCOMA-ASSOCIATED<br />

HERPESVIRUS SEROCONVERSION AND<br />

SEROREVERSION IN A MATERNAL-<br />

INFANT COHORT IN ZAMBIA<br />

54 DISTRIBUTION OF KSHV MICRORNA<br />

POLYMORPHISMS IN AIDS-KS,<br />

CLASSICAL KS, AND MULTICENTRIC<br />

CASTLEMAN’S DISEASE<br />

55 EARLY CHILDHOOD INFECTION BY<br />

KAPOSI’S SARCOMA-ASSOCIATED<br />

HERPESVIRUS AND EPSTEIN-BARR VIRUS<br />

IN ZAMBIA<br />

4:00 pm – 5:30 pm<br />

Session 9: Round Table<br />

Topic <strong>and</strong> chairs to be confirmed<br />

Ashley L. Fowlkes, Cedric Brown, Minal M.<br />

Amin, John Roback, Robert Downing, Esau<br />

Nzaro, Jonathan Mermin, Wolfgang Hladik,<br />

Sheila C. Dollard<br />

Darya Bubman, Utthara Nayar, Balasz<br />

Csernus, Ilaria Guasparri, Ethel Cesarman<br />

Debasmita Roy, Sang-Hoon Sin, Ling Wang,<br />

Blossom A. Damania, Dirk P. Dittmer<br />

Silvia de Sanjose, Georgina Mbisa, Sussana<br />

Perez, Sukhon Sukvirach, Nguyen Trong<br />

Hieu, Hai-Rim Shin, Pham Thi Hoang Anh,<br />

Jaiye O Thomas, Eduardo Lazcano, Elena<br />

Matos, Rol<strong>and</strong>o Herrero, Nubia Muñoz, Silvia<br />

Franceschi, Denise Whitby<br />

Lei YANG, Xiao-Hua TAN, Jiang-Mei QING,<br />

Feng LI, Shu-Xia GUO, Jian-Xin XIE, Jin<br />

HUANG, Dong-Mei LI, Yan ZENG<br />

Veenu Minhas, Kay L. Crabtree, Janet<br />

Wojcicki, Tendai J. M’soka, Chipepo Kankasa,<br />

Charles D. Mitchell <strong>and</strong> Charles Wood<br />

Vickie Marshall, Eliza Martró, Elizabeth<br />

Brown, Dian Wang, Alex Ray, Maria<br />

Nazzarena Labo, The Classical Kaposi’s<br />

Sarcoma Working Group, Robert Yarchoan,<br />

Jordi Casabona, Rolfe Renne <strong>and</strong> Denise<br />

Whitby<br />

Veenu Minhas, Brad P Brayfield, Tendai J.<br />

M’soka, Chipepo Kankasa, Charles D. Mitchell<br />

<strong>and</strong> Charles Wood<br />

17


Saturday July 26 th<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 10: Virus-Cell Interactions II<br />

8:45 am – 10:45 am<br />

Chair: John Nicholas<br />

Abstract # Title Authors<br />

56 INTRACELLULAR LOCALIZATION MAP OF<br />

HHV-8 PROTEINS<br />

57 KSHV INTERFERON REGULATORY FACTOR<br />

4, A NOVEL CBF1 INTERACTION PARTNER<br />

58 CHARACTERISATION OF ENDOPLASMIC<br />

RETICULUM (ER) STRESS INDUCERS THAT<br />

LEAD TO KSHV REACTIVATION FROM<br />

59<br />

LATENCY; IDENTIFYING PHYSIOLOGICAL<br />

TRIGGERS?<br />

INTEGRIN αVβ3 BINDS TO THE RGD MOTIF<br />

OF THE GLYCOPROTEIN B OF KAPOSI’S<br />

SARCOMA-ASSOCIATED HERPESVIRUS<br />

(KSHV/HHV8) AND FUNCTIONS AS AN<br />

RGD-DEPENDENT ENTRY RECEPTOR<br />

60 KAPOSI’S SARCOMA ASSOCIATED<br />

HERPESVIRUS (KSHV/HHV-8) FORMS A<br />

MULTI-MOLECULAR COMPLEX OF<br />

INTEGRINS (αVβ5, αVβ3 AND α3β1), CD98<br />

AND XCT DURING INFECTION OF HUMAN<br />

DERMAL MICROVASCULAR ENDOTHELIAL<br />

(HMVEC-D) CELLS<br />

61 MURINE GAMMAHERPESVIRUS 68<br />

ESTABLISHES A PERSISTENT INFECTION<br />

IN ENDOTHELIAL CELLS<br />

62 TARGETING EXTRACELLULAR HSP90<br />

REDUCES KSHV GENE EXPRESSION<br />

DURING DE NOVO INFECTION BY<br />

INHIBITING<br />

ACTIVATION<br />

MAPK PATHWAY<br />

63 FAK AND SHP2 ARE REQUIRED FOR KSHV<br />

VGPCR SIGNALING<br />

Gaby S<strong>and</strong>er, Andreas Konrad, Mathias<br />

Thurau, Effi Wies, Rene Leubert, Elisabeth<br />

Kremmer, Holger Dinkel, Thomas Schulz,<br />

Frank Neipel <strong>and</strong> Michael Stürzl<br />

Katharina Heinzelmann, Barbara Scholz,<br />

Elisabeth Kremmer, Jürgen Haas, Even<br />

Fossum, Bettina Kempkes<br />

Lucy Dalton-Griffin, Ed Tsao <strong>and</strong> Paul Kellam<br />

H. Jacques Garrigues, Yelena E.<br />

Rubinchikova, C. Michael DiPersio <strong>and</strong><br />

Timothy M. Rose<br />

Mohanan Valiya Veettil, Fu-Zhang Wang,<br />

Sathish Sadagopan, Neelam Sharma-Walia,<br />

Hari Raghu, Laszlo Varga <strong>and</strong> Bala Ch<strong>and</strong>ran<br />

A.L. Suárez <strong>and</strong> L.F. van Dyk<br />

Zhiqiang Qin, Jennifer Isaacs <strong>and</strong> Chris<br />

Parsons<br />

Thomas Bakken, Chris Bosh<strong>of</strong>f <strong>and</strong> Mark<br />

Cannon<br />

Session 11: Gene Expression II<br />

11:15 am – 12:15 pm<br />

Chair: Dirk Dittmer<br />

Abstract # Title Authors<br />

64 DEVELOPMENT OF A COMPREHENSIVE Linda Persson, Scott Millman <strong>and</strong> Angus<br />

TRANSCRIPTIONAL NETWORK FOR KSHV Wilson<br />

65 KSHV ORF57 AND RNA EXPORT FACTORS:<br />

ROLES OF UAP56, URH49, RBM15, AND<br />

OTT3 IN ORF57 EXPRESSION AND<br />

66<br />

FUNCTION<br />

DYNAMICS OF K-RTA RECRUITMENT ON<br />

THE KSHV GENOME REVEAL NOVEL<br />

67<br />

REGULATION BY NF-KB<br />

TYPE I INTERFERONS SUPPRESS MURINE<br />

GAMMAHERPESVIRUS-68 LYTIC<br />

INFECTION AT THE VIRUS<br />

TRANSCRIPTOME LEVEL VIA DIRECT<br />

MODULATION<br />

ACTIVITY<br />

OF RTA PROMOTER<br />

Merlyn Deng, Vladimir Majerciak, Barbara K.<br />

Felber, <strong>and</strong> Zhi-Ming Zheng<br />

Thomas J. Ellison, Chie Izumiya, Paul A.<br />

Luciw, Hsing-Jien Kung, Yoshihiro Izumiya<br />

L. Roaden, B. Manso, B. Lane, E. Arico <strong>and</strong><br />

B. Ebrahimi<br />

18


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Abstracts<br />

19


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 1 abstracts 1-7:<br />

Latency<br />

20


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Latency Abstract 1<br />

DELETION OF LANA FROM RHESUS RHADINOVIRUS (RRV) GENERATES A<br />

HIGHLY LYTIC RECOMBINANT VIRUS<br />

Kwun Wah Wen, Chelsey Hilscher, Dirk P. Dittmer <strong>and</strong> Blossom Damania<br />

Department <strong>of</strong> Microbiology <strong>and</strong> Immunology <strong>and</strong> Lineberger Cancer Center, <strong>University</strong> <strong>of</strong><br />

North Carolina at Chapel Hill, North Carolina 27599.<br />

Abstract<br />

Rhesus monkey rhadinovirus (RRV) is a gamma herpesvirus that is closely related to the<br />

human Kaposi's sarcoma-associated herpesvirus (KSHV/HHV-8). RRV serves as an in<br />

vitro <strong>and</strong> an in vivo model for KSHV. RRV can be grown to high titers on rhesus<br />

fibroblasts <strong>and</strong> the availability <strong>of</strong> the RRV lytic system facilitates analysis <strong>of</strong> viral<br />

replication <strong>and</strong> the contribution <strong>of</strong> individual open-reading frames to viral fitness. We<br />

had previously reported that RRV LANA (R-LANA) can suppress lytic viral replication <strong>and</strong><br />

that R-LANA inhibits Rta/Orf50 transactivation <strong>of</strong> lytic promoters resulting in lower viral<br />

titers (Dewire <strong>and</strong> Damania, J. Virology, 2005). Here we present data on the construction<br />

<strong>of</strong> a RRVΔLANA/GFP recombinant virus by homologous recombination. Integrity <strong>of</strong> the<br />

recombinant virus was confirmed by Southern blot analysis, restriction digest <strong>and</strong> PCR.<br />

We compared replication kinetics <strong>of</strong> RRVΔLANA/GFP, RRV-GFP, wild-type (WT) RRV, <strong>and</strong><br />

a revertant virus (RRVREV) using one-step growth curves. Viral replication was quantitated<br />

using traditional plaque assays as well as real-time PCR based genome quantification<br />

assay. We found that the RRVΔLANA/GFP recombinant virus exhibits highly lytic<br />

replicative properties compared to RRV-GFP, WT RRV, <strong>and</strong> the revertant virus. We also<br />

employed a quantitative real time PCR-based RRV viral array to transcriptionally pr<strong>of</strong>ile<br />

lytic gene expression during de novo infection using RRVΔLANA/GFP <strong>and</strong> RRV-GFP<br />

recombinant viruses. The RRVΔLANA/GFP virus displayed increased lytic gene expression<br />

at all time points post-infection compared to RRV-GFP.<br />

Presenting author E-mail: kenwen@med.unc.edu<br />

21


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Latency Abstract 2<br />

DIVERGENCE OF THE NUCLEAR LOCALIZATION SIGNAL WITHIN THE ORF73<br />

LATENCY-ASSOCIATED NUCLEAR ANTIGENS (LANA) OF KSHV AND THE<br />

MACAQUE RV1 AND RV2 RHADINOVIRUSES<br />

Kellie L. Burnside 1 <strong>and</strong> Timothy M. Rose 1,2<br />

1 Seattle Children’s Hospital Research Institute <strong>and</strong> 2 <strong>University</strong> <strong>of</strong> Washington, Seattle WA<br />

Abstract<br />

KSHV LANA is targeted to the nucleus <strong>of</strong> infected cells by a discrete lysine/arginine-rich<br />

nuclear localization signal (NLS; Region I), which allows LANA to enter the nucleus <strong>and</strong><br />

mediate viral episome persistence, interact with cellular proteins <strong>and</strong> play a role in<br />

latency <strong>and</strong> tumorigenesis. Conserved homologs <strong>of</strong> LANA that also exhibit nuclear<br />

localization have been identified in KSHV-like macaque herpesviruses belonging to the<br />

RV1 <strong>and</strong> RV2 lineages <strong>of</strong> Old World primate rhadinoviruses. To characterize the signals<br />

mediating nuclear localization, the N-terminal domains <strong>of</strong> the LANA homologs <strong>of</strong> the<br />

macaque RV1 (RFHVMn) <strong>and</strong> RV2 (RRV <strong>and</strong> MneRV2) rhadinoviruses were structurally<br />

<strong>and</strong> functionally compared to the KSHV NLS. A distinct lysine/arginine-rich NLS motif<br />

was identified in RFHVMn LANA (Region II) that was adjacent but evolutionarily unrelated<br />

to the KSHV NLS. An extended glycine/arginine-rich NLS, spanning 46 residues across<br />

Regions I <strong>and</strong> II, was identified within the RRV <strong>and</strong> MneRV2 LANAs. Although<br />

evolutionarily distinct, the NLS motifs <strong>of</strong> the human <strong>and</strong> macaque RV1 rhadinoviruses,<br />

KSHV <strong>and</strong> RFHVMn, both conform to the consensus motif for classical NLSs which interact<br />

with importin alpha during nuclear localization. In contrast, the NLSs <strong>of</strong> the more<br />

distantly-related macaque RV2 rhadinoviruses conform to the structure <strong>of</strong> a number <strong>of</strong><br />

glycine/arginine-rich NLS motifs which bind directly to importin B during nuclear<br />

transport. Like other proteins containing glycine/arginine-rich NLSs, the RV2 LANAs are<br />

targeted to the nucleolus. Our findings suggest that the LANA homologs <strong>of</strong> the RV1 <strong>and</strong><br />

RV2 rhadinoviruses may be differentially transported <strong>and</strong> function in different subnuclear<br />

compartments.<br />

Presenting author Email: timothy.rose@seattlechildrens.org<br />

22


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Latency Abstract 3<br />

NUCLEOPHOSMIN IS A NOVEL REGULATOR OF KSHV REPLICATION VIA<br />

FUNCTIONAL INTERACTIONS WITH VIRAL CYCLIN AND LANA<br />

Grzegorz Sarek 1 , Annika Järviluoma 1 , Henna Syrjäkari 2 , Sari Tojk<strong>and</strong>er 2 , Salla Vartia 1 ,<br />

Marikki Laiho 2,3 , <strong>and</strong> Päivi M. Ojala 1<br />

1 Genome-Scale Biology Program, Biomedicum Helsinki & Institute <strong>of</strong> Biomedicine, <strong>and</strong><br />

the Foundation for the Finnish Cancer Institute, 2 Molecular Cancer Biology Program,<br />

Biomedicum Helsinki & Haartman Institute; <strong>University</strong> <strong>of</strong> Helsinki, P.O. Box 63, 00014-<br />

Univ. <strong>of</strong> Helsinki, Finl<strong>and</strong>; 3 Department <strong>of</strong> Radiation Oncology <strong>and</strong> Molecular Radiation<br />

<strong>Sciences</strong>, The Johns Hopkins <strong>University</strong> School <strong>of</strong> Medicine, Baltimore, MD 21231<br />

Abstract<br />

During Kaposi's sarcoma herpesvirus (KSHV) latency, viral transcription is restricted to a<br />

subset <strong>of</strong> latent genes. The latency-associated nuclear antigen (LANA) interacts with<br />

interphase chromatin together with a variety <strong>of</strong> cellular factors <strong>and</strong> it actively represses<br />

transcription <strong>of</strong> the KSHV lytic genes. Viral cyclin (v-cyclin), another latent KSHV gene,<br />

transcribed from the same promoter as LANA, is structurally similar to cellular D-type<br />

cyclins <strong>and</strong> forms an active kinase complex with cellular CDK6. Nucleophosmin (NPM) is a<br />

multifunctional nuclear phosphoprotein implicated in chromatin organization <strong>and</strong><br />

transcription control. We have previously demonstrated that exogenous expression <strong>of</strong> vcyclin<br />

causes NPM redistribution from the nucleolus to the nucleoplasm in human<br />

osteosarcoma cells, suggesting that v-cyclin <strong>and</strong> NPM may have a functional relationship.<br />

Here, we present evidence that NPM interacts with LANA <strong>and</strong> is a novel substrate for vcyclin-CDK6<br />

kinase complex in primary effusion lymphoma (PEL) cells. Intriguingly, we<br />

establish the first functional link between latent proteins LANA <strong>and</strong> v-cyclin by<br />

demonstrating that phosphorylation <strong>of</strong> NPM by v-cyclin-CDK6 facilitates interaction<br />

between NPM <strong>and</strong> LANA. Silencing <strong>of</strong> NPM expression in PEL cells induces an increase in<br />

the acetylation <strong>of</strong> LANA, decreases its association with chromatin, <strong>and</strong> leads to<br />

spontaneous viral lytic reactivation. In addition, we show that NPM depletion in PEL cells<br />

abolishes interaction between HDAC1 <strong>and</strong> core histones suggesting that NPM is involved<br />

in the recruitment <strong>of</strong> HDAC1 during KSHV latent infection. This work establishes NPM as a<br />

novel regulator <strong>of</strong> KSHV replication, which maintains the transcriptional silencing <strong>of</strong> KSHV<br />

lytic genes.<br />

Presenting author Email: grzegorz.sarek@helsinki.fi<br />

23


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Latency Abstract 4<br />

ROLE OF BET PROTEINS IN THE FUNCTION OF KSHV AND MHV68 LANA<br />

Magdalena Weidner-Glunde, Matthias Ottinger, Daniel Pliquet, Ronald Frank* <strong>and</strong><br />

Thomas F. Schulz<br />

Institute <strong>of</strong> Virology, Hannover <strong>Medical</strong> School, Germany<br />

*HZI Braunschweig, Germany<br />

Abstract<br />

KSHV LANA-1 is a multifunctional protein known to be involved in KSHV genome<br />

maintenance, replication <strong>and</strong> transcriptional regulation. Its homologue in murine<br />

herpesvirus 68 has been shown to contribute to establishing latency. Two members <strong>of</strong><br />

the BET protein family, Brd2/RING3 <strong>and</strong> BRD4/HUNK, which bind to chromatin via<br />

acetylated histones, were found to interact with KSHV LANA-1<strong>and</strong> MHV68 orf73 protein.<br />

In view <strong>of</strong> the participation <strong>of</strong> Brd4 in the Mediator transcriptional co-activator complex<br />

<strong>and</strong> its implication in HPV-E2-mediated transcriptional regulation, we tested whether<br />

these two proteins are involved in LANA-1 dependent transcriptional activation <strong>and</strong> KSHV<br />

episome replication.<br />

Using a peptide array covering the MHV68 orf73 we identified aa226-231 as residues<br />

interacting with recombinant Brd2/RING3. MHV68 orf73 mutants with alanine<br />

substitutions in this region failed to activate a range <strong>of</strong> cellular promoters (cyclin E, D1,<br />

D2). They also showed an altered interaction with cellular chromatin.<br />

For KSHV LANA-1 we found that siRNAs specific for Brd2 or Brd4 reduced the KSHV<br />

LANA-1 dependent replication <strong>of</strong> a plasmid containing the latent origin. In addition, a<br />

Brd2 fragment comprising the ET domain inhibited episomal replication when<br />

overexpressed transiently.<br />

Our data suggest that (I) members <strong>of</strong> the BRD/BET protein family are involved in<br />

transcriptional regulation mediated by KSHV LANA-1 <strong>and</strong> its MHV68 homologue <strong>and</strong> that<br />

(II) Brd2/RING3 plays a role in LANA-1-mediated episomal replication. These<br />

observations could serve as a basis for the development <strong>of</strong> a peptide inhibitor <strong>of</strong> the<br />

interaction between LANA-1 <strong>and</strong> Brd2 <strong>and</strong> possibly also <strong>of</strong> the KSHV latent replication.<br />

Presenting author Email: weidnerg@allmail.mh-hannover.de<br />

24


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Latency Abstract 5<br />

INTERACTION OF KSHV LANA-1 WITH USP7: IMPLICATIONS FOR P53<br />

FUNCTION<br />

W. Albrecht (1), E. Gellermann (1), A. Viejo-Borbolla (2), T. F. Schulz (1)<br />

(1) Institute <strong>of</strong> Virology, Hanover <strong>Medical</strong> School, Hanover, Germany<br />

(2) Dept. <strong>of</strong> Cell <strong>and</strong> Molecular Biology, Centro Nacional de Biotecnología, Madrid, Spain<br />

Abstract<br />

LANA-1 is constitutively expressed in all latently KSHV-infected cells <strong>and</strong> displays several<br />

functions in the persistence <strong>and</strong> replication <strong>of</strong> the viral episome.<br />

We found the ubiquitin-specific protease 7 (USP7) to be a new interaction partner <strong>of</strong><br />

LANA-1. USP7 was originally identified in association with the HSV-1 protein ICP0, an E3<br />

ligase, <strong>and</strong> is thought to prevent the proteasomal degradation <strong>of</strong> autoubiquitinated ICP0.<br />

The EBV EBNA-1 protein, which is functionally related to LANA-1, also recruits USP7 <strong>and</strong><br />

is thought to absorb USP7 from p53, one <strong>of</strong> its physiological targets, thereby leading to<br />

the destabilization <strong>of</strong> p53.<br />

USP7 binds to LANA-1 via a sequence motif resembling the USP7-binding site in EBNA-1.<br />

However, LANA-1 mutants lacking this binding site were not compromised in their ability<br />

to antagonize p53-mediated transcriptional activation. LANA-1 interacts directly with p53<br />

<strong>and</strong> we mapped the binding site to three amino acids in the C-terminus <strong>of</strong> LANA-1.<br />

Mutation <strong>of</strong> the p53-binding site eliminates the ability <strong>of</strong> LANA-1 to antagonize p53dependent<br />

transcriptional activation.<br />

Furthermore, LANA-1 mutants lacking the USP7-binding site showed evidence <strong>of</strong><br />

increased ubiquitination.<br />

These findings indicate that the model <strong>of</strong> p53 stabilization by ‚squelching‘ <strong>of</strong> USP7, as<br />

developed for EBNA-1, does not apply to LANA-1 <strong>and</strong> that direct binding <strong>of</strong> p53 to<br />

LANA-1 is required for its ability to antagonize p53 function. In contrast, binding <strong>of</strong> USP7<br />

by LANA-1 may modulate ubiquitination <strong>of</strong> LANA-1.<br />

We currently investigate the functional importance <strong>of</strong> LANA-1 ubiquitination <strong>and</strong> the<br />

properties <strong>of</strong> LANA-1 mutants deficient in USP7 binding.<br />

Presenting author Email: albrecht.wiebke@mh-hannover.de<br />

25<br />

Comment [W1]: ?<br />

Comment [W2]: ?


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Latency Abstract 6<br />

KSHV ENCODED LANA INTERACTS WITH THE NUCLEAR MITOTIC APPARATUS<br />

PROTEIN TO REGULATE GENOME MAINTENANCE AND SEGREGATION<br />

Huaxin Si 1 , Subhash C. Verma 1 , Michael Lampson 2 , Qiliang Cai 1 , Erle S. Robertson 1<br />

1Department<br />

<strong>of</strong> Microbiology <strong>and</strong> the Tumor Virology Program <strong>of</strong> the Abramson<br />

Comprehensive Cancer Center, <strong>University</strong> <strong>of</strong> Pennsylvania, School <strong>of</strong> Medicine,<br />

Philadelphia, Pennsylvania, 19104<br />

2<br />

Department <strong>of</strong> Biology, <strong>University</strong> <strong>of</strong> Pennsylvania, School <strong>of</strong> Medicine, Philadelphia,<br />

Pennsylvania, 19104<br />

Abstract<br />

Kaposi’s sarcoma associated herpesvirus (KSHV) genomes are tethered to the host<br />

chromosomes <strong>and</strong> partitioned faithfully into daughter cells with the host chromosomes.<br />

The latency associated nuclear antigen (LANA) is important for segregation <strong>of</strong> the newly<br />

synthesized viral genomes to the daughter nuclei. Here we report that the Nuclear Mitotic<br />

Apparatus protein (NuMA) <strong>and</strong> LANA can associate in KSHV infected cells. In<br />

synchronized cells, NuMA <strong>and</strong> LANA are colocalized in interphase cells <strong>and</strong> separate<br />

during mitosis at the beginning <strong>of</strong> prophase, reassociating again at the end telophase <strong>and</strong><br />

cytokinesis. Silencing <strong>of</strong> NuMA expression by siRNA <strong>and</strong> expression <strong>of</strong> LGN <strong>and</strong> a<br />

dominant negative <strong>of</strong> dynactin (P150-CC1) which disrupts the association <strong>of</strong> NuMA with<br />

microtubule resulted in the loss <strong>of</strong> KSHV terminal repeats plasmids containing the major<br />

latent origin. Thus, NuMA is required for persistence <strong>of</strong> the KSHV episomes to daughter<br />

cells. This interaction between NuMA <strong>and</strong> LANA is critical for segregation <strong>and</strong><br />

maintenance <strong>of</strong> the KSHV episomes through a temporally controlled mechanism <strong>of</strong><br />

binding <strong>and</strong> release during specific phases <strong>of</strong> mitosis.<br />

Presenting author Email: vermas@mail.med.upenn.edu<br />

26


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Latency Abstract 7<br />

MULTIPLE DOMAINS CONTRIBUTE TO THE ASSOCIATION OF LANA WITH HOST<br />

CHROMATIN COMPONENTS<br />

Ryo Nasu, Satoko Matsumura, Naoko Tanese <strong>and</strong> Angus Wilson<br />

Department <strong>of</strong> Microbiology & NYU Cancer Institute, New York <strong>University</strong> School <strong>of</strong><br />

Medicine, New York, NY 10016, USA<br />

Abstract<br />

The latency-associated nuclear antigen (LANA, LANA1, LNA1) encoded by ORF73 is<br />

expressed in all KSHV infected cells <strong>and</strong> performs a variety <strong>of</strong> functions associated with<br />

maintenance <strong>of</strong> the viral genome <strong>and</strong> proliferation <strong>of</strong> the infected host cell. As its name<br />

indicates, LANA accumulates in the nucleus where it is associated with host chromosomes<br />

throughout the cell cycle. Direct interactions with a number <strong>of</strong> chromatin components<br />

have been described but the functional significance <strong>of</strong> these interactions is not fully<br />

understood. We have focused on the association <strong>of</strong> LANA with methyl-CpG binding<br />

protein 2 (MeCP2) <strong>and</strong> core histones. Evidence will be presented showing that both the<br />

N- <strong>and</strong> C-terminal domains <strong>of</strong> LANA contribute to interaction with MeCP2 <strong>and</strong> this can<br />

direct LANA to regions <strong>of</strong> pericentric heterochromatin. Localization is dependent on the<br />

methyl-CpG binding (MBD) <strong>and</strong> adjacent trans-repression (TRD) domains <strong>of</strong> MeCP2. The<br />

chromatin binding motif (CBM) at the extreme N-terminus <strong>of</strong> LANA (residues 5-11)<br />

influences subcellular localization but is not strictly required for MeCP2 binding. Instead,<br />

the CBM recognizes the four histones that form the nucleosome core. In addition to<br />

interactions with histones H2A/H2B previously described by the Kaye <strong>and</strong> Luger<br />

laboratories, we will present evidence for additional interactions with histones H3 <strong>and</strong> H4.<br />

Sedimentation studies employing recombinant proteins show that the LANA CBM is<br />

capable <strong>of</strong> assembling isolated nucleosomes into compacted structures that may serve to<br />

regulate host <strong>and</strong> viral gene expression.<br />

Presenting author Email: wilsoa02@med.nyu.edu<br />

27


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 2 abstracts 8-13:<br />

Immunology I<br />

28


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology I Abstract 8<br />

INDUCTION OF CYTOKINES BY HHV-8 IN MYELOID DENDRITIC CELLS AND B<br />

CELLS<br />

Emilee Knowlton, Giovanna Rappocciolo, Paolo Piazza, Heather Hensler, Frank J. Jenkins,<br />

Mariel Jais, Charles R. Rinaldo<br />

Dept <strong>of</strong> Infectious Diseases <strong>and</strong> Microbiology, Graduate School <strong>of</strong> Public Health,<br />

<strong>University</strong> <strong>of</strong> Pittsburgh, PA<br />

Abstract<br />

Objectives<br />

Myeloid dendritic cells (DC) <strong>and</strong> activated B lymphocytes are pr<strong>of</strong>essional antigen<br />

presenting cells susceptible to HHV-8 infection following DC-SIGN receptor binding.<br />

However, the virus undergoes productive infection in B cells but not in DC. We examined<br />

whether these different pathways taken by the virus were related to differential cytokine<br />

production in DC <strong>and</strong> B cells.<br />

Methods<br />

Monocyte derived DC <strong>and</strong> activated B cells were infected with purified HHV-8 <strong>and</strong><br />

supernatants were examined for cytokines at 0, 6, 12, 18, 24, <strong>and</strong> 48 hours postinfection<br />

by the Cytometric Bead Array (BD).<br />

Results<br />

Peak levels <strong>of</strong> TNF-α, IL-6, IL-8, IL-10, MIP-1α <strong>and</strong> MIP-1β <strong>of</strong> 30 (IL-10) to 2.6 x 10 4<br />

(MIP-1β) pg/ml were detected in B cell cultures by 24-48h <strong>of</strong> infection, which were 4 to<br />

22 fold higher than in uninfected B cell cultures. In contrast, DC produced peak amounts<br />

<strong>of</strong> IP-10, RANTES, MIP-1α <strong>and</strong> MIP-1β <strong>of</strong> 20 to 3.5 x 10 4 pg/ml by 24-48h, which were<br />

1.8-18 fold higher than in uninfected DC. Moreover, infected DC did not produce higher<br />

levels <strong>of</strong> IL-8, IL-12p70 or TNF compared to uninfected DC.<br />

Conclusions<br />

Elevated levels <strong>of</strong> several inflammatory cytokines <strong>and</strong> MIP chemokines were detected in<br />

HHV-8 infected B cells. In contrast, HHV-8 infected DC produced MIP chemokines,<br />

RANTES <strong>and</strong> IP-10, but not IL-12 p70. Distinct cytokine production by HHV-8 infected B<br />

cells <strong>and</strong> DC could play a role in viral replication <strong>and</strong> functional abilities in these antigen<br />

presenting cells.<br />

Presenting author Email: ERK21@pitt.edu<br />

29


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology I Abstract 9<br />

KSHV K5 COUNTERACTS TETHERIN, A NOVEL COMPONENT OF INNATE<br />

ANTIVIRAL IMMUNITY, TO FACILITATE VIRUS RELEASE<br />

Edward Tsao 1 , Sam Wilson 1 , Claire Pardieu 1 , Ben Webb, Imogen Lai, Stuart Neil 2 , Greg<br />

Towers 1 , <strong>and</strong> Paul Kellam 1<br />

1. MRC Centre for <strong>Medical</strong> Molecular Virology, Department <strong>of</strong> Infection, UCL, 46<br />

Clevel<strong>and</strong> Street, London W1T 4JF; 2. Peter Gorer Department <strong>of</strong> Immunobiology, 2nd<br />

Floor, Borough Wing, Guy's, King's <strong>and</strong> St Thomas' Hospitals, King's <strong>College</strong> London,<br />

Great Maze Pond, London SE1 9RT<br />

Abstract<br />

Tetherin (also called CD317, BST2, or HM1.24) has been identified as an interferoninducible<br />

human factor that inhibits the release <strong>of</strong> retroviruses, <strong>and</strong> its antiviral effect can<br />

be antagonised by HIV-1 vpu. It has been postulated that tetherin, a membraneassociated<br />

protein, is present both on plasma membranes <strong>and</strong> viral envelopes (acquired<br />

from the host membrane through viral budding). The interaction between virion- <strong>and</strong><br />

cell-associated tetherin molecules not only prevents virus release, but may also lead to<br />

endocytotic internalisation <strong>of</strong> the virions <strong>and</strong> their subsequent degradation. This model<br />

suggests that tetherin would be effective against all enveloped viruses. A recent<br />

proteomic study revealed that the levels <strong>of</strong> tetherin were strongly reduced in HeLa cells<br />

expressing KSHV K5. Here we show that 1) tetherin is expressed in PELs, that 2) KSHV<br />

K5 is required for the efficient release <strong>of</strong> virions from cells, <strong>and</strong> 3) in the absence <strong>of</strong> K5,<br />

the reduction <strong>of</strong> tetherin expression using shRNA rescues virus release. This is the first<br />

study to show that tetherin is active against enveloped DNA viruses, supporting the<br />

notion that it is an indiscriminate antiviral factor acting against enveloped viruses.<br />

Presenting author Email: e.tsao@ucl.ac.uk<br />

30


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology I Abstract 10<br />

MOLECULAR MECHANISM OF BST2/TETHERIN DOWNREGULATION BY KSHV-K5<br />

M<strong>and</strong>ana Mansouri, Janet Douglas, Kasinath Viswanathan, Jean Gustin, Ashlee Moses <strong>and</strong><br />

Klaus Früh<br />

Vaccine <strong>and</strong> Gene Therapy Institute, Oregon Health <strong>and</strong> Science <strong>University</strong>, 505<br />

NW185th Ave, Beaverton, OR, 97001, USA<br />

Abstract<br />

K5 (MIR2) targets cellular immunostimulatory proteins for degradation by ubiquitinating<br />

their cytoplasmic tails. Using quantitative membrane proteomics we previously showed<br />

that bone marrow stromal antigen 2 (BST2) is downregulated by K5 (Bartee, 2006, PLoS<br />

Pathogens 2:e107). In similar experiments, we also observed that BST2 is<br />

downregulated by the HIV-1 immunomodulator Vpu. Recent reports showed that BST2 is<br />

an interferon-induced transmembrane glycoprotein that prevents egress <strong>of</strong> mature HIV-1<br />

virions by tethering them to the cell membrane, an antiviral activity that is overcome by<br />

Vpu (Neil, 2008, Nature 451). We determined the molecular mechanisms through which<br />

KSHV-K5 downregulates BST2/Tetherin in comparison to Vpu. In dermal microvascular<br />

endothelial cells, IFN-induced expression <strong>of</strong> BST2 is inhibited by K5. Upon infection with<br />

KSHV, BST2 expression is further increased in the presence <strong>of</strong> K5-specific siRNA<br />

suggesting that K5 downregulates virus-induced BST2 during de novo infection. We<br />

further show that Vpu downregulates BST2 from the cell surface by sequestering it in the<br />

Golgi whereas K5 targets BST2 for degradation since steady state levels <strong>of</strong> total BST2 or<br />

cell surface BST2 are dramatically reduced in K5-expressing cells. Taken together with<br />

other data to be presented, we conclude that BST2 is a bona-fide target <strong>of</strong> K5. The role<br />

<strong>of</strong> BST2/Tetherin in modulating KSHV-infection is unknown. We will present data to<br />

address whether, in the absence <strong>of</strong> K5, BST2 interferes with egress <strong>of</strong> KSHV virions. Our<br />

data suggest that BST2 is part <strong>of</strong> the innate antiviral response to viral infection <strong>and</strong> that<br />

viruses <strong>of</strong> different genera have developed distinct countermeasures against this protein.<br />

Presenting author Email: dougljan@ohsu.edu<br />

31


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology I Abstract 11<br />

UPREGULATION OF THE TLR3 PATHWAY BY KSHV DURING PRIMARY INFECTION<br />

OF MONOCYTES<br />

John West, Sean Gregory <strong>and</strong> Blossom Damania<br />

Department <strong>of</strong> Microbiology <strong>and</strong> Immunology <strong>and</strong> Lineberger Cancer Center, <strong>University</strong> <strong>of</strong><br />

North Carolina at Chapel Hill, North Carolina 27599<br />

Abstract<br />

Kaposi’s sarcoma-associated herpesvirus (KSHV) is associated with several different<br />

human malignancies including Kaposi’s sarcoma, primary effusion lymphoma, <strong>and</strong><br />

multicentric Castleman’s disease. KSHV establishes lifelong latency in the host <strong>and</strong><br />

modulates the host immune response. Innate immunity is critical for controlling de novo<br />

viral infection. Toll like receptors (TLRs) are key components <strong>of</strong> the innate immune<br />

system <strong>and</strong> they serve as pathogen recognition receptors that stimulate the host antiviral<br />

response. In particular, TLR3 has been implicated in RNA virus recognition.<br />

Currently there is no information on how KSHV infection modulates any TLR pathway. We<br />

report the first evidence showing that KSHV upregulates TLR3 expression in human<br />

monocytes sixteen hours post-infection. This is also the first demonstration <strong>of</strong> a human<br />

DNA tumor virus upregulating TLR3, a TLR that thus far has been associated with the<br />

recognition <strong>of</strong> RNA viruses. We found that KSHV upregulates the TLR3 pathway <strong>and</strong><br />

induces TLR3-specific cytokines <strong>and</strong> chemokines, including IFN-β1 <strong>and</strong> CXCL10 (IP-10).<br />

SiRNAs directed against TLR3 greatly reduced the ability <strong>of</strong> KSHV to upregulate IFN-β1<br />

<strong>and</strong> CXCL10 upon infection. TLR3 upregulation was monitored at different time-points<br />

post-infection. We found that TLR3 expression levels were highest 16 hours postinfection<br />

<strong>and</strong> slowly declined thereafter returning to baseline by 120 hours post-infection<br />

as the virus established latency.<br />

Presenting author Email: john_west@med.unc.edu<br />

32


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology I Abstract 12<br />

KSHV EVADES INNATE IMMUNITY BY SUPPRESSION OF TLR4 EXPRESSION<br />

Dimitris Lagos 1 , Richard James Vart 1 , Fiona Gratrix 1 , Samantha Jane Westrop 2 , Ping-Pui<br />

Wong 1 , Nesrina Imami 2 , Mark Bower 3 , Frances Gotch 2 <strong>and</strong> Chris Bosh<strong>of</strong>f 1<br />

1<br />

Cancer Research UK Viral Oncology Group, UCL Cancer Institute, Paul O’Gorman<br />

Building, Huntley Street, <strong>University</strong> <strong>College</strong> London, WC1E 6BT, London, U.K.<br />

2<br />

Department <strong>of</strong> Immunology, Chelsea <strong>and</strong> Westminster Hospital, Imperial <strong>College</strong><br />

London, London, U.K.<br />

3<br />

Imperial <strong>College</strong> School <strong>of</strong> Medicine, Chelsea <strong>and</strong> Westminster Hospital, Imperial <strong>College</strong><br />

London, London, UK.<br />

Abstract<br />

Toll-like receptors (TLRs) play a critical role in innate immune responses against invading<br />

pathogens. However, the role <strong>and</strong> regulation <strong>of</strong> TLRs during KSHV infection remain poorly<br />

understood. We demonstrate that TLR4 mediates innate response against KSHV <strong>and</strong> that<br />

KSHV targets TLR4 expression as a mechanism <strong>of</strong> immune evasion. In vitro, mouse<br />

macrophages or human lymphatic endothelial cells lacking TLR4 are more susceptible to<br />

KSHV infection, whereas activation <strong>of</strong> TLR4-mediated signalling with bacterial<br />

lipopolysaccharide protects cells from KSHV infection. In vivo, HIV-1-infected individuals<br />

who carry a non-functional TLR4 allele appear more likely to have multicentric<br />

Castleman’s disease, a lymphoproliferation associated with enhanced KSHV replication.<br />

Furthermore, we show that KSHV has evolved mechanisms to evade the TLR4-mediated<br />

response. The KSHV-encoded vIRF1 <strong>and</strong> vGPCR co-operate to suppress TLR4 expression<br />

in human lymphatic endothelial cells. Notably, ERK activation by vGPCR or by viral<br />

attachment to the cell membrane results in rapid TLR4 down-regulation. Our findings<br />

reveal the first example <strong>of</strong> viral immune evasion through suppression <strong>of</strong> TLR4 expression<br />

<strong>and</strong> suggest the potential use <strong>of</strong> TLR4 agonists for treatment <strong>of</strong> KSHV-related neoplasms.<br />

Presenting author Email: d.lagos@ucl.ac.uk<br />

33


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology I Abstract 13<br />

THE KSHV LYTIC PROTEIN VOX2 AND ITS CELLULAR COUNTERPART, CD200,<br />

INHIBIT EPITOPE-SPECIFIC T CELL RESPONSES<br />

Rachel Colman, Karen Misstear, Heather Long, Omar Quereshi^, David Sansom^,<br />

Andrew Hislop, David J. Blackbourn<br />

CRUK Institute for Cancer Studies <strong>and</strong> ^Division <strong>of</strong> Immunity <strong>and</strong> Infection, <strong>University</strong> <strong>of</strong><br />

Birmingham, Birmingham B15 2TT, UK<br />

Abstract<br />

Background & Objectives: The KSHV lytic protein vOX2 shares 36% identity with<br />

human cellular CD200. An engineered soluble derivative <strong>of</strong> vOX2 inhibited neutrophil<br />

activity in vitro <strong>and</strong> had anti-inflammatory activity in vivo, suggesting the protein inhibits<br />

innate immune responses. In the present study we determined whether adaptive<br />

immune responses were modulated by native, membrane-bound vOX2 <strong>and</strong> CD200.<br />

Methods: Antigen presenting cells (APC) were engineered to express either vOX2 or<br />

CD200 on their surface by retroviral transduction <strong>and</strong> membrane expression was<br />

confirmed by flow cytometry <strong>and</strong> IFA. APC loaded with epitope peptides were incubated<br />

with appropriate epitope-specific CD4+ or CD8+ T cell clones <strong>and</strong> T cell responses were<br />

measured by IFN-γ release. Ex-vivo restimulation <strong>of</strong> epitope-specific CD8+ T cells was<br />

performed by coculturing peripheral blood mononuclear cells (PBMC) with autologous APC<br />

engineered to express either vOX2 or CD200 on their surface. T cell function was<br />

measured by IFN-γ release.<br />

Results: Expression <strong>of</strong> vOX2 or CD200 on the surface <strong>of</strong> APCs inhibited IFN-γ secretion<br />

by co-cultured CD4+ <strong>and</strong> CD8+ T cell clones up to 50% when compared to vector only<br />

transduced APCs. Restimulation <strong>of</strong> PBMC was similarly inhibited. Neither HLA class I, nor<br />

CD80 <strong>and</strong> CD86 costimulatory molecule expression levels were downregulated on APCs<br />

by vOX2 or CD200 expression.<br />

Conclusion: These data demonstrate that both KSHV vOX2 <strong>and</strong> cellular CD200 inhibit<br />

antigen-specific T-lymphocyte activity. They suggest vOX2 suppresses adaptive immune<br />

responses against KSHV lytically infected cells, facilitating virus replication <strong>and</strong><br />

dissemination in vivo.<br />

Presenting author Email: r.colman@bham.ac.uk<br />

34


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 3 abstracts 14-21:<br />

Pathogenesis<br />

35


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Pathogenesis Abstract 14<br />

GENERATION OF RHESUS RHADINOVIRUS LACKING EXPRESSION OF VGPCR<br />

AND VCD200 FOR THE ASSESSMENT OF THEIR ROLES IN DISEASE<br />

DEVELOPMENT IN A RHESUS MACAQUE MODEL OF KSHV INFECTION<br />

Ryan D. Estep 1<br />

, Elisa Cardenas 3 , <strong>and</strong> Scott W. Wong 1,2,3<br />

Vaccine <strong>and</strong> Gene Therapy Institute1 , Oregon Health & Science <strong>University</strong> West Campus,<br />

Division <strong>of</strong> Pathobiology <strong>and</strong> Immunology2 , Oregon National Primate Research Center,<br />

Beaverton, Oregon 97006;<br />

Department <strong>of</strong> Molecular Microbiology <strong>and</strong> Immunology3 , Oregon Health & Science<br />

<strong>University</strong>, Portl<strong>and</strong>, Oregon 97201.<br />

Abstract<br />

RRV strain 17577, the rhesus macaque homologue <strong>of</strong> KSHV, was recently cloned as an<br />

infectious Bacterial Artificial Chromosome (BAC). The RRV BAC system allows for the<br />

rapid genetic manipulation <strong>of</strong> the RRV genome in bacteria, <strong>and</strong> the subsequent<br />

production <strong>of</strong> modified forms <strong>of</strong> RRV. Currently, we are using the RRV BAC system to<br />

modify genes that have been proposed to be important in the development <strong>of</strong> disease<br />

associated with KSHV infection in humans. KSHV <strong>and</strong> RRV both encode a viral G proteincoupled<br />

receptor (vGPCR) <strong>and</strong> viral CD200 homologue (vCD200), proteins which possess<br />

oncogenic <strong>and</strong> immunomodulatory functions, respectively. Although extensively analyzed<br />

in vitro, the roles that the KSHV <strong>and</strong> RRV vGPCR <strong>and</strong> vCD200 proteins play during a de<br />

novo infection in vivo are currently unknown. Therefore, we have used the RRV BAC<br />

system to disrupt expression <strong>of</strong> the vGPCR <strong>and</strong> vCD200 proteins in RRV, through the<br />

insertion <strong>of</strong> specific stop mutations in ORF74 <strong>and</strong> R15. Thus far, we have successfully<br />

generated viruses that no longer express these proteins during infection, <strong>and</strong> preliminary<br />

analyses indicate that viruses devoid <strong>of</strong> vGPCR or vCD200 expression possess similar<br />

growth properties to wild-type RRV in vitro. Other ongoing studies with these viruses<br />

include examination <strong>of</strong> the effects <strong>of</strong> vGPCR <strong>and</strong>/or vCD200 expression on virus-induced<br />

signaling <strong>and</strong> immunoregulatory properties in vitro, <strong>and</strong> importantly, how lack <strong>of</strong> these<br />

proteins may affect the development <strong>of</strong> viral-induced disease in vivo. Further plans<br />

include the generation <strong>of</strong> chimeric versions <strong>of</strong> RRV expressing KSHV vGPCR <strong>and</strong>/or<br />

vCD200.<br />

Presenting author Email: estepr@ohsu.edu<br />

36


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Pathogenesis Abstract 15<br />

GENERATION OF VFLIP TRANSGENIC MICE: A MODEL TO STUDY KSHV-<br />

ASSOCIATED LYMPHOMAGENESIS<br />

Gianna Ballon 1 , Amy Chadburn 1 , Yi-Fang Liu 1 , Yoshiteru Sasaki 2 , Klaus Rajewsky 2 , Ethel<br />

Cesarman 1<br />

1<br />

Department <strong>of</strong> Pathology <strong>and</strong> Laboratory Medicine, Weill Cornell <strong>Medical</strong> <strong>College</strong>, New<br />

York, NY 10021, USA<br />

2<br />

CBR Institute for Biomedical Research, Harvard <strong>Medical</strong> School, Boston, MA 02115, USA<br />

Abstract<br />

Primary effusion lymphoma (PEL) is a distinct subtype <strong>of</strong> aggressive non-Hodgkin’s<br />

lymphoma (NHL), specifically associated with infection by Kaposi's sarcoma-associated<br />

herpesvirus (KSHV). Several in vitro observations suggest that vFLIP, a protein<br />

expressed during latency, is an important viral oncogene. It is essential for the survival<br />

<strong>of</strong> KSHV-infected PEL cells, mainly by constitutively activating the NF-kB pathway. In<br />

order to assess the role <strong>of</strong> vFLIP in the pathogenesis <strong>of</strong> PEL, we developed transgenic<br />

mouse models expressing vFLIP in B-cells. The experimental approach used has been a<br />

conditional recombinant activation <strong>of</strong> vFLIP, by using the ROSA26 knock-in system. A<br />

specifically restricted expression <strong>of</strong> the transgene in CD19+ B-cells has been achieved by<br />

crossing the ROSA26.vFLIP knock-in mice with other mice expressing cre recombinase<br />

under the control <strong>of</strong> the CD19 promoter. These mice have also been crossed with the<br />

LANA transgenic mice to assess a potential synergistic effect between these two KSHV<br />

latent proteins in the lymphomagenic process <strong>of</strong> PEL. vFLIP expression in the CD19+ Bcells<br />

results in splenomegaly, with an increase in both T <strong>and</strong> B-cells, <strong>and</strong> with a relative<br />

increase <strong>of</strong> the T versus B-cell ratio. Although primary follicles were enlarged, the<br />

expression <strong>of</strong> vFLIP in the CD19+ B-cells results in lack <strong>of</strong> germinal center formation in<br />

the spleen, lymph nodes <strong>and</strong> intestine, <strong>and</strong> in partially impaired class-switching<br />

recombination. These results indicate that, by constitutively activating the NF-kB<br />

pathway in pre-germinal center B-cells expressing CD19, the normal B-cell differentiation<br />

is impaired, <strong>and</strong> provide clues about possible aberrant differentiation in PEL cells.<br />

Presenting author Email: gib2004@med.cornell.edu<br />

37


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Pathogenesis Abstract 16<br />

DEVELOPMENT OF ANIMAL MODELS OF KSHV INFECTION AND AIDS-<br />

ASSOCIATED DISEASE BASED ON MACAQUE RHADINOVIRUSES<br />

A. Gregory Bruce 1 , Jonathan T. Ryan 1 , Courtney Gravett 1,2 , <strong>and</strong> Timothy M. Rose 1,2 ,<br />

1 Seattle Children’s Hospital Research Institute <strong>and</strong> 2 <strong>University</strong> <strong>of</strong> Washington, Seattle WA<br />

Abstract<br />

Two related viral lineages within the Rhadinovirus genus <strong>of</strong> gammaherpesviruses have<br />

been identified in macaques <strong>and</strong> other Old World non-human primates. In macaques,<br />

the prototypes <strong>of</strong> these lineages were first identified in the rhesus macaque (Macaca<br />

mulatta); retroperitoneal fibromatosis-associated herpesvirus (RFHVMm) <strong>and</strong> rhesus<br />

rhadinovirus (RRV) members <strong>of</strong> the Rhadinovirus-1 (RV1) <strong>and</strong> RV2 lineages, respectively.<br />

Each macaque species is host to a distinct pair <strong>of</strong> RV1 <strong>and</strong> RV2 rhadinoviruses,<br />

distinguishable by molecular sequence. Both RV1 <strong>and</strong> RV2 rhadinoviruses have been<br />

implicated in diseases in macaques that closely resemble cancers <strong>and</strong> proliferative<br />

diseases associated with KSHV in humans, including Kaposi sarcoma (KS), lymphoma<br />

<strong>and</strong> multicentric Castleman’s disease. Thus, the characterization <strong>of</strong> the macaque<br />

rhadinoviruses, including the development <strong>of</strong> specific reagents <strong>and</strong> assays that enable<br />

the evaluation <strong>of</strong> their biology <strong>and</strong> life cycles, is an important aspect in the development<br />

<strong>of</strong> KSHV-related macaque models <strong>of</strong> disease. We have developed protocols for<br />

experimental infection <strong>of</strong> macaques with both RV1 <strong>and</strong> RV2 macaque rhadinoviruses, <strong>and</strong><br />

are developing reagents <strong>and</strong> analytical approaches to follow the resulting infections <strong>and</strong><br />

host responses to infection in vivo. We will present the current status <strong>of</strong> the first small<br />

pilot study examining the experimental infection <strong>of</strong> naïve macaques with infectious<br />

rhadinoviruses from saliva <strong>of</strong> infected macaques <strong>and</strong> subsequent chemical <strong>and</strong> viral<br />

treatments to activate virus by inducing immunosuppression. These studies will provide<br />

the groundwork for establishing relevant animal models <strong>of</strong> human disease associated<br />

with KSHV.<br />

Presenting author Email: greg.bruce@seattlechildrens.org<br />

38


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Pathogenesis Abstract 17<br />

β ARRESTINS REGULATE KSHV GPCR ACTIVITY AND ARE MODULATED BY<br />

CANNABINOIDS<br />

Xuefeng Zhang, Yehoshua Maor, Jerome E. Groopman<br />

Division <strong>of</strong> Experimental Medicine, Beth Israel Deaconess <strong>Medical</strong> Center, Harvard<br />

<strong>Medical</strong> School, Boston, MA 02115 USA<br />

Abstract<br />

Kaposi’s sarcoma (KS) is a frequent neoplasm among immuno-compromised patients<br />

such as those with AIDS <strong>and</strong> after organ transplantation. Among certain populations,<br />

recreational use <strong>of</strong> cannabinoids is common <strong>and</strong> medicinal use <strong>of</strong> cannabinoids is<br />

increasing as well. Cannabinoids bind to CB1 <strong>and</strong> CB2 GPCRs on the endothelial cell<br />

surface. In our previous study, we found that low doses <strong>of</strong> Δ 9 -tetrahydrocannabinol (Δ 9 -<br />

THC), the major psychoactive ingredient <strong>of</strong> marijuana, induced KSHV GPCR expression<br />

<strong>and</strong> endothelial transformation. Using immunoprecipitation <strong>and</strong> confocal microscopy, we<br />

then found that KSHV GPCR was constitutively associated with β arrestins. β arrestins are<br />

key molecules contributing to desensitization <strong>of</strong> GPCRs. We now report that knockdown<br />

<strong>of</strong> β arrestins using RNAi technology increased KSHV GPCR expression. This resulted in<br />

phosphorylation <strong>of</strong> focal adhesion kinase (FAK) <strong>and</strong> inhibition <strong>of</strong> p53 expression. Such<br />

changes in FAK <strong>and</strong> p53 may contribute to endothelial transformation by KSHV GPCR. We<br />

also found that knockdown <strong>of</strong> β arrestins significantly induced NFκB (p65) expression in<br />

KSHV-infected HMVEC. The NFκB pathway has been observed to participate in KSHV<br />

GPCR-induced transformation. Our results indicate that β arrestins may exert inhibitory<br />

effects on KSHV GPCR activity, <strong>and</strong> when KSHV-infected cells are exposed to a strong<br />

GPCR stimulus like cannabinoids, cytosolic arrestins are preferentially recruited to<br />

activated GPCRs like CB1 <strong>and</strong> CB2. This would significantly decrease association <strong>of</strong> KSHV<br />

GPCR with β arrestins, increasing activity <strong>of</strong> KSHV GPCR <strong>and</strong> enhancing KS<br />

transformation.<br />

Presenting author Email: xzhang3@bidmc.harvard.edu<br />

39


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Pathogenesis Abstract 18<br />

Activation <strong>of</strong> NF-κB by KSHV K15 involves recruitment <strong>of</strong> NIK (NF-κB inducing<br />

kinase)<br />

Anika Hävemeier, Macel Pietrek <strong>and</strong> Thomas F. Schulz<br />

Institute <strong>of</strong> Virology, <strong>Medical</strong> School Hannover, Hannover, Germany<br />

Abstract<br />

ORF K15 is the positional homologue <strong>of</strong> EBV LMP2A. Like LMP2A, the K15 protein<br />

contains 12 predicted transmembrane <strong>and</strong> a cytoplasmic domain involved in signaling.<br />

The cytoplasmic domain interacts with several cellular proteins, including TRAF <strong>and</strong> Src<br />

kinases, <strong>and</strong> activates NF-kB as well as the MAPKs c-Jun-N-terminal kinase (JNK) <strong>and</strong><br />

extracellular signal-regulated kinase (Erk). Mutation <strong>of</strong> tyrosine 481 in the cytoplasmic<br />

tail <strong>of</strong> K15 (K15 Y 481 F) abolishes the ability <strong>of</strong> K15 to induce these pathways.<br />

Activation <strong>of</strong> NF-kB by several stimuli occurs via two distinct pathways: the classical<br />

(canonical) or the alternative (noncanonical) pathway. We found that K15 induces the<br />

alternative NF-kB pathway via NIK, leading to the processing <strong>of</strong> p100 <strong>and</strong> the<br />

translocation <strong>of</strong> p52 into the nucleus. Transfection <strong>of</strong> a dominant negative NIK <strong>and</strong> NIK<br />

siRNA decreases the K15 mediated induction <strong>of</strong> NF-kB, shown by western blot analysis or<br />

luciferase reporter experiments. Using alanin scanning mutants the binding site for NIK<br />

localised to six aminoacids near the last transmembrane domain <strong>of</strong> K15. NIK-binding<br />

deficient K15 mutants show a decreased NF-kB activation, but appear to retain the ability<br />

to activate other K15-induced promoters.<br />

Direct recruitment <strong>of</strong> NIK represents a novel way for a viral protein to activate NF-kB. In<br />

activating NF-kB via NIK, KSHV K15 shows functional similarities to the Lymphotoxin<br />

beta-receptor, rather than the B-cell receptor, whose function is thought to be mimicked<br />

by EBV LMP2A.<br />

Presenting author Email: haevemeier.anika@mh-hannover.de<br />

40


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Pathogenesis Abstract 19<br />

KAPOSI'S SARCOMA ASSOCIATED HERPES VIRUS (KSHV/HHV-8) INDUCES<br />

ANGIOGENIN DURING INFECTION OF HUMAN DERMAL MICROVASCULAR<br />

ENDOTHELIAL (HMVEC-D) CELLS THAT IS CRITICAL FOR ANTI-APOPTOSIS,<br />

CELL PROLIFERATION AND ANGIOGENESIS<br />

Sathish Sadagopan, Neelam-Sharma Walia, Mohanan Valiya Veettil, Virginie Bottero, Rita<br />

Levine <strong>and</strong> Bala Ch<strong>and</strong>ran<br />

H.M. Bligh cancer research laboratories, Department <strong>of</strong> Microbiology <strong>and</strong> Immunology,<br />

Chicago <strong>Medical</strong> School, Rosalind Franklin <strong>University</strong> <strong>of</strong> Medicine <strong>and</strong> Science, Chicago, IL<br />

Abstract<br />

De novo KSHV infection <strong>of</strong> HMVEC-d cells results in increased expression <strong>and</strong> secretion <strong>of</strong><br />

angiogenin, a multifunctional angiogenic protein. KS tissue sections were positive for<br />

angiogenin highlighting the importance <strong>of</strong> angiogenin in KS pathogenesis. Viral gene<br />

expression was critical for the sustained angiogenin secretion during primary infection.<br />

KSHV ORF 73 (latent) <strong>and</strong> ORF 74 (lytic) genes induced an increased secretion <strong>of</strong><br />

angiogenein. TIVE cells latently infected with KSHV secreted high levels <strong>of</strong> angiogenin.<br />

Angiogenin bound to surface actin, internalized in a microtubule independent manner,<br />

translocated into the nucleus when the cells were semi-confluent <strong>and</strong> became nucleolar<br />

in sub-confluent cells. In the nucleolus, angiogenin bound to the upstream sequence <strong>of</strong><br />

45SrRNA promoter <strong>and</strong> increased 45SrRNA transcription. Angiogenin increased the antiapoptosis<br />

<strong>and</strong> cell proliferation <strong>of</strong> KSHV infected endothelial cells, <strong>and</strong> these activities<br />

were blocked by neomycin which blocked the nuclear translocation <strong>of</strong> angiogenin.<br />

Upregulation <strong>of</strong> angiogenin lead to an increased activation <strong>of</strong> urokinase plasminogen<br />

activator <strong>and</strong> generation <strong>of</strong> active plasmin from inactive plasminogen. Plasmin generation<br />

aided the migration <strong>of</strong> endothelial cells towards chemoattractant including angiogenin,<br />

<strong>and</strong> chemotaxis was prevented by the inhibition <strong>of</strong> angiogenin nuclear translocation.<br />

Tube formation <strong>of</strong> endothelial cells was significantly inhibited by treating infected cell<br />

supernatants with anti-angiogenin antibodies. Inhibition <strong>of</strong> nuclear translocation <strong>of</strong><br />

angiogenin also blocked VEGF-C expression. Collectively, these results suggest that KSHV<br />

induced angiogenin plays multiple roles during infection such as increase in 45SrRNA<br />

synthesis, proliferation, cell migration <strong>and</strong> angiogenesis, <strong>and</strong> blocking angiogenin could<br />

have a therapeutic value in treating KSHV infection <strong>and</strong> KS.<br />

Presenting author Email: bala.ch<strong>and</strong>ran@rosalindfranklin.edu<br />

41


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Pathogenesis Abstract 20<br />

ROLE OF K15 IN KAPOSI’S SARCOMA HERPESVIRUS INDUCED ENDOTHELIAL<br />

CELL MIGRATION<br />

Bernd Hillenbr<strong>and</strong>, Antje Bürger, Irina Fischer, Khaled Alkharsah <strong>and</strong> Thomas F. Schulz<br />

Institute <strong>of</strong> Virology, Hanover <strong>Medical</strong> School, Hanover, 30625, Germany<br />

Abstract<br />

Kaposi’s sarcoma herpesvirus (KSHV) is the cause <strong>of</strong> Kaposi’s sarcoma, a highly<br />

vascularized tumour. Others have previously proposed a role for IL-8 in endothelial cell<br />

migration induced by KSHV-infected cells (Lane et al., J.Virol. 2002; 76:11570-11583).<br />

We previously showed that the K15 protein <strong>of</strong> KSHV induces increased IL-8 secretion in<br />

epithelial cells. In this study we addressed the role <strong>of</strong> K15 for virus induced endothelial<br />

cell migration in the context <strong>of</strong> the whole viral genome. We constructed a K15 deficient<br />

recombinant virus genome (KSHVΔK15) by using a bacterial artificial chromosome (BAC)<br />

system. We generated stable HEK293 cells carrying the BAC36 (wt) or BAC ΔK15.<br />

Following the induction <strong>of</strong> the lytic replication cycle reduced IL-8 mRNA levels were found<br />

by gene expression microarray in BAC ΔK15 cells in comparison to BAC36 cells. This<br />

reduction was confirmed by IL-8 specific ELISA <strong>of</strong> BAC36 <strong>and</strong> BAC ΔK15 conditioned<br />

media. In endothelial cell migration assays the migration <strong>of</strong> primary human vascular<br />

endothelial cells (HUVEC) in response to conditioned culture supernatants from KSHV<br />

ΔK15-infected cells was significantly reduced in comparison to supernatants from<br />

KSHVwt-infected cells. In contrast, supernatants from cells infected with a recombinant<br />

KSHV lacking the viral FLIP (BAC ΔFLIP), like K15 an inducer <strong>of</strong> NFκB, induced the same<br />

degree <strong>of</strong> endothelial cell migration as KSHVwt–infected cells. These results suggest an<br />

involvement <strong>of</strong> K15 in KSHV induced endothelial cell migration <strong>and</strong> possibly angiogenesis.<br />

Presenting author Email: hillenbr<strong>and</strong>.bernd@mh-hannover.de<br />

42


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Pathogenesis Abstract 21<br />

DOWNREGULATION OF GALECTIN-3 IN KSHV INFECTED DMVEC CELLS AND<br />

KAPOSI’S SARCOMA: IMPLICATIONS FOR TUMORIGENESIS<br />

Donald J. Alcendor 1 Wen Qui Zhu 1 Prashant Desai 2 <strong>and</strong> Gary S. Hayward 2<br />

1 Meharry <strong>Medical</strong> <strong>College</strong>, School <strong>of</strong> Medicine, Center for AIDS Health Disparities<br />

Research <strong>and</strong> Department <strong>of</strong> Microbial Pathogenesis <strong>and</strong> Immune Response, 1005 Dr.<br />

D.B. Todd Jr. Blvd., Hubbard Hospital, Rm. 5025, Nashville, Tennessee, 37208-3599,<br />

USA, 2 Department <strong>of</strong> Viral Oncology <strong>and</strong> Pharmacology, Molecular Virology Laboratories,<br />

Sidney-Kimmel Cancer Research Center, Johns Hopkins <strong>University</strong> School <strong>of</strong> Medicine,<br />

Baltimore, Maryl<strong>and</strong> 21231-1000 USA<br />

Abstract<br />

The Galectins are a family <strong>of</strong> proteins that share an affinity for b-galactoside containing<br />

glycoconjugates. Galectin-3 has been implicated in tumor progression <strong>and</strong> metastasis.<br />

Down regulation <strong>of</strong> Galectin-3 is associated with malignancy in prostate, ovarian <strong>and</strong><br />

breast cancer. Kaposi’s sarcoma (KS) is characterized as an angioproliferative tumor <strong>of</strong><br />

vascular endothelial cells <strong>and</strong> produces rare B cell lymphoproliferative diseases in the<br />

form <strong>of</strong> primary effusion lymphomas (PEL) <strong>and</strong> some forms <strong>of</strong> Multicentric Castleman’s<br />

Disease (MDC). We have observed suppression <strong>of</strong> galectin-3 expression in KSHV infected<br />

dermal microvascular endothelial cells (DMVEC) <strong>and</strong> KS tumor tissue. Here we<br />

demonstrate that galectin-3 protein expression is downregulated 20-fold in KSHV<br />

infected DMVEC cells by immunoblot analysis. In infected DMVEC, we show absence <strong>of</strong><br />

galectin-3 staining by dual labeled immun<strong>of</strong>luorescence in latency associated nuclear<br />

antigen (LANA) positive spindle cells. There is also transcriptional suppression <strong>of</strong><br />

Galectin-3 message in KSHV infected DMVEC cells as observed by RT-PCR. Furthermore<br />

we find reduced levels <strong>of</strong> galectin-3 expression in LANA positive spindle cell regions in<br />

archival KS tissue by dual labeled immunohistochemistry. Transfection studies reveal<br />

KSHV vFLIP is likely the viral gene that targets galectin-3 downregulation in HeLa cells.<br />

Finally, in uninduced PEL cells lines galectin-3 expression levels vary with levels <strong>of</strong> KSHV<br />

replication. The search for novel host cell factors that may contribute to the overall<br />

pathogenesis <strong>of</strong> KS is essential for early detection <strong>of</strong> KS <strong>and</strong> development <strong>of</strong> innovative<br />

therapies for treatment.<br />

Presenting author Email: dalcendor@mmc.edu<br />

43


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 4 abstracts 22-28:<br />

Virus-Cell Interactions I<br />

44


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions I Abstract 22<br />

ANIMAL MODELS OF KAPOSI'S SARCOMA REVEAL A ROLE OF KSHV VGPCR AND<br />

RAC1 IN ANGIOGENESIS AND GENETIC INSTABILITY<br />

Lucas Cavallin 1,2 , Qi Ma 1 , Rui Zhang 1,2 , E. Margarita Duran 1,2 , Pascal J. Goldschmidt-<br />

Clermont 1 <strong>and</strong> Enrique A. Mesri 1,2<br />

1 <strong>University</strong> <strong>of</strong> Miami Miller School <strong>of</strong> Medicine <strong>and</strong> 2 Viral Oncology Program, Sylvester<br />

Comprehensive Cancer Center. Miami, FL 33136<br />

Abstract<br />

Kaposi’s sarcoma (KS) is a viral malignancy characterized by angiogenesis <strong>and</strong><br />

proliferation <strong>of</strong> KSHV-infected spindle cells. Spindle cells explanted from tumors lose<br />

KSHV <strong>and</strong> are not tumorigenic. However, tumorigenic KS cells could be isolated from<br />

advanced tumors where is more frequent to find cellular oncogenic alterations. We<br />

described a new animal model <strong>of</strong> KS (Mutlu et al. 2007. Cancer Cell 11:245-258):<br />

KSHVBac36-tranfected mouse endothelial lineage cells (mECK36) induce KSHV-infected<br />

KS-like tumors in nude mice, while mECK36 that lose the KSHV episome lose their<br />

tumorigenicity. shRNA silencing showed that vGPCR, a KSHV angiogenic gene<br />

upregulated in mECK36 tumors, is essential for angiogenesis <strong>and</strong> tumorigenicity. We<br />

found that mECK36 explanted from tumors that lose the KSHV episome are tumorigenic,<br />

indicating that, as KSHV tumorigenesis progresses in vivo, the malignant phenotype<br />

becomes irreversible <strong>and</strong> KSHV-independent. Explanted mECK36 displayed 3 times more<br />

foci <strong>of</strong> DNA repair than cells never grown in tumors indicating that DNA damage is<br />

increased in vivo. We now find that expression <strong>of</strong> constitutively activated Rac1, a<br />

mediator <strong>of</strong> vGPCR pathogenesis over-expressed in KS lesions, is sufficient to induce KSlike<br />

tumors in transgenic mice by a mechanism involving PTEN inactivation <strong>and</strong> R.O.S.mediated<br />

oxidative DNA damage. These results points to a key role for Rac1 induced<br />

R.O.S. production in KS pathogenesis. They indicate that in addition to angiogenesis,<br />

vGPCR activation <strong>of</strong> Rac1 can increase mutation rate to foster a multi-step carcinogenesis<br />

process driven by oxidative genetic damage.<br />

Presenting author Email: emesri@med.miami.edu<br />

45


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions I Abstract 23<br />

INSIGHTS INTO KSHV ACTIVATION OF THE IKK SIGNALOSOME: CRYSTAL<br />

STRUCTURE OF A VFLIP-IKKγ COMPLEX.<br />

Claire Bagnéris, Alex<strong>and</strong>er V Ageichik, Nora Cronin, Bonnie Wallace, Mary Collins, Chris<br />

Bosh<strong>of</strong>f, Gabriel Waksman <strong>and</strong> Tracey Barrett 1<br />

1<br />

Institute <strong>of</strong> Structural <strong>and</strong> Molecular Biology, School <strong>of</strong> Crystallography, Birkbeck<br />

<strong>College</strong>, Malet Street, London WC1E 7HX, UK.<br />

Abstract<br />

During the latent phase <strong>of</strong> Kaposi Sarcoma Herpes virus (KSHV) infection, a limited<br />

number <strong>of</strong> genes are expressed that are pivotal to viral propagation <strong>and</strong> survival.<br />

Amongst them is ks-vFLIP whose over-expression has been directly linked to Kaposi<br />

Sarcoma (KS) <strong>and</strong> other lymphoproliferative malignancies. ks-vFLIP shares considerable<br />

homology with the human cellular FLIPs that have a key anti-apoptotic role, but by<br />

contrast operates via a mechanism that involves subversion <strong>of</strong> the canonical NF-κB<br />

transcriptional pathway (implicated in diverse cellular processes that include immune<br />

activation <strong>and</strong> cellular growth). This is achieved through a direct interaction with IKKγ,<br />

the regulatory component <strong>of</strong> the IKK complex or signalosome, <strong>and</strong> results in the normally<br />

tightly regulated pathway being rendered constitutively active. As a consequence, the<br />

expression <strong>of</strong> genes associated with anti-apoptosis, inflammation <strong>and</strong> oncogenesis are<br />

aberrantly up-regulated. In order to establish the molecular basis underpinning ks-vFLIP<br />

constitutive activation <strong>of</strong> the canonical NF-κB pathway, we have determined the crystal<br />

structure <strong>of</strong> ks-vFLIP bound to an IKKγ fragment incorporating the ks-vFLIP recognition<br />

motif. This first structure <strong>of</strong> a ks-vFLIP-IKKγ complex reveals an extensive ks-vFLIP-IKKγ<br />

interface that has been probed using site directed mutagenesis to confirm the roles <strong>of</strong><br />

key amino acids in mediating the ks-vFLIP-IKKγ interaction. In addition, these have been<br />

shown to be essential for ks-vFLIP induced activation <strong>of</strong> the canonical NF-κB pathway.<br />

Our structure thus provides a framework for the design <strong>of</strong> novel therapeutics aimed at<br />

the treatment <strong>of</strong> KS <strong>and</strong> related pathologies.<br />

Presenting author Email: t.barrett@mail.cryst.bbk.ac.uk<br />

46


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions I Abstract 24<br />

THE VIRAL INHIBITOR OF APOPTOSIS VFLIP/K13 PROTECTS ENDOTHELIAL<br />

CELLS AGAINST SUPEROXIDE - INDUCED CELL DEATH<br />

Michael Stürzl(1), Gaby S<strong>and</strong>er(1), Nathalie Gonin-Laurent(1), Kristina Weinländer(1),<br />

Elisabeth Naschberger(1), Ramona Jochmann(1), Khaled R. Alkharsah(2), Thomas F.<br />

Schulz(2), Margot Thome(3), Frank Neipel(4) <strong>and</strong> Mathias Thurau(1)<br />

(1) Department <strong>of</strong> Surgery, Division <strong>of</strong> Molecular <strong>and</strong> Experimental Surgery, <strong>University</strong> <strong>of</strong><br />

Erlangen-Nuremberg, Erlangen, Germany. (2) <strong>Medical</strong> School Hannover, Department <strong>of</strong><br />

Virology, Hannover, Germany. (3) Department <strong>of</strong> Biochemistry, <strong>University</strong> <strong>of</strong> Lausanne,<br />

Epalinges, Switzerl<strong>and</strong>. (4) Institute for Clinical <strong>and</strong> Molecular Virology, <strong>University</strong> <strong>of</strong><br />

Erlangen-Nuremberg, Erlangen, Germany<br />

Abstract<br />

Human herpesvirus-8 (HHV-8) is the etiological agent <strong>of</strong> Kaposi’s sarcoma (KS). The<br />

gene K13 <strong>of</strong> HHV-8 encodes for the anti-apoptotic viral FLICE-inhibitory protein<br />

vFLIP/K13. It is expressed in the latent phase <strong>of</strong> the viral life cycle <strong>and</strong> its expression in<br />

KS lesions is inversely related to the rate <strong>of</strong> apoptosis. The mechanisms by which<br />

vFLIP/K13 inhibits cell death have to be elucidated. By using a comparative proteome<br />

analysis we identified manganese superoxide dismutase (MnSOD), a mitochondrial<br />

antioxidant <strong>and</strong> an important anti-apoptotic enzyme, as the most strongly up-regulated<br />

protein by vFLIP/K13 in endothelial cells. MnSOD expression was also up-regulated in<br />

endothelial cells upon infection with HHV-8. The induction <strong>of</strong> MnSOD expression by<br />

vFLIP/K13 was dependent on NF-kappaB activation, occurred in a cell intrinsic manner,<br />

<strong>and</strong> correlated with decreased intracellular superoxide accumulation <strong>and</strong> increased<br />

resistance <strong>of</strong> endothelial cells against superoxide-induced death. These findings show a<br />

novel function <strong>of</strong> vFLIP/K13 in cell death inhibition <strong>and</strong> in regulation <strong>of</strong> mitochondrial<br />

redox-balance via the upregulation <strong>of</strong> MnSOD expression.<br />

Presenting author Email: michael.stuerzl@uk-erlangen.de<br />

47


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions I Abstract 25<br />

ROLE OF DEFECTIVE OCT-2 AND OCA-B EXPRESSION IN IMMUNOGLOBULIN<br />

PRODUCTION AND KSHV LYTIC REACTIVATION IN PRIMARY EFFUSION<br />

LYMPHOMA<br />

Daniel DiBartolo 1 , Elizabeth Hyjek 1 , Shannon Keller 1 , Ilaria Guasparri 1 , Ren Sun 2 , Amy<br />

Chadburn 1 , Daniel M Knowles 1 , <strong>and</strong> Ethel Cesarman 1<br />

Department <strong>of</strong> Pathology <strong>and</strong> Laboratory Medicine, Weill Cornell <strong>Medical</strong> <strong>College</strong>, New<br />

York, NY, 1 Molecular Biology IDP, <strong>University</strong> <strong>of</strong> California at Los Angeles, Los Angeles,<br />

CA 2<br />

Abstract<br />

PEL cells do not express surface or cytoplasmic immunoglobulin (Ig) despite having a<br />

genotype <strong>and</strong> gene expression signature <strong>of</strong> highly differentiated B cells. We show the<br />

lack <strong>of</strong> Oct-2 <strong>and</strong> OCA-B transcription factors to be responsible, at least in part, for this<br />

defect in Ig production. ORF50/Rta, the major regulator <strong>of</strong> KSHV lytic reactivation,<br />

contains an octamer motif within its promoter. Thus, we also examined the impact that<br />

the lack <strong>of</strong> Oct-2 <strong>and</strong> OCA-B have on lytic reactivation mediated by ORF50. Previous<br />

studies had shown that binding <strong>of</strong> Oct-1 to the ORF50 promoter significantly enhances<br />

ORF50 transactivation. However, we found that Oct-2, on the other h<strong>and</strong>, inhibits ORF50<br />

expression <strong>and</strong> consequently lytic reactivation. Chromatin immunoprecipitation assays<br />

showed that endogenous Oct-1 associates with ORF50 promoter in uninduced cells <strong>and</strong><br />

transfection <strong>of</strong> ORF50 further enhances its binding. Using PEL cells with inducible Oct-2<br />

expression we found that induction <strong>of</strong> Oct-2 results in Oct-2 association with the ORF50<br />

promoter <strong>and</strong> decreased Oct-1 binding suggesting that Oct-2 competes with Oct-1 for the<br />

octamer site in the ORF50 promoter, explaining mechanistically the observed inhibitory<br />

effect on lytic reactivation. The consistent absence <strong>of</strong> Oct-2 in PEL leads us to speculate<br />

that this deficiency is selected for in order to maintain the ability to efficiently transition<br />

from latency to lytic reactivation.<br />

Presenting author Email: ecesarm@med.cornell.edu<br />

48


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions I Abstract 26<br />

KAPOSI’S SARCOMA ASSOCIATED HERPESVIRUS (KSHV) INDUCES<br />

TUMORIGENIC CELLULAR MIRNAS IN LATENTLY INFECTED ENDOTHELIAL CELLS<br />

1Rebecca L. Skalsky 2 Wendell Miley, 2 Rachel Bagni, 1 Soo-Jin Han 1 Jianhong Hu, Chang<br />

Hee Kim 3 , 1 Rolf Renne <strong>and</strong> 2* Denise Whitby<br />

1. Department <strong>of</strong> Molecular Genetics <strong>and</strong> Microbiology, <strong>University</strong> <strong>of</strong> Florida, Gainesville,<br />

Florida. 2. Viral Oncology Section, AIDS <strong>and</strong> Cancer Virus Program, SAIC-Frederick, NCI-<br />

Frederick, Frederick MD. 3. Laboratory <strong>of</strong> Molecular Technology, Advanced Technology<br />

Program, SAIC-Frederick, NCI-Frederick, Frederick, MD<br />

Abstract<br />

MicroRNAs are small non-coding RNA molecules which post-transcriptionally regulate<br />

gene expression by binding to 3’UTRs <strong>of</strong> target genes thereby contributing to important<br />

biological processes. Recently, a subset <strong>of</strong> the more than 450 human miRNAs has been<br />

shown to be aberrantly expressed in many human tumors. Kaposi’s sarcoma associated<br />

herpesvirus (KSHV) encodes 12 miRNAs, some <strong>of</strong> which target host cellular genes<br />

thereby affecting angiogenesis, apoptosis, <strong>and</strong> B cell development. So far reports on<br />

KSHV-encoded miRNAs have focused solely on lymphoma cells <strong>and</strong> not on endothelial<br />

cells from which KS tumours arise.<br />

To address this question, we utilized a custom miRNA array to investigate both viral <strong>and</strong><br />

the host cellular miRNA expression pattern in endothelial cells in the presence <strong>and</strong><br />

absence <strong>of</strong> latent KSHV infection.<br />

We detected a limited set <strong>of</strong> KSHV miRNAs in endothelial cells from two different lineages<br />

- overall viral miRNA expression was significantly lower in endothelial cells compared to<br />

PEL cell lines. Additionally, we observed significant changes in host cellular miRNA<br />

expression in latently infected cells. Importantly, the hsa-miR 17/92 cluster, hsa miR16,<br />

hsa-miR155 <strong>and</strong> several let-7 family members were significantly up-regulated. Aberrant,<br />

expression <strong>of</strong> miR155 <strong>and</strong> the miR17/92 cluster has been reported in many lymphoid <strong>and</strong><br />

solid tumours. Moreover, these cellular miRNAs were the first for which oncogenic activity<br />

was demonstrated in transgenic mouse models.<br />

Our data suggests that KSHV usurps miRNA pathways not only by expression <strong>of</strong> viral<br />

miRNAs but also by inducing significant changes in the host cellular miRNA expression<br />

pattern.<br />

Presenting author Email: whitbyd@ncifcrf.gov<br />

49


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions I Abstract 27<br />

KSHV AND CELLULAR MIRNA EXPRESSION DURING LATENCY AND LYTIC<br />

REACTIVATION IN PEL CELLS<br />

*1Soo-Jin Han * 1 Jianhong Hu, * 1 Karlie Plaisance * 2 Wendell Miley 2 Rachel Bagni, Chang<br />

Hee Kim 3 , 2 Denise Whitby <strong>and</strong> 2 Rolf Renne<br />

1. Department <strong>of</strong> Molecular Genetics <strong>and</strong> Microbiology, <strong>University</strong> <strong>of</strong> Florida, Gainesville,<br />

Florida 32610, 2.Viral Oncology Section, AIDS <strong>and</strong> Cancer Virus Program, SAIC-<br />

Frederick, NCI-Frederick, Frederick MD 21702 3. Laboratory <strong>of</strong> Molecular Technology,<br />

Advanced Technology Program, SAIC-Frederick, NCI-Frederick, Frederick, MD 21702<br />

(*equal contribution).<br />

Abstract<br />

MicroRNAs are small non-coding RNA molecules which post-transcriptionally regulate<br />

gene expression by binding to 3’UTRs <strong>of</strong> target genes thereby contributing to important<br />

biological processes. Kaposi’s sarcoma associated herpesvirus (KSHV) encodes 12<br />

miRNAs within the KSHV latency associated region (KLAR), some <strong>of</strong> which target host<br />

cellular genes thereby affecting angiogenesis, apoptosis, <strong>and</strong> B cell development.<br />

Originally, KSHV miRNAs were cloned from BCBL-1 cells in the presence <strong>and</strong> absence <strong>of</strong><br />

TPA induction, suggesting that miRNAs may contribute to both phases <strong>of</strong> infection.<br />

The KLAR region is expressed from at least three different promoters giving rise to singly<br />

<strong>and</strong> multiply spliced transcripts. To further elucidate the complexity <strong>of</strong> this locus with<br />

emphasis on transcripts that could serve as pri-miRNAs, we performed a detailed RT-PCR<br />

<strong>and</strong> RNase protection analysis <strong>and</strong> identified several new transcripts that could give rise<br />

to miRNA expression. We identified novel multiply-spliced transcripts originating from the<br />

LANAp <strong>and</strong> LTI promoters upstream <strong>of</strong> LANA, which contained 3’ exons within the<br />

Kaposin locus. Furthermore, the observed splicing patterns from these long KLAR<br />

encompassing transcripts changed during lytic replication, suggesting that both LANAp<br />

<strong>and</strong> LTI could drive miRNA expression.<br />

Next, we utilized a custom miRNA array containing both viral <strong>and</strong> human miRNAs to<br />

investigate miRNA output in PEL cells during latency <strong>and</strong> reactivated by Ad-ORF50.<br />

Surprisingly, while we identified marked changes in the splicing pattern in this region,<br />

viral miRNA expression was not significantly altered in PEL cells 48 hours post<br />

reactivation. Cellular miRNA expression signatures during both conditions will also be<br />

discussed.<br />

Presenting author Email: rrenne@ufscc.ufl.edu<br />

50


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions I Abstract 28<br />

KAPOSI’S SARCOMA ASSOCIATED HERPESVIRUS (KSHV/HHV-8) UTILIZES<br />

MACROPINOCYTIC PATHWAY TO ENTER HUMAN DERMAL MICROVASCULAR<br />

ENDOTHELIAL (HMVEC-D) AND HUMAN UMBILICAL VEIN ENDOTHELIAL<br />

(HUVEC) CELLS<br />

Hari Raghu, Neelam Sharma Walia, Mohanan Valiya Veettil, Sathish Sadagopan <strong>and</strong> Bala<br />

Ch<strong>and</strong>ran<br />

H.M. Bligh cancer research laboratories, Department <strong>of</strong> Microbiology <strong>and</strong> Immunology,<br />

H.M. Bligh Cancer Research Laboratories, Chicago <strong>Medical</strong> School, Rosalind Franklin<br />

<strong>University</strong> <strong>of</strong> Medicine <strong>and</strong> Science, 3333 Green Bay Road, North Chicago, IL. USA<br />

Abstract<br />

KSHV utilizes the clathrin mediated endocytic pathway for its infectious entry into human<br />

foreskin fibroblast (HFF) cells (J.Virology. 2003. 77: 7978-7990). Here, we characterized<br />

KSHV entry in primary HMVEC-d <strong>and</strong> HUVEC cells. Similar to HMVEC-d cells, KSHV<br />

infection <strong>of</strong> HUVEC cells also resulted in the initial high level lytic ORF50 <strong>and</strong> K8 gene<br />

expression <strong>and</strong> subsequent decline, while the latent gene expression persisted. In<br />

contrast to HFF cells, cytochalasin D affecting actin polymerization significantly blocked<br />

virus entry <strong>and</strong> gene expression in both endothelial cell types tested. Chlorpromazine, a<br />

clathrin mediated endocytosis inhibitor which inhibited KSHV entry in HFF cells did not<br />

have any effect on KSHV binding, internalization <strong>and</strong> gene expression in HMVEC-d <strong>and</strong><br />

HUVEC cells. No significant inhibition was observed in both the endothelial cell types with<br />

filipin, a caveolar endocytosis inhibitor. In contrast, internalization <strong>and</strong> gene expression<br />

was significantly inhibited in both the endothelial cell types by macropinocytosis<br />

inhibitors EIPA <strong>and</strong> Rottlerin. Internalized virus particles enclosed in large vesicles were<br />

seen by electron microscopy. Confocal microscopy localized the viral capsid with KSHV<br />

envelope glycoprotein gpK8.1A at 5’ <strong>and</strong> 10’ post infection. Inhibition <strong>of</strong> macropinocytosis<br />

resulted in the distribution <strong>of</strong> viral capsids at the cell periphery <strong>and</strong> very little association<br />

with microtubules. Internalized viral glycoprotein gpK8.1A was associated with dextran, a<br />

marker for macropinocytosis, <strong>and</strong> KSHV entry in HMVEC-d cells was dynamin<br />

independent. Although KSHV was not associated with the early endosome marker EEA-1,<br />

internalized KSHV was associated with Rab5 <strong>and</strong> Rab34 GTPases that are known to<br />

regulate macropinocytosis. Taken together, these findings suggest that for its infectious<br />

entry in HMVEC-d <strong>and</strong> HUVEC cells, KSHV utilizes a macropinocytic pathway.<br />

Presenting author Email: bala.ch<strong>and</strong>ran@rosalindfranklin.edu<br />

51


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 5 abstracts 29-34:<br />

Gene Expression I<br />

52


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gene Expression I Abstract 29<br />

SCREENING FOR XINJIANG KS ASSOCIATED GENES AND THE STUDY ON THEIR<br />

MECHANISMS<br />

Lei YANG, Yan ZENG, Dong-Mei LI,Ling-ling XIAN,Xiao –Fei ZHOU,Xiao-Hua TAN,Jin<br />

HUANG,Feng LI,Jian-Xin XIE,Hui ZHANG,Sheng Wang,Xian-Dao LUO<br />

Key Laboratory <strong>of</strong> Xinjiang Endemic <strong>and</strong> Ethnic Disease, Shihezi <strong>University</strong>,Shihezi,<br />

XinJiang, 832002, China<br />

Abstract<br />

In China, classical KS mainly occurs in Uygur <strong>and</strong> Kazak ethic group in Xinjiang<br />

Autonomous Region. The occurrence <strong>and</strong> development <strong>of</strong> sarcoma is linked with the<br />

abnormality <strong>of</strong> genes. For comprehensive clarifying the pathogenesis <strong>of</strong> KS in Xinjiang,<br />

We screened the differentially expressed genes between KS’ tissues <strong>and</strong> normal skin<br />

tissues from an classical patient <strong>of</strong> Uygur by Suppression subtractive hybridization (SSH).<br />

We cloned <strong>and</strong> sequenced them <strong>and</strong> established differential expression cDNA library on<br />

the base <strong>of</strong> SSH.Then we enlarged the samples <strong>and</strong> selected 4 classic patients to do the<br />

further screening by human genome U133 plus 2.0 microarray (Affymetrix). According to<br />

the bioinformatics analysis,we got the congenerous differentially expressed genes<br />

nearly 80. The function <strong>of</strong> them are focused on signal transduction <strong>and</strong> correlated with<br />

apoptosis or tumor occurrence. These studies identified a large number <strong>of</strong> genes whose<br />

expression was altered in KS. we have verified some <strong>of</strong> these differentially expressed<br />

genes by Real-time PCR <strong>and</strong> Immunohistochemistry, it’s consistent with the SSH <strong>and</strong><br />

gene chip results.We have carried on some functional study on these genes,such as 14-<br />

3-3β, HO-1, Neuritin, EGFR, MMP14, LYN, et al. We detected the expression level <strong>of</strong> KS<br />

associated genes in BC-3 cells <strong>and</strong> Human umbilical vein endothelial cells to analyze if<br />

infection <strong>of</strong> HHV8 have effect on the expression <strong>of</strong> these genes .Most <strong>of</strong> these genes were<br />

related with the infection <strong>of</strong> HHV8 <strong>and</strong> participate in cell proliferation, apoptosis <strong>and</strong> this<br />

might show particular relation in the pathogenesis <strong>of</strong> KS.<br />

Presenting author Email: zengyan910@yahoo.com.cn<br />

53


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gene Expression I Abstract 30<br />

HOST CELL AND VIRAL MICRORNA (MIRNA) EXPRESSION IN B CELL<br />

LYMPHOMAS<br />

Eve M Coulter, Dan Frampton, Ed Tsao, Paul Kellam<br />

MRC Centre for <strong>Medical</strong> Molecular Virology, Department <strong>of</strong> Infection, <strong>University</strong> <strong>College</strong><br />

London, 46 Clevel<strong>and</strong> Street, London, Engl<strong>and</strong> W1T 4JF<br />

Abstract<br />

Micro RNAs (miRNAs) are a class <strong>of</strong> approximately 21- nucleotide non-coding small RNAs<br />

that post-transcriptionally down-regulate the expression <strong>of</strong> mRNAs bearing<br />

complementary target sequences. Several hundred miRNAs have been predicted within<br />

the human genome <strong>and</strong> their key role as regulators <strong>of</strong> important biological pathways<br />

such as development, proliferation, apoptosis <strong>and</strong> viral infection is becoming clear.<br />

Recent discovery <strong>of</strong> virus-encoded miRNAs, especially miRNAs encoded by herpesviruses,<br />

indicates that viruses also use this fundamental mode <strong>of</strong> gene regulation. Using miRNA<br />

microarray technology, (470 human <strong>and</strong> 64 human viral miRNAs represented), we have<br />

determined the pattern <strong>of</strong> expression <strong>of</strong> human, Epstein Barr virus (EBV) <strong>and</strong> Kaposi’s<br />

sarcoma virus (KSHV) encoded miRNAs in B cell tumour cell lines associated with<br />

different stages <strong>of</strong> normal B cell development. Here we show that only a small proportion<br />

<strong>of</strong> human miRNAs are expressed in different B cell tumours cell lines <strong>and</strong> that difference<br />

in viral miRNA expression are evident between different herpesvirus positive tumour<br />

types. miR-155 <strong>and</strong> other human miRNAs are differentially expressed across different Bcell<br />

lymphomas including PEL. This suggests potential functional roles for their target<br />

genes in PEL biology. These data show that like virus <strong>and</strong> host cell gene expression, virus<br />

<strong>and</strong> host cell miRNA expression is associated with the underlying cell type.<br />

Presenting author Email: rebmeco@ucl.ac.uk<br />

54


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gene Expression I Abstract 31<br />

CELLULAR AND VIRAL GENE EXPRESSION PROFILING IN KSHV-INFECTED<br />

MONOCYTES<br />

Sean Gregory, Ling Wang, John West, Chelsey Hilscher, Dirk P. Dittmer <strong>and</strong> Blossom<br />

Damania<br />

Department <strong>of</strong> Microbiology <strong>and</strong> Immunology <strong>and</strong> Lineberger Cancer Center, <strong>University</strong> <strong>of</strong><br />

North Carolina at Chapel Hill, North Carolina 27599.<br />

Abstract<br />

Kaposi's sarcoma (KS)-associated herpesvirus is the etiologic agent <strong>of</strong> Kaposi's sarcoma,<br />

primary effusion lymphoma <strong>and</strong> multicentric Castleman's disease. KSHV can infect a<br />

number <strong>of</strong> cell types including B cells, endothelial cells, epithelial cells <strong>and</strong> monocytes,<br />

macrophages <strong>and</strong> dendritic cells. We report the establishment <strong>of</strong> a monocytic cell line<br />

that is latently infected with KSHV (KSHV-THP-1). To study the host response to KSHV<br />

infection, we compared cellular gene expression in KSHV-THP-1 cells to uninfected THP-1<br />

cells. We found that approximately six percent <strong>of</strong> cellular genes demonstrated a greater<br />

than two fold increase or decrease in gene expression in the KSHV-THP-1 cells compared<br />

to uninfected controls. Differences in gene expression <strong>of</strong> a select panel <strong>of</strong> genes were<br />

also confirmed by quantitative RT-PCR. We found that several genes involved in the host<br />

immune response were selectively downregulated in the KSHV-infected monocytes.<br />

These included tumor necrosis factor (TNF), interleukin-1b, CXCL10 <strong>and</strong> CCL4.<br />

Interestingly, the activation markers, CD86 <strong>and</strong> CD83 were also downregulated in KSHVinfected<br />

monocytes. Additionally we found several factors involved in gene expression<br />

<strong>and</strong> signal transduction to be upregulated in the KSHV-THP1 cells compared to the<br />

uninfected cells. We also pr<strong>of</strong>iled eighty-four viral genes in the KSHV-THP-1 cells, <strong>and</strong><br />

found that KSHV predominantly exhibited a latent gene pr<strong>of</strong>ile. Latency associated genes<br />

such as LANA, v-Cyclin <strong>and</strong> v-FLIP were expressed in the KSHV-infected monocytes.<br />

Presenting author Email: sgregory@med.unc.edu<br />

55


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gene Expression I Abstract 32<br />

PROFILING OF CELLULAR AND VIRAL MICRORNAS IN KAPOSI SARCOMA AND<br />

VIRAL-ASSOCIATED LYMPHOMA<br />

Andrea J. O’Hara*, Bruce J. Dezube # , William Harrington Jr.^, Blossom Damania*, <strong>and</strong><br />

Dirk P. Dittmer^*<br />

*Department <strong>of</strong> Microbiology <strong>and</strong> Immunology, Lineberger Comprehensive Cancer<br />

Center, <strong>and</strong> Curriculum in Genetics <strong>and</strong> Molecular Biology, <strong>University</strong> <strong>of</strong> North Carolina at<br />

Chapel Hill, Chapel Hill, NC 27599; # Beth Israel Deaconess <strong>Medical</strong> Center; ^Sylvester<br />

Comprehensive Cancer Center, <strong>University</strong> <strong>of</strong> Miami.<br />

Abstract<br />

MicroRNAs are regulated by gene alteration at the DNA level, transcriptional regulation,<br />

<strong>and</strong> mature miRNA processing via Dicer. Thus far, few studies have simultaneously<br />

assessed all three levels <strong>of</strong> regulation. Using real-time quantitative polymerase chain<br />

reaction (QPCR)-based arrays, changes in gene copy number, pre-miRNA <strong>and</strong> mature<br />

miRNA levels for a large set <strong>of</strong> primary effusion lymphomas (PELs) <strong>and</strong> primary Kaposi<br />

Sarcoma biopsies (KS) has been determined; this includes miRNA gene alterations <strong>and</strong><br />

concordant changes in pre-miRNA <strong>and</strong> mature miRNA expression levels. The real-time<br />

QPCR based approach confirmed many <strong>of</strong> the KSHV viral <strong>and</strong> cellular miRNAs previously<br />

cloned from PEL. However, array-based pr<strong>of</strong>iling also uncovered many novel PEL–specific<br />

miRNAs, since cloning-based approaches are not always saturating. The miRNA<br />

expression pattern for neither viral nor cellular miRNAs has hitherto been determined for<br />

KS. Furthermore, comprehensive SNP analysis <strong>of</strong> the viral miRNAs has been pr<strong>of</strong>iled to<br />

further examine the effects <strong>of</strong> sequence <strong>and</strong> processing. This defines the miRNA<br />

signature <strong>of</strong> PEL, KS, KSHV-infected cancers <strong>and</strong> experimental models. It shows that the<br />

transcriptional regulation <strong>of</strong> pre-miRNA as well as mature miRNA levels contribute nonredundant<br />

information that can be used in the classification <strong>of</strong> human tumors.<br />

Presenting author Email: ohara@unc.edu<br />

56


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gene Expression I Abstract 33<br />

INEFFICIENT CODON USAGE IN V-FLIP MRNA LEADS TO TRANSCRIPT<br />

INSTABILITY<br />

Priya Bellare, Andrew T Dufresne <strong>and</strong> Don Ganem<br />

HHMI <strong>and</strong> G.W. Hooper Foundation, <strong>University</strong> <strong>of</strong> California, San Francisco, California<br />

94143-0552, USA<br />

Abstract<br />

KSHV v-FLIP is a latent gene product that activates the NF-κB pathway, prolonging the<br />

survival <strong>of</strong> PEL cells <strong>and</strong> causing the characteristic spindled morphology <strong>of</strong> latently<br />

infected endothelial cells. The expression <strong>of</strong> v-FLIP is unusual in two regards. (i) The<br />

gene is expressed from a bi or tricistronic message that also encodes v-cyclin <strong>and</strong> LANA<br />

genes; to date, no monocistronic v-FLIP mRNA has been consistently detected in infected<br />

cells. (ii) The level <strong>of</strong> v-FLIP protein in infected or transfected cells is extremely low, even<br />

when a monocistronic mRNA is artificially engineered. Here we show that the low<br />

abundance <strong>of</strong> v-FLIP mRNA can be attributed to instability <strong>of</strong> the v-FLIP message, which<br />

in turn is a consequence <strong>of</strong> its sub-optimal codon usage. We transfected 293 cells with a<br />

vector designed to produce a monocistronic v-FLIP mRNA expressing Flag-tagged v-FLIP.<br />

As expected, v-FLIP protein levels were nearly undetectable; surprisingly, however, v-<br />

FLIP mRNA levels were also extremely low. Examination <strong>of</strong> codon usage in v-FLIP showed<br />

it to be dominated by poorly used codons throughout the length <strong>of</strong> the ORF. Expression<br />

<strong>of</strong> a re-engineered v-FLIP gene with efficient codon usage not only generated abundant<br />

v-FLIP protein but also restored mRNA accumulation to levels readily detected by<br />

Northern blotting. Analysis <strong>of</strong> chimeras <strong>of</strong> optimal <strong>and</strong> sub-optimal v-FLIP coding<br />

sequences reveals that mRNA stability cannot be mapped to a unique region <strong>of</strong> the<br />

mRNA; however, when an unstable v-FLIP construct is placed 3’ to a well expressed ORF,<br />

the resulting bicistronic mRNA is stable. These results explain many, but perhaps not all,<br />

<strong>of</strong> the prior conundrums <strong>of</strong> v-FLIP expression, <strong>and</strong> provide a striking example <strong>of</strong><br />

translational regulation <strong>of</strong> RNA accumulation, the mechanism <strong>of</strong> which is now under<br />

study.<br />

Presenting author Email: Priya.Bellare@ucsf.edu<br />

57


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gene Expression II Abstract 34<br />

ROLE OF MURINE GAMMAHERPESVIRUS-68 ORF37 IN MEDIATING INHIBITION<br />

OF HOST GENE EXPRESSION<br />

L. Roaden 1 , V. Sheridan 1 , B. Lane 1 , R. Sun 2 , B. Dutia 3 <strong>and</strong> B. Ebrahimi 1<br />

1 Division <strong>of</strong> <strong>Medical</strong> Microbiology, School <strong>of</strong> Infection & Host Defence, <strong>University</strong> <strong>of</strong><br />

Liverpool, Liverpool L69 3GA, UK, 2 Molecular Biology Institute, <strong>University</strong> <strong>of</strong> California,<br />

Los Angeles, California, USA, <strong>and</strong> 3 Centre for Infectious Diseases, Division <strong>of</strong> Veterinary<br />

Biomedical <strong>Sciences</strong>, <strong>University</strong> <strong>of</strong> Edinburgh, UK.<br />

Abstract<br />

In lytic infection, Murine gammaherpesvirus-68 (MHV-68) <strong>and</strong> Kaposi’s sarcoma<br />

associated herpesvirus (KSHV) <strong>and</strong> Epstein-Barr virus have been shown to mediate a<br />

global inhibition <strong>of</strong> host gene expression. g-herpesviruses, unlike a-herpesviruses, do<br />

not encode RNases. Recently, however, the KSHV ORF37 (an alkaline DNA exonuclease,<br />

also known as SOX) was shown to promote this shutdown by enhancing the degradation<br />

<strong>of</strong> cellular mRNAs. Similar observations have been made with the EBV homologue <strong>of</strong><br />

SOX. Interestingly, SOX does not appear to possess any apparent RNase activity. The<br />

murine gammaherpesvirus-68 (MHV-68) encodes an ORF37 highly homologous to KSHV<br />

ORF37 both at DNA (55% identity) <strong>and</strong> protein (44% identity) levels. Here we show that<br />

MHV-68 is capable <strong>of</strong> orchestrating a global repression <strong>of</strong> cellular gene expression. Using<br />

transient transfection assays <strong>and</strong> engineered reporters with very rapid half-lives, we<br />

show that ORF37 can independently cause rapid loss <strong>of</strong> these transcripts when expressed<br />

in transient transfection assays. Using point mutations, we have been able to dissect key<br />

amino acids which may relate to the RNase activity <strong>of</strong> MHV-68 ORF37. Furthermore, we<br />

have generated a stop mutant <strong>of</strong> MHV-68 ORF37 to assess the potential shutdown<br />

properties <strong>of</strong> this viral protein during lytic infection in vitro <strong>and</strong> in vivo <strong>and</strong> its potential<br />

role in host gene shut-<strong>of</strong>f during latency <strong>and</strong> reactivation from latency in vivo.<br />

Presenting author Email: ebrahimi@liv.ac.uk<br />

58


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 6 abstracts 35-41:<br />

Lytic Replication<br />

59


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Lytic Replication Abstract 35<br />

FUNCTIONAL CHARACTERIZATION OF KAPOSI'S SARCOMA–ASSOCIATED<br />

HERPESVIRUS ORF K8 BY BAC-BASED MUTAGENESIS<br />

Yan Wang, Charles Hollow <strong>and</strong> Yan Yuan<br />

Department <strong>of</strong> Microbiology, <strong>University</strong> <strong>of</strong> Pennsylvania School <strong>of</strong> <strong>Dental</strong> Medicine,<br />

Philadelphia, Pennsylvania 19104<br />

Abstract<br />

The open reading frame K8 <strong>of</strong> KSHV encodes a bZip protein, namely K8 protein or<br />

replication-associated protein (RAP) or K-bZip. Three functions have been reportedly<br />

associated with this protein. (i) K8 / RAP was found to bind to the origin <strong>of</strong> viral lytic DNA<br />

replication (ori-Lyt) <strong>of</strong> KSHV <strong>and</strong> its binding is absolutely required for the DNA<br />

replication. (ii) K8 / RAP causes cell cycle arrest at G1 phase through induction <strong>of</strong> C/EBP<br />

<strong>and</strong> p21. (iii) K8 / RAP interacts with virally encoded replication <strong>and</strong> transcription<br />

activator (RTA) <strong>and</strong> represses the transcription <strong>of</strong> viral delayed-early genes by RTA. But<br />

the mechanisms behind these activities <strong>of</strong> K8 still remain elusive. Especially most <strong>of</strong> data<br />

regarding K8 function were obtained in transient assays but not in the context <strong>of</strong> the<br />

virus. To study the role <strong>of</strong> K8 in viral life cycle, we generated K8-null recombinant viruses<br />

with bacterial artificial chromosome (BAC) <strong>and</strong> the recombineering technique. Stable<br />

293T cells carrying BAC-cloned wild type KSHV (BAC-36) <strong>and</strong> K8-null viral genomes<br />

(BAC- K8 <strong>and</strong> BAC-stopK8) were generated. When monolayers <strong>of</strong> 293T-BAC36, 293T-<br />

BAC- K8 <strong>and</strong> 293T-BAC-stop45 cells were induced with tetradecanoyl-phorbol-13acetate,<br />

no significant difference was found between them in overall viral gene<br />

expression. To our surprise, lytic DNA replication <strong>and</strong> extracellular virion particles were<br />

detected in the K8-null mutant viruses in the levels comparable to the wild type,<br />

suggesting that K8 is not absolutely required for viral DNA replication <strong>and</strong> viral<br />

propagation. The effects <strong>of</strong> the K8 mutations on host gene expression were also<br />

investigated.<br />

Presenting author Email: yuan2@pobox.upenn.edu<br />

60


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Lytic Replication Abstract 36<br />

APPLICATION OF ACTIVE KINOME COLLECTION FOR IDENTIFICATION OF A<br />

NOVEL KINASE FAMILY INVOLVED IN REACTIVATION OF KSHV<br />

Fang Cheng 1 , Markku Varjosalo 1 , Anne Lehtonen 1 , Magdalena Weidner-Glunde 2 , Päivi J.<br />

Koskinen 3 , Thomas Schulz 2 , Jussi Taipale 1 , <strong>and</strong> Päivi M. Ojala 1 .<br />

1 Genome-Scale Biology Program, Biomedicum Helsinki & Institute <strong>of</strong> Biomedicine,<br />

<strong>University</strong> <strong>of</strong> Helsinki, Helsinki, Finl<strong>and</strong>, 2 Institute <strong>of</strong> Virology, Hannover <strong>Medical</strong> School,<br />

Hannover, 3 Turku Center for Biotechnology, Turku, Finl<strong>and</strong><br />

Abstract<br />

Infection by KSHV displays two different phases: latent <strong>and</strong> lytic. During latency, the<br />

viral genome is episomal, with only few viral genes expressed. Upon induction <strong>of</strong> the lytic<br />

cycle, extensive viral DNA replication <strong>and</strong> viral gene expression is initiated (viral<br />

reactivation), which leads to production <strong>of</strong> new viral particles. Host signal-transduction<br />

pathways are involved in the switch between latency <strong>and</strong> productive infection. By using a<br />

gain-<strong>of</strong>-function human kinome screen, we identified two new kinases, Pim-1 <strong>and</strong> -3, to<br />

be involved in KSHV reactivation. Ectopic expression <strong>of</strong> Pim-1 <strong>and</strong> Pim-3 Kinases induces<br />

viral lytic replication <strong>and</strong> production <strong>of</strong> progeny viruses. Silencing <strong>of</strong> Pim-1 <strong>and</strong> Pim-3 by<br />

RNA interference demonstrate that Pim-1 <strong>and</strong> Pim-3 are required for KSHV lytic<br />

reactivation. By performing experiments in both de novo <strong>and</strong> naturally infected KSHV cell<br />

models we were able to identify the molecular mechanism for Pim-1/3 induced viral<br />

reactivation. The identification <strong>of</strong> this novel cellular kinase family regulating the<br />

gammaherpesvirus life cycle will facilitate a deeper underst<strong>and</strong>ing <strong>of</strong> KSHV reactivation<br />

<strong>and</strong> could represent a potential novel target for therapeutic intervention.<br />

Presenting author Email: cheng.fang@helsinki.fi<br />

61


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Lytic Replication Abstract 37<br />

NOVEL FUNCTIONS OF K-RTA AND K-BZIP AS SUMO-TARGETING LIGASES AND<br />

EPIGENETIC REGULATORS<br />

Hsing-Jien Kung, Pei-Ching Chang, Latricia Fitzgerald, Tom Ellison, Paul Luciw* <strong>and</strong> Yoshi<br />

Izumiya<br />

Dept. <strong>of</strong> Biochemistry <strong>and</strong> Molecular Medicine, *Dept <strong>of</strong> Pathology, UC Davis Cancer<br />

Center, Sacramento CA 95864<br />

Abstract<br />

SUMO modification emerges as a major post-translational modification <strong>of</strong> viral <strong>and</strong><br />

cellular proteins. Sumoylation in concert with histone methylation at H3K9 are hallmarks<br />

<strong>of</strong> heterochromatin. Like phospho-tyrosines, sumoylated lysines also serve as signals<br />

linking components <strong>of</strong> signal transduction. SUMO interacting motifs (SIM) have been<br />

identified in a number <strong>of</strong> cellular regulatory proteins including PML, DAXX <strong>and</strong> RNF4.<br />

RNF4, a SUMO-targeting ubiquitin ligase, preferentially facilitates the degradation <strong>of</strong><br />

sumoylated proteins. Here we report the identification <strong>of</strong> K-Rta <strong>and</strong> K-bZIP as SUMO<br />

binding proteins, <strong>and</strong> have mapped the SIMs in these molecules. We have previously<br />

shown that K-bZIP behaves like a SUMO-ligase, via its ability to bind Ubc9 E2 SUMO<br />

conjugation enzyme (which is itself sumoylated). Studies from the labs <strong>of</strong> Hayward <strong>and</strong><br />

Wood showed that K-Rta behaves like a ubiquitin ligase. We reported that K-Rta<br />

decreases the overall level <strong>of</strong> sumoylated proteins, <strong>and</strong> is counteracted by K-bZIP. We<br />

now show that K-Rta preferentially degrades sumoylated interacting protein. Taken in the<br />

present context, these data are consistent with the notion that K-bZIP is a SUMOtargeting<br />

SUMO ligase, <strong>and</strong> K-Rta, a SUMO-targeting Ubiquitin ligase. If true, they are<br />

the first SUMO-targeting viral ligases identified. The ability <strong>of</strong> K-Rta <strong>and</strong> K-bZIP to<br />

modulate the activity <strong>and</strong> stability <strong>of</strong> sumoylated proteins raises the possibility <strong>of</strong> their<br />

roles in the epigenetic regulation <strong>of</strong> viral latency. Data will be presented that JMJD2A, a<br />

H3K9me3 histone demethylase, is sumoylated <strong>and</strong> its demethylase activity is directly<br />

blocked by K-bZIP.<br />

Presenting author Email: yizumiya@ucdavis.edu<br />

62


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Lytic Replication Abstract 38<br />

X-BOX BINDING PROTEIN 1 DOES NOT INDUCE EBV LYTIC REPLICATION IN<br />

PRIMARY EFFUSION LYMPHOMA<br />

Imogen Yi-Chun Lai <strong>and</strong> Paul Kellam<br />

MRC Centre for <strong>Medical</strong> Molecular Virology, Department <strong>of</strong> Infection, <strong>University</strong> <strong>College</strong><br />

London. 46 Clevel<strong>and</strong> Street, London W1T 4JF<br />

Abstract<br />

The plasma cell differentiation factor XBP-1 has been shown to transactivate the KSHV<br />

ORF50 promoter in primary effusion lymphoma (PEL) cells, leading to reactivation <strong>of</strong><br />

KSHV from latency. PEL is a plasmablast lymphoma, arrested immediately before<br />

terminal differentiation into antibody secreting plasma cells. PEL are negative for the<br />

spliced transcriptional active form <strong>of</strong> XBP-1, namely XBP-1s. All PEL cells are infected<br />

with Karposi’s Sarcoma-associated Herpesvirus (KSHV), with about 75% also co-infected<br />

with Epstein - Barr virus (EBV). However little is known about the reactivation <strong>of</strong> EBV<br />

from latency in PEL cells. XBP-1s has been suggested to transactivate the immediate<br />

early gene BZLF-1 <strong>of</strong> EBV. Here we investigate the effect <strong>of</strong> different inducers <strong>of</strong> EBV<br />

lytic reactivation in PEL cells. By using Western blot, we show that XBP-1s does not<br />

induce the lytic reactivation <strong>of</strong> EBV defined by lack <strong>of</strong> expression <strong>of</strong> BZLF-1, despite<br />

inducing lytic reactivation <strong>of</strong> KSHV.<br />

Presenting author Email: imogen.lai@ucl.ac.uk<br />

63


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Lytic Replication Abstract 39<br />

RECRUITMENT OF THE COMPLETE HTREX COMPLEX IS REQUIRED FOR KSHV<br />

INTRONLESS MRNA NUCLEAR EXPORT AND VIRUS REPLICATION<br />

James R. Boyne <strong>and</strong> Adrian Whitehouse<br />

Institute <strong>of</strong> Molecular <strong>and</strong> Cellular Biology, <strong>University</strong> <strong>of</strong> Leeds, Leeds, LS2 9JT, United<br />

Kingdom<br />

Abstract<br />

The majority <strong>of</strong> pre-mRNAs in higher eukaryotes contain introns <strong>and</strong> are exported via<br />

recruitment <strong>of</strong> the conserved export machinery, TREX, which occurs in a splicingdependent<br />

manner. In contrast, most Kaposi’s sarcoma associated herpesvirus (KSHV)<br />

mRNAs lack introns <strong>and</strong> are exported by the KSHV ORF57 protein. Herein, we show that<br />

ORF57 functions to recruit the human TREX (hTREX) complex to intronless viral mRNAs<br />

<strong>and</strong> that this recruitment, but not recruitment <strong>of</strong> the exon-junction complex, is essential<br />

for nuclear export. The formation <strong>of</strong> the ORF57-hTREX complex is mediated by a direct<br />

interaction between ORF57 <strong>and</strong> the export adapter, Aly. We show that a point mutation<br />

in ORF57 which disrupts the ORF57-Aly interaction leads to a failure in both the ORF57mediated<br />

recruitment <strong>of</strong> hTREX to viral mRNA <strong>and</strong> their subsequent nuclear export.<br />

Furthermore, using a KSHV replication system we demonstrate that expression <strong>of</strong> a<br />

trans-dominant Aly mutant disrupts hTREX assembly but retains an ORF57-Aly-TAP<br />

complex on the viral mRNA. Strikingly, in the absence <strong>of</strong> UAP56 <strong>and</strong> hTHO-complex we<br />

observed a dramatic decrease in intronless viral mRNA export <strong>and</strong> virus replication.<br />

These data provide the first direct evidence that the entire hTREX complex is essential for<br />

the export <strong>of</strong> viral intronless mRNAs <strong>and</strong> in turn, KSHV replication.<br />

Presenting author Email: bmbjb@bmb.leeds.ac.uk<br />

64


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Lytic Replication Abstract 40<br />

DOMAIN STRUCTURE AND FUNCTION OF KSHV ORF57 PROTEIN IN PROTEIN-<br />

PROTEIN AND PROTEIN-RNA INTERACTIONS<br />

Vladimir Majerciak <strong>and</strong> Zhi-Ming Zheng<br />

HIV <strong>and</strong> AIDS Malignancy Branch, National Cancer Institute, National Institutes <strong>of</strong> Health,<br />

10 Central Dr. 6N106, Bethesda, MD 20892-1868, USA<br />

Abstract<br />

Kaposi’s sarcoma-associated herpesvirus (KSHV) ORF57 is a multifunctional regulator in<br />

the expression <strong>of</strong> viral lytic genes. ORF57 enhances the expression <strong>of</strong> both intronless <strong>and</strong><br />

intron-containing viral genes <strong>and</strong> is essential for virus replication. However, the<br />

molecular mechanism <strong>of</strong> ORF57 action remains unclear. Our analysis <strong>of</strong> ORF57 amino<br />

acid (aa) sequence identified two distinctive parts <strong>of</strong> ORF57 based on secondary structure<br />

prediction: the N-terminal part has no obvious secondary structure (coil), but bears a<br />

strong antigenic epitope; the C-terminal part consists <strong>of</strong> several alpha-helixes <strong>and</strong> betasheets,<br />

with all weak antigenic epitopes. In vivo ORF57 was found in a multiprotein<br />

complex enriched in RNA-binding proteins including RNA helicase A, hnRNP U, nucleolin,<br />

<strong>and</strong> Aly/REF in association with RNA. The relaxed N-terminal region, in particular the<br />

nuclear localization signal 2 (NLS2, aa 121-130) that overlaps the antigenic epitope, was<br />

mapped to mediate these interactions. The interaction <strong>of</strong> ORF57 with nucleolin depends<br />

on RNAs. In lyticaly infected B cells, KSHV ORF57 interacts with nucleolin in nucleoplasm,<br />

but not colocalizes with nucleolin concentrated in nucleoli. The C-terminal structural<br />

region <strong>of</strong> ORF57 contains a domain for ORF57 dimerization. The dimer formation between<br />

mutant <strong>and</strong> wild-type ORF57 leads the mt ORF57 normally localized in the cytoplasm to<br />

translocate in the nucleus in co-transfected cells. Data indicate that KSHV ORF57<br />

associates with many protein partners through its different domains to diversify its<br />

functions in regulating expression <strong>of</strong> viral intronless or intron-containing genes at<br />

posttranscriptonal level.<br />

Presenting author Email: majerciv@mail.nih.gov<br />

65


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Lytic Replication Abstract 41<br />

KSHV ORF57 ENHANCES TRANSLATION OF VIRAL INTRONLESS MRNAS<br />

James R. Boyne, Adam Taylor <strong>and</strong> Adrian Whitehouse<br />

Institute <strong>of</strong> Molecular <strong>and</strong> Cellular Biology, <strong>University</strong> <strong>of</strong> Leeds, Leeds, LS2 9JT, United<br />

Kingdom<br />

Abstract<br />

The majority <strong>of</strong> Kaposi’s sarcoma associated herpesvirus (KSHV) mRNAs lack introns <strong>and</strong><br />

are exported by the ORF57 protein. We have shown that ORF57 binds specifically to<br />

intronless viral mRNAs <strong>and</strong> functions to recruit the human TREX (hTREX) complex, in<br />

contrast to the exon-junction complex, allowing efficient nuclear export <strong>of</strong> the viral<br />

intronless mRNAs. The failure <strong>of</strong> ORF57 to recruit EJC components to viral intronless<br />

mRNAs presents KSHV with an intriguing problem, as the EJC enhances translation <strong>of</strong><br />

cellular spliced mRNAs. The EJC specifically recruits the translation machinery onto the<br />

mRNA via an interaction with the cellular protein PYM, thereby enhancing mRNA<br />

translation. Herein, we demonstrate that KSHV ORF57 functions in translation<br />

enhancement, bypassing the EJC, by recruiting essential cellular translation factors. We<br />

shown, using an in vitro translation assay, that recombinant KSHV ORF57 can enhance<br />

translation <strong>of</strong> an in vitro transcribed KSHV intronless RNA. Moreover, coimmunoprecipitation<br />

assays demonstrate that KSHV ORF57 interacts with PYM <strong>and</strong> 48S<br />

preinitiation components in an RNA-independent manner in both transfected <strong>and</strong><br />

reactivated BCBL cells. Furthermore, GST-pulldown analysis shows that ORF57 only<br />

associates directly with PYM <strong>and</strong> no other members <strong>of</strong> the 48S preinitiation complex. This<br />

suggests that KSHV ORF57 enhances translation by recruiting the 48S preinitiation<br />

complex onto a viral intronless mRNA lacking an EJC, via its direct interaction with PYM.<br />

This data highlights a new role for the ORF57 protein in the KSHV lytic replication cycle.<br />

Presenting author Email: A.Whitehouse@leeds.ac.uk<br />

66


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 7 abstracts 42-47:<br />

Immunology II<br />

67


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology II Abstract 42<br />

MODULATION BY KSHV OF LIGANDS THAT MEDIATE NK CELL RECOGNITION<br />

Alexis Madrid <strong>and</strong> Don Ganem<br />

HHMI <strong>and</strong> Dept <strong>of</strong> Microbiology, UCSF, San Francisco, CA 94143-0552<br />

Abstract<br />

Natural Killer (NK) cells express an array <strong>of</strong> activating <strong>and</strong> inhibitory receptors on their<br />

surfaces. The inhibitory receptors generally recognize MHC class I (MHC-I) molecules on<br />

healthy cells, which are therefore not targeted for destruction. Activating receptors<br />

recognize a variety <strong>of</strong> cellular lig<strong>and</strong>s, many <strong>of</strong> which are upregulated in pathologic<br />

conditions. The balance <strong>of</strong> activating <strong>and</strong> inhibitory signals within the NK cell determines<br />

whether a target cell will be destroyed <strong>and</strong> an immune response mounted. KSHV<br />

encodes proteins that downregulate MHC-I on infected cells, thus potentially exposing<br />

these cells to NK cell mediated lysis. Therefore we hypothesized that KSHV would also<br />

have evolved mechanisms to evade killing by NK cells, <strong>and</strong> one potential way to evade<br />

killing is through downregulation <strong>of</strong> NK activating lig<strong>and</strong>s. To test this hypothesis, we<br />

infected several human cell lines with KSHV, <strong>and</strong> measured the cell surface levels <strong>of</strong> the<br />

various NK activating lig<strong>and</strong>s. The surface display <strong>of</strong> most NK lig<strong>and</strong>s tested was not<br />

affected during infection. However, we found that one NK activating lig<strong>and</strong>, NKp44-L, is<br />

consistently downregulated during both latent <strong>and</strong> lytic KSHV infection. We then<br />

screened a KSHV ORF library in order to search for the viral gene product responsible for<br />

this activity, <strong>and</strong> identified this protein as Kaposin B. However, Nkp44-L downregulation<br />

is not due to Kaposin B’s known function in activating the p38/MK2 signaling pathway,<br />

<strong>and</strong> likely reflects an additional activity <strong>of</strong> the molecule. Studies <strong>of</strong> the mechanism by<br />

which Kaposin B downregulates NKp44-L are ongoing <strong>and</strong> will be reported.<br />

Presenting author Email: Alexis.madrid@ucsf.edu<br />

68


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology II Abstract 43<br />

SCREENING FOR NATURALLY OCCURRING ANTI-KCP (KSHV COMPLEMENT<br />

CONTROL PROTEIN; ORF4) ANTIBODIES IN KSHV-INFECTED PATIENTS AND<br />

GENERATION OF SPECIFIC MONOCLONAL ANTIBODIES FOR PATHOGENESIS<br />

STUDIES<br />

Anna M. Blom 1a , Marcin Okroj, 1a Linda Mark, 1a Zoltan Korodi, 1b Rosamaria Tedeschi, 2<br />

Joakim Dillner 1b <strong>and</strong> O. Brad Spiller 3<br />

a Department <strong>of</strong> Laboratory Medicine, b Department <strong>of</strong> <strong>Medical</strong> Microbiology, 1 Lund<br />

<strong>University</strong>, <strong>University</strong> Hospital Malmö, S-205 02, Malmö, Sweden. 2 Department <strong>of</strong><br />

Microbiology, Oncological Center, Aviano 33081, Italy. 3 Department <strong>of</strong> Child Health,<br />

Cardiff <strong>University</strong>, School <strong>of</strong> Medicine, Cardiff CF14 4XN, United Kingdom.<br />

Abstract<br />

ORF4 encodes a complement regulating protein expressed in the late virus replication<br />

phase, referred to as the KSHV Complement Control Protein (KCP). KCP is expressed on<br />

the virion surface <strong>and</strong> can be induced on the surface <strong>of</strong> KSHV-positive primary effusion<br />

lymphoma (PEL) B-lymphocytes. KCP mediates protection from complement attack <strong>and</strong><br />

an integral heparin binding motif enables virion KCP to enhance virus infection through<br />

extracellular matrix binding. Using recombinant soluble KCP <strong>and</strong> transfected cells hyperexpressing<br />

various recombinant portions <strong>of</strong> KCP, serum from different KSHV-infected<br />

patient groups were investigated for anti-KCP reactivity. Specific anti-KCP IgG, but not<br />

IgM, was found predominantly in patients that had high titres <strong>of</strong> antibodies against other<br />

lytic KSHV proteins <strong>and</strong> the second (CCP2) <strong>of</strong> the 4 complement control domains in KCP<br />

was found to be the dominant epitope recognised. Patient anti-KCP antibodies enhanced<br />

complement attack on KCP-expressing CHO cells, suggesting neutralisation. Anti-KCP<br />

antibodies were found in all KSHV-infected lymphoma patients except one examined, but<br />

only 3/16 KS patients, suggesting differential KCP expression between these two<br />

diseases. Recombinant soluble <strong>and</strong> cell-expressed KCP were used to generate <strong>and</strong><br />

characterise monoclonal anti-KCP antibodies against each CCP domain. These<br />

monoclonal antibodies differentially regulated decay-accelerating factor activity, c<strong>of</strong>actor<br />

activity <strong>and</strong> heparin binding. Primary conjugated anti-KCP monoclonal antibodies<br />

labelled with FITC, PE, APC or HRP have now been created <strong>and</strong> await appropriate KSHV<br />

patient groups for longitudinal flow cytometric <strong>and</strong> histological assessment through<br />

disease progression <strong>and</strong> treatment.<br />

Presenting author Email: spillerb@Cardiff.ac.uk<br />

69


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology II Abstract 44<br />

A KSHV VIRAL HOMOLOGUE OF CELLULAR CD200 (VOX2) INDIRECTLY<br />

SUPPRESSES GRANULOCYTE OXIDATIVE ACTIVITY IN HUMAN BLOOD<br />

Karen Misstear 1 , Rachel Colman 1 , Hema Chahal 2 , Janet Lord 2 , David Blackbourn 1<br />

CRUK Institute for Cancer Studies 1 <strong>and</strong> Department <strong>of</strong> Immunity <strong>and</strong> Infection 2 ,<br />

<strong>University</strong> <strong>of</strong> Birmingham, Edgbaston, Birmingham B15 2TT, UK<br />

Abstract<br />

Background: Kaposi’s sarcoma-associated herpesvirus (KSHV) encodes a homologue<br />

(vOX2) <strong>of</strong> CD200, a transmembrane protein with known immunoregulatory activities<br />

exerted via its receptor, CD200R. vOX2 shares approximately 36% identity with CD200,<br />

suggesting similar roles, though vOX2 functions are incompletely understood. In vivo, an<br />

engineered, soluble vOX2:Fc reduced inflammation in the murine carrageenan footpad<br />

model, suggesting it suppresses neutrophil activity. vOX2 <strong>and</strong> CD200 bind CD200R with<br />

similar affinity, though a putative integrin binding domain indicates a larger receptor<br />

repertoire for vOX2 is likely.<br />

Methods: vOX2, CD200 <strong>and</strong> an inactive KSHV protein, KCPmut, were expressed with an<br />

in-frame carboxyl-terminal Fc domain <strong>of</strong> human IgG1. Human primary neutrophil<br />

activation was quantified by superoxide <strong>and</strong> myeloperoxidase release. vOX2:Fc <strong>and</strong><br />

CD200:Fc activity in whole blood was determined by flow cytometric analysis <strong>of</strong><br />

granulocyte oxidative activity. CD200R expression by leukocyte subpopulations was<br />

quantified by flow cytometry.<br />

Results <strong>and</strong> conclusions: vOX2:Fc <strong>and</strong> CD200:Fc exerted no effect upon isolated<br />

neutrophils, but suppressed granulocytic oxidative activity in whole blood by 19.13% <strong>and</strong><br />

24.34% respectively. These results suggest that vOX2:Fc acts upon granulocytes<br />

indirectly, via another leukocyte population(s). Natively expressed vOX2 <strong>and</strong> CD200<br />

inhibited the secretion <strong>of</strong> IL-8, a potent neutrophil chemoattractant, from a monocytic<br />

cell line. The expression <strong>of</strong> CD200R on major leukocyte subpopulations, including<br />

granulocytes, suggests that although vOX2:Fc ligates CD200R, it may not invoke<br />

signalling by this receptor.<br />

Presenting author Email: kxm694@bham.ac.uk<br />

70


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology II Abstract 45<br />

CD8+ T CELL RECOGNITION OF THE KSHV LATENT PROTEIN LANA1<br />

Shereen Sabbah & Andrew D Hislop<br />

CRUK Institute for Cancer Studies, The <strong>University</strong> <strong>of</strong> Birmingham, Edgbaston,<br />

Birmingham, B15 2TT, UK<br />

Abstract<br />

Kaposi’s sarcoma-associated herpesvirus (KSHV) has been implicated in the development<br />

<strong>of</strong> the endothelial malignancy, Kaposi’s sarcoma, <strong>and</strong> the B cell malignancies primary<br />

effusion lymphoma <strong>and</strong> multicentric Castleman’s disease. In these malignancies as well<br />

as KSHV infected cells, the viral genome maintenance protein latency associated nuclear<br />

antigen 1 (LANA1) is expressed. Potentially then, LANA1 is an attractive immune target.<br />

Similar to what is seen in the genome maintenance protein <strong>of</strong> the other human gammaherpesvirus,<br />

Epstein-Barr virus EBNA1, LANA1 contains extensive amino acid repeat<br />

sequences. In the case <strong>of</strong> EBNA1, these repeats reduce the efficiency <strong>of</strong> presentation <strong>of</strong><br />

EBNA1 derived epitopes to CD8+ T cells. To determine whether the repeats <strong>of</strong> LANA1<br />

afford a similar protection, we established a model system to examine the ability <strong>of</strong><br />

CD8+ T cells to recognise target cells expressing either LANA1 or a LANA1 construct<br />

deleted <strong>of</strong> the repeat region (LANA1-delta), both <strong>of</strong> which encode an inserted human<br />

CD8+ T cell epitope. We find that cells expressing the LANA1-delta protein express<br />

higher levels <strong>of</strong> this protein compared to the full length protein, yet CD8+ T cells<br />

recognise both target cells types efficiently. In this model system then, the LANA1<br />

repeat sequences give no protection against CD8+ T cell recognition. We are now<br />

mapping CD8+ T cell epitopes contained in LANA1 <strong>and</strong> will establish CD8+ T cell clones<br />

specific for identified epitopes. These T cell clones will be used to examine their ability to<br />

recognise natively processed <strong>and</strong> presented LANA1 epitopes on KSHV-infected cells.<br />

Presenting author Email: sxs463@bham.ac.uk<br />

71


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology II Abstract 46<br />

CYTOKINE RESPONSES BY CD8 T LYMPHOCYTES TO KSHV/HHV8 LYTIC AND<br />

LATENCY PROTEINS<br />

Lauren Lepone, Giovanna Rappocciolo, Emilee Knowlton, Paolo Piazza, Mariel Jais, Frank<br />

J. Jenkins <strong>and</strong> Charles R. Rinaldo<br />

Dept <strong>of</strong> Infectious Diseases <strong>and</strong> Microbiology, Graduate School <strong>of</strong> Public Health,<br />

<strong>University</strong> <strong>of</strong> Pittsburgh, Pittsburgh, PA<br />

Abstract<br />

Objectives: CD8 T cells producing more than one cytokine or cytotoxicity factor, i.e.<br />

polyfunctional, have been shown to relate to control <strong>of</strong> HIV-1 infection. Based on our<br />

previous findings that dendritic cells (DC) are optimally required as antigen presenting<br />

cells to reveal MHC class I epitopes <strong>of</strong> KSHV/HHV-8 glycoprotein B (gB) (Blood 99:3360,<br />

2002), we determined CD8 T cell cytokine responses to multiple lytic <strong>and</strong> latency viral<br />

proteins.<br />

Methods: T cells were stimulated with DC from HHV-8 seropositive, HLA A*0201 donors<br />

that had been loaded with overlapping peptides derived from gB <strong>and</strong> K8.1 lytic proteins,<br />

<strong>and</strong> K12 <strong>and</strong> LANA latency proteins, <strong>and</strong> screened for IFNγ production by ELISPOT.<br />

Positive peptides were assessed for IFNγ, IL2, TNFα, MIP1β, <strong>and</strong> CD107a production by<br />

flow cytometry.<br />

Results: ELISPOT <strong>and</strong> flow cytometry revealed new, potential HLA A*0201 9mer<br />

epitopes for gB (5), K8.1 (4), K12 (2) <strong>and</strong> LANA (5). In HHV-8 seropositive healthy<br />

donors controlling infection, the predominant CD8 T cell response to protein epitopes was<br />

mon<strong>of</strong>unctional, with a portion <strong>of</strong> CD8 T cells being polyfunctional.<br />

Conclusions: This demonstrates that HHV-8 proteins predominantly induce single<br />

cytokine or cytotoxicity factors in CD8 T cells from healthy, HHV-8 seropositive, HIV-1<br />

negative adults, with a portion <strong>of</strong> T cells producing 2-3 <strong>of</strong> these factors. These<br />

immunogenic regions <strong>of</strong> the viral proteins could be important in immunopathogenesis <strong>of</strong><br />

KSHV/HHV-8 infection <strong>and</strong> progression to Kaposi’s sarcoma.<br />

Presenting author Email: LML33@pitt.edu<br />

72


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Immunology II Abstract 47<br />

INFECTION OF LYMPHOID CELLS BY KSHV IN CULTURED PRIMARY HUMAN<br />

TONSILLAR LYMPHOID CELLS EX VIVO<br />

Jinjong Myoung <strong>and</strong> Don Ganem<br />

HHMI <strong>and</strong> Departments <strong>of</strong> Microbiology <strong>and</strong> Medicine, UCSF<br />

Abstract<br />

KSHV is known to infect B cells in vivo <strong>and</strong> is a causative agent <strong>of</strong> rare human B cell<br />

lymphomas (PELs). Paradoxically, however, established B cell lines are resistant to<br />

infection in vitro, which has greatly retarded progress in underst<strong>and</strong>ing the biology <strong>of</strong><br />

KSHV in its natural lymphoid compartment. We prepared primary human lymphoid<br />

aggregate cultures (HLACs) from tonsils, <strong>and</strong> examined their infectability by KSHV.<br />

Recombinant virus (rKSHV.219) expressing GFP under the EF1α promoter was prepared<br />

from induced Vero cells latently infected with this virus. Tonsil cells were infected with<br />

rKSHV, then subjected to flow cytometric analysis. When tonsil cells were infected with<br />

an MOI <strong>of</strong> 0.75, 15% <strong>of</strong> CD19+ cells became GFP-positive. Surprisingly, in the same<br />

culture many more T cells (54% <strong>of</strong> CD3+ cells) were shown to be GFP+; CD8+ T cells<br />

were consistently more susceptible than CD4+ T cells. This difference in infectivity in T<br />

cell subsets became more prominent when PHA was used to activate the T cells prior to<br />

infection. When exposed to chemical inducers <strong>of</strong> lytic growth, infected HLAC cultures<br />

displayed enhanced production <strong>of</strong> infectious KSHV.214, demonstrating that HLACs are<br />

competent to support the complete lytic cycle <strong>of</strong> KSHV. No immortalization or blastic<br />

transformation has yet been observed in infected HLACs. However, GFP+ B cells were<br />

significantly enriched when assessed at d13 post-infection, while the proportion <strong>of</strong> GFP+<br />

T cells decreased significantly by d13 PI. These data suggest that KSHV infection<br />

provides survival advantage in B cells, but not in T cells.<br />

Presenting author Email: jinjong.myoung@ucsf.edu<br />

73


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 8 abstracts 48-55:<br />

Clinical & Epidemiology<br />

74


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Clinical & Epidemiology Abstract 48<br />

PASSIVE TRANSFER OF HHV-8 ANTIBODIES FROM BLOOD DONORS TO<br />

TRANSFUSION RECIPIENTS AND POSSIBLE PROTECTION FROM HHV-8<br />

INFECTION<br />

Ashley L. Fowlkes 1 , Cedric Brown 1 , Minal M. Amin 1 , John Roback 2 , Robert Downing 3 , Esau<br />

Nzaro 4 , Jonathan Mermin 5 , Wolfgang Hladik 3 , Sheila C. Dollard 1<br />

1Centers for Disease Control <strong>and</strong> Prevention (CDC), Atlanta, GA. 2 Emory <strong>University</strong><br />

School <strong>of</strong> Medicine, Atlanta, GA, 3 Global AIDS Program, CDC, Ug<strong>and</strong>a; 4 Mulago Hospital,<br />

Kampala, Ug<strong>and</strong>a 5 Coordinating Office for Global Health, CDC Kenya<br />

Abstract<br />

Human herpesvirus 8 (HHV-8) has been found to be transmissible by blood transfusion.<br />

Examination <strong>of</strong> serially collected serum specimens from patients following blood<br />

transfusion with HHV-8 seropositive blood revealed that antibodies to HHV-8 appeared in<br />

several patients following transfusion then declined rapidly over a matter <strong>of</strong> weeks. The<br />

source <strong>of</strong> this antibody was determined to be the transfused blood. We compared the<br />

frequency <strong>of</strong> passive antibody transfer by donor serostatus, the frequency <strong>of</strong> HHV-8<br />

infection in the absence <strong>of</strong> passive antibody, <strong>and</strong> evaluated the risk <strong>of</strong> infection as a<br />

function <strong>of</strong> the number <strong>of</strong> seropositive transfusions <strong>and</strong> recipient antibody levels. Of 542<br />

recipients included, passive antibody was found in 69% <strong>of</strong> patients transfused with high<br />

antibody titer blood. Patients who received multiple HHV-8 seropositive transfusions <strong>and</strong><br />

those with highest levels <strong>of</strong> anti-HHV-8 antibodies were least likely to contract HHV-8<br />

infection from transfusion. Our study indicates that HHV-8 antibody is passively<br />

transferred by blood transfusion, waning within 40 days. Furthermore, higher levels <strong>of</strong><br />

HHV-8 antibody in donor blood may have conferred protection from infection in<br />

transfused patients, or indicated donors less likely to have infectious virus in the blood.<br />

Presenting author Email: sgd5@cdc.gov<br />

75


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Clinical & Epidemiology Abstract 49<br />

HEIGHTENED REDOX STATUS OF PRIMARY EFFUSION LYMPHOMA CELLS CAN BE<br />

EXPLOITED THERAPEUTICALLY BY DECREASING RESISTANCE TO OXIDATIVE<br />

STRESS<br />

Darya Bubman 1 , Utthara Nayar 2 , Balasz Csernus 3 , Ilaria Guasparri 3 , Ethel Cesarman 3<br />

1= Program in Pharmacology, Weill Cornell Graduate School <strong>of</strong> <strong>Medical</strong> <strong>Sciences</strong>, 2=<br />

Program in Immunology <strong>and</strong> Microbial Pathogenesis, Weill Cornell Graduate School <strong>of</strong><br />

<strong>Medical</strong> <strong>Sciences</strong>, 3= Department <strong>of</strong> Pathology <strong>and</strong> Laboratory Medicine, Weill Cornell<br />

<strong>Medical</strong> <strong>College</strong><br />

Abstract<br />

Intracellular reactive oxygen species (ROS) levels are maintained in a fine balance; high<br />

levels can induce an oxidative stress response <strong>and</strong> cell death, while chronic moderately<br />

high levels may promote neoplastic growth <strong>and</strong> proliferation. Specifically, a high level <strong>of</strong><br />

the superoxide (O2 - ) species is associated with cellular transformation. We have shown<br />

that PEL cells have higher levels <strong>of</strong> ROS, particularly superoxide, than KSHV-negative<br />

cells. Furthermore, we showed that vFLIP is responsible for these high levels <strong>of</strong><br />

superoxide, by both overexpression <strong>and</strong> knockdown approaches. PEL cells also express<br />

high levels <strong>of</strong> superoxide dismutases (SODs), specifically MnSOD, which is probably<br />

necessary for PEL cells to eliminate superoxide <strong>and</strong> avoid oxidative stress. We therefore<br />

determined whether oxidative stress could be used as an approach for the treatment <strong>of</strong><br />

PEL. 2-Methoxyestradiol (2ME2), or Panzem (Entremed), is an endogenous metabolite <strong>of</strong><br />

estrogen that has antiangiogenic <strong>and</strong> antitumor effects, <strong>and</strong> is currently in early clinical<br />

studies for a variety <strong>of</strong> neoplasms. While 2ME2 acts through multiple mechanisms <strong>of</strong><br />

action, these include inhibition <strong>of</strong> SOD activity <strong>and</strong> disruption <strong>of</strong> angiogenesis through the<br />

inhibition <strong>of</strong> hypoxia inducible factor-1 alpha (HIF-1α), a protein required for<br />

angiogenesis <strong>and</strong> cell survival under hypoxic <strong>and</strong> oxidative stress. We therefore treated<br />

cells with 2ME2 in vitro, <strong>and</strong> demonstrated that PELs, but not normal lymphocytes, were<br />

extremely sensitive to drug. We also determined whether this drug is effective in vivo, by<br />

using a mouse PEL xenograft model with in vivo imaging, following oral administration <strong>of</strong><br />

clinically formulated 2ME2.<br />

Presenting author Email: utn2001@med.cornell.edu<br />

76


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Clinical & Epidemiology Abstract 50<br />

RAPAMYCIN IS EFFECTIVE AGAINST PTEN POSITIVE AIDS-DEFINING<br />

MALIGNANCIES<br />

Debasmita Roy, Sang-Hoon Sin, Ling Wang, Blossom A. Damania, Dirk P. Dittmer<br />

<strong>University</strong> <strong>of</strong> North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA<br />

Abstract<br />

Primary Effusion Lymphoma (PEL), a B-cell lymphoma characteristic <strong>of</strong> AIDS <strong>and</strong><br />

immune-compromised patients, is tightly correlated with Kaposi’s Sarcoma-associated<br />

Herpes Virus (KSHV) infection. In this study we investigate the effect <strong>of</strong> inhibition <strong>of</strong><br />

mTor signaling (mammalian target <strong>of</strong> Rapamycin) in PEL. The anti-tumor potential <strong>of</strong><br />

Rapamycin has been the subject <strong>of</strong> intense investigations <strong>and</strong> we have shown previously<br />

that it is effective against PEL both in culture <strong>and</strong> our murine xenograft model via downregulation<br />

<strong>of</strong> cytokines, IL-6 <strong>and</strong> IL-10 (Sin et. al. Blood 2007. 109(5)). Here we show<br />

that the PEL-specific inhibitory effect <strong>of</strong> Rapamycin is also mediated by reduction <strong>of</strong> proangiogenic<br />

Vascular Endothelial Growth Factor (VEGF). We further find that the genetic<br />

status <strong>of</strong> the mTOR signaling cascade is intact in PEL cells, without mutations in negative<br />

regulators such as Phosphatase <strong>and</strong> Tensin Homolog (PTEN), Tuberous Sclerosis (TSC)<br />

Factor-1 <strong>and</strong> TSC-2. Typically, Akt activation results from deletion <strong>of</strong> the PTEN gene<br />

resulting in hyperactivation <strong>of</strong> mTor; instead we observe that Akt is activated <strong>and</strong> PTEN<br />

inactivated post-translationally, presumably by viral factors. Specifically, we show the<br />

presence <strong>of</strong> wildtype PTEN, but that it is silenced epigenetically, the mechanism <strong>of</strong> which<br />

differs between PEL <strong>and</strong> KSHV-infected endothelial cells. This suggests that in the special<br />

case <strong>of</strong> HIV-AIDS defining malignancies, Rapamycin can be effective in PTEN wild type<br />

tumors, contrary to previous reports <strong>of</strong> Rapamycin being effective exclusively in PTEN<br />

deleted tumors.<br />

Presenting author Email: debasmita_roy@med.unc.edu<br />

77


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Clinical & Epidemiology Abstract 51<br />

GEOGRAPHIC VARIATION OF THE PREVALENCE OF KAPOSI’S SARCOMA-<br />

ASSOCIATED HERPESVIRUS AND RISK FACTORS FOR TRANSMISSION IN<br />

WOMEN FROM 8 COUNTRIES IN FOUR CONTINENTS<br />

Silvia de Sanjose 1 , Georgina Mbisa 2 , Sussana Perez 1 , Sukhon Sukvirach 3 , Nguyen Trong<br />

Hieu 4 , Hai-Rim Shin 5 , Pham Thi Hoang Anh 4 , Jaiye O Thomas 6 , Eduardo Lazcano 7 , Elena<br />

Matos 8 , Rol<strong>and</strong>o Herrero 9 , Nubia Muñoz 10 , Silvia Franceschi 5 , Denise Whitby 2<br />

1. Institut Català d’Oncologia, Barcelona, Spain; 2. NCI-Frederick, Frederick, MD, USA; 3.<br />

Nacional Institute, Bangkok; 4. Hung Vuong Hospital, Ho Chi Minh City, Vietnam; 5.<br />

IARC, Lyon, France; 6. <strong>University</strong> <strong>of</strong> Ibadan, Ibadan, Nigeria; 7. Instituto Nacional de<br />

Salud Publica, Cuernavaca, Mexico; 8. Instituto de Oncologia Angel H R<strong>of</strong>fo, Universidad<br />

de Buenos Aires, Argentina; 9. Proyecto Epidemiologico Guanacaste, Fundación<br />

INCIENSA, San José, Costa Rica; 10. Instituto Nacional de Cancerologia, Bogota,<br />

Colombia<br />

Abstract<br />

Transmission routes <strong>of</strong> Kaposi’s sarcoma-associated herpes virus (KSHV) in the general<br />

population are poorly understood. Sexual transmission appears to be common in<br />

homosexual men but heterosexual transmission has not been clearly documented. This<br />

study aims to estimate the prevalence <strong>of</strong> KSHV in the female general populations <strong>of</strong><br />

Argentina, Colombia, Costa Rica, Nigeria, Spain, Vietnam, Thail<strong>and</strong> <strong>and</strong> Korea to explore<br />

geographical variation <strong>and</strong> potential heterosexual transmission. Samples <strong>and</strong><br />

questionnaire data were available from a study organized by the International Agency for<br />

Research on Cancer (IARC) to estimate the prevalence <strong>of</strong> distinct sexually transmitted<br />

infections. The study includes 10963 women from 10 centers with questionnaire<br />

information available on socio-demographic, reproductive <strong>and</strong> sexual lifetime<br />

experiences, smoking habits. HPV DNA detection was previously measured .Antibodies<br />

against KSHV encoded K8.1 <strong>and</strong> orf73 were determined. Prevalence <strong>of</strong> antibodies to any<br />

<strong>of</strong> the two antigens k8.1 or orf73 was 13.9% with an important geographical variation<br />

(range= Nigeria 46%- 3.8% in Spain). Antibodies increased with increasing age<br />

particularly in high prevalent countries such as Nigeria, Colombia <strong>and</strong> Costa Rica. KSHV<br />

was not related to education, age at first sexual intercourse, number <strong>of</strong> sexual partners,<br />

number <strong>of</strong> children, patterns <strong>of</strong> use <strong>of</strong> oral contraceptives or presence <strong>of</strong> cervical HPV<br />

DNA. A decreased prevalence was observed with increasing number <strong>of</strong> cigarettes smoked<br />

per day ( p=0.000).<br />

The study provides reliable <strong>and</strong> comparable estimates <strong>of</strong> KSHV in diverse cultural settings<br />

across four continents <strong>and</strong> provides a powerful indication <strong>of</strong> absence <strong>of</strong> heterosexual<br />

transmission <strong>of</strong> KSHV.<br />

Presenting author Email: whitbyd@ncifcrf.gov<br />

78


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Clinical & Epidemiology Abstract 52<br />

SERUM EPIDEMIOLOGICAL STUDIES ON KAPOSI'S SARCOMA IN XINJIANG<br />

Lei YANG, Xiao-Hua TAN, Jiang-Mei QING, Feng LI, Shu-Xia GUO, Jian-Xin XIE, Jin<br />

HUANG, Dong-Mei LI,Yan ZENG<br />

Key Laboratory <strong>of</strong> Xinjiang Endemic <strong>and</strong> Ethnic Disease, Shihezi <strong>University</strong>,Shihezi,<br />

XinJiang, 832002, China<br />

Abstract<br />

Kaposi’s Sarcoma (KS) is a multiple malignant idiopathic pigmented sarcoma. KS has<br />

obvious characteristic <strong>of</strong> endemicity <strong>and</strong> ethnology. In China, Xinjiang Uygur Autonomous<br />

Region is one <strong>of</strong> the highest incidence regions <strong>of</strong> KS.A 1:4 matched case-control study<br />

was conducted to assess the role <strong>of</strong> environment <strong>and</strong> life style factor on the pathogenesis<br />

<strong>of</strong> KS in Xinjiang. We collected blood samples from17 cases <strong>of</strong> KS patients <strong>and</strong> 68 cases<br />

<strong>of</strong> non-KS patients as control. Infectious factors, some cytokines’ serum level <strong>and</strong><br />

immune state IL-6, TNF-α, IFN-γ, VEGF, Neopterrin, β2-MG were tested for both group<br />

using ELISA. We tested the infection rate <strong>of</strong> Human herpesvirus 8(HHV-8) by nested<br />

PCR. The strength <strong>of</strong> association <strong>of</strong> risk factors was determined using x 2 test. We found<br />

KS show no relationship to area <strong>of</strong> residence <strong>and</strong> environment factors. The level <strong>of</strong> VEGF,<br />

TNF-α, IL-6, neopterin <strong>and</strong> β2-miroglobin were significantly higher among cases than<br />

control. The positive infection rate <strong>of</strong> HHV-8 in KS patient (91.66%) was significantly<br />

higher (p


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Clinical & Epidemiology Abstract 53<br />

KAPOSI’S SARCOMA-ASSOCIATED HERPESVIRUS SEROCONVERSION AND<br />

SEROREVERSION IN A MATERNAL-INFANT COHORT IN ZAMBIA<br />

Veenu Minhas, 1 Kay L. Crabtree, 1 Janet Wojcicki, Tendai J. M’soka, 2 Chipepo Kankasa, 2<br />

Charles D. Mitchell, 3 <strong>and</strong> Charles Wood 1<br />

1 Nebraska Center for Virology, School <strong>of</strong> Biological <strong>Sciences</strong>, <strong>University</strong> <strong>of</strong> Nebraska<br />

Lincoln, Lincoln, NE USA; 2 Department <strong>of</strong> Paediatrics <strong>and</strong> Child Health, <strong>University</strong><br />

Teaching Hospital, Lusaka, Zambia; 3 Department <strong>of</strong> Pediatrics, <strong>University</strong> <strong>of</strong> Miami<br />

School <strong>of</strong> Medicine, Miami, FL USA.<br />

Abstract<br />

Zambia is currently experiencing an HIV-1 epidemic leading to an increase in the<br />

incidence <strong>of</strong> Kaposi’s sarcoma among adults <strong>and</strong> children. This is a prospective study<br />

designed to investigate Kaposi’s sarcoma-associated herpesvirus (KSHV) seroincidence<br />

<strong>and</strong> associated risk factors for KSHV seroconversion <strong>and</strong> seroreversion in a mother-infant<br />

cohort, by following them for 48 months after delivery. Our results show that overall<br />

KSHV seroincidence in KSHV negative Zambian women was 0.7 per 100 person-months<br />

which was lower as compared to children followed in the same cohort. We observed that<br />

up to 40% <strong>of</strong> Zambian children in this cohort may be infected by 48 months <strong>of</strong> age,<br />

indicating that primary infection occurs mostly in early childhood. HIV-1 infection was<br />

associated with higher risk for seroconversion in women <strong>and</strong> children. In adult women,<br />

KSHV seroconversion was not associated with most socio-economic factors, sexual habits<br />

or medical history. This indicates that sexual transmission during adulthood may not be<br />

an important route <strong>of</strong> transmission in this cohort.<br />

Our laboratory has earlier reported that KSHV antibodies may not persist over time in<br />

children <strong>and</strong> that seroreversion leading to partial or complete loss <strong>of</strong> KSHV antibodies is<br />

prevalent. We observed seroreversion in up to 55% <strong>of</strong> women <strong>and</strong> this was more<br />

common in single women <strong>and</strong> in women delivering for the first time. These results<br />

demonstrate that multiple longitudinal samples per patient may be required to accurately<br />

determine KSHV infection status.<br />

Presenting author Email: veenu@bigred.unl.edu<br />

80


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Clinical & Epidemiology Abstract 54<br />

DISTRIBUTION OF KSHV MICRORNA POLYMORPHISMS IN AIDS-KS, CLASSICAL<br />

KS, AND MULTICENTRIC CASTLEMAN’S DISEASE<br />

Vickie Marshall, 1 Eliza Martró, 2 Elizabeth Brown, 3 Dian Wang, 1 Alex Ray, 1 Maria<br />

Nazzarena Labo, 1 The Classical Kaposi’s Sarcoma Working Group, Robert Yarchoan, 4<br />

Jordi Casabona, 2 Rolfe Renne, 5 <strong>and</strong> Denise Whitby 1<br />

1 NCI-Frederick, Frederick, MD, USA; 2 Hospital Universitari Germans Trias i Pujol,<br />

Badalona, Spain; 3 <strong>University</strong> <strong>of</strong> Alabama at Birmingham, Birmingham, AL, USA; 4 Division<br />

<strong>of</strong> Clinical Services, NCI, NIH, Bethesda, MD, USA; 5 <strong>University</strong> <strong>of</strong> Florida Sh<strong>and</strong>s Cancer<br />

Center, Gainsville, FL, USA<br />

Abstract<br />

MicroRNAs are short regulatory RNAs <strong>of</strong> approximately 19-24 nucleotides in length that<br />

bind specifically to mRNA in a process that interferes with translation or promotes<br />

degradation. KSHV encodes twelve microRNAs, all located within the latency associated<br />

transcript which is highly expressed in KSHV-associated malignancies.<br />

We amplified, cloned, <strong>and</strong> sequenced a 2,712 bp non-coding region containing a cluster<br />

<strong>of</strong> 10 microRNAs <strong>and</strong> a 646 bp fragment <strong>of</strong> the K12/T0.7 gene which contains two<br />

microRNAs. We successfully characterized KSHV microRNA sequences from lymphocytes<br />

collected as part <strong>of</strong> two Kaposi’s sarcoma case-control studies (one AIDS-KS, one<br />

Classic–KS) <strong>and</strong> MCD clinic attendees from the NCI. We have completely or partly<br />

characterized all twelve KSHV microRNAs from a total <strong>of</strong> 37 Kaposi sarcoma cases, 21<br />

KSHV positive controls, <strong>and</strong> 8 MCD patients.<br />

Phylogenetic analysis <strong>of</strong> both regions demonstrates extreme conservation <strong>of</strong> microRNAs<br />

overall. However, several distinct polymorphisms were discovered in this study, some<br />

occurring within the mature microRNA transcript. Phylogenetic characterization <strong>of</strong> the<br />

KSHV K1 <strong>and</strong> viral microRNA sequences in this study indicated all were subtypes A/C.<br />

However, we did detect four microRNA polymorphisms we previously described as<br />

specific to KSHV viral subtypes A5/B. Some polymorphisms appear to be more common<br />

in Kaposi’s sarcoma <strong>and</strong> MCD cases than in KSHV positive controls.<br />

Our data demonstrate that most KSHV microRNAs are highly conserved suggesting that<br />

they contribute to the biological activity <strong>and</strong> possibly viral pathogenesis <strong>of</strong> KSHV.<br />

However, some KSHV microRNAs have distinct polymorphisms which may affect<br />

processing, function, <strong>and</strong> mRNA target recognition.<br />

Presenting author Email: marshall@ncifcrf.gov<br />

81


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Clinical & Epidemiology Abstract 55<br />

EARLY CHILDHOOD INFECTION BY KAPOSI’S SARCOMA-ASSOCIATED<br />

HERPESVIRUS AND EPSTEIN-BARR VIRUS IN ZAMBIA<br />

Veenu Minhas, 1* Brad P Brayfield, 1 Tendai J. M’soka, 2 Chipepo Kankasa, 2 Charles D.<br />

Mitchell, 3 <strong>and</strong> Charles Wood, 1<br />

1 Nebraska Center for Virology, School <strong>of</strong> Biological <strong>Sciences</strong>, <strong>University</strong> <strong>of</strong> Nebraska<br />

Lincoln, Lincoln, NE USA; 2 Department <strong>of</strong> Paediatrics <strong>and</strong> Child Health, <strong>University</strong><br />

Teaching Hospital, Lusaka, Zambia; 3 Department <strong>of</strong> Pediatrics, <strong>University</strong> <strong>of</strong> Miami<br />

School <strong>of</strong> Medicine, Miami, FL USA.<br />

Abstract<br />

Significant differences exist between developed <strong>and</strong> developing countries in the<br />

prevalence rate, age <strong>of</strong> infection <strong>and</strong> the observed clinical manifestations for Kaposi’s<br />

sarcoma-associated herpesvirus (KSHV) <strong>and</strong> Epstein-Barr virus (EBV) infections. Both are<br />

oncogenic gamma-herpesviruses that have been associated with Kaposi’s sarcoma <strong>and</strong><br />

AIDS-related non-Hodgkin lymphomas, respectively. Therefore, it is <strong>of</strong> interest to<br />

underst<strong>and</strong> the natural history <strong>of</strong> infection <strong>of</strong> these two gamma-herpesviruses in an<br />

endemic area, their potential interaction <strong>and</strong> the role <strong>of</strong> human immunodeficiency virus<br />

(HIV) infection <strong>and</strong> other risk factors that may be associated with early childhood<br />

infection. This study was conducted on a cohort <strong>of</strong> 12 month old infants from Lusaka,<br />

Zambia. A set <strong>of</strong> pre-tested st<strong>and</strong>ardized questionnaires was used to gather data from<br />

the caregivers <strong>and</strong> multivariate analysis was performed to assess the association <strong>of</strong> a<br />

range <strong>of</strong> risk factors to seroprevalence. We observed a significantly higher<br />

seroprevalence for EBV (60.7%) as compared to KSHV (15.4%). HIV infected children<br />

were at a significantly higher risk <strong>of</strong> being infected with KSHV. The HIV positive status <strong>of</strong><br />

their mothers was a significant risk factor for increased risk <strong>of</strong> EBV but not KSHV<br />

transmission to children. Presence <strong>of</strong> rashes was significantly associated with higher risk<br />

<strong>of</strong> seroconversion for KSHV <strong>and</strong> EBV. Most other medical <strong>and</strong> socio-economic factors<br />

were not associated with increased risk. High viral burdens during early childhood<br />

especially in HIV infected children may be contributing to higher morbidity, mortality or<br />

risk <strong>of</strong> developing childhood cancers in young children.<br />

Presenting author Email: veenu@bigred.unl.edu<br />

82


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 10 abstracts 56-63:<br />

Virus-Cell Interactions II<br />

83


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions II Abstract 56<br />

INTRACELLULAR LOCALIZATION MAP OF HHV-8 PROTEINS<br />

Gaby S<strong>and</strong>er 1 , Andreas Konrad 1 , Mathias Thurau 1 , Effi Wies 2 , Rene Leubert 3 , Elisabeth<br />

Kremmer 4 , Holger Dinkel 5 , Thomas Schulz 6 , Frank Neipel 2 <strong>and</strong> Michael Stürzl 1<br />

1 Division <strong>of</strong> Molecular <strong>and</strong> Experimental Surgery, Department <strong>of</strong> Surgery, <strong>University</strong> <strong>of</strong><br />

Erlangen-Nuremberg, Schwabachanlage 10, D-91054 Erlangen, Germany; 2 Institute <strong>of</strong><br />

Clinical <strong>and</strong> Molecular Virology, <strong>University</strong> <strong>of</strong> Erlangen-Nuremberg, Schlossgarten 4, D-<br />

91054 Erlangen, Germany; 3 Department <strong>of</strong> Virus-induced Vasculopathy, GSF-National<br />

Research Center for Environment <strong>and</strong> Health, Ingolstädter L<strong>and</strong>strasse 1, D-85764<br />

Neuherberg, Germany; 4 GSF-Service Unit Monoclonal Antibodies <strong>and</strong> Cell Sorting, GSF-<br />

National Research Center for Environment <strong>and</strong> Health, Marchioninistr. 25, D-81377<br />

Munich, Germany; 5 Division <strong>of</strong> Bioinformatics, Institute <strong>of</strong> Biochemistry, <strong>University</strong> <strong>of</strong><br />

Erlangen-Nuremberg, Fahrstrasse 17, D-91054 Erlangen, Germany; 6 <strong>Medical</strong> School<br />

Hannover, Department <strong>of</strong> Virology, Carl-Neubergstrasse 1, D-30625 Hannover, Germany<br />

Abstract<br />

Human herpesvirus-8 (HHV-8) is the etiological agent <strong>of</strong> Kaposi’s sarcoma. We present a<br />

localization map <strong>of</strong> 85 HHV-8-encoded proteins in mammalian cells. Viral open reading<br />

frames were cloned with a Myc-tag in expression plasmids, confirmed by full-length<br />

sequencing, <strong>and</strong> expressed in HeLa cells. Protein localizations were analyzed by<br />

immun<strong>of</strong>luorescence microscopy. 51% <strong>of</strong> all proteins were localized in the cytoplasm,<br />

22% in the nucleus, <strong>and</strong> 27% were found in both compartments. Surprisingly, we<br />

detected vFLIP in the nucleus <strong>and</strong> in the cytoplasm, whereas cellular FLIPs are generally<br />

localized exclusively in the cytoplasm. This suggested that vFLIP may exert additional or<br />

alternative functions as compared to cellular FLIPs. In addition, it has been shown<br />

recently that K10 can bind to at least 15 different HHV-8 proteins. We noticed that K10<br />

but only five <strong>of</strong> its 15 putative binding factors were localized in the nucleus when<br />

expressed in HeLa cells individually. Interestingly, in co-expression experiments K10 colocalized<br />

with 87% (13 <strong>of</strong> 15) <strong>of</strong> its putative binding partners. Co-localization was<br />

induced by translocation <strong>of</strong> either K10 alone or <strong>of</strong> both proteins. These results indicate<br />

active intracellular translocation processes in virus infected cells. Specifically in this<br />

framework, the localization map may provide a useful reference to further elucidate the<br />

function <strong>of</strong> HHV-8-encoded genes in human diseases.<br />

Presenting author Email: gaby.s<strong>and</strong>er@uk-erlangen.de<br />

84


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions II Abstract 57<br />

KSHV INTERFERON REGULATORY FACTOR 4, A NOVEL CBF1 INTERACTION<br />

PARTNER<br />

Katharina Heinzelmann 1+ , Barbara Scholz 1*+ , Elisabeth Kremmer 2 , Jürgen Haas 3 , Even<br />

Fossum 3 , Bettina Kempkes 1<br />

Helmholtz Zentrum München, Institute <strong>of</strong> Clinical Molecular Biology <strong>and</strong> Tumor Genetics 1 ,<br />

Institute <strong>of</strong> Molecular Immunology 2 , Munich, Germany; Max-von-Pettenk<strong>of</strong>er-Institute 3 ,<br />

Ludwig-Maximilians-<strong>University</strong>, Munich, Germany;<br />

+ These authors contributed equally.<br />

Abstract<br />

Activation <strong>of</strong> the Notch transmembrane receptor by lig<strong>and</strong> binding induces the proteolytic<br />

cleavage <strong>and</strong> release <strong>of</strong> an intracellular Notch fragment which can bind to the CBF1<br />

protein. CBF1 (also named CSL, Su(H), RBP-Jk) is a sequence specific DNA binding<br />

protein, which can recruit corepressor complexes to target genes <strong>and</strong> thereby repress<br />

transcription. Notch binding results in corepressor release <strong>and</strong> assembly <strong>of</strong> a coactivator<br />

complex. The KSHV proteins RTA <strong>and</strong> LANA-1 have been recently shown to bind to CBF1.<br />

CBF1/RTA interactions are critical for the lytic life cycle <strong>of</strong> the virus, while the<br />

CBF1/LANA-1 interaction might establish a regulatory loop which controls lytic<br />

reactivation.<br />

We have identified a novel CBF1 interacting KSHV protein, the viral interferon regulatory<br />

factor 4 (vIRF4). The KSHV genome encodes four viral proteins, which share homology to<br />

cellular interferon regulatory (cIRF) transcrition factors. Although vIRFs in contrast to<br />

cIRFs lack DNA binding functions, three vIRFs (vIRF1-3) can still modulate the interferon<br />

response by diverse mechanisms. For vIRF4 such activities have not been reported. We<br />

show, that vIRF4 is the only CBF1 interacting vIRF. Within vIRF4 we have mapped two<br />

distinct CBF1 binding regions. CBF1 mutants, which are deficient for Notch binding, no<br />

longer bind vIRF4 suggesting that Notch <strong>and</strong> vIRF4 binding sites might overlap. In Gal4fusion<br />

based reporter gene assays vIRF4 scores as a repressor. Since vIRF4 is induced<br />

upon lytic cycle reactivation, the CBF1/vIRF4 interaction might be an additional critical<br />

factor controlling the balance <strong>of</strong> lytic <strong>and</strong> latent life cycle <strong>of</strong> KSHV.<br />

Presenting author Email: kempkes@helmholtz-muenchen.de<br />

85


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions II Abstract 58<br />

CHARACTERISATION OF ENDOPLASMIC RETICULUM (ER) STRESS INDUCERS<br />

THAT LEAD TO KSHV REACTIVATION FROM LATENCY; IDENTIFYING<br />

PHYSIOLOGICAL TRIGGERS?<br />

Lucy Dalton-Griffin, Ed Tsao <strong>and</strong> Paul Kellam<br />

MRC centre for <strong>Medical</strong> Molecular virology, Department <strong>of</strong> Infection,<br />

<strong>University</strong> <strong>College</strong> London, 46 Clevel<strong>and</strong> Street, London, W1T 4JF, UK<br />

Abstract<br />

Kaposi’s Sarcoma-Associated Herpesvirus (KSHV) like other herpesviruses has two stages<br />

to its life cycle; latency <strong>and</strong> lytic replication. KSHV is known to be required for<br />

development <strong>of</strong> Kaposi’s Sarcoma, a tumour <strong>of</strong> endothelial origin <strong>and</strong> is associated with<br />

the B-cell tumours, Primary Effusion Lymphomas (PELs) <strong>and</strong> the B-cell polyclonal<br />

expansion Multicentric Castleman’s Disease (MCD). Recently we <strong>and</strong> others have shown<br />

that the transcription factor X-box binding protein-1 (XBP-1) is a physiological trigger <strong>of</strong><br />

KSHV lytic reactivation in PEL. XBP-1 is responsible for the terminal differentiation <strong>of</strong> Bcells<br />

into plasma cells (PCs) as it is a major regulator <strong>of</strong> the unfolded protein response<br />

(UPR) <strong>and</strong> therefore allows production <strong>of</strong> large quantities <strong>of</strong> immunoglobulin in these<br />

cells. We have previously shown that PEL are blocked in differentiation by the absence <strong>of</strong><br />

active XBP-1; when provided the PEL cells differentiate towards a PC <strong>and</strong> induce KSHV<br />

lytic replication. Using an RFP lytic reactivation reporter assay we show that the ER stress<br />

inducer DTT activates the KSHV lytic cycle in an XBP-1 dependent manner. Recent<br />

studies have also indicated a role for hypoxia in reactivation via the hypoxia inducible<br />

factors (HIFs). Hypoxia can also result in XBP-1 activation as it is an ER stress inducer.<br />

Here we have investigated the roles <strong>of</strong> HIF-1α, HIF-2α <strong>and</strong> XBP-1 in activating the KSHV<br />

lytic cycle.<br />

Presenting author Email: ucbclda@ucl.ac.uk<br />

86


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions II Abstract 59<br />

INTEGRIN αVβ3 BINDS TO THE RGD MOTIF OF THE GLYCOPROTEIN B OF<br />

KAPOSI’S SARCOMA-ASSOCIATED HERPESVIRUS (KSHV/HHV8) AND<br />

FUNCTIONS AS AN RGD-DEPENDENT ENTRY RECEPTOR<br />

H. Jacques Garrigues 1 , Yelena E. Rubinchikova 1 , C. Michael DiPersio 2<br />

<strong>and</strong> Timothy M. Rose 1,3<br />

1 Seattle Children’s Hospital Research Institute, Seattle WA, 2 Albany <strong>Medical</strong> <strong>College</strong>,<br />

Albany, New York, <strong>and</strong> 3 <strong>University</strong> <strong>of</strong> Washington, Seattle WA<br />

Abstract<br />

The virion-associated glycoprotein B (gB) <strong>of</strong> KSHV is involved in the initial steps <strong>of</strong><br />

binding cells during KSHV infection. Glycoprotein B contains an RGD motif reported to<br />

bind the integrin α3β1 during virus entry. Although the lig<strong>and</strong> specificity <strong>of</strong> α3β1 has been<br />

controversial, current literature indicates that α3β1 lig<strong>and</strong> recognition is independent <strong>of</strong><br />

RGD. We compared α3β1 to αVβ3, a known RGD-binding integrin, for binding to envelopeassociated<br />

gB <strong>and</strong> a peptide, gB(RGD), containing the RGD motif. Adhesion assays<br />

demonstrated that β3-CHO cells overexpressing αVβ3 specifically bound gB(RGD), whereas<br />

α3-CHO cells overexpressing α3β1 did not. Function-blocking antibodies to αVβ3 inhibited<br />

adhesion <strong>of</strong> HT1080 fibrosarcoma cells to gB(RGD), while antibodies to α3β1 did not.<br />

Using affinity-purified integrins <strong>and</strong> confocal-microscopy, we determined that αVβ3 bound<br />

to gB(RGD) <strong>and</strong> KSHV virions demonstrating direct receptor-lig<strong>and</strong> interactions. Specific<br />

αVβ3 antagonists, including cyclic <strong>and</strong> di-cyclic RGD peptides <strong>and</strong> αVβ3 function-blocking<br />

antibodies, strongly inhibited KSHV infection. Keratinocytes from α3-null mice lacking<br />

α3β1 were fully competent for infection by KSHV <strong>and</strong> reconstitution <strong>of</strong> α3β1 function by<br />

transfection with α3 cDNA reduced KSHV infectivity. Additional inhibitory effects <strong>of</strong> α3β1<br />

were detected on the expression <strong>of</strong> αVβ3 <strong>and</strong> on αVβ3-mediated adhesion <strong>of</strong> α3-CHO cells<br />

overexpressing α3β1, consistent with previous reports <strong>of</strong> transdominant inhibition <strong>of</strong><br />

αVβ3function by α3β1. These observations may explain previous reports <strong>of</strong> an inhibition <strong>of</strong><br />

KSHV infection by soluble α3β1. Our studies demonstrate that αVβ3 is a cellular receptor<br />

mediating both cell adhesion <strong>and</strong> entry <strong>of</strong> KSHV into target cells through binding the RGD<br />

motif <strong>of</strong> the virion-associated gB.<br />

Presenting author Email: timothy.rose@seattlechildrens.org<br />

87


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions II Abstract 60<br />

KAPOSI’S SARCOMA ASSOCIATED HERPESVIRUS (KSHV/HHV-8) FORMS A<br />

MULTI-MOLECULAR COMPLEX OF INTEGRINS (αVβ5, αVβ3 AND α3β1), CD98 AND<br />

XCT DURING INFECTION OF HUMAN DERMAL MICROVASCULAR ENDOTHELIAL<br />

(HMVEC-D) CELLS<br />

Mohanan Valiya Veettil, Fu-Zhang Wang, Sathish Sadagopan, Neelam Sharma-Walia,<br />

Hari Raghu, Laszlo Varga <strong>and</strong> Bala Ch<strong>and</strong>ran<br />

Presenting author. H.M. Bligh cancer research laboratories, Department <strong>of</strong> Microbiology<br />

<strong>and</strong> Immunology, Chicago <strong>Medical</strong> School, Rosalind Franklin <strong>University</strong> <strong>of</strong> Medicine <strong>and</strong><br />

Science, 3333 Green Bay Road, North Chicago, IL 60064, USA<br />

Abstract<br />

Baculovirus expressed purified KSHV-gB mediated the adhesion <strong>of</strong> HMVEC-d, HFF, CV-1<br />

<strong>and</strong> HT-1080 cells. Anti-αV <strong>and</strong> β1 integrin antibodies inhibited the cell adhesion.<br />

Variable levels <strong>of</strong> neutralization <strong>of</strong> HMVEC-d <strong>and</strong> HFF cell infection was observed with<br />

anti- αVβ3 <strong>and</strong> αVβ5 integrin antibodies, <strong>and</strong> variable levels <strong>of</strong> inhibition <strong>of</strong> virus entry in<br />

four adherent cell types was observed with soluble integrins. Virus binding <strong>and</strong> DNA<br />

internalization studies suggest that αVβ3 <strong>and</strong> αVβ5 integrins play roles in KSHV entry.<br />

We observed time dependent temporal KSHV interactions with HMVEC-d cell integrins,<br />

CD98 <strong>and</strong> xCT. Integrin αvβ5 interaction with CD98 predominantly occurred by 1’ postinfection<br />

(PI) <strong>and</strong> dissociated at 10’ PI, whereas α3β1-CD98 interaction was maximum at<br />

10’ PI which dissociated at 30’ PI, <strong>and</strong> αvβ3 -CD98 interaction was maximum at 10’ PI<br />

<strong>and</strong> retained at the observed 30’ PI. Confocal microscopy studies demonstrated the<br />

association <strong>of</strong> CD98/xCT with α3β1 <strong>and</strong> KSHV. Pre-incubation <strong>of</strong> KSHV with soluble<br />

heparin <strong>and</strong> α3β1 significantly inhibited this association. KSHV infection activated FAK<br />

<strong>and</strong> Src co-localized with CD98 at the plasma membranes <strong>of</strong> infected cells. Anti-CD98<br />

<strong>and</strong> xCT antibodies did not block virus binding <strong>and</strong> entry; however, viral gene expression<br />

was significantly inhibited suggesting that CD98-xCT play roles in post entry-stage <strong>of</strong><br />

infection. Together, these studies suggest that KSHV interacts with functionally related<br />

integrins (αvβ5, α3β1 <strong>and</strong> αvβ3) <strong>and</strong> CD98/xCT molecules to form a multi-molecular<br />

complex during the early stages <strong>of</strong> endothelial cell infection probably mediating multiple<br />

roles in entry, signal transduction <strong>and</strong> viral gene expression.<br />

Presenting author Email: bala.ch<strong>and</strong>ran@rosalindfranklin.edu<br />

88


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions II Abstract 61<br />

MURINE GAMMAHERPESVIRUS 68 ESTABLISHES A PERSISTENT INFECTION IN<br />

ENDOTHELIAL CELLS<br />

A.L. Suárez <strong>and</strong> L.F. van Dyk<br />

<strong>University</strong> <strong>of</strong> Colorado Denver School <strong>of</strong> Medicine, Department <strong>of</strong> Microbiology, 12800 E<br />

19 th Ave, RC1 North Bldg P18-9401A, Aurora, CO 80010, USA<br />

Abstract<br />

Gammaherpesvirus associated diseases occur predominately in immunocompromised<br />

individuals. Gammaherpesvirus infection <strong>of</strong> cells usually results either in productive lytic<br />

infection, characterized by expression <strong>of</strong> all viral genes <strong>and</strong> complete cell lysis, or in<br />

latent infection, in which few viral genes are expressed <strong>and</strong> no cell lysis occurs.<br />

Herpesvirus infections must traverse endothelial cell (EC) barriers, <strong>and</strong> infection <strong>of</strong> ECs is<br />

important to the pathogenesis <strong>of</strong> both human cytomegalovirus <strong>and</strong> Kaposi’ sarcoma<br />

associated virus. The role <strong>of</strong> ECs in the mouse model system, gammaherpesvirus 68<br />

(gHV68), has not been previously characterized. Recently, we demonstrated that ECs<br />

were unique in that a significant proportion <strong>of</strong> the cells escaped lysis, remained viable<br />

<strong>and</strong> proliferated in culture, but continued to produce infectious virus. Infected ECs were<br />

altered in adherence, morphology <strong>and</strong> surface marker expression. However, infected ECs<br />

maintained MHC class I expression, suggesting that EC contribution to pathogenesis is<br />

likely limited to immunodeficient states. Primary ECs also supported productive infection<br />

with enhanced survival, <strong>and</strong> we are now investigating ECs as a source <strong>of</strong> persistent<br />

infection in vivo. From a panel <strong>of</strong> gHV68 mutants, we identified several viral oncogenes<br />

which promote survival <strong>of</strong> infected ECs. Intriguingly, the viral genes which promote EC<br />

survival have previously been characterized as important in latency, but dispensable for<br />

lytic infection. Our data reveal a cell type specific outcome <strong>of</strong> gHV68 infection which can<br />

provide an ongoing source <strong>of</strong> virus production. gHV68 infection <strong>of</strong> ECs provides a facile<br />

screen for viral persistence genes <strong>and</strong> investigation <strong>of</strong> EC infection in pathogenesis.<br />

Presenting author Email: <strong>and</strong>rea.suarez@uchsc.edu<br />

89


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions II Abstract 62<br />

TARGETING EXTRACELLULAR HSP90 REDUCES KSHV GENE EXPRESSION<br />

DURING DE NOVO INFECTION BY INHIBITING MAPK PATHWAY ACTIVATION<br />

Zhiqiang Qin 1 , Jennifer Isaacs 2 , <strong>and</strong> Chris Parsons 1,3<br />

Departments <strong>of</strong> 1 Medicine, 2 Pharmacology <strong>and</strong> 3 Microbiology/Immunology<br />

Hollings Cancer Center, <strong>Medical</strong> <strong>University</strong> <strong>of</strong> South Carolina<br />

Charleston, SC, USA<br />

Abstract<br />

Heat shock protein 90 (Hsp90) acts as a molecular chaperone orchestrating the folding <strong>of</strong><br />

intracellular signaling proteins. Although extracellular (EC) domains for Hsp90 have been<br />

identified, the functional consequences <strong>of</strong> engaging EC Hsp90 are largely unknown.<br />

Signal transduction induced during KSHV receptor binding <strong>and</strong> entry is critical for de novo<br />

viral gene expression. Using PCR <strong>and</strong> immun<strong>of</strong>luorescence assays, we found that<br />

treatment <strong>of</strong> HeLa cells with a non-permeable compound targeting EC Hsp90, DNo,<br />

inhibited de novo expression <strong>of</strong> latent KSHV transcripts <strong>and</strong> LANA. Pre-treatment <strong>of</strong> cells<br />

with DNo did not reduce intracellular KSHV DNA content, <strong>and</strong> DNo maintained an<br />

inhibitory effect when treatment was initiated following viral incubation. In addition, no<br />

effect was observed following KSHV pre-incubation with two soluble Hsp90 protein<br />

is<strong>of</strong>orms or a mAb targeting a different EC Hsp90 domain. These results suggest that<br />

DNo interferes with post-entry events important for viral gene expression. Immunoblot<br />

analyses <strong>of</strong> signal transduction intermediates indicated that mitogen-activated protein<br />

kinase kinase (MEK) <strong>and</strong> extracellular signal-regulated kinase (ERK) phosphorylation<br />

were selectively inhibited in a dose- <strong>and</strong> time-dependent manner by DNo. Furthermore,<br />

DNo blocked KSHV-mediated induction <strong>of</strong> MEK <strong>and</strong> ERK phosphorylation corresponding to<br />

a reduction in LANA expression. Finally, constructs conferring constitutive expression <strong>of</strong><br />

P-MEK <strong>and</strong> P-ERK, but not an ERK dominant negative construct, rescued KSHV gene<br />

expression in DNo-treated cells. These results suggest that EC Hsp90 plays an important<br />

role in KSHV-initiated signal transduction events <strong>and</strong> viral gene expression <strong>and</strong> that<br />

strategies targeting EC Hsp90 may have therapeutic benefit for KSHV-related disease.<br />

Presenting author Email: parsonch@musc.edu<br />

90


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Virus-Cell Interactions II Abstract 63<br />

FAK AND SHP2 ARE REQUIRED FOR KSHV VGPCR SIGNALING<br />

Thomas Bakken*, Chris Bosh<strong>of</strong>f**, Mark Cannon*<br />

*CIDMTR, Department <strong>of</strong> Medicine, <strong>University</strong> <strong>of</strong> Minnesota, ** C.R.U.K. Viral Oncology<br />

Group, <strong>University</strong> <strong>College</strong> London<br />

Abstract<br />

The KSHV vGPCR is a constitutively active homologue <strong>of</strong> CXCR1 <strong>and</strong> CXCR2. It shows<br />

potential both in vitro <strong>and</strong> in vivo to contribute to the proliferative, angiogenic <strong>and</strong><br />

inflammatory components <strong>of</strong> KS biology. In transgenic mice, vGPCR causes KS-like<br />

tumors <strong>and</strong> potentiates the tumorgenicity <strong>of</strong> other viral proteins. These effects are likely<br />

a combination <strong>of</strong> direct vGPCR signaling <strong>and</strong> paracrine effects via vGPCR-induced<br />

elaboration <strong>of</strong> various growth <strong>and</strong> angiogenic factors. Inhibition <strong>of</strong> vGPCR function is a<br />

promising anti-KSHV strategy.<br />

We focus on identifying host cell pathways that are disrupted by vGPCR <strong>and</strong> could be<br />

targeted to interfere with vGPCR function. The protein phosphatase, Shp2, <strong>and</strong> the focal<br />

adhesion-associated kinase (FAK) are signal integration proteins with adaptor <strong>and</strong><br />

enzymatic functions. Up-regulation <strong>of</strong> both is associated with various malignancies.<br />

Furthermore, both are the target <strong>of</strong> newly developed pharmacologic inhibitors (FAK<br />

inhibition in particular has shown promise in a mouse ovarian cancer model). Using<br />

shRNA knock-down <strong>and</strong> pharmacologic inhibition in HEK293, we find that vGPCR<br />

activation <strong>of</strong> MEK requires both FAK <strong>and</strong> Shp2. vGPCR-induced NFκB <strong>and</strong> AP-1 activation<br />

are also FAK-dependent, although only the former requires Shp2. These studies are<br />

being taken into primary endothelial cells in which the role <strong>of</strong> Shp2 <strong>and</strong> FAK in vGPCRmediated<br />

signaling <strong>and</strong> phenotypic changes will be assessed.<br />

Interfering with pathways that are upregulated by vGPCR is a promising anti-KSHV<br />

approach. We have identified two signaling integration proteins that are receiving<br />

attention in the medicinal chemistry field <strong>and</strong> that are required for important vGPCRmediated<br />

events.<br />

Presenting author Email: canno101@umn.edu<br />

91


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Session 11 abstracts 64-67:<br />

Gene Expression II<br />

92


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gene Expression II Abstract 64<br />

DEVELOPMENT OF A COMPREHENSIVE TRANSCRIPTIONAL NETWORK FOR KSHV<br />

Linda Persson, Scott Millman <strong>and</strong> Angus Wilson<br />

Department <strong>of</strong> Microbiology & NYU Cancer Institute, New York <strong>University</strong> School <strong>of</strong><br />

Medicine, New York, NY 10016, USA<br />

Abstract<br />

Viral transcription factor RTA (product <strong>of</strong> ORF50) plays an essential <strong>and</strong> far-reaching role<br />

in control <strong>of</strong> KSHV replication <strong>and</strong> pathogenesis. In latently infected cells, the silent<br />

ORF50 promoter responds to a variety <strong>of</strong> reactivation stimuli, leading to rapid<br />

accumulation <strong>of</strong> RTA protein <strong>and</strong> initiation <strong>of</strong> the lytic program. A principal function <strong>of</strong><br />

RTA is to direct the transcription <strong>of</strong> more than 80 genes required for the lytic replication<br />

<strong>and</strong> virion assembly. Several RTA targets encode positive <strong>and</strong> negative regulators which<br />

further mold the basic program into a cascade <strong>of</strong> sequential viral gene expression.<br />

Paradoxically, RTA may also contribute to the establishment <strong>of</strong> latency during de novo<br />

infection, acting through an inducible promoter (LTi) upstream <strong>of</strong> the K12-ORF73 latency<br />

gene cluster. To better underst<strong>and</strong> the intricate network <strong>of</strong> regulatory interactions that<br />

control lytic replication <strong>and</strong> determine how the program can be modified by<br />

environmental parameters, we embarked on a comprehensive analysis <strong>of</strong> the RTAdirected<br />

transcriptome. We began with a functional screen to identify RTA-responsive<br />

sequences irrespective <strong>of</strong> their position in the genome. A pilot study covering 37,300-bp<br />

between K8.1 <strong>and</strong> ORF66 revealed 8 separate RTA-responsive elements, corresponding<br />

to a mix <strong>of</strong> new <strong>and</strong> known lytic promoters. Currently, this unbiased mapping approach is<br />

being applied to the entire KSHV genome. Mutational analysis <strong>and</strong> bioinformatics<br />

methods are being used to dissect the individual response elements <strong>and</strong> identify points at<br />

which cellular signaling pathways intersect with RTA to fine tune the lytic program or<br />

express only a subset <strong>of</strong> viral genes.<br />

Presenting author Email: wilsoa02@med.nyu.edu<br />

93


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gene Expression I Abstract 65<br />

KSHV ORF57 AND RNA EXPORT FACTORS: ROLES OF UAP56, URH49, RBM15,<br />

AND OTT3 IN ORF57 EXPRESSION AND FUNCTION<br />

Merlyn Deng 1 , Vladimir Majerciak 1 , Barbara K. Felber 2 , <strong>and</strong> Zhi-Ming Zheng 1<br />

1HIV<br />

<strong>and</strong> AIDS Malignancy Branch, National Cancer Institute, NIH, Bethesda, MD. USA.<br />

2<br />

Human Retrovirus Pathogenesis Section, Vaccine Branch, National Cancer Institute,<br />

Frederick, MD, USA<br />

Abstract<br />

KSHV ORF57 promotes the expression <strong>of</strong> a subset <strong>of</strong> viral lytic genes at<br />

posttranscriptional level <strong>and</strong> is essential for virus production. To investigate how ORF57<br />

might function to promote RNA export <strong>of</strong> its targets, we examined the cellular export<br />

factors UAP56, URH49, RBM15 <strong>and</strong> OTT3 for their possible roles in ORF57-enhanced<br />

expression <strong>of</strong> KSHV ORF59. We found that reducing the expression <strong>of</strong> each <strong>of</strong> these<br />

proteins with gene-specific siRNAs significantly reduced the expression <strong>of</strong> ORF59 in<br />

cotransfection experiments. Surprisingly, we also found that all four proteins affected<br />

ORF57 expression, indicating that the reduced ORF59 expression was likely due to the<br />

reduction <strong>of</strong> ORF57. The effect became most severe when two genes were knocked down<br />

simultaneously. To investigate whether the downregulation <strong>of</strong> ORF57 expression was due<br />

to a reduction <strong>of</strong> ORF57 mRNA export, we fractionated the nuclear RNA from the<br />

cytoplasmic RNA <strong>of</strong> cells transfected with an ORF57 expression vector <strong>and</strong> UAP56<br />

<strong>and</strong>/URH49 siRNAs. A substantial reduction <strong>of</strong> cytoplasmic ORF57 RNA was observed<br />

along with substantial accumulation <strong>of</strong> nuclear ORF57 RNA. A greater reduction <strong>of</strong><br />

cytoplasmic ORF57 RNA was noticed in double-knockdown cells. Although overexpression<br />

<strong>of</strong> UAP56 had no effect on ORF57 or ORF59, the presence <strong>of</strong> RBM15 or OTT3 greatly<br />

increased expression <strong>of</strong> ORF59, but suppressed ORF57 expression. Collectively, our data<br />

suggest that ORF57 expression is controlled by the four RNA export factors analyzed.<br />

The role <strong>of</strong> ORF57 is likely to substitute cellular RBM15 <strong>and</strong> OTT3 in promotion <strong>of</strong> ORF59<br />

expression by sequestering them from RNA export machinery.<br />

Presenting author Email: zhengt@exchange.nih.gov<br />

94


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gene Expression II Abstract 66<br />

DYNAMICS OF K-RTA RECRUITMENT ON THE KSHV GENOME REVEAL NOVEL<br />

REGULATION BY NF-KB<br />

Thomas J. Ellison 1 , Chie Izumiya 1 , Paul A. Luciw , Hsing-Jien Kung 1 , Yoshihiro Izumiya 1<br />

1 nd 2<br />

UC Davis Cancer Center, 4645 2 Ave. Sacramento, California 95817, USA.<br />

Department <strong>of</strong> Pathology, UC Davis, 1 Sheilds Ave. Davis, Davis, California 95616, USA.<br />

Abstract<br />

KSHV is regulated epigenetically <strong>and</strong> transcriptionally, subject to cellular factors<br />

including NF-kB. K-Rta <strong>and</strong> K-bZIP are two key factors that control reactivation <strong>and</strong> lytic<br />

replication. In this work, we performed genome-wide chromatin immunoprecipitation<br />

anaylsis with a viral promoter-chip (ChIP-on-Vchip) containing all 83 putative KSHV<br />

promoter regions. The recruitment <strong>of</strong> K-Rta <strong>and</strong> K-bZIP were examined in BCBL-1, as<br />

well as association with acetylated histone 3 as a marker for chromatin state. K12 <strong>and</strong><br />

Ori-RNA promoters were major sites for recruitment <strong>of</strong> K-Rta, <strong>and</strong> a number <strong>of</strong> K-bZIP<br />

binding sites were also identified. To examine the dynamics <strong>of</strong> recruitment, ten viral<br />

promoters were selected for a time-course. K-Rta recruitment was most evident at<br />

intermediate-strength target promoters, whereas the primary sites <strong>of</strong> K-bZIP binding<br />

were its repression targets, to which it was co-recruited with K-Rta by 4-12 hours post<br />

induction. Because NF-kB has been implicated in K-Rta transactivation, it was also<br />

examined using the viral promoter library. Interestingly, the only two viral promoters<br />

not responsive to NF-kB mediated inhibition were the K-Rta major binding promoters.<br />

Overexpression <strong>of</strong> NF-kB strongly inhibited recruitment <strong>of</strong> K-Rta to the ORF57 <strong>and</strong> KbZIP<br />

promoters but not the K12 promoter during viral reactivation. These results were<br />

further tested by in vitro DNA binding assay using RBPjk, RelA, NF-kB1, <strong>and</strong> K-Rta. NFkB<br />

sequestered RBP-jk from the ORF57 promoter, <strong>and</strong> was found to form a complex with<br />

RBP-jk through their Rel homology domains. These studies set the stage for further<br />

analysis <strong>of</strong> regulation <strong>of</strong> KSHV reactivation.<br />

Presenting author Email: yizumiya@ucdavis.edu<br />

95


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gene Expression II Abstract 67<br />

TYPE I INTERFERONS SUPPRESS MURINE GAMMAHERPESVIRUS-68 LYTIC<br />

INFECTION AT THE VIRUS TRANSCRIPTOME LEVEL VIA DIRECT MODULATION<br />

OF RTA PROMOTER ACTIVITY<br />

L. Roaden 1 , B. Manso 1 , B. Lane 1 , E. Arico 2 <strong>and</strong> B. Ebrahimi 1<br />

1 Division <strong>of</strong> <strong>Medical</strong> Microbiology, School <strong>of</strong> Infection & Host Defence, <strong>University</strong> <strong>of</strong><br />

Liverpool, Duncan Building, Daulby Street, Liverpool L69 3GA, UK, <strong>and</strong> 2 Laboratory <strong>of</strong><br />

Virology, Istituto Superiore di Sanità, Viale Regina Elena n.299 0161 Rome, Italy<br />

Abstract<br />

Type I interferons (IFN) are potent suppressors <strong>of</strong> herpesvirus lytic infections, however,<br />

their mode <strong>of</strong> action during early stages post-infection <strong>and</strong> reactivation from latency is<br />

not entirely clear. This is partly because IFN modulate a number <strong>of</strong> cellular pathways<br />

which interfere with viral infections. In this study, we used the murine<br />

gammaherpesvirus (MHV-68) as a model to underst<strong>and</strong> how type I IFNs control<br />

productive phase <strong>of</strong> MHV-68 at different stages immediately post infection. IFN<br />

treatment prevented the formation <strong>of</strong> infectious virus particles. Although IFN treatment<br />

was capable <strong>of</strong> reducing virus entry, the virus was still capable to enter cells <strong>and</strong> uncoat<br />

<strong>and</strong> gain entry into the nucleus as evidenced by virus genomic DNA copy numbers <strong>and</strong><br />

electron microscopy. IFN-mediated suppression was also evident at the level <strong>of</strong> viral<br />

protein expression; viral lytic antigens were absent in IFN-treated cells. Using a virusspecific<br />

microarray platform, IFN-mediated suppression was most pr<strong>of</strong>ound at the level <strong>of</strong><br />

virus transcriptome including viral tRNAs <strong>and</strong> miRNAs. Bioinformatics analysis<br />

highlighted a number <strong>of</strong> key IFN-related transcription factor binding sites within the<br />

ORF50 promoter. Using transient transfection assays, we show that direct suppression <strong>of</strong><br />

ORF50 promoter by IFN is a major regulatory mechanism in suppression <strong>of</strong> MHV-68 lytic<br />

infection. Since the ORF50 protein product, Rta, is a key lytic cycle switch in MHV-68<br />

<strong>and</strong> KSHV, direct manipulation <strong>of</strong> its promoter would explain virus transcriptome shutdown<br />

by IFN. The implications <strong>of</strong> these findings in relation to other gammaherpesviruses<br />

are discussed.<br />

Presenting author Email: ebrahimi@liv.ac.uk<br />

96


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster session abstracts<br />

P1-P18<br />

97


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P1<br />

THE LOCALIZATION OF LANA TO THE NUCLEAR MATRIX FRACTION IS LINKED<br />

TO THE KSHV GENOME REPLICATION<br />

Eriko Ohsaki, Tohru Suzuki, Keiji Ueda<br />

Department <strong>of</strong> Infectious Diseases, <strong>University</strong> <strong>of</strong> Hamamatsu School <strong>of</strong> Medicine, 1-20-1<br />

H<strong>and</strong>ayama, Hamamatsu, Shizuoka 431-3192, Japan.<br />

Abstract<br />

Kaposi’s sarcoma-associated herpesvirus (KSHV) has mechanisms <strong>of</strong> the replication that<br />

synchronizes a cell cycle <strong>and</strong> <strong>of</strong> the genome segregation during the cell division. Latent<br />

replication <strong>of</strong> the KSHV genome requires the latency-associated nuclear antigen (LANA)<br />

<strong>and</strong> terminal repeat (TR) as a replication origin (Ori-P), <strong>and</strong> it is thought that prereplication<br />

complexes (pre-RCs) are recruited to the TR region in a LANA-dependent<br />

manner.<br />

In this report, cell fractionation experiments demonstrated that LANA was localized to the<br />

insoluble fraction <strong>and</strong> TR <strong>and</strong> pre-RCs were also accumulated in this fraction at G1 phase.<br />

We tried to identify the regions <strong>of</strong> LANA involved in the localization to the insoluble<br />

fraction. The N-terminally deleted LANA, which maintains domain for dimerization <strong>and</strong><br />

DNA binding, lost the ability to localize to the insoluble fraction. We investigated whether<br />

these LANA mutants could replicate the TR-containing plasmid using a transient<br />

replication assay. All N-terminally deleted LANA including a mutant keeping only<br />

dimerization <strong>and</strong> DNA binding domains dramatically decreased the efficiency <strong>of</strong><br />

replication.<br />

The insoluble fraction is DNase I- <strong>and</strong> high salt-resistant, <strong>and</strong> contains mainly nuclear<br />

matrix components. It has been reported that the DNA replication associated with the<br />

nuclear matrix. In recent study, a real-time imaging showed that replication foci are<br />

stably anchored in the nucleus. Taken together, it is possible that the specific<br />

localization <strong>of</strong> LANA is important process for the KSHV replication in latency <strong>and</strong> the<br />

KSHV genome replication occurs in the nuclear matrix although further study is needed<br />

to determine what kinds <strong>of</strong> nuclear matrix proteins LANA specifically interacts with.<br />

Presenting author Email: eohsaki@hama-med.ac.jp<br />

98


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P2<br />

MODULATION OF INTERFERON β GENE EXPRESSION BY KAPOSI’S SARCOMA<br />

ASSOCIATED HERPESVIRUS (KSHV) LATENCY PROTEINS<br />

Nathalie Cloutier <strong>and</strong> Louis Flam<strong>and</strong>.<br />

Laboratory <strong>of</strong> Virology, Rheumatology <strong>and</strong> Immunology Research Center, CHUQ Research<br />

Center <strong>and</strong> Faculty <strong>of</strong> Medicine, Laval <strong>University</strong>, Quebec, Canada, G1V 4G2.<br />

Abstract<br />

The interferon (IFN) system represents a potent antiviral defense mechanism. KSHV is an<br />

oncogenic virus associated with KS, Primary Effusion Lymphoma <strong>and</strong> Multicentric<br />

Castleman’s Disease. Latency proteins are important for immune evasion by KSHV from<br />

the host <strong>and</strong> ensuring viral persistence. Our objective is to determine the impact <strong>of</strong> v-<br />

FLIP, v-Cyclin <strong>and</strong> LANA expression on interferon-b synthesis.<br />

Considering that the ifn-b gene is regulated partly through NF-kB, we sought to<br />

determine whether v-FLIP could activate the ifn-b gene. By itself, v-FLIP protein has no<br />

effect on ifn-b gene activation but when combined with IFN-b inducers, a synergistic<br />

activation occurs. This effect is strictly dependent on NF-kB <strong>and</strong> is mediated through the<br />

positive regulatory domain II <strong>of</strong> the IFN-b promoter. v-Cyclin has no impact on ifn-b<br />

gene activation. Our preliminary results show that LANA inhibits ifn-b activation.<br />

v-FLIP activates NF-κB <strong>and</strong> is essential for the survival <strong>of</strong> infected cells. During the lytic<br />

cycle, dsRNA <strong>and</strong> dsDNA molecules produced by KSHV replication will activate the<br />

interferon pathway <strong>and</strong> v-FLIP is likely to potentiate this effect. The synergy between v-<br />

FLIP <strong>and</strong> the IFN-b inducers will favor IFN-b production by lytically infected cells. On the<br />

other h<strong>and</strong>, LANA expression dampens this effect by suppressing ifn-b expression.<br />

Studies with infected cells will be important to underst<strong>and</strong> the interplay between latency<br />

proteins <strong>and</strong> IFN-b production during the replication <strong>of</strong> KSHV. A balance between<br />

activation <strong>and</strong> inhibition <strong>of</strong> the ifn-b gene must occur for infected cells to persist <strong>and</strong><br />

avoid elimination by the host immune system defense mechanisms.<br />

Presenting author Email: Nathalie.Cloutier@crchul.ulaval.ca<br />

99


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P3<br />

SEVERE CHANGES OF PHENOTYPE AND CYTOKINE EXPRESSION IN KSHV-<br />

INFECTED B-LYMPHOCYTES<br />

Guergana Iotzova 1 , Georg Malterer 1 , Kai Bratke 2 , Werner Luttmann 2 , Antoine Gessain 3 ,<br />

Christine S. Falk 4 , Kevin Robertson 5 , Peter Ghazal 5 <strong>and</strong> Jürgen Haas 1,5<br />

1 Max-von-Pettenk<strong>of</strong>er Institute, Ludwig-Maximilians-Universität München; 2 Department<br />

<strong>of</strong> Pneumology, <strong>University</strong> Hospital, Rostock; 3 Unit <strong>of</strong> Epidemiology <strong>and</strong> Physiopathology<br />

<strong>of</strong> Oncogenic Viruses, Institut Pasteur, Paris; 4 Institute for Molecular Immunology,<br />

Helmholtz Zentrum München; 5 Division <strong>of</strong> Pathway Medicine, <strong>University</strong> <strong>of</strong> Edinburgh,<br />

Edinburgh<br />

Abstract<br />

KSHV is involved in the pathogenesis <strong>of</strong> Kaposi´s sarcoma (KS) <strong>and</strong> B-cell derived<br />

primary effusion lymphomas (PEL). The aim <strong>of</strong> this study was to systematically<br />

investigate the influence <strong>of</strong> KSHV infection on the expression <strong>of</strong> cellular genes in B<br />

lymphocytes. To study these changes, a microarray analysis was performed with<br />

persistently in vitro KSHV-infected B-cells from several donors. A considerable number <strong>of</strong><br />

genes (408) were found to be modulated more than 4-fold by KSHV infection: 67.4% <strong>of</strong><br />

these genes were downregulated <strong>and</strong> 32.6% upregulated. Intriguingly, many<br />

downregulated genes encoded for B-cell surface markers <strong>and</strong> B-cell specific transcription<br />

factors (PAX-5, Oct-2 <strong>and</strong> Spi-B), which was confirmed on the protein level. The massive<br />

loss <strong>of</strong> B-cell surface markers or “null” phenotype was similar to PEL tumor cells<br />

suggesting that the loss <strong>of</strong> B-cell identity is caused by KSHV rather than cellular factors.<br />

KSHV-infected cells could not be lysed by allo-reactive cytotoxic T-cells, indicating that<br />

the null phenotype is associated with immune evasion. The downregulation <strong>of</strong> the Tolllike<br />

receptors TLR7, TLR9 <strong>and</strong> TLR10, which was confirmed by real-time PCR, might<br />

contribute to the immune escape <strong>of</strong> KSHV-infected B-cells. On the other h<strong>and</strong>, we<br />

detected a marked upregulation <strong>of</strong> Granzyme A (GzmA) transcripts in KSHV-infected<br />

cells, as well as exceedingly high levels <strong>of</strong> secreted GzmA in supernatants <strong>of</strong> KSHVinfected<br />

B-cells <strong>and</strong> in effusion fluids <strong>of</strong> PEL patients. shRNA knockdown studies revealed<br />

that GzmA participitates in a parakrine cytokine by inducing cellular IL-10, indicating a<br />

potential role <strong>of</strong> GzmA in the pathogenesis <strong>of</strong> KSHV-related PEL.<br />

Presenting author Email: malterer@mvp.uni-muenchen.de<br />

100


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P4<br />

STUDIES ON THE ROLE OF MICROENVIRONMENT IN THE DEVELOPMENT OF<br />

PRIMARY EFFUSION LYMPHOMA<br />

Calabrò ML, 1 Gasperini P, 2 Di Gangi IM, 1 Indraccolo S, 1 Amadori A, 1 Chieco-Bianchi L. 2<br />

1 Immunology <strong>and</strong> Diagnostic Molecular Oncology, Istituto Oncologico Veneto, IRCCS,<br />

Padova; 2 Department <strong>of</strong> Oncology <strong>and</strong> Surgical <strong>Sciences</strong>, Oncology Section, <strong>University</strong> <strong>of</strong><br />

Padova, Italy.<br />

Abstract<br />

We used a SCID mouse model <strong>of</strong> primary effusion lymphoma (PEL) that mimics the<br />

aggressive course <strong>of</strong> this human KSHV-induced lymphoma to dissect the contribution <strong>of</strong><br />

the host microenvironment in PEL development. The activity <strong>of</strong> a murine, i.e. hostspecific,<br />

interferon (IFN)-α1-expressing lentiviral vector (mIFN-α1-LV) was compared to<br />

that <strong>of</strong> a hIFN-α2b-LV on the in vitro <strong>and</strong> in vivo growth <strong>of</strong> PEL cells. A control EGFP-LV<br />

was used in parallel. Lentiviral vectors efficiently delivered the transgene to PEL cells <strong>and</strong><br />

conferred long-term transgene expression both in vitro <strong>and</strong> in vivo. Treatment <strong>of</strong> PELinjected<br />

SCID mice with hIFN-α2b-LV was found to significantly prolong the survival in<br />

the majority <strong>of</strong> animals compared to control treatments, <strong>and</strong> was associated with a<br />

remarkable reduction in ascites formation. Interestingly, the delivery <strong>of</strong> a host-specific<br />

cytokine showed an anti-neoplastic activity comparable to that observed with the hIFNα2b-LV,<br />

thus suggesting that the specific targeting <strong>of</strong> the host microenvironment may<br />

impair PEL cell growth in vivo.<br />

Presenting author Email: lcalabro@unipd.it<br />

101


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P5<br />

ENDOTHELIAL PROGENITORS FROM THE PERIPHERAL BLOOD OF PATIENTS<br />

WITH CLASSIC KAPOSI’S SARCOMA ARE PERSISTENTLY INFECTED BY KSHV<br />

Taddeo A 1 , Della Bella S, 1 Colombo E, 1 Brambilla L, 2 Bergamo E, 3 Calabrò ML. 3<br />

1Laboratory <strong>of</strong> Immunology, Dipartimento di Scienze e Tecnologie Biomediche, Università<br />

degli Studi di Milano; 2 Departiment di Dermatology, IRCCS Ospedale Maggiore,<br />

Milano; 3 Immunology <strong>and</strong> Diagnostic Molecular Oncology, Istituto Oncologico Veneto,<br />

IRCCS, Padova, Italy.<br />

Abstract<br />

Accumulating evidence indicates that tumor angiogenesis is supported by the<br />

mobilization <strong>and</strong> incorporation <strong>of</strong> endothelial progenitor cells (EPCs), highly proliferative<br />

precursors <strong>of</strong> bone marrow origin. Our recent demonstration that EPCs are increased in<br />

the peripheral blood <strong>of</strong> patients with Kaposi’s sarcoma (KS), together with the intrinsic<br />

biologic properties <strong>of</strong> these cells, strongly suggests that EPCs could be involved in the<br />

pathogenesis <strong>of</strong> KS. The fact that the characteristic spindle cells share many markers<br />

with vascular endothelial cells <strong>and</strong> are thought to be <strong>of</strong> endothelial origin further supports<br />

this hypothesis. A possible scenario may be that EPCs may act as preferential KSHV<br />

reservoirs <strong>and</strong>, whether infected, may home to permissive sites <strong>and</strong> propagate to<br />

produce KS lesions. Novel insights into the state <strong>of</strong> KSHV infection <strong>of</strong> EPCs could greatly<br />

improve the comprehension <strong>of</strong> KS pathogenesis. Therefore, we investigated KSHV<br />

infection <strong>of</strong> ex-vivo cultured late-EPCs that, among other cell populations with endothelial<br />

features, contribute more directly to neovascularization <strong>and</strong> might represent a major<br />

source <strong>of</strong> endothelial progenitors in vivo. We found that late-EPCs from KS patients<br />

harbor KSHV DNA <strong>and</strong> retain the virus after multiple passages. Lytic phase induction or<br />

hypoxia could amplify the virus in cells <strong>and</strong> supernatants, indicating that late-EPCs<br />

support KSHV productive replication. EPCs appear therefore to represent potential virus<br />

reservoirs <strong>and</strong> putative precursors <strong>of</strong> KS spindle cells. The biological mechanisms that<br />

govern the infection <strong>of</strong> EPC by KSHV are currently under investigation.<br />

Presenting author Email: lcalabro@unipd.it<br />

102


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P6<br />

KSHV infection <strong>of</strong> human placental histocultures<br />

Di Stefano M, 1 Fiore JR 2 , Di Gangi IM, 3 Chieco-Bianchi L, 4 Greco P, 5 Gesualdo L, 1 Menu E, 6<br />

Calabrò ML. 3<br />

1 Laboratory <strong>of</strong> Molecular Medicine, <strong>University</strong> <strong>of</strong> Foggia, Foggia, Italy; 2 Department <strong>of</strong><br />

Clinical <strong>and</strong> Occupational Health, <strong>University</strong> <strong>of</strong> Foggia, Foggia, Italy; 3 Immunology <strong>and</strong><br />

Diagnostic Molecular Oncology, Istituto Oncologico Veneto, IRCCS, Padova, Italy;<br />

4 Department <strong>of</strong> Oncology <strong>and</strong> Surgical <strong>Sciences</strong>, Oncology Section, <strong>University</strong> <strong>of</strong> Padova,<br />

Padova, Italy; 5 Department <strong>of</strong> Surgical <strong>Sciences</strong>, <strong>University</strong> <strong>of</strong> Foggia, Foggia, Italy;<br />

6 Unité de Régulation des Infections Rétrovirales, Institut Pasteur, Paris, France.<br />

Abstract<br />

A placenta histoculture system was used to analyse the susceptibility <strong>of</strong> placental cells to<br />

in vitro KSHV infection. KSHV quantitative detection was performed by real-time PCR in<br />

cultured villi <strong>and</strong> supernatants, <strong>and</strong> virus expression was determined by<br />

immunohistochemistry for latent <strong>and</strong> lytic KSHV antigens. Increasing amounts <strong>of</strong> KSHV<br />

DNA were detected in placental tissues <strong>and</strong> culture supernatants. Immunohistochemistry<br />

analyses showed that both cyto- <strong>and</strong> syncitiotrophoblasts, as well as endothelial cells,<br />

expressed latent <strong>and</strong> lytic antigens. Moreover, relevant apoptotic phenomena were<br />

observed in infected histocultures.<br />

These data indicate that placental cells may be productively infected in vitro by KSHV,<br />

<strong>and</strong> suggest that this phenomenon might influence vertical transmission <strong>and</strong> pregnancy<br />

outcome in KSHV-infected women.<br />

Presenting author Email: lcalabro@unipd.it<br />

103


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P7<br />

SUMO-MODIFICATION MODULATES THE SUBCELLULAR LOCALIZATION AND<br />

ACTIVITY OF THE KSHV LATENT PROTEIN LANA2<br />

1 2 3 3 2<br />

Laura Marcos-Villar, Pedro Gallego, Fern<strong>and</strong>o Lopitz, Manuel S Rodriguez & Carmen<br />

Rivas<br />

1Dpt Microbiologia II, Fac Farmacia, Universidad Complutense de Madrid, Madrid, Spain;<br />

2 Dpt Biología Molecular y Celular, Centro Nacional de Biotecnología, CSIC, Madrid, Spain;<br />

3 Ubiquitin-like proteins & Cancer Group, Proteomics Unit. CIC-BioGUNE, Derio, Spain.<br />

Abstract<br />

Small ubiquitin-related modifier (SUMO) family proteins function by becoming covalently<br />

attached to other proteins as post-translational modifications. SUMO modifies many<br />

proteins that participate in diverse processes, including transcriptional regulation, nuclear<br />

transport, maintenance <strong>of</strong> genome integrity, <strong>and</strong> signal transduction. The functional<br />

consequences <strong>of</strong> SUMO attachment vary greatly from substrate to substrate; however,<br />

two general modes <strong>of</strong> action have been suggested. Sumoylation may alter protein<br />

stability or may alter protein-protein interactions that could result in a variety <strong>of</strong><br />

consequences, including changes in cellular localization.<br />

LANA2 is a multifunctional protein exclusively expressed in KSHV infected B cells that is<br />

required for the survival <strong>of</strong> KSHV-infected primary effusion lymphoma cells. Studies<br />

carried out in our laboratory demonstrate that LANA2 is covalently modified by SUMO.<br />

Sumoylation <strong>of</strong> LANA2 does not affect protein stability but modulates its subcellular<br />

localization <strong>and</strong> protein activity.<br />

Presenting author Email: lmarcos@cnb.csic.es<br />

104


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P8<br />

HHV8-ASSOCIATED K5/MIR2 EXPRESSION IN VIVO CORRELATES WITH<br />

REDUCED COEXPRESSION OF CD31, MHC-1, AND ®-CATENIN IN KAPOSI’S<br />

SARCOMA<br />

Liron Pantanowitz1, M<strong>and</strong>ana Mansouri2, Sharon Marconi1, Eric Bartee2, Ashlee V.<br />

Moses2, Klaus Früh2<br />

1 Department <strong>of</strong> Pathology, Baystate <strong>Medical</strong> Center, Tufts <strong>University</strong> School <strong>of</strong> Medicine,<br />

Springfield, MA; 2 Vaccine <strong>and</strong> Gene Therapy Institute, Oregon Health & Science<br />

<strong>University</strong>, 505 NW185th Ave, Beaverton, OR, 97006, USA<br />

Abstract<br />

K5/MIR2 eliminates MHC-I, CD31 <strong>and</strong> the VE-Cadherin/β-catenin complex from KSHVinfected<br />

endothelial cells in vitro, thereby reducing their immune recognition, cell<br />

adhesion <strong>and</strong> migration. Expression <strong>of</strong> K5 in vivo <strong>and</strong> downregulation <strong>of</strong> K5-target<br />

proteins in Kaposi’s sarcoma (KS) has not been studied. Therefore, we investigated K5 in<br />

patient procured KS lesions, <strong>and</strong> sought to determine whether this expression correlated<br />

with loss <strong>of</strong> CD31, MHC-1, <strong>and</strong> β-Catenin. Immunohistochemistry was performed on<br />

biopsied KS lesions <strong>of</strong> various stages (patch 2, plaque 10, tumor 8) using novel<br />

antibodies to K5, as well as antibodies to CD31, CD34, MHC-I, β-Catenin, LNA-1, <strong>and</strong> D2-<br />

40. All cases were immunoreactive with CD34, D2-40 <strong>and</strong> LNA-1. K5-specific staining<br />

was observed in 100% <strong>of</strong> tumor stage lesions <strong>and</strong> 40% <strong>of</strong> KS plaques. While only a few<br />

spindle cells were K5 positive (3+) in plaque lesions, up to 90% <strong>of</strong> lesional cells were K5<br />

positive in KS tumors. In only 10% <strong>of</strong> cases with K5 staining was concomitant expression<br />

<strong>of</strong> K5-targeted molecules (CD31, MHC-1, <strong>and</strong> β-Catenin) noted, whereas 50% <strong>of</strong> K5negative<br />

cases stained for K5-targeted molecules. These data suggest that K5 expression<br />

in vivo is more wide-spread than currently assumed, particularly in advanced KS lesions.<br />

Moreover, K5 expression in KS tumors appears to be associated with an absence <strong>of</strong> the<br />

K5-targeted molecules CD31, MHC-1, <strong>and</strong> β-Catenin in KSHV-infected lymphatic<br />

endothelial lesional cells. This striking inverse correlation between K5 <strong>and</strong> CD31/MHC-<br />

1/β-Catenin expression supports a crucial role for K5 in modulating tumor cell function in<br />

proliferating KS lesions.<br />

Presenting author Email: fruehk@ohsu.edu<br />

105


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P9<br />

KBZIP SERVES AS A GLOBAL TRANSCRIPTIONAL REPRESSOR VIA<br />

SUMOYLATION AND DIRECT INHIBITION OF JMJD2A, A HISTONE DEMETHYLASE<br />

Latricia, Fitzgerald, D 1 , Yoshihiro, Izumiya 1 ; Cliff, Tepper, G. 1 ; Datsun, Hsia 1 ; Chang,<br />

Pei-Ching 1 ; Ellison, Tom 1 ; Luciw, Paul A. 2 ; Hsing-Jien, Kung 1<br />

UC Davis Cancer Center, Sacramento, CA, United States 1 ; <strong>University</strong> <strong>of</strong> California at<br />

Davis, Davis, California, United States 2<br />

Abstract<br />

Sumoylation <strong>and</strong> histone H3K9 methylation are hallmarks <strong>of</strong> heterochromatin <strong>and</strong><br />

silenced genes. Polycomb complex, a global gene silencer, contains both SUMO ligase<br />

<strong>and</strong> histone methylase. KbZIP is an early gene expressed in the KSHV genome upon lytic<br />

activation. It is a strong transcriptional repressor <strong>and</strong> a moderate transcriptional<br />

activator. In the absence <strong>of</strong> other viral proteins, K-bZIP is a general repressor <strong>of</strong> host<br />

genes <strong>and</strong> an effective inducer <strong>of</strong> H3K9 trimethylation. K-bZIP is sumoylated at lysine<br />

158, when this site is mutated K-bZIP no longer serves as a global repressor. K-bZIP<br />

also represses transcription through its interaction with transcriptional machinery, such<br />

as JMJD2a, a histone demethylase. We were able to demonstrate that K-bZIP interacts<br />

with JMJD2a <strong>and</strong> this interaction leads to inhibition <strong>of</strong> JMJD2A activity. As K-bZIP is a<br />

SUMO-binding protein <strong>and</strong> behaves like a SUMO-ligase/adaptor, we asked whether<br />

JMJD2A is sumoylated. The results showed that JMJD2A is sumoylated <strong>and</strong> colocalized<br />

with SUMO1 foci. In addition, JMJD2A is transciptionally downmodulated by K-bZIP. Thus,<br />

K-bZIP utilizes multiple ways to shut <strong>of</strong>f JMJD2A which was observed that overexpression<br />

<strong>of</strong> JMJD2A in BCBL-1 increases the level <strong>of</strong> reactivation <strong>of</strong> latent genome. consistent with<br />

the notion that chromosomal remodeling plays a pivotal part <strong>of</strong> latency switch. These<br />

data implicates K-bZIP in the establishment <strong>of</strong> latency by K-Rta inhibition <strong>and</strong> modulating<br />

the chromatin structure <strong>of</strong> the infecting genome.<br />

Presenting author Email: lfitzgerald@ucdavis.edu<br />

106


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P10<br />

ANALYSIS METHODS FOR THE RELIABLE PREDICTION OF VIRUS AND HOST<br />

MIRNA EXPRESSION FROM THE AGILENT MIRNA MICROARRAY<br />

Eve Coulter, Dan Frampton, Paul Kellam<br />

MRC Centre for <strong>Medical</strong> Molecular Virology, Department <strong>of</strong> Infection, UCL, 46 Clevel<strong>and</strong><br />

Street, London, W1T 4JF<br />

Abstract<br />

MicroRNAs play an important role in the down-regulation <strong>of</strong> gene expression by blocking<br />

translation <strong>of</strong> their mRNA targets. Hundreds <strong>of</strong> miRNAs have been identified within the<br />

human genome, many <strong>of</strong> which are predicted to be involved in the regulation <strong>of</strong> major<br />

cellular processes such as cell development <strong>and</strong> apoptosis. It is thought their abnormal<br />

expression may contribute to cancer development <strong>and</strong> proliferation. In addition, miRNAs<br />

have been shown to be present in every herpesvirus examined to date suggesting they<br />

play a similar role in viral gene regulation.<br />

The development <strong>of</strong> suitable miRNA array technology allows us to detect the presence or<br />

absence <strong>of</strong> both human <strong>and</strong> viral miRNAs <strong>and</strong> to use miRNA pr<strong>of</strong>iles to differentiate<br />

between virus-infected <strong>and</strong> non-infected cell types. However, there is some uncertainty<br />

over the design <strong>of</strong> reliable probes for viral miRNAs, with several different probes being<br />

used to measure the level expression <strong>of</strong> a given miRNA. We have used the Agilent miR<br />

array platform <strong>and</strong> a panel <strong>of</strong> herpesvirus positive B-cell lymphomas to develop an<br />

accurate data analysis method which discards unreliable probes <strong>and</strong> show it is better able<br />

to discriminate between KSHV- <strong>and</strong> EBV-infected B-cell tumour cell lines than the<br />

st<strong>and</strong>ard Agilent method. Statistical validation was performed using ROC analysis <strong>of</strong> a<br />

large dataset <strong>of</strong> B cell tumour cell line miRNA arrays <strong>and</strong> predicted differentially<br />

expressed miRNAs confirmed by real time PCR.<br />

Presenting author Email: d.frampton@ucl.ac.uk<br />

107


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P11<br />

KAPOSI’S SARCOMA-ASSOCIATED HERPESVIRUS (KSHV) INFECTED PRIMARY<br />

EFFUSION LYMPHOMA CELLS SHOWS A DISTINCT GENE EXPRESSION PROFILE<br />

FROM BURKITT LYMPHOMA CELLS EITHER WITH OR WITHOUT EPSTEIN-BARR<br />

VIRUS AND THE OTHER T CELL LYMPHOBLASTIC LEUKEMIA CELLS<br />

Keiji Ueda, Eriko Ohsaki, Tohru Suzuki <strong>and</strong> Masato Karayama<br />

Dept. <strong>of</strong> Infectious Disease, Hamamatsu <strong>University</strong> School <strong>of</strong> Medicine,<br />

1-20-1 H<strong>and</strong>ayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan<br />

Abstract<br />

Analysis on a gene expression pr<strong>of</strong>ile using various kinds <strong>of</strong> normal or abnormal tissues,<br />

including tumors, <strong>and</strong> cell lines etc., is very informative to know the nature <strong>of</strong> the cells,<br />

which leads to further analyses <strong>and</strong> exploring new methods to treat diseased cells.<br />

Though many reports have already published on gene expression pr<strong>of</strong>iles <strong>of</strong> KSHV<br />

infected tissues, either in vitro or in vivo, we will report here gene expression pr<strong>of</strong>iles <strong>of</strong><br />

cultured cell lines; KSHV infected primary effusion lymphoma cell lines such as BC1, BC3,<br />

BCBL1 <strong>and</strong> TY1, Burkitt cell lines such as BJAB, Ramos, which are not infected with<br />

Epstein-Barr virus (EBV) <strong>and</strong> Daudi, Raji, Akaka, which are infected with EBV, <strong>and</strong> T<br />

lymphoblastic cell lines as Sup T1, Molt 3 <strong>and</strong> Jurkat, <strong>and</strong> HIV-1 infected MT4.<br />

What we found is that each categorized group <strong>of</strong> cell lines showed distinct gene<br />

expression pr<strong>of</strong>iles <strong>and</strong> Burkitt cell lines infected with or without EBV showed a similar<br />

pr<strong>of</strong>ile. Since all PEL cell lines are infected with KSHV <strong>and</strong> BC1 is infected with both EBV<br />

<strong>and</strong> KSHV, we can not conclude that KSHV infection governs the gene expression pr<strong>of</strong>ile<br />

but it is likely that EBV status does not affect the pr<strong>of</strong>ile.<br />

We are further studying how a typical gene is activated in the PEL cell lines <strong>and</strong> will<br />

discuss about it.<br />

Presenting author Email: kueda@hama-med.ac.jp<br />

108


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P12<br />

THE M TYPE K15 PROTEIN OF KAPOSI'S SARCOMA-ASSOCIATED HERPESVIRUS<br />

INDUCES CELL MIGRATION, INVASION AND REGULATES MICRORNA<br />

EXPRESSION VIA ITS SH2-BINDING MOTIF<br />

Yu-Hsuan Wu 1 , Min-Fen Wu 1 , Yuan-Hau Tsai 1 , Su-Fang Lin 3 , Tyson V. Sharp 4 <strong>and</strong> Hsei-<br />

Wei Wang 1,2,5<br />

1 Institute <strong>of</strong> Microbiology <strong>and</strong> Immunology, 2 Institute <strong>of</strong> Clinical Medicine, National Yang-<br />

Ming <strong>University</strong>; 3 Division <strong>of</strong> Clinical Research, National Health Research Institute,<br />

Taiwan; 4 School <strong>of</strong> Biomedical <strong>Sciences</strong>, <strong>University</strong> <strong>of</strong> Nottingham <strong>Medical</strong> School,<br />

Queen’s <strong>Medical</strong> Centre, Nottingham, UK; 5 Department <strong>of</strong> Teaching <strong>and</strong> Research,Taipei<br />

City Hospital, Taipei, Taiwan<br />

Abstract<br />

Kaposi’s sarcoma (KS) associated herpesvirus (KSHV) is the etiological agent <strong>of</strong> KS. In<br />

vivo KS is a tumor capable <strong>of</strong> spreading throughout the body, <strong>and</strong> pulmonary metastasis<br />

is observed clinically. In vitro KSHV induces the invasiveness <strong>of</strong> endothelial cells.<br />

However, no viral gene has yet been implicated in cell invasion. The KSHV ORF K15 is a<br />

KSHV-specific gene encoding a transmembrane protein. Two highly divergent forms <strong>of</strong><br />

K15, the predominant (P) <strong>and</strong> minor (M) forms (K15P <strong>and</strong> K15M, respectively) have been<br />

identified in different KSHV strains. K15 resembles the LMP2A gene <strong>of</strong> Epstein-Barr virus<br />

(EBV) in their genomic locations <strong>and</strong> protein topology. Also, both K15 proteins have<br />

similar motifs to those found in EBV LMP1 protein. K15 therefore appears to be a hybrid<br />

<strong>of</strong> a distant evolutionary relative <strong>of</strong> both EBV LMP1 <strong>and</strong> 2A. Since both LMP1 <strong>and</strong> LMP2A<br />

proteins are capable <strong>of</strong> inducing cell motility <strong>and</strong> have been linked to NPC metastasis, we<br />

questioned whether K15 also possesses similar abilities. In this study, we show by the<br />

use <strong>of</strong> a K15M-specific mAb <strong>and</strong> PCR, that K15M is latently expressed in KSHV positive<br />

PEL cells. K15M localizes on lysosomal membrane <strong>and</strong> is capable <strong>of</strong> activating the NF-kB<br />

transcription factor via its SH2-binding motif. K15M induces cell migration, invasion <strong>and</strong><br />

the expression <strong>of</strong> microRNAs miR-21 <strong>and</strong> miR-31 via this conserved motif. K15M<br />

therefore may contribute to KSHV-mediated tumor metastasis <strong>and</strong> angiogenesis.<br />

Targeting <strong>of</strong> K15 proteins or their downstream micorRNAs for therapy may represent a<br />

novel avenue <strong>of</strong> treatment for KSHV-associated neoplasia.<br />

Presenting author Email: b881625@life.nthu.edu.tw<br />

109


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P13<br />

IDENTIFICATION OF NOVEL CONSERVED GAMMAHERPESVIRUS MICRORNAS<br />

Nicole Walz , Thomas Christalla <strong>and</strong> Adam Grundh<strong>of</strong>f<br />

Heinrich-Pette Institute for Experimental Virology <strong>and</strong> Immunology, D-20251 Hamburg,<br />

Germany<br />

Abstract<br />

microRNAs (miRNAs) are small (~22 nt.), non-coding RNA molecules which posttranscriptionally<br />

regulate mRNA expression <strong>and</strong> play important roles in the regulation <strong>of</strong><br />

diverse cellular processes. The current release <strong>of</strong> the miRNA database (miRBase) lists a<br />

total <strong>of</strong> 132 known viral miRNAs, the vast majority <strong>of</strong> which are encoded by members <strong>of</strong><br />

the herpesvirus family. While the propensity to encode miRNAs appears to be a<br />

conserved feature <strong>of</strong> many herpesviruses, the miRNAs themselves show little<br />

conservation: with the exception <strong>of</strong> 7 miRNAs known to be shared between Epstein-Barr<br />

Virus (EBV) <strong>and</strong> its close relative Rhesus Lymphocryptovirus (rLCV), all other herpesvirus<br />

miRNAs appear unrelated in sequence. We have recently developped VMir, a<br />

computational ab initio prediction method to identify viral miRNAs, <strong>and</strong> have employed<br />

the program to identify miRNAs in SV40, KSHV <strong>and</strong> EBV. We have modified the algorithm<br />

to allow consideration <strong>of</strong> evolutionary conservation <strong>and</strong> conducted an extended analysis<br />

<strong>of</strong> the herpesvirus family. We were able to identify <strong>and</strong> experimentally verify 2 novel<br />

EBV- <strong>and</strong> 17 novel rLCV-encoded miRNAs; with a total <strong>of</strong> 33 miRNAs rLCV is now the<br />

virus with the highest number <strong>of</strong> known miRNAs. In contrast, we have found little<br />

evidence for the existence <strong>of</strong> hitherto unknown conserved miRNAs shared by other<br />

herpesviruses, including KSHV <strong>and</strong> the closely related Rhesus Rhadinovirus (RRV). While<br />

EBV <strong>and</strong> rLCV thus share even more miRNAs than previously thought, our results<br />

underline the notion that, in contrast to their cellular counterparts, viral miRNAs show an<br />

almost total lack <strong>of</strong> sequence conservation.<br />

Presenting author Email: Adam.Grundh<strong>of</strong>f@hpi.uni-hamburg.de<br />

110


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P14<br />

LYTIC INDUCTION OF KAPOSI'S SARCOMA-ASSOCIATED HERPESVIRUS IN<br />

PRIMARY EFFUSION LYMPHOMA CELLS WITH NON-TOXIC NATURAL PRODUCTS<br />

Hye-Jeong Cho 1 , Fuqu Yu 2 , Ren Sun 2 , Dongho Lee 1 <strong>and</strong> Moon Jung Song 1<br />

1 Division <strong>of</strong> Biotechnology, <strong>College</strong> <strong>of</strong> Life <strong>Sciences</strong> <strong>and</strong> Biotechnology, Korea <strong>University</strong>,<br />

Seoul 136-713, Republic <strong>of</strong> Korea, <strong>and</strong> 2 Department <strong>of</strong> Molecular <strong>and</strong> <strong>Medical</strong><br />

Pharmacology, <strong>University</strong> <strong>of</strong> California at Los Angeles, Los Angeles, CA90095, U.S.A.<br />

Abstract<br />

Kaposi’s sarcoma-associated herpesvirus (KSHV) has been linked to Kaposi’s sarcoma,<br />

primary effusion lymphoma (PEL), <strong>and</strong> multicentric Castleman’s disease. Intentional lytic<br />

induction <strong>of</strong> gammaherpesviruses in the presence <strong>of</strong> antiviral drugs is thought to be an<br />

effective treatment option for gammaherpesvirus-related tumors. In this study, we used<br />

a cell-based fluorescence bioassay system in which a KSHV-infected PEL cell line was<br />

stably transfected with a potent viral promoter-driven reporter gene to identify effective<br />

non-toxic reagents capable <strong>of</strong> inducing latent KSHV. Among 400 plant extracts screened,<br />

three extracts increased reporter gene expression in a dose-dependent manner. The<br />

three extracts activated the RTA promoter <strong>and</strong> induced expressions <strong>of</strong> lytic genes in the<br />

endogenous viral genomes <strong>of</strong> KSHV-infected tumor cells. Furthermore, these extracts<br />

were also capable <strong>of</strong> inducing lytic replication <strong>of</strong> Epstein-Barr virus in B95.8 cells,<br />

suggesting a conserved reactivation mechanism(s) among gammaherpesviruses.<br />

Together, our results demonstrate the effectiveness <strong>of</strong> the screening system to identify<br />

natural products capable <strong>of</strong> inducing KSHV reactivation, thereby facilitating the<br />

development <strong>of</strong> novel therapeutic agents for KSHV-associated malignancies.<br />

Presenting author Email: chohyejeong@korea.ac.kr<br />

111


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P15<br />

KSHV DNA QUANTIFICATION IN MATCHED PLASMA AND PBMCs SAMPLES OF<br />

HIV+ PATIENTS WITH KSHV-RELATED LYMPHOPROLIFERATIVE DISEASES<br />

Rosamaria Tedeschi 1 , Alessia Marus 1 , Ettore Bidoli 2 , Cecilia Simonelli 3 , <strong>and</strong> Paolo De<br />

Paoli 1<br />

1 Microbiology-Immunology <strong>and</strong> Virology Unit; 2 Epidemiology Unit; 3 <strong>Medical</strong> Oncology<br />

Dept.; Centro di Riferimento Oncologico, IRCCS, Aviano, Italy<br />

Abstract<br />

Background: The quantitative evaluation <strong>of</strong> Kaposi’s sarcoma Herpesvirus (KSHV) viral<br />

load is not well described in the clinical management <strong>of</strong> KSHV-related lymphoproliferative<br />

diseases.<br />

Aim: To evaluate <strong>and</strong> to compare KSHV viral load in different blood compartments from<br />

HIV+ patients with Multicentric Castleman’s disease (MCD), Primary Effusion Lymphoma<br />

(PEL) <strong>and</strong> virus-associated solid lymphoma (SLY) <strong>and</strong> to establish which clinical sample<br />

would be preferable for KSHV DNA testing.<br />

Methods: We assessed KSHV DNA in plasma <strong>and</strong> PBMCs paired samples from 7 PEL, 8<br />

MCD, 2 SLY HIV+ patients at the diagnosis <strong>and</strong> during the course <strong>of</strong> the illness by using a<br />

real time PCR assay. EBV DNA, HIV RNA <strong>and</strong> CD4 cell counts were also measured.<br />

Results: KSHV viremia was always detectable at diagnosis among the three groups <strong>of</strong><br />

patients. KSHV DNA levels were correlated in matched pairs <strong>of</strong> samples at diagnosis <strong>and</strong><br />

also during follow-up (Spearman correlation coefficient: r=0.73; p


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P16<br />

PATHOLOGY OF RITUXIMAB-INDUCED KAPOSI SARCOMA FLARE<br />

Liron Pantanowitz1, Bruce J. Dezube1, Sharon Marconi1, Ashlee V. Moses2, Klaus Früh2<br />

Department <strong>of</strong> Pathology, Baystate <strong>Medical</strong> Center, Tufts <strong>University</strong> School <strong>of</strong> Medicine,<br />

Springfield, MA; 2 Vaccine <strong>and</strong> Gene Therapy Institute, Oregon Health & Science<br />

<strong>University</strong>, 505 NW185th Ave, Beaverton, OR, 97006, USA<br />

Abstract<br />

Background: Kaposi sarcoma (KS) flare may occur following therapy with<br />

corticosteroids, as part <strong>of</strong> the immune reconstitution inflammatory syndrome seen with<br />

highly active antiretroviral therapy (HAART), <strong>and</strong> after rituximab therapy. The exact<br />

mechanism responsible for iatrogenic KS flare is unclear.<br />

Methods: A case <strong>of</strong> AIDS-associated cutaneous KS flare following rituximab therapy was<br />

compared to similar controls by means <strong>of</strong> immunohistochemistry using vascular makers<br />

(CD34, CD31), monoclonal antibodies to Human Herpesvirus 8 (HHV8) gene products<br />

(LNA-1, K5), as well as B-lymphocyte (CD20) <strong>and</strong> T-lymphocyte (CD3, CD4, CD8)<br />

markers.<br />

Results: CD20+ B-cell depletion with rituximab in KS flare occurred concomitantly with<br />

activation <strong>of</strong> the HHV8 immediate early gene protein K5. KS flare in this patient was<br />

successfully treated with liposomal doxorubicin <strong>and</strong> valganciclovir.<br />

Conclusion: Rituximab-induced KS flare appears to be related to HHV8 activation.<br />

Effective management <strong>of</strong> iatrogenic KS flare therefore depends upon the control <strong>of</strong> HHV8<br />

viremia in conjunction with specific chemotherapy for KS.<br />

Presenting author Email: fruehk@ohsu.edu<br />

113


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P17<br />

C-MAF: A KSHV MICRORNA TARGET<br />

Amy Hansen 1 , Dimitris Lagos 1 , Stephen Henderson 1 , Vicky Emuss 1 Fiona Gratrix 1 , Rolf<br />

Renne 2 <strong>and</strong> Chris Bosh<strong>of</strong>f 1<br />

1 Cancer Research UK Viral Oncology Group, UCL Cancer Institute, Paul O’Gorman<br />

Building, Huntley Street, <strong>University</strong> <strong>College</strong> London, WC1E 6BT, London, U.K.<br />

Abstract<br />

MicroRNAs are small non-coding RNA molecules which post-transcriptionally regulate<br />

gene expression by either blocking translation or inducing mRNA degradation. KSHV<br />

encodes 12 microRNAs located within the latency-associated region <strong>of</strong> the genome; ten<br />

microRNAs are clustered <strong>and</strong> co-expressed. Despite being identified 3 years ago, few<br />

KSHV microRNA targets have been identified. We present microRNA pr<strong>of</strong>iling data which<br />

confirms the expression <strong>of</strong> viral microRNAs in our in vitro endothelial cell model <strong>of</strong><br />

primary infection <strong>and</strong> in vivo within KS lesions. We sought to identify cellular targets <strong>of</strong><br />

the KSHV microRNA cluster in human lymphatic endothelial cells (LECs). Gene expression<br />

microarray pr<strong>of</strong>iling <strong>of</strong> LECs infected with lentivirus expressing either microRNA cluster or<br />

empty vector identified cellular targets silenced by microRNA induced mRNA degradation.<br />

Several genes were significantly deregulated; amongst these was the transcription factor<br />

c-Maf, an oncogenic avian retrovirus homologue. C-maf overexpression transforms B <strong>and</strong><br />

T cells, however its function in endothelial cells is poorly characterised. In silico<br />

prediction analysis identified several potential KSHV microRNA target sites within the c-<br />

Maf 3’UTR. We have subsequently identified miR-K12-11 <strong>and</strong> miR-K12-6 as the principle<br />

silencers <strong>of</strong> c-Maf. In addition we show that silencing is mediated by direct microRNA<br />

interaction with the c-Maf 3’UTR. This work identifies <strong>and</strong> experimentally validates c-Maf<br />

as the first endothelial-specific KSHV microRNA target.<br />

Presenting author Email: amy.hansen@wibr.ucl.ac.uk<br />

114


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Poster Session Abstract P18<br />

KSHV KAPOSIN B SUPPRESSES THE HOST INTERFERON RESPONSE BY<br />

PREVENTING STAT1 PHOSPHORYLATION<br />

Christine A King, Jennifer A Corcoran <strong>and</strong> Craig McCormick<br />

Dalhousie <strong>University</strong>, Department <strong>of</strong> Microbiology <strong>and</strong> Immunology, Halifax, Nova Scotia,<br />

B3H 1X5, Canada.<br />

Abstract<br />

A central component <strong>of</strong> host antiviral defence is the interferon (IFN)-mediated antiviral<br />

pathway. Detection <strong>of</strong> virus by infected cells triggers the secretion <strong>of</strong> type I IFN (IFN-α or<br />

IFN-β) that engages specific receptors on target cells, resulting in activation <strong>of</strong> two<br />

receptor-associated tyrosine kinases, Jak1 <strong>and</strong> Tyk2. This is followed by tyrosine<br />

phosphorylation <strong>and</strong> heterodimerization <strong>of</strong> the STAT1 <strong>and</strong> STAT2 proteins.<br />

Phosphorylated STAT1 <strong>and</strong> STAT2 combine with IRF-9 to form the ISGF-3 complex,<br />

which, upon translocation to the nucleus, binds to the cis element ISRE (IFN-stimulated<br />

response element), to drive transcription <strong>of</strong> IFN responsive genes. To ensure viral<br />

persistence <strong>and</strong> productive infection, KSHV has evolved a variety <strong>of</strong> countermeasures to<br />

disrupt IFN signal transduction. Here we show that the latent KSHV kaposin B protein<br />

potently inhibits transcription from the interferon stimulated response element (ISRE) in<br />

response to IFN-α. Furthermore, primary human umbilical vein endothelial cells<br />

(HUVECs) expressing kaposin B fail to accumulate phosphorylated STAT1 in response to<br />

IFN-α <strong>and</strong> harbor decreased levels <strong>of</strong> total STAT1. Our data suggest that kaposin B<br />

interferes with the host IFN response thereby contributing to successful immune evasion<br />

by KSHV.<br />

Presenting author Email: caking@dal.ca<br />

115


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Delegate Contact Details<br />

116


Adam Grundhh<strong>of</strong>f<br />

Friedensallee 63<br />

Hamburg<br />

22763<br />

Germany<br />

Adam.Grundh<strong>of</strong>f@hpi.uni-hamburg.de<br />

Akira Shimizu,<br />

Division <strong>of</strong> Infection & Immunity,<br />

UCL,<br />

46 Clevel<strong>and</strong> Street,<br />

London,<br />

W1T 4JF<br />

ashimizu@med.gunma-u.ac.jp<br />

Amy Hansen<br />

Viral Oncology Team<br />

UCL Cancer Institute<br />

Paul O'Gorman Building<br />

<strong>University</strong> <strong>College</strong> London<br />

72 Huntley Street<br />

WC1E 6BT<br />

United Kingdom<br />

a.hansen@ucl.ac.uk<br />

Andrea Suarez<br />

12800 E 19th Ave MS 8333<br />

PO Box 6511<br />

Aurora Colorado<br />

80045<br />

United States<br />

thomas.shallow@uchsc.edu<br />

Angus Wilson<br />

NYU School <strong>of</strong> Medicine<br />

Dept. Microbiology<br />

550 First Avenue<br />

New York<br />

NY 10016<br />

United States<br />

wilsoa02@med.nyu.edu<br />

Bahram Ebrahimi<br />

Division <strong>of</strong> <strong>Medical</strong> Microbiology<br />

<strong>University</strong> <strong>of</strong> Liverpool<br />

Daulby Street<br />

Liverpool<br />

L69 3GA<br />

United Kingdom<br />

ebrahimi@liv.ac.uk<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Adrian Whitehouse<br />

Institute <strong>of</strong> Molecular <strong>and</strong> Cellular Biology<br />

<strong>University</strong> <strong>of</strong> Leeds<br />

Leeds<br />

LS2 9JT<br />

United Kingdom<br />

a.whitehouse@leeds.ac.uk<br />

Alexis Madrid<br />

Ganem Lab<br />

HSW1501<br />

513 Parnassus Ave<br />

San Francisco<br />

94143-0552<br />

United States<br />

alexis.madrid@ucsf.edu<br />

Andrea O’Hara<br />

3 Georgetown Ct<br />

Durham<br />

NC<br />

27705<br />

United States<br />

ohara@unc.edu<br />

Andrew Hislop<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Institute for Cancer Studies<br />

Edgbaston<br />

Birmingham<br />

West Midl<strong>and</strong>s<br />

B15 2TT<br />

United Kingdom<br />

a.d.hislop@bham.ac.uk<br />

Anika Haevemeier<br />

Hannover <strong>Medical</strong> School<br />

Institute <strong>of</strong> Virology OE5230<br />

Carl-Neuberg-Str.1<br />

Hannover<br />

30625<br />

Germany<br />

anikahaevemeier@web.de<br />

Bala Ch<strong>and</strong>ran<br />

Department <strong>of</strong> Microbiology <strong>and</strong> Immunology<br />

RFUMS<br />

3333 Green Bay Road<br />

North Chicago IL<br />

60064<br />

United States<br />

bala.ch<strong>and</strong>ran@rosalindfranklin.edu<br />

117


Bettina Kempkes<br />

Helmholtz Center Munich<br />

Institute <strong>of</strong> Clinical Molecular Biology<br />

Marchioninistr. 25<br />

Munich<br />

81377<br />

Germany<br />

kempkes@helmholtz-muenchen.de<br />

Brad Spiller<br />

Department <strong>of</strong> Child Health<br />

Cardiff <strong>University</strong> School <strong>of</strong> Medicine<br />

5th Floor <strong>University</strong> Hospital <strong>of</strong> Wales<br />

Cardiff<br />

South Glamorgan<br />

CF144XN<br />

United Kingdom<br />

spillerb@cf.ac.uk<br />

Charles Rinaldo<br />

A419C Crabtree Hall- GSPH<br />

130 DeSoto Street<br />

<strong>University</strong> <strong>of</strong> Pittsburgh<br />

Pittsburgh<br />

15261<br />

United States<br />

rinaldo@pitt.edu<br />

Chris Parsons<br />

86 Jonathan Lucas St<br />

Hollings Cancer Center- Room HO506<br />

Charleston South Carolina<br />

29425<br />

United States<br />

parsonch@musc.edu<br />

Craig McCormick<br />

15 Silverwood Terrace<br />

Halifax<br />

Nova Scotia<br />

B3M2Y9<br />

Canada<br />

craig.mccormick@dal.ca<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Blossom Damania<br />

Lineberger Cancer Ctr RM 31-353<br />

UNC-Chapel Hill CB7295<br />

Chapel Hill<br />

27599<br />

United States<br />

damania@med.unc.edu<br />

Bridget Robinson<br />

831 SW Vista Ave<br />

Apt 201<br />

Portl<strong>and</strong><br />

USA<br />

97205<br />

United States<br />

robinsob@ohsu.edu<br />

Charles Wood<br />

<strong>University</strong> <strong>of</strong> Nebraska - Lincoln<br />

102C Morrison Center<br />

Lincoln<br />

Nebraska<br />

68583-0900<br />

United States<br />

cwood1@unl.edu<br />

Christine A King<br />

121 John Stewart Dr<br />

Dartmouth<br />

B2W5W8<br />

Canada<br />

caking@dal.ca<br />

Cristina Areste<br />

Institute for Cancer Studies<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Edgbaston<br />

Birmingham<br />

West Midl<strong>and</strong>s<br />

B15 2TT<br />

United Kingdom<br />

m.c.arestecalero@bham.ac.uk<br />

118


Dan Frampton<br />

Department <strong>of</strong> Infection<br />

<strong>University</strong> <strong>College</strong> London<br />

46 Clevel<strong>and</strong> Street<br />

London<br />

W1T 4JF<br />

United Kingdom<br />

dan.frampton@gmail.com<br />

Dean Kedes<br />

1300 Jefferson Park Ave.<br />

Dept. <strong>of</strong> Microbiology<br />

<strong>University</strong> <strong>of</strong> Virginia<br />

Charlottesville<br />

22908<br />

United States<br />

kedes@virginia.edu<br />

Denise Whitby<br />

SAIC-Frederick, Inc<br />

National Cancer Institute<br />

P.O. Box B, Bldg. 535<br />

Frederick<br />

MD<br />

21702<br />

United States<br />

whitbyd@ncifcrf.gov<br />

Dirk Dittmer<br />

72005 Wilkinson<br />

Chapel Hill<br />

NC<br />

27517<br />

United States<br />

ddittmer@med.unc.edu<br />

Dr Bernd Hillenbr<strong>and</strong><br />

Engelhardstr. 7<br />

Hannover<br />

30173<br />

Germany<br />

hillenbr<strong>and</strong>.bernd@mh-hannover.de<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

David Blackbourn<br />

Cancer Research UK Institute for Cancer Studies<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Vincent Drive<br />

Edgbaston<br />

Birmingham<br />

B15 2TT<br />

United Kingdom<br />

d.j.blackbourn@bham.ac.uk<br />

Debasmita Roy<br />

140 BPW Club Road Apt B18<br />

Carrboro<br />

Orange<br />

NC27510<br />

United States<br />

debasmita_roy@med.unc.edu<br />

Denys Khaperskyy<br />

12-6059 Shirley St<br />

Halifax<br />

B3H 2M9<br />

Canada<br />

D.Khaperskyy@dal.ca<br />

Donald Alcendor<br />

1005 Dr. D.B. Todd Jr. Blvd.<br />

Hubbard Hospital 5th Floor Rm 5025<br />

Nashville TN<br />

Davidson<br />

37208<br />

United States<br />

dalcendor@mmc.edu<br />

Dr Dimitris Lagos<br />

Cancer Research UK Viral Oncology Group<br />

<strong>University</strong> <strong>College</strong> London<br />

Paul O'Gorman Building<br />

72 Huntley Street<br />

WC1E 6BT<br />

United Kingdom<br />

d.lagos@ucl.ac.uk<br />

119


Dr Janet Douglas<br />

505 NW 185th Ave<br />

Beaverton<br />

97006<br />

United States<br />

dougljan@ohsu.edu<br />

Edward Tsao<br />

Centre for Virology (Bloomsbury)<br />

<strong>University</strong> <strong>College</strong> London<br />

46 Clevel<strong>and</strong> Street<br />

London<br />

W1T 4JF<br />

United Kingdom<br />

e.tsao@ucl.ac.uk<br />

Emilee Knowlton<br />

312 Lehigh Avenue<br />

Apt 3<br />

Pittsburgh<br />

15232<br />

United States<br />

erk21@pitt.edu<br />

Eriko Osaki<br />

H<strong>and</strong>ayama Higashi-ku Hamamatsu-shi<br />

1-20-1<br />

Hamamatsu<br />

Shizuoka<br />

431-3129<br />

Japan<br />

eohsaki@hama-med.ac.jp<br />

Ethel Cesarman<br />

820 Park Ave<br />

Hoboken NJ<br />

7030<br />

United States<br />

ecesarm@med.cornell.edu<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Dr Lynn Butler<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Edgbaston<br />

Birmingham<br />

B15 2TT<br />

butlerlm@adf.bham.ac.uk<br />

Elizabeth Read-Connole<br />

6130 Executive Blvd.<br />

Rockville MD 20852<br />

Rockville<br />

Montgomery<br />

20852<br />

United States<br />

econnole@mail.nih.gov<br />

Enrique Mesri<br />

<strong>University</strong> <strong>of</strong> Miami Sylvester Cancer Center<br />

Microbiology <strong>and</strong> Immunology<br />

1550 NW 10th Ave Pap Bldg Room 109<br />

Miami<br />

33136<br />

United States<br />

emesri@med.miami.edu<br />

Erle Robertson<br />

3610 Hamilton Walk<br />

201E Johnson Pavilion<br />

Philadelphia<br />

Pennsylvania<br />

19104<br />

United States<br />

amyho2@mail.med.upenn.edu<br />

Eve Coulter<br />

Department <strong>of</strong> Infection<br />

<strong>University</strong> <strong>College</strong> London<br />

46 Clevel<strong>and</strong> Street<br />

London<br />

W1T 4JF<br />

United Kingdom<br />

evecoulter@hotmail.com<br />

120


Fang Cheng<br />

Vuolukiventie 1b g145<br />

Helsinki<br />

710<br />

Finl<strong>and</strong><br />

cheng.fang@helsinki.fi<br />

Gary Hayward<br />

1026 Cowpens Ave<br />

Baltimore<br />

MD<br />

21286<br />

United States<br />

ghayward@jhmi.edu<br />

Gianna Ballon<br />

21-59 45th Street<br />

Astoria<br />

New York<br />

11105<br />

United States<br />

gib2004@med.cornell.edu<br />

Grzegorz Sarek<br />

Ahventie 5A11<br />

Espoo<br />

2170<br />

Finl<strong>and</strong><br />

grzegorz.sarek@helsinki.fi<br />

Hye-Jeong Cho<br />

Anam-dong Seongbuk-Gu<br />

Division <strong>of</strong> Biotechnology <strong>College</strong> <strong>of</strong> Life<br />

<strong>Sciences</strong> <strong>and</strong> Biotechnology<br />

Korea <strong>University</strong><br />

Seoul<br />

136-713<br />

Republic <strong>of</strong> Korea<br />

chohyejeong@korea.ac.kr<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Gaby S<strong>and</strong>er<br />

Molecular <strong>and</strong> Experimental Surgery<br />

Schwabachanlage 10<br />

Erlangen<br />

Bavaria<br />

91054<br />

Germany<br />

gaby.s<strong>and</strong>er@uk-erlangen.de<br />

Georg Malterer<br />

Max von Pettenk<strong>of</strong>er Institut<br />

Dept. <strong>of</strong> Virology<br />

Pettenk<strong>of</strong>er Strasse 9a<br />

Muenchen<br />

80336<br />

Germany<br />

malterer@mvp.uni-muenchen.de<br />

Greg Bruce<br />

9756 Lakeshore Blvd NE<br />

Seattle<br />

Washington<br />

98115<br />

United States<br />

greg.bruce@seattlechildrens.org<br />

Imogen Lai<br />

<strong>University</strong> <strong>College</strong> London<br />

46 Clevel<strong>and</strong> Street<br />

London<br />

W1T 4JF<br />

United Kingdom<br />

imogen.lai@ucl.ac.uk<br />

121


James Boyne<br />

Institute <strong>of</strong> Molecular <strong>and</strong> Cellular Biology<br />

Garstang Building<br />

<strong>University</strong> <strong>of</strong> Leeds<br />

Leeds<br />

LS2 9JT<br />

United Kingdom<br />

j.r.boyne@leeds.ac.uk<br />

Jennifer Corcoran<br />

Room 7F Tupper 5850 <strong>College</strong> Street<br />

Dept. Microbiology <strong>and</strong> Immunology<br />

Dalhousie <strong>University</strong><br />

Halifax NS<br />

B3H 1X5<br />

Canada<br />

jcorcora@dal.ca<br />

Jinjong Myoung<br />

1322 3rd Ave<br />

Rm1<br />

San Francisco<br />

94122<br />

United States<br />

jinjong.myoung@ucsf.edu<br />

John Nicholas<br />

Johns Hopkins Oncology Center<br />

1650 Orleans Street<br />

CRB-I Room 309<br />

Baltimore<br />

Maryl<strong>and</strong><br />

21231<br />

United States<br />

nichojo@jhmi.edu<br />

John West<br />

5803 Sentinel Dr.<br />

Raleigh<br />

27609<br />

United States<br />

john_west@med.unc.edu<br />

Karen Misstear<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Division <strong>of</strong> Cancer Studies<br />

Birmingham<br />

West Midl<strong>and</strong>s<br />

B30 1TR<br />

United Kingdom<br />

kxm694@bham.ac.uk<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Jean Gustin<br />

VGTI<br />

505 NW 185th Avenue<br />

Beaverton<br />

Washington<br />

97006<br />

United States<br />

gustinj@ohsu.edu<br />

Jiang-Mei Qin<br />

12 Blenheim Crescent<br />

Leeds<br />

LS2 9AY<br />

United Kingdom<br />

qinjiangmei@yahoo.com.cn<br />

Johanna Viiliäinen<br />

Genome-Scale Biology Research Program<br />

Institute <strong>of</strong> Biomedicine, Biomedicum Helsinki<br />

A530b1<br />

P.O Box 63 (Haartmaninkatu 8)<br />

FIN-00014 <strong>University</strong> <strong>of</strong> Helsinki<br />

Finl<strong>and</strong><br />

johanna.viiliainen@helsinki.fi<br />

Johnan Kaleeba<br />

4301 Jones Bridge Road<br />

Bethesda MD<br />

20814<br />

United States<br />

jkaleeba@usuhs.mil<br />

Karlie Plaisance<br />

1376 Mowry Rd<br />

Renne Lab Room 375E<br />

Gainesville<br />

Florida<br />

32610<br />

United States<br />

kplais4@ufl.edu<br />

122


Keiji Ueda<br />

Dept.<strong>of</strong> Infectious Disease<br />

Hamamatsu Univ. School <strong>of</strong> Medicine<br />

1-20-1 H<strong>and</strong>ayama Higashi-ku Hamamatsu<br />

Shizuoka<br />

431-3192<br />

Japan<br />

kueda@hama-med.ac.jp<br />

Kwun Wah Wen<br />

126 Marlowe Ct<br />

Carrboro<br />

27510<br />

United States<br />

kenwen@med.unc.edu<br />

Lauren Lepone<br />

721 Copel<strong>and</strong> St<br />

Apt 2<br />

Pittsburgh<br />

PA<br />

15232<br />

United States<br />

lml33@pitt.edu<br />

Lucy Dalton-Griffin<br />

<strong>University</strong> <strong>College</strong> London<br />

Windeyer Institute <strong>of</strong> <strong>Medical</strong> <strong>Sciences</strong><br />

46 Clevel<strong>and</strong> Street<br />

London<br />

W1T 4JF<br />

United Kingdom<br />

ucbclda@ucl.ac.uk<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Maria Luisa Calabro'<br />

Department <strong>of</strong> Oncology <strong>and</strong> Surgical <strong>Sciences</strong><br />

Oncology Section<br />

via Gattamelata 64<br />

Padova<br />

35128<br />

Italy<br />

lcalabro@unipd.it<br />

Klaus Frueh<br />

505 NW 185th Ave<br />

Beaverton<br />

Oregon<br />

97006<br />

United States<br />

fruehk@ohsu.edu<br />

Laura Marcos<br />

Centro Nacional de Biotecnologia CSIC<br />

Darwin 3 Campus Universidad Autonoma<br />

Madrid<br />

28049<br />

Spain<br />

lmarcos@farm.ucm.es<br />

Lei Yang<br />

Key Laboratory <strong>of</strong> Xinjiang Endemic & Ethnic<br />

Disease<br />

Shihezi <strong>University</strong><br />

Shihezi<br />

XinJiang<br />

832002<br />

China<br />

Lyang@shzu.edu.cn<br />

Magdalena Weidner-Glunde<br />

Hannover <strong>Medical</strong> School<br />

Institute <strong>of</strong> Virology<br />

Carl-Neuberg.-Str.1 Bldg.I6<br />

Hannover<br />

30625<br />

Germany<br />

weidnerg@allmail.mh-hannover.de<br />

Maria Nazzarena Labo<br />

SAIC-Frederick, Inc<br />

National Cancer Institute<br />

P.O. Box B, Bldg. 535<br />

Frederick<br />

MD<br />

21702<br />

United States<br />

labon@mail.nih.gov<br />

123


Mark Cannon<br />

2016 Fremont Ave S<br />

Minneapolis<br />

55405<br />

United States<br />

canno101@umn.edu<br />

Michael Stuerzl<br />

Schwabachanlage 10<br />

Erlangen<br />

91054<br />

Germany<br />

Michael.Stuerzl@uk-erlangen.de<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Nathalie Cloutier<br />

Rhumatology <strong>and</strong> Immunology Research Center<br />

2705 Laurier blvd<br />

Room T1-49<br />

Sainte-Foy<br />

Quebec<br />

G1V 4G2<br />

Canada<br />

nathalie.cloutier@crchul.ulaval.ca<br />

Paul Kellam<br />

Centre for Virology (Bloomsbury)<br />

<strong>University</strong> <strong>College</strong> London<br />

46 Clevel<strong>and</strong> Street<br />

London<br />

W1T 4JF<br />

United Kingdom<br />

p.kellam@ucl.ac.uk<br />

Michael Lagun<strong>of</strong>f<br />

8816 8th ave N.E.<br />

Seattle<br />

Washington<br />

98115<br />

United States<br />

lagun<strong>of</strong>f@u.washington.edu<br />

Moon Jung Song<br />

1 Anam-dong<br />

Divison <strong>of</strong> Biotechnology <strong>College</strong> <strong>of</strong> Life <strong>Sciences</strong><br />

<strong>and</strong> Biotechnology<br />

Korea Univeristy<br />

Seoul<br />

Seongbuk-gu<br />

136-713<br />

Republic <strong>of</strong> Korea<br />

moonsong@korea.ac.kr<br />

Paivi Ojala<br />

Haartmaninkatu 8<br />

<strong>University</strong> <strong>of</strong> Helsinki<br />

14<br />

Finl<strong>and</strong><br />

Paivi.Ojala@helsinki.fi<br />

Priya Bellare<br />

355 Crestmont Drive<br />

San Francisco<br />

California<br />

94131<br />

United States<br />

Priya.Bellare@ucsf.edu<br />

124


Pr<strong>of</strong> Juergen Haas<br />

Max-von-Pettenk<strong>of</strong>er Institute<br />

Pettenk<strong>of</strong>erstrasse 9a<br />

Muenchen<br />

80336<br />

Germany<br />

haas@lmb.uni-muenchen.de<br />

Rachel Wheat<br />

Univeristy <strong>of</strong> Birmingham<br />

Division <strong>of</strong> Cancer Studies<br />

Birmingham<br />

West Midl<strong>and</strong>s<br />

B30 1TR<br />

United Kingdom<br />

r.l.wheat@bham.ac.uk<br />

Rosamaria Tedeschi<br />

Centro di Riferimento Oncologico<br />

via F Gallini 2<br />

Aviano<br />

33081<br />

Italy<br />

rtedeschi@cro.it<br />

Scott Wong<br />

Vaccine & Gene Therapy Institute<br />

505 NW 185th Avenue<br />

Beaverton Oregon<br />

97006<br />

United States<br />

wongs@ohsu.edu<br />

Sally Watterson<br />

Division <strong>of</strong> Infection & Immunity,<br />

UCL,<br />

46 Clevel<strong>and</strong> Street,<br />

London,<br />

W1T 4JF<br />

sally_watterson@hotmail.com<br />

Sheila Dollard<br />

1600 Clifton Rd<br />

Atlanta<br />

30333<br />

United States<br />

sgd5@cdc.gov<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Rachel Colman<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Institute <strong>of</strong> Cancer Studies<br />

Edgbaston<br />

Birmingham<br />

West Midl<strong>and</strong>s<br />

B15 2TT<br />

United Kingdom<br />

r.colman@bham.ac.uk<br />

Rolf Renne<br />

1376 Mowry Road<br />

PO BOX 103633<br />

Gainesville<br />

Florida<br />

32610<br />

United States<br />

rrenne@ufl.edu<br />

Ryan Estep<br />

505 NW 185th Ave<br />

Beaverton OR<br />

97006<br />

United States<br />

estepr@ohsu.edu<br />

Sean Gregory<br />

140 BPW Club Rd<br />

Apt E7<br />

Carrboro<br />

27510<br />

United States<br />

sgregory@med.unc.edu<br />

Sherren Sabbah<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Institute for Cancer Studies<br />

Edgbaston<br />

Birmingham<br />

West Midl<strong>and</strong>s<br />

B15 2TT<br />

United Kingdom<br />

SXS463@bham.ac.uk<br />

125


Simon Chanas<br />

<strong>University</strong> <strong>of</strong> Birmingham<br />

Institute for Cancer Studies<br />

Edgbaston<br />

Birmingham<br />

West Midl<strong>and</strong>s<br />

B15 2TT<br />

United Kingdom<br />

s.a.chanas@bham.ac.uk<br />

Terri DiMaio<br />

4038 Stone Way N. 303<br />

Seattle<br />

King<br />

98103<br />

United States<br />

tdimaio@u.washington.edu<br />

Timothy Rose<br />

9514 Lake Shore Blvd NE<br />

Seattle<br />

Washington<br />

98115<br />

United States<br />

timothy.rose@seattlechildrens.org<br />

Utthara Nayar<br />

1233 York Ave Apt 12B<br />

New York<br />

NY<br />

10065<br />

United States<br />

utn2001@med.cornell.edu<br />

Vickie Marshall<br />

SAIC-Frederick, Inc<br />

National Cancer Institute<br />

P.O. Box B, Bldg. 535<br />

Frederick<br />

MD<br />

21702<br />

United States<br />

marshall@ncifcrf.gov<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Subhash Verma<br />

201E Johnson Pavilion<br />

3610 Hamilton Walk<br />

Philadelphia<br />

PA<br />

19104<br />

United States<br />

vermas@mail.med.upenn.edu<br />

Thomas Schulz<br />

Institute <strong>of</strong> Virology<br />

Hannover <strong>Medical</strong> School<br />

Carl-Neuberg-Str. 1<br />

Hannover<br />

30625<br />

Germany<br />

schulz.thomas@mh-hannover.de<br />

Tracey Barrett<br />

School <strong>of</strong> Crystallography<br />

Birkbeck <strong>College</strong><br />

Malet Street<br />

London<br />

WC1E 7HX<br />

United Kingdom<br />

t.barrett@mail.cryst.bbk.ac.uk<br />

Veenu Minhas<br />

UNL Nebraska Center for Virology<br />

102A Morrison Ctr<br />

4240 Fair Street<br />

Lincoln<br />

Nebraska<br />

68583<br />

United States<br />

veenu9@yahoo.com<br />

Vicky Emuss<br />

UCL Cancer Institute<br />

74 Huntley Sreett<br />

Paul O’Gorman Building<br />

London<br />

WC1E 6BT<br />

United Kingdom<br />

v.emuss@ucl.ac.uk<br />

126


Vladimir Majerciak<br />

4817 36th Street NW<br />

Apt. 208<br />

Washington<br />

DC<br />

20008<br />

United States<br />

majerciv@mail.nih.gov<br />

Wiebke Albrecht<br />

<strong>Medical</strong> School Hanover<br />

Institute <strong>of</strong> Virology OE5230<br />

Carl-Neuberg-Str.1<br />

Hannover<br />

30625<br />

Germany<br />

albrecht.wiebke@mh-hannover.de<br />

Yan Yuan<br />

Department <strong>of</strong> Microbiology<br />

<strong>University</strong> <strong>of</strong> Pennsylvania <strong>Dental</strong> School<br />

240 S. 40th Street<br />

Philadelphia<br />

Pennsylvania<br />

19104<br />

United States<br />

yuan2@pobox.upenn.edu<br />

Yoshihiro Izumiya<br />

UCDMC Research III Rm2400<br />

4645 2nd Ave.<br />

Sacramento<br />

95817<br />

United States<br />

yizumiya@ucdavis.edu<br />

Zhi-Ming Zheng<br />

14507 Cartwright Way<br />

North Potomac MD<br />

Montgomery<br />

20878<br />

United States<br />

zhengt@exchange.nih.gov<br />

The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Wendall Miley<br />

SAIC-Frederick, Inc<br />

National Cancer Institute<br />

P.O. Box B, Bldg. 535<br />

Frederick<br />

MD<br />

21702<br />

United States<br />

miley@ncifcrf.gov<br />

Xuefeng Zhang<br />

264 Grove Street Apt. 1<br />

Newton<br />

2466<br />

United States<br />

xzhang3@bidmc.harvard.edu<br />

Yan Zeng<br />

Key Laboratory <strong>of</strong> Xinjiang Endemic & Ethnic<br />

Disease<br />

Shihezi <strong>University</strong><br />

Shihezi<br />

XinJiang<br />

832002<br />

China<br />

zengyan910@yahoo.com.cn<br />

Yu-Hsuan Wu<br />

No.155- Sec.2- Linong Street- Taipei- 112<br />

Taiwan (ROC)<br />

Institute <strong>of</strong> Microbiology & Immunology<br />

Room 411<br />

Taipei<br />

112<br />

Taiwan<br />

b881625@life.nthu.edu.tw<br />

127


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Author index<br />

128


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Author Abstract # Author Abstract #<br />

Ageichik, Alex<strong>and</strong>er V. 23 Chahal, Hema 44<br />

Albrecht, W. 5 Ch<strong>and</strong>ran, Bala 19, 28, 60<br />

Alcendor, Donald J. 21 Chang, Pei-Ching 37, P9<br />

Alkharsah, Khaled R. 20, 24 Cheng, Fang 36<br />

Amadori, A. P4 Chieco-Bianchi, L. P4, P6<br />

Amin, Minal M. 48 Cho, Hye-Jeong P14<br />

Anh, Pham Thi Hoang 51 Christalla, Thomas P13<br />

Arico, E. 67 Cloutier, Nathalie P2<br />

Bagnéris, Claire 23 Collins, Mary 23<br />

Bagni, Rachel 26, 27 Colman, Rachel 13, 44<br />

Bakken, Thomas 63 Colombo, E. P5<br />

Ballon, Gianna 15 Corcoran, Jennifer A. P18<br />

Barrett, Tracey 23 Coulter, Eve M. 30, P10<br />

Bartee, Eric P8 Crabtree, Kay L. 53<br />

Bellare, Priya 33 Cronin, Nora 23<br />

Bergamo, E. P5 Csernus, Balasz 49<br />

Bidoli, Ettore P15 Dalton-Griffin, Lucy 58<br />

Blackbourn, David J. 13, 44 Damania, Blossom A. 1, 11, 31, 32, 50<br />

Blom, Anna M. 43 De Paoli, Paolo P15<br />

Bosh<strong>of</strong>f, Chris 12, 23, 63, P17 De Sanjose, Silvia 51<br />

Bottero, Virginie 19 Della Bella, S. P5<br />

Bower, Mark 12 Deng, Merlyn 65<br />

Boyne, James R. 39, 41 Desai, Prashant 21<br />

Brambilla, L. P5 Dezube, Bruce J. 32, P16<br />

Bratke, Kai P3 Di Gangi, I.M. P4, P6<br />

Brayfield, Brad P. 55 Di Stefano, M. P6<br />

Brown, Cedric 48 DiBartolo, Daniel 25<br />

Brown, Elizabeth 54 Dillner, Joakim 43<br />

Bruce, A. Gregory 16 Dinkel, Holger 56<br />

Bubman, Darya 49 DiPersio, C. Michael 59<br />

Bürger, Antje 20 Dittmer, Dirk P. 1, 31, 32, 50<br />

Burnside, Kellie L. 2 Dollard, Sheila C. 48<br />

Cai, Qiliang 6 Douglas, Janet 10<br />

Calabrò, M.L. P4, P5, P6 Downing, Robert 48<br />

Cannon, Mark 63 Dufresne, Andrew T. 33<br />

Cardenas, Elisa 14 Duran, E. Margarita 22<br />

Casabona, Jordi 54 Dutia, B. 34<br />

Cavallin, Lucas 22 Ebrahimi, B. 34, 67<br />

Cesarman, Ethel 15, 25, 49 Ellison, Thomas J. 37, 66, P9<br />

Chadburn, Amy 15, 25 Emuss, Vicky P17<br />

129


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Estep, Ryan D. 14 Hensler, Heather 8<br />

Falk, Christine S. P3 Herrero, Rol<strong>and</strong>o 51<br />

Felber, Barbara K. 65 Hieu, Nguyen Trong 51<br />

Fiore, J.R. P6 Hillenbr<strong>and</strong>, Bernd 20<br />

Fischer, Irina 20 Hilscher, Chelsey 1, 31<br />

Fitzgerald, Latricia D. 37, P9 Hislop, Andrew D. 13, 45<br />

Flam<strong>and</strong>, Louis P2 Hladik, Wolfgang 48<br />

Fossum, Even 57 Hollow, Charles 35<br />

Fowlkes, Ashley L. 48 Hsia, Datsun P9<br />

Frampton, Dan 30, P10 Hu, Soo-Jin Han Jianhong 26, 27<br />

Franceschi, Silvia 51 Huang, Jin 29, 52<br />

Frank, Ronald 4 Hyjek, Elizabeth 25<br />

Früh, Klaus 10, P8, P16 Imami, Nesrina 12<br />

Gallego, Pedro P7 Indraccolo, S. P4<br />

Ganem, Don 33, 42, 47 Iotzova, Guergana P3<br />

Garrigues, H. Jacques 59 Isaacs, Jennifer 62<br />

Gasperini, P. P4 Izumiya, Chie 66<br />

Gellermann, E. 5 Izumiya, Yoshihiro 37, 66, P9<br />

Gessain, Antoine P3 Jais, Mariel 8, 46<br />

Gesualdo, L. P6 Järviluoma, Annika 3<br />

Ghazal, Peter P3 Jenkins, Frank J. 8, 46<br />

Goldschmidt-Clermont, Pascal J. 22 Jochmann, Ramona 24<br />

Gonin-Laurent, Nathalie 24 Kankasa, Chipepo 53, 55<br />

Gotch, Frances 12 Karayama, Masato P11<br />

Gratrix, Fiona 12, P17 Kellam, Paul 9, 30, 38, 58, P10<br />

Gravett, Courtney 16 Keller, Shannon 25<br />

Greco, P. P6 Kempkes, Bettina 57<br />

Gregory, Sean 11, 31 King, Christine A. P18<br />

Groopman, Jerome E. 17 Kim, Chang Hee 26, 27<br />

Grundh<strong>of</strong>f, Adam P13 Knowles, Daniel M. 25<br />

Guasparri, Ilaria 25, 49 Knowlton, Emilee 8, 46<br />

Guo, Shu-Xia 52 Konrad, Andreas 56<br />

Gustin, Jean 10 Korodi, Zoltan 43<br />

Haas, Jürgen 57, P3 Koskinen, Päivi J. 36<br />

Hansen, Amy P17 Kremmer, Elisabeth 56, 57<br />

Harrington Jr., William 32 Kung, Hsing-Jien 37, 66, P9<br />

Hävemeier, Anika 18 Labo, Maria Nazzarena 54<br />

Hayward, Gary S. 21 Lagos, Dimitris 12, P17<br />

Heinzelmann, Katharina 57 Lai, Imogen Yi-Chun 9, 38<br />

Henderson, Stephen P17 Laiho, Marikki 3<br />

130


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Lampson, Michael 6 Millman, Scott 64<br />

Lane, B. 34, 67 Minhas, Veenu 53, 55<br />

Lazcano, Eduardo 51 Misstear, Karen 13, 44<br />

Lee, Dongho P14 Mitchell, Charles D. 53, 55<br />

Lehtonen, Anne 36 Moses, Ashlee V. 10, P8, P16<br />

Lepone, Lauren 46 Muñoz, Nubia 51<br />

Leubert, Rene 56 Myoung, Jinjong 47<br />

Levine, Rita 19 Naschberger, Elisabeth 24<br />

Li, Dong-Mei 29, 52 Nasu, Ryo 7<br />

Li, Feng 29, 52 Nayar, Utthara 49<br />

Lin, Su-Fang P12 Neil, Stuart 9<br />

Liu, Yi-Fang 15 Neipel, Frank 24, 56<br />

Long, Heather 13 Nzaro, Esau 48<br />

Lopitz, Fern<strong>and</strong>o P7 O’Hara, Andrea J. 32, P11<br />

Lord, Janet 44 Ohsaki, Eriko P1<br />

Luciw, Paul A. 37, 66, P9 Ojala, Päivi M 3, 36<br />

Luo, Xian-Dao 29 Okroj, Marcin 43<br />

Luttmann, Werner P3 Ottinger, Matthias 4<br />

M’soka, Tendai J. 53, 55 Pantanowitz, Liron P8, P16<br />

Ma, Qi 22 Pardieu, Claire 9<br />

Madrid, Alexis 42 Parsons, Chris 62<br />

Majerciak, Vladimir 40, 65 Perez, Sussana 51<br />

Malterer, Georg P3 Persson, Linda 64<br />

Manso, B. 67 Piazza, Paolo 8, 46<br />

Mansouri, M<strong>and</strong>ana 10, P8 Pietrek, Macel 18<br />

Maor, Yehoshua 17 Plaisance, Karlie 27<br />

Marconi, Sharon P8, P16 Pliquet, Daniel 4<br />

Marcos-Villar, Laura P7 Qin, Zhiqiang 62<br />

Mark, Linda 43 Qing, Jiang-Mei 52<br />

Marshall, Vickie 54 Quereshi, Omar 13<br />

Martró, Eliza 54 Raghu, Hari 28, 60<br />

Marus, Alessia P15 Rajewsky, Klaus 15<br />

Matos, Elena 51 Rappocciolo, Giovanna 8, 46<br />

Matsumura, Satoko 7 Ray, Alex 54<br />

Mbisa, Georgina 51 Renne, Rolf 26, 27, 54, P17<br />

McCormick, Craig P18 Rinaldo, Charles R. 8, 46<br />

Menu, E. P6 Rivas, Carmen P7<br />

Mermin, Jonathan 48 Roaden, L. 34, 67<br />

Mesri, Enrique A. 22 Roback, John 48<br />

Miley, Wendell 26, 27 Robertson, Erle S. 6<br />

131


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Robertson, Kevin P3 Thurau, Mathias 24, 56<br />

Rodriguez, Manuel S. P7 Tojk<strong>and</strong>er, Sari 3<br />

Rose, Timothy M. 2, 16, 59 Towers, Greg 9<br />

Roy, Debasmita 50 Tsai, Yuan-Hau P12<br />

Rubinchikova, Yelena E. 59 Tsao, Edward 9, 30, 58<br />

Ryan, Jonathan T. 16 Ueda, Keiji P1, P11<br />

Sabbah, Shereen 45 van Dyk, L.F. 61<br />

Sadagopan, Sathish 19, 28, 60 Varga, Laszlo 60<br />

S<strong>and</strong>er, Gaby 24, 56 Varjosalo, Markku 36<br />

Sansom, David 13 Vart, Richard James 12<br />

Sarek, Grzegorz 3 Vartia, Salla 3<br />

Sasaki, Yoshiteru 15 Veettil, Mohanan Valiya 19, 28, 60<br />

Scholz, Barbara 57 Verma, Subhash C. 6<br />

Schulz, Thomas F. 4, 5, 18, 20, 24, Viejo-Borbolla, A.<br />

5<br />

36, 56<br />

Viswanathan, Kasinath 10<br />

Sharma-Walia, Neelam 19, 28, 60 Waksman, Gabriel 23<br />

Sharp, Tyson V P12 Wallace, Bonnie 23<br />

Sheridan, V. 34 Walz, Nicole P13<br />

Shin, Hai-Rim 51 Wang, Dian 54<br />

Si, Huaxin 6 Wang, Fu-Zhang 60<br />

Simonelli, Cecilia P15 Wang, Hsei-Wei P12<br />

Sin, Sang-Hoon 50 Wang, Ling 31, 50<br />

Skalsky, Rebecca L. 26 Wang, Sheng 29<br />

Song, Moon Jung P14 Wang, Yan 35<br />

Spiller, O. Brad 43 Webb, Ben 9<br />

Stürzl, Michael<br />

24, 56<br />

Weidner-Glunde,<br />

Suárez, A.L.<br />

61<br />

Magdalena<br />

4, 36<br />

Sukvirach, Sukhon 51 Weinländer, Kristina 24<br />

Sun, Ren 25, 34, P14 Wen, Kwun Wah 1<br />

Suzuki, Tohru P1, P11 West, John 11, 31<br />

Syrjäkari, Henna 3 Westrop, Samantha Jane 12<br />

Taddeo, A. P5 Whitby, Denise 26, 27, 51, 54<br />

Taipale, Jussi 36 Whitehouse, Adrian 39, 41<br />

Tan, Xiao-Hua 29, 52 Wies, Effi 56<br />

Tanese, Naoko 7 Wilson, Angus 7, 64<br />

Taylor, Adam 41 Wilson, Sam 9<br />

Tedeschi, Rosamaria 43, P15 Wojcicki, Janet 53<br />

Thome, Margot 24 Wong, Ping-Pui 12<br />

Tepper, Cliff G. P9 Wong, Scott W. 14<br />

Thomas, Jaiye O. 51 Wood, Charles 53, 55<br />

132


The 11 th International Workshop on KSHV & Related Agents, Birmingham, UK<br />

Wu, Min-Fen P12 Zeng, Yan 29, 52<br />

Wu, Yu-Hsuan P12 Zhang, Hui 29<br />

Xian, Ling-ling 29 Zhang, Rui 22<br />

Xie, Jian-Xin 29, 52 Zhang, Xuefeng 17<br />

Yang, Lei 29, 52 Zheng, Zhi-Ming 40, 65<br />

Yarchoan, Robert 54 Zhou, Xiao–Fei 29<br />

Yu, Fuqu P14 Zhu, Wen Qui 21<br />

Yuan, Yan 35<br />

133

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!