28.04.2015 Views

View - Blood

View - Blood

View - Blood

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

~~<br />

Loss of Attachment to Fibronectin With Terminal Human Erythroid Differentiation<br />

By M.H. Vuillet-Gaugler, J. Breton-Gorius, W. Vainchenker, J. Guichard, C. Leroy, G. Tchernia, and L. Coulombel<br />

Human erythroblastic progenitors (colony-forming uniterythroid<br />

[CFU-E] and burst-forming unit-erythroid [BFU-<br />

E]) have been shown to attach to fibronectin (Fn), a<br />

property that might be involved in the local regulation of<br />

erythropoiesis. In this study, we have investigated changes<br />

in cell attachment to Fn upon terminal erythroid differentiation.<br />

We first purified CFU-E from human marrow by<br />

avidin-biotin immune rosetting. This negative selection<br />

procedure yielded a cell population containing -80% blasts<br />

that, after characterization by colony-assays and electron<br />

microscopy, appeared to consist of CFU-E (10% to 15%)<br />

and their immediate progeny (85% to 90%). here defined as<br />

“preproerythroblasts.” In the presence of erythropoietin,<br />

purified cells differentiated into reticulocytes in 7 to 10<br />

days. Cell attachment to Fn was inversely correlated to the<br />

N MANY TISSUES acquisition of differentiated func-<br />

I tions is induced by either cell-cell or cell-matrix<br />

interactions.’.* In the bone marrow also, hematopoietic stem<br />

cells depend for their differentiation upon interactions with a<br />

complex environment composed of nonhematopoietic stromal<br />

cells and their extracellular matrix (ECM).3-6 However,<br />

there is little known about the molecular mechanisms involved<br />

in these interactions. Recently attention has been<br />

focused on the role of growth factors that are synthesized by<br />

stromal cells and act either in a soluble or bound to<br />

the cell-surface9 or to ECM components.” However, most of<br />

these studies have been performed with fibroblasts and<br />

endothelial cells from extramedullary sources that may not<br />

help to elucidate lineage- or differentiation-specific interactions<br />

between hematopoietic and stromal cells in the marrow.<br />

These may be accounted for by multiple specialized marrow<br />

stromal cells. Alternatively, hematopoietic cells themselves<br />

may modulate, as differentiation proceeds, the expression on<br />

their surface of receptors involved in stromal cell or matrix<br />

recognition. Likely candidate molecules for such a mechanism<br />

are integrins.” These glycoproteins exist as heterodimers<br />

of noncovalently associated LY and @ transmembrane<br />

subunits and are widely distributed. Three different @<br />

chains exist, which define three integrin families.” In each<br />

family, @ associates with multiple a chains, resulting in<br />

different ligand specificities. Thus, the al-6 81 family, or<br />

VLA family, includes most of the cell surface receptors for<br />

Fn and ECM proteins.”.’*<br />

There are at least two arguments to implicate integrins<br />

and cell-matrix adhesion in the regulation of hematopoietic<br />

differentiation. First, the ECM produced by marrow stromal<br />

cells has been shown to contribute to the function of the<br />

adherent layer in long-term marrow culture^.'^^'^ Second,<br />

we’’ and ~thersl~.’~ have demonstrated direct adhesion of<br />

progenitor cells to ECM proteins. Thus, colony-forming<br />

units-erythroid (CFU-E) and some burst forming unitserythroid<br />

(BFU-E) show significant attachment to Fn,”.I6<br />

whereas CFU granulocyte-macrophage (GM) interact with<br />

a different ECM component.” Furthermore, in the erythroblastic<br />

lineage, adhesion to Fn is a differentiation-regulated<br />

event, as shown by the higher capacity of CFU-E than<br />

BFU-E to bind Fn,” and by the loss of Fn-adhesion and of<br />

stage of differentiation of the erythroid cell: more than 50%<br />

of the CFU-E population reproducibly adhered to Fn,<br />

whereas at most 30% of the preproerythroblasts had the<br />

same capacity. Adhesion was further lost at late maturation<br />

stages, and a constant finding was the inability of<br />

reticulocytes to adhere to Fn. Finally, CFU-E adhesion to Fn<br />

was blocked by polyclonal IgG raised against the Fn<br />

receptor and by a monoclonal antibody against VLA-I.<br />

These results demonstrate that adhesion to Fn is developmentally<br />

regulated during normal human erythropoiesis.<br />

Restriction of its expression to CFU-E and its first divisions<br />

strikingly correlates with the migratory capacity of these<br />

cells.<br />

0 1990 by The American Society of Hematology.<br />

the Fn receptor in murine erythroleukemic cells when induced<br />

by dimethyl sulfoxide<br />

In the present study, we have investigated how adhesion to<br />

Fn is modulated during terminal erythroid differentiation.<br />

For this purpose, we developed a procedure for obtaining a<br />

highly enriched population of very immature erythroid cells,<br />

ie, CFU-E and their immediate progeny. These purified cells<br />

were then induced to differentiate in vitro, and during this<br />

process, we investigated their changing ability to attach to<br />

Fn. We found that CFU-E attach strongly to Fn, and that<br />

this property is lost at specific stages of terminal erythroid<br />

maturation. Moreover, the binding of CFU-E to Fn is<br />

mediated by a receptor bearing some similarities to the<br />

integrin family.<br />

MATERIALS AND METHODS<br />

Bone marrow samples. Normal marrow samples were obtained<br />

after informed consent from hematologically normal patients undergoing<br />

hip surgery. Cells were flushed from the bone fragments with a<br />

medium containing 100 to 200 pg/mL DNAse (type I, Sigma<br />

Chemical Co, St Louis, MO). Cells were separated on Ficoll-<br />

Hypaque (1.077 g/mL) (Eurobio, Paris, France). Light-density<br />

mononuclear cells were washed twice, resuspended at a concentration<br />

of 3 x lo6 cells/mL in a-medium supplemented with 10% fetal<br />

From the Hematology Laboratory and University Paris XI,<br />

H6pitaI BicBtre, Kremlin-BicZtre: and INSERM U.91, H6pital<br />

Henri-Mondor, CrLteil, France.<br />

Submitted April 24,1989; accepted October 24. 1989.<br />

Supported by Grants No. CRE 862007 from INSERM (L.C.);<br />

Grant No. ARC 6532 from the Association pour la Recherche<br />

contre le Cancer; and a grant from Ligue contre le Cancer (Comitt<br />

Dkpartemental des Hauls-de-Seine). M.H. K-G. is the recipient of a<br />

fellowship from the French Ministere de I’lndustrie et de la<br />

Recherche.<br />

Address reprint requests to Laure Coulombel, MD, PhD, Institut<br />

de Pathologie Cellulaire, H6pital Bici?tre, 94275 Kremlin-Bici?tre,<br />

France.<br />

The publication costs of this article were defrayed in part by page<br />

charge payment. This article must therefore be hereby marked<br />

“advertisement” in accordance with 18 U.S.C. section 1734 solely to<br />

indicate this fact.<br />

6 1990 by The American Society of Hematology.<br />

0006-4971/90/7504-0028$3.00/0<br />

<strong>Blood</strong>, Vol 75, No 4 (February 15), 1990: pp 865-873 865


866 VUILLET-GAUGLER ET AL<br />

calf serum (FCS) (Flow Laboratories, Paris, France), and incubated<br />

in 75 cm’ flasks for 1 hour at 37OC in a 5% CO, atmosphere in air to<br />

remove plastic-adherent cells. The nonadherent mononuclear cells<br />

(NA-MNC) were subsequently fractionated by immune rosetting<br />

(see below).<br />

Antibodies. Monoclonal antibodies (MoAbs) against human<br />

HLA-DR (OKIal), human T cells (OKT3), and human CALLA<br />

(CD IO; OKB-CALLA) were purchased from Ortho Diagnostic<br />

Systems (Roissy, France). MoAb recognizing HPCA (CD34) was<br />

from Becton Dickinson (Mountain <strong>View</strong>, CA), BI (anti-B cells)<br />

from Coulter Immunology (Hialeah, FL). MoAbs CH3 (IgM<br />

recognizing granulocytes and CFU-GM), CS4 (recognizing glycophorin<br />

A) and CK26 (binding to lymphoid and monocytic cells) have<br />

been previously described,20 and were generously provided by Dr<br />

Berthier (INSERM U.217, Grenoble, France). Anti-human HLA-<br />

DR K5 MoAb was a gift from Dr Fermand (INSERM U108,<br />

Hdpital St Louis, France). FA6-152 MoAb,,’ which recognizes an<br />

88 Kd glycoprotein on platelets, monocytes, and erythroid cells and<br />

belongs to the CD36, was a gift from L. Edelman (Institut Pasteur,<br />

Paris, France). Anti-platelet GPIIIa C17 MoAb and anti-glycophorin<br />

A (GPA) plyclonal antibody used in electron microscopy<br />

were kind gifts from Drs F. Tetteroo22 and L.C. Ander~son.~’<br />

Biotin-conjugated affinity-purified goat anti-mouse IgM (p specific)<br />

and IgG (H -t L) F(ab’)2 fragments were obtained from Cappel-<br />

Cooper Biomedical (Malvern, PA). A rabbit antiserum prepared<br />

against the purified human placental fibronectin receptor (HFnR-<br />

Ab) was purchased from Telios Pharmaceuticals (La Jolla, CA).<br />

This antibody recognizes mainly the 115 Kd 81 subunit of the<br />

receptor, and is less reactive with the 160 Kd a5 subunit.24 A rat<br />

MoAb BlE5, which is directed against the a5 subunit of the human<br />

Fn receptor and which selectively inhibits cell attachment to Fn was<br />

kindly provided by C. Damsky (University of California, San<br />

Francisco, CA) and used as hybridoma s~pernatant.~’<br />

Enrichment of erythroblastic cells by immune rosetting. Cell<br />

purification was performed by negative selection using avidin-biotin<br />

immune rosetting according to the original technique described by<br />

Fiirfang and Thierfelder.26 Briefly, 1.2 mg avidin (Vector Laboratories<br />

Inc, Burlingame, CA) was covalently coupled to 2 x lo9<br />

glutaraldehyde-fixed sheep red blood cells (SRBC-A) as described in<br />

detail elsewhere.26 SRBC-A can be kept at 4OC in sterile conditions<br />

for at least 4 to 6 months without loss of reactivity. The purification<br />

was performed in two steps: NA-MNC (usually 0.5 to 3 x IO8 cells)<br />

were first mixed with a 10-fold excess of SRBC-A. The mixture was<br />

centrifuged 6 minutes at 200g, and the pellet was incubated 45<br />

minutes at 4OC. Cells were then gently resuspended at a concentration<br />

of 1 x lo7 cells/mL in a-medium supplemented with 5% FCS<br />

((~5%) and layered over Ficoll-Hypaque. Mature granulocytes that<br />

had phagocytized SRBC-A were eliminated in the pellet (fraction<br />

P-1). Cells harvested at the interface (fraction 1-1) were washed<br />

twice, resuspended at a concentration of 4 x lo7 cells/mL, and<br />

incubated 45 minutes at 4OC with an excess of the following MoAbs:<br />

OKIal, OKT3, OKB-CALLA, B1, anti-HPCA, K5, CH3, CS4,<br />

CK26. At the end of the incubation period, cells were washed twice,<br />

resuspended at a concentration of 4 x lo7 cells/mL in a5% and<br />

incubated with a 1/40 dilution of a mixture of biotin-conjugated<br />

goat anti-mouse IgM and IgG for 45 minutes at 4°C. Excess<br />

biotinylated immunoglobulin was removed by three washes in 015%.<br />

Finally, labeled cells were incubated with SRBC-A in the proportion<br />

of l:lO, as described above, and rosettes were allowed to form. Cells<br />

were then gently resuspended and adjusted to the initial concentration.<br />

Rosettes were removed by centrifugation over Ficoll-Hypaque<br />

(fraction P-2), and unlabeled cells were recovered at the interface<br />

layer (fraction 1-2). Cells recovered in each fraction (1-1, P-I, 1-2,<br />

P-2) were counted and the number of rosettes was determined.<br />

Purified cells (1-2) were characterized morphologically on May-<br />

Griinwald-Giemsa (MGG) stained cytocentrifuged slides, by electron<br />

microscopy (see below), and by their ability to form colonies in<br />

methylcellulose colony-assays.<br />

Electron microscopy. The simultaneous detection of specific<br />

membrane markers (GPIIIa and GPA) by immunogold and intracellular<br />

peroxidatic activities was determined on the purified erythroid<br />

cell population (1-2) as previously described in detaiL2’ Briefly,<br />

immunolabeling was first performed by incubating cells with specific<br />

antibodies recognizing either GPIIIa or GPA, and then with a<br />

second antibody coupled with 15 nm and 5 nm gold particles,<br />

respectively. Cells were then fixed in 1.25% glutaraldehyde in Gey’s<br />

buffer, washed, incubated in diaminobenzidine medium,28 post-fixed<br />

with osmium tetroxide, dehydrated, and embedded in Epn. Sections<br />

were examined with a Philips electron microscope CM 10 (Eindhoven,<br />

The Netherlands) without staining in order to easily identify<br />

ferritin molecules by their own contrast.<br />

Suspension culture of purijed erythroblastic cells. Purified<br />

cells from the 1-2 fraction were cultured at a concentration of 1 x lo6<br />

cells/mL in a-medium containing 20% FCS and 3 U/mL human<br />

urinary erythropoietin (Ep). The batch of FCS used during the study<br />

contained less than 10 pg of Fn per milliliter as measured by an<br />

enzyme-linked immunosorbent assay (ELISA). Cultures were initiated<br />

in 96-flat bottom microtiter plate wells (Falcon, Becton-<br />

Dickinson, France, No. 3072; 200 pL per well) and incubated at<br />

37OC in a water saturated air atmosphere with 5% CO,. Cells were<br />

harvested regularly, and their number, viability (dye exclusion),<br />

morphology, plating efficiency, and ability to adhere to Fn were<br />

determined. Three stages of maturation were defined after MGG<br />

staining: immature (proerythroblasts and early basophilic erythroblasts),<br />

intermediate (late basophilic and polychromatophilic), and<br />

mature (acidophilic and enucleations). Reticulocytes were counted<br />

separately .<br />

Hematopoietic progenitor cell colony-assays. Cells from the<br />

P-1, 1-1, P-2, and 1-2 fractions were plated in a-medium containing<br />

0.8% methylcellulose, 30% FCS, 1% deionized bovine serum albumin<br />

(BSA), mol/L 8-mercaptoethanol, 3 U/mL Ep and 10%<br />

agar leukocyte conditioned medium. Final concentration of cells per<br />

dish was 1.5 x IO’ for NA-MNC and P-1 and 1 to 3 x lo4 for 1-1,<br />

1-2, and P-2. Erythroid colonies were scored as previously described<br />

in detail7<br />

Adhesion assay. The capacity of erythroid cells to adhere onto<br />

Fn was assessed in two different ways: purified 1-2 cells and cells<br />

grown during various periods of time in suspension (see above) were<br />

incubated on Fn-coated 2.5 cm2 tissue culture wells (2 to 4 x 10’<br />

cells per well) in (Y 5% for 2 hours at 37OC in a 5% CO, atmosphere.<br />

Nonadherent cells were then harvested, weakly adherent cells were<br />

removed by two mild washings, and strongly adherent cells were<br />

detached by vigorous pipetting or short trypsinization. Adherent and<br />

nonadherent cells were counted, stained with MGG for morphologic<br />

examination, and plated in methylcellulose colony-assays at 1.5 x<br />

lo4 cells per dish to assess the number of CFU-E in each fraction. In<br />

the second method, cells were grown as described above for suspension<br />

culture, except that they were incubated from the start on<br />

Fn-coated 96-flat bottom microtiter plate wells (1 x lo6 cells/mL,<br />

200 pL per well) and kept on that substrate for the whole culture<br />

period. Every 2 days during the 6 to 10 day-culture period, one well<br />

was sacrificed and nonadherent and adherent cells were harvested<br />

separately, counted, and stained with MGG. Cell viability of both<br />

fractions was checked by dye exclusion (Nigrosin). In both methods,<br />

tissue-culture wells were precoated with 50 to 80 pg/mL of Fn<br />

purified from human plasma by gelatin affinity chromatography, or<br />

human collagens I, 111, and IV in the monomeric form or laminin.<br />

Nonspecific binding sites were blocked by 1-hour incubation in 10<br />

mg/mL BSA.”


I<br />

ERYTHROBLAST ADHESION TO FIBRONECTIN 867<br />

L A<br />

- {<br />

A<br />

Fig 1. Photomicrographs of MGG-stained 1-2 cells before culture (A), and after 7 days (6). and 10 days (C), in suspension culture in the<br />

presence of Ep. There was no difference in the morphology of cells grown in suspension or on an Fn 8ubstrate. (Original<br />

magnification, xs00)<br />

In some experiments, 1-2 cells were first incubated (45 minutes at<br />

4OC) with a 1/10 dilution of the HFnR-Ab or undiluted BlE5<br />

hybridoma supernatant, and then incubated as described above onto<br />

Fn-coated wells for 2 hours in the continuous presence of the<br />

antibody. Negative controls included incubation of cells either with<br />

nonimmune rabbit IgG or with anti-CD36 (FA6-152) or anti-GPA<br />

hybridoma supernatant (CS4).<br />

Indirect immuncfluoresence labeling. Purified erythroblasts were<br />

stained with the HFnR-Ab by indirect immunofluorescence. For<br />

that purpose, 0.5 to 1 x lo6 cells in 100 pL were incubated 30<br />

minutes at 4% with a 1/10 dilution of the HFnR-Ab or control<br />

nonimmune rabbit IgG. Cells were subsequently washed twice and<br />

resuspended in a 1 /50 dilution of fluorescein-labeled goat anti-rabbit<br />

IgG F(ab')2. Incubation was continued for 30 minutes at 4OC. Cells<br />

were washed twice, resuspended at a final concentration of 1 x IO6<br />

cells/mL in phosphate buffered saline (PBS) containing 1% BSA<br />

and examined under a Zeiss microscope equipped for epifluorescence.<br />

RESULTS<br />

Purification to homogeneity of very immature human<br />

marrow erythroblasts. The purification procedure de-<br />

scribed under Materials and Methods yielded in 20 experiments<br />

a population (1-2) containing 79% f 3% of blasts that<br />

were HLA-DR, CD34, and GPA negative by indirect immunofluorescence<br />

(Fig 1). Positive labeling with the FA6-152<br />

MoAb whose expression is restricted to platelets, monocytes,<br />

and erythroid cells suggested the erythroid origin of these<br />

~elIs.2~<br />

The erythroid origin of purified 1-2 cells was clearly<br />

demonstrated by the 1-2 cells' ultrastructural characteristics<br />

and immunophenotype. Eighty percent of 1-2 cells were<br />

classified as erythroid and included only two maturation<br />

stages (Fig 2): the most immature phenotype represented<br />

10% to 15% of this cell fraction. Despite the absence of GPA<br />

and rhopheocytosis, the population was recognized as erythroid<br />

by the presence of Theta granules (8-Gr) and of a few<br />

ferritin molecules located in the 8-Gr and scattered through<br />

the cytoplasm (Fig 2A, B). These cells were presumably<br />

CFU-E, since the 1-2 cell fraction contained the same<br />

proportion of progenitors giving rise to 1-2 hemoglobinized<br />

clusters (see below). Other erythroid cells in the 1-2 fraction<br />

were more mature, as shown by the presence of GPA (Fig<br />

Fig 5. Photomicrographs of<br />

MGG-stained adherent (A) and<br />

nonadherent (6) erythroblasts<br />

observed in a typical experiment<br />

where purified 1-2 cells<br />

were cultured in Fn-coated<br />

wells for 7 days.<br />

t<br />

A


868 VUILLET-GAUGLER ET AL<br />

.<br />

Fig 2. Ultrastructural analysis<br />

of purified 1-2 cells. (A) Pre<br />

sumptive CFU-E. (inset left) At<br />

low magnification (original magnification,<br />

~3,300) a very large<br />

nucleolus is saen in tho nucleus.<br />

Cytoplasmic vacuoles and<br />

granules are clustered in the<br />

golgi zone. At higher magnification<br />

(original magnification,<br />

x 52.600). the structura of 0-Gr<br />

is clearly distinguished. In all of<br />

them, a few ferritin molecules<br />

are identified. There are no gold<br />

particles on the plasma mambrane,<br />

indicating the absence<br />

of GPA. (inset right) High magnification<br />

(original, x 108,300)<br />

of a 0-Gr with its dense rod and<br />

some dense periodic structure.<br />

The typical configuration of ferritin<br />

molecules (f) is recognized<br />

in the cytoplasm and in 0-Gr.<br />

(E) Preproerythroblast. Weak<br />

labaling for GPA is detected at<br />

one place of the cell membrane<br />

(arrow). Two rhopheocytosis invaginations<br />

(r), also called<br />

coated pits, are indicated. In<br />

one of them, ferritin molecules<br />

(f) are presant. Nota also the<br />

presence of intracytoplasmic<br />

free ferritin (f)(original magnification,<br />

~68,800). (C) A more<br />

matura preproarythroblast as<br />

judged by the heavier labeling<br />

(arrow) by GPA-Ab and by the<br />

increased number of free ferritin<br />

molecules (1). and ferritin<br />

molecules associated with theta<br />

granules 0-Gr (original magnification.<br />

~58,800).<br />

2B). However, GPA density was below the threshold of<br />

sensitivity expected for conventional immunofluorescence or<br />

avidin-biotin labeling, which explains why these cells were<br />

not eliminated by immune rosetting. One or 2 sites of<br />

rhopheocytosis were observed (Fig 2B), and the concentration<br />

of ferritin in the 0-Gr and in the cytoplasm was increased<br />

in comparison with the more immature cdls (Fig 2C).<br />

Hemoglobin was absent, as judged by the negative dense<br />

peroxidase reaction. These properties, together with the low<br />

expression of rhopheocytosis and GPA, indicated that this<br />

population was less differentiated than the classical<br />

proerythroblast,*’ and we have therefore designated these<br />

cells “preproerythroblasts” as they appear to represent a<br />

stage intermediate between CFU-E and proerythroblasts.<br />

The 1-2 fraction contained on average 65% preproerythroblasts.<br />

Twenty to twenty-five percent of 1-2 cells were<br />

nonerythroid, as recognized by electron microscopy. Most of<br />

these belong to the megakaryocytic lineage, and a minority<br />

were mast cells, basophil myelocytes, and promyelocytes<br />

(data not shown).<br />

Clonogenic properties of purified 1-2 cells were assessed in<br />

methylcellulose colony-assays (Table 1). If we define a<br />

CFU-E as a progenitor giving rise to a cluster of at least 10 to<br />

15 hemoglobinized cells at day 9 to 10, 11% * 1% of 1-2 cells<br />

were CFU-E (n = 20), whereas preproerythroblasts variably<br />

gave rise to clusters of less than 10 cells that usually had<br />

lysed by day 9. The proportion of CFU-E matched the<br />

proportion of very early GPA negative erythroid cells recognized<br />

by electron microscopy. CFU-E enrichment was 36 2<br />

4-fold relative to NA-MNC and over 50 to 60-fold compared<br />

with native marrow. The enrichment of preproerythroblasts<br />

could not be calculated, as these cells cannot be accurately<br />

quantitated in unpurified marrow cells either by morphologic<br />

or colony-assays criteria. Recovery of CFU-E in the 1-2


ERYTHROBLAST ADHESION TO FIBRONECTIN 869<br />

Table 1. Progenitor Cell Enrichment During the Different Steps of the Purification Procedure<br />

Fraction<br />

NA-MNC<br />

I- 1<br />

P- 1<br />

1-2<br />

P-2<br />

No. Progenitors/l.B x lo5 Cells Plated<br />

CFU-E mBFU-E IBFU-E CFU-GM<br />

490 f 60<br />

1,609 k 314<br />

(3)<br />

62 f 16<br />

17,571 k 1,658<br />

(36)<br />

586 k 123<br />

155 f 24 12 f 2 328 f 54<br />

427 f 57 43 f 7 1,308 f 113<br />

(2.7) (3.5) (4)<br />

27 k 13 924 90 k 26<br />

1,262 f 276 26 f 8 898 f 286<br />

(8) (2) (2.7)<br />

634 f 74 57 f 12 1,316 f 263<br />

Each number represents the mean f SEM of seven different experiments. Numbers in brackets represent the enrichment factor relative to NA-MNC in<br />

the 1-1 and 1-2 fractions.<br />

Abbreviations: mBFU-E, mature BFU-E; iBFU-E, immature BFU-E.<br />

fraction was 41% 3%. In seven experiments where the<br />

different fractions obtained during the purification were<br />

cloned in methylcellulose, 10% CFU-E were found in the P-2<br />

fraction (Table 1). The latter may represent a subset of<br />

HLA-DR+, CD34+ CFU-E. Our results also showed that<br />

contamination by other progenitor cells was negligible since<br />

in 20 experiments, BFU-E and CFU-GM represented 0.7%<br />

and 0.6% of 1-2 cells, respectively (Table 1).<br />

In order to assess the proliferative and differentiative<br />

behavior of the purified 1-2 cells in vitro, we cultured them in<br />

suspension in the presence of 3 U/mL Ep (but no other<br />

hematopoietic growth factors). For the first 2 to 3 days, there<br />

was no net change in the number of cells, whereas during the<br />

next 7 to 9 days, there was a four- to ninefold increase over<br />

the initial number of cells, reflecting the proliferation of the<br />

maturing erythroblasts (Fig 3). Morphologic examination<br />

showed that 1-2 cells differentiated synchronously up to the<br />

enucleation stage, with the production of many reticulocytes<br />

(Fig 1). In the absence of Ep, 1-2 cells did not survive beyond<br />

48 hours. Nonerythroid cells were not maintained in these<br />

cultures.<br />

Adhesion of purified erythroblasts to Fn during their<br />

diferentiation. CFU-E and preproerythroblasts adhesion<br />

to Fn was evaluated in 19 separate experiments using 1-2<br />

cells. The proportion of Fn-adherent CFU-E was precisely<br />

quantitated by colony-assays of adherent and nonadherent<br />

cells and that of Fn-adherent preproerythroblasts was estimated<br />

from the proportion of total Fn-adherent 1-2 cells. The<br />

mean (+SEM) proportion of Fn-adherent CFU-E was<br />

58% f 5%. It was over 65% in 10 of 19 experiments, and<br />

below 30% in only three experiments. In contrast to the high<br />

proportion of adherent CFU-E, only 31% 3% of total<br />

nucleated 1-2 cells were Fn-adherent. As CFU-E represents<br />

10% to 15% of the 1-2 population, and preproerythroblasts<br />

65%, this gives a proportion of Fn-adherent preproerythroblasts<br />

below 25%. This difference in the ability of CFU-E<br />

and preproerythroblasts to attach to Fn was seen in each<br />

experiment, and suggests loss of adhesion to Fn as CFU-E<br />

matures in preproerythroblasts. No significant attachment of<br />

CFU-E and preproerythroblasts occurred onto either human<br />

collagen I, 111, IV or laminin.<br />

The effect of differentiation on the adherence to Fn was<br />

next investigated: seven experiments were performed using<br />

cells removed after varying periods of time from 7 to 10<br />

day-cultures of highly CFU-E-enriched population, and six<br />

experiments were performed by culturing cells 7 to 10 days<br />

on an Fn substrate. Results from these 11 experiments have<br />

been pooled in Fig 4. In each experiment, adhesion to Fn<br />

decreased with maturation and was completely lost at late<br />

maturation stages. Even though the rate of decline varied<br />

between experiments, the pattern observed was similar in<br />

each experiment, ie, in the purified 1-2 population before<br />

culture (day 0), the proportion of adherent CFU-E was<br />

higher (58%) than that of total purified cells (30%) and thus<br />

preproerythroblasts, and slowly declined over the next 8 days<br />

in culture to reach 20% at day 8 to 9 (polychromatophilic<br />

stage). At that stage, the capacity to adhere to Fn appeared<br />

to rapidly decline and was completely lost at the enucleation<br />

- 10-<br />

E<br />

\ 9-<br />

(D<br />

-; 8-<br />

z 7-<br />

6-<br />

LL<br />

s 5-<br />

W<br />

z 4-<br />

0<br />

3-<br />

W 2 2-<br />

1 1 1 1 1 1 1 1 1 1 ,<br />

1 2 3 4 5 6 7 8 9 10 11<br />

DAYS IN CULTURE<br />

2 Imm 81 87 30 3 0<br />

0 0 60 59 22<br />

Mat 0 0 10 38 78<br />

2 1 inter<br />

c<br />

Fig 3. Changes in the number and cytology of cells present<br />

after varying periods of time when purified 1-2 cells were cultured<br />

in suspension in the presence of Ep. Each point represents the<br />

mean f SEM of the cell concentration observed in three to seven<br />

experiments. The SEM is not indicated for the day 10 point since<br />

only two experiments were available. Morphologic examination of<br />

the cells was performed after MGG staining at the indicated day<br />

(see Materials and Methods).<br />

1


870 VUILLET-GAUGLER ET AL<br />

adhere to Fn was paralleled by the disappearance of a<br />

cell-surface receptor (data not shown).<br />

2<br />

W<br />

V<br />

5 40-<br />

W<br />

K W<br />

8 30-<br />

a<br />

I-<br />

Z<br />

W<br />

p 20- Purified 1-2<br />

W 0<br />

10 - cells<br />

DAYS IN CULTURE<br />

Fig 4. Loss of adhesion to Fn with terminal erythroid differentiation.<br />

The percentage of adherent cells and CFU-E was first<br />

determined in the 1-2 population at day 0. Cells were then cultured<br />

either in suspension (n = 7) or on Fn-coated wells (n = 6). and<br />

the proportion of adherent cells was measured sequentially as<br />

described in the text. Each point represents the mean c SEM of<br />

the percent adherent cells. 1-2 cells differentiated synchronously<br />

up to day 7. However, beyond that point, the population was<br />

heterogeneous, and reticulocytes never represented 100% of the<br />

cell population. Therefore, the day 11 point and the dashed line<br />

represent the percent adhesion expected if 100% of day 7 cells<br />

had matured into reticulocytes.<br />

stage. Reticulocytes were always found exclusively in the<br />

nonadherent fraction. This was best appreciated in experiments<br />

in which purified 1-2 cells were cultured on Fn during<br />

several days in the presence of Ep. In a representative<br />

experiment, shown in Fig 5, 20% to 30% of the day 7 cells<br />

were still strongly adherent to Fn. Most of these adherent<br />

cells had a phenotype of polychromatophilic erythroblasts,<br />

and no reticulocytes were seen (Fig 5). However, some<br />

reticulocytes were already present in this culture, but they<br />

had been released from the Fn substrate and represented<br />

25% of the nonadherent population (Fig 5). The same<br />

partitioning was seen for enucleating cells. Failure of enucleating<br />

erythroblasts and reticulocytes produced in culture<br />

(kFn) to attach to Fn was not attributable to any change in<br />

their viability as assessed by dye exclusion. Furthermore,<br />

assessment of enriched reticulocytes, freshly isolated from<br />

the peripheral blood from three patients with hemolytic<br />

anemia showed that these also did not attach to Fn in our<br />

assay (data not shown).<br />

Effect of anti-HFnR antibodies on the adhesion of purified<br />

erythroblasts to Fn. In seven separate experiments,<br />

CFU-E and preproerythroblasts (1-2 cells) were first preincubated<br />

with either a 1/10 dilution of polyclonal IgG against<br />

HFnR or with undiluted B1E5 hybridoma supernatant and<br />

then incubated in Fn-coated wells. As shown in Fig 6, both<br />

antibodies specifically inhibited CFU-E and 1-2 cells (ie,<br />

preproerythroblast) adhesion to Fn, as nonimmune rabbit<br />

IgG, anti-CD 36 (FA6-152), and anti-GPA had no effect.<br />

HFnR-Ab labeled 100% of erythroid 1-2 cells by indirect<br />

immunofluorescence, whereas enucleating cells and reticulocytes<br />

stained negatively, suggesting that the disappearance<br />

of the functional capacity of differentiating erythroblasts to<br />

DISCUSSION<br />

One of the major questions regarding hematopoiesis is the<br />

understanding of the mechanisms whereby progenitor cells<br />

interact with their marrow environment. Our recent<br />

observationls that human marrow CFU-E and mature BFU-<br />

E selectively attach to ECM prepared from marrow fibroblasts<br />

and to purified Fn suggests that progenitor cellsmatrix<br />

interactions might represent one such mechanism. In<br />

the present study, we have demonstrated that attachment to<br />

Fn is a property of most CFU-E and early erythroblasts in<br />

normal human marrow, and that this property is subsequently<br />

lost upon terminal erythroid differentiation.<br />

We have approached this question by designing an in vitro<br />

model that reproduces the successive steps followed by a<br />

CFU-E when it differentiates into reticulocytes. Our strategy<br />

was first to purify human CFU-E directly from fresh marrow<br />

samples. Negative selection by avidin-biotin immune rosetting<br />

reproducibly yielded a homogenous population of very<br />

immature cells identified as erythroid by their positive<br />

labeling with FA6-1 52,29 and by ultrastructural criteria. The<br />

procedure described was nontoxic, and more powerful and<br />

reliable in our hands than panning.30s31 Moreover, large<br />

numbers of marrow cells could be easily processed within a<br />

few hours with minimal cell loss, in contrast to cell sorting.'*<br />

The availability of a highly enriched population of very<br />

immature normal erythroblasts gave us the opportunity to<br />

delineate the ultrastructural changes that are associated with<br />

human erythroid cell differentiation. Previous information<br />

has relied on analyses of cells from patients with erythroblastic<br />

leukemia or of partially purified populations of normal<br />

CFU-E.31 The earliest markers detected in 1-2 cells were<br />

0-Gr and ferritin molecules (Fig 2). Most likely, cells that<br />

exhibited these two features only represent CFU-E. This<br />

4 60<br />

W<br />

I<br />

5 30<br />

g 20<br />

: 10<br />

a.<br />

CFU-E<br />

T<br />

T<br />

PURIFIED CELLS<br />

Fig 6. Inhibition of CFU-E and 1-2 cell attachment to Fn by<br />

anti-HFnR antibodies. 1-2 cells were incubated 2 hours on Fncoated<br />

wells after preincubation with either polyclonal IgG against<br />

the human Fn receptor (a5Dl; ) or B1E5 MoAb against the a5<br />

chain (W). Controls included preincubation of the cells with no<br />

antibody (0). nonimmune rabbit IgG (&I, or with hybridoma<br />

supernatant from an unrelated erythroid-specific antibody (B).<br />

Histograms show the mean * SEM of the percentage of adherent<br />

cells (n = 6).


ERYTHROBLAST ADHESION TO FIBRONECTIN 87 1<br />

conclusion is based on our finding that this morphology<br />

identified the same proportion of cells in the purified population<br />

as were detected by colony-assay as CFU-E. Surprisingly,<br />

these cells did not express platelet-peroxidase-like<br />

activity previously described in presumptive CFU-E.27,31<br />

However, after a few hours in culture, this activity was<br />

recovered (data not shown), suggesting its possible inactivation<br />

by the purification procedure. Cell-surface GPA and<br />

images of rhopheocytosis were seen on the other 1-2 erythroid<br />

cells, but at a much lower density than what has been<br />

described for proerythr~blasts.~~.~~ These cells, or “preproerythroblasts,”<br />

therefore appear to represent an intermediate<br />

stage between CFU-E and proerythroblasts.<br />

Human CFU-E have been purified only indire~tly~’.~~~~~<br />

through the proliferation of partially purified blood or<br />

marrow BFU-E cultured 7 to 9 days either in liquid<br />

su~pension,~~ or in semisolid assay” in the presence of Ep.<br />

This approach also yielded 80% pure populations of very<br />

immature erythroblasts containing up to 50% CFU-E. However,<br />

in contrast to cells directly purified from marrow by<br />

avidin-biotin immune rosetting, which appear homogeneous<br />

with respect to their stage of maturation, BFU-E-derived<br />

cells are more heterogeneous and include many hemoglobin-<br />

ized proerythrobla~ts.~~ CFU-E and early erythroblasts have<br />

also been purified from either thiamphenicol-treated,36 or<br />

Friend virus-infected mice spleen cells,37 but it is difficult to<br />

compare them with human cells.<br />

We have previously shown that the primitive BFU-E,<br />

which give rise to multiclustered colonies, do not attach to<br />

Fn, whereas 30% of CFU-E, which represents the most<br />

mature type of progenitor, express this property.” In this<br />

study, we assessed the attachment to Fn of purified CFU-E,<br />

preproerythroblasts, and their progeny, generated in cultures<br />

that support their terminal erythroid differentiation. Interestingly,<br />

the majority of CFU-E recovered in the purified<br />

population selected for these studies showed a higher proportion<br />

of Fn-adhering members. Although a direct comparison<br />

of Fn-adherence for purified and unpurified CFU-E was not<br />

undertaken, it is possible that cells defined as CFU-E by<br />

colony-assays are heterogeneous with respect to their Fnadherence<br />

properties, and that our purification procedure<br />

isolated a subset of CFU-E with higher Fn-adherence potential.<br />

Alternatively, it is possible that the adhesion assay may<br />

give different absolute results according to the composition of<br />

the population being tested. What the present studies have<br />

revealed is that adherence to Fn also clearly changes during<br />

the later stages of erythroid cell differentiation: (1) CFU-E<br />

attached to Fn in the highest proportion, close to 60% (Fig 4),<br />

(2) the proportion of adherent preproerythroblasts was<br />

consistently half that observed with the copurified CFU-E,<br />

and (3) enucleating erythroblasts and reticulocytes derived<br />

from these immature erythroblasts were unable to attach to<br />

Fn. These data demonstrate that the Fn-adhesion property is<br />

lost in differentiating erythroblasts in two sequential stages:<br />

an initial decline during the transition from CFU-E to<br />

preproerythroblasts, with a subsequent loss of all remaining<br />

adhesive properties at the time of enucleation.<br />

Expression of Fn-adhesion during human erythroid differentiation<br />

appears to be strikingly correlated to the migratory<br />

capacity of these cells. In semisolid culture media, all BFU-E<br />

exhibit an extensive migratory capacity, which is abruptly<br />

lost at the CFU-E level.38 In vivo, marrow cell egress occurs<br />

at the reticulocyte stage, and young reticulocytes are motile,<br />

whereas very few CFU-E circulate. It is thus tempting to<br />

speculate that these changes in erythroid cell motility reflect<br />

changes in cell attachment to Fn and possibly other adhesive<br />

glycoproteins of the marrow ECM. This is strengthened by<br />

the recent observation that glycoprotein IV, an 88 Kd<br />

glycoprotein identified as a thrombospondin re~eptor,’~ is<br />

present on mature BFU-E, CFU-E and erythroblasts, and is<br />

similarly lost during terminal erythroid differentiati~n.’~<br />

Loss of attachment to Fn with terminal differentiation has<br />

been previously observed in DMSO-induced murine erythroleukemic<br />

cells,” and hemin-induced human K 562 cells,”<br />

and ascribed to the loss of the 140 Kd Fn receptor molecule<br />

identified as VLA-5 on uninduced K562.12,40*41 Results obtained<br />

with human reticulocytes isolated from patients with<br />

hemolytic anemia or recently splenectomized have shown<br />

that these cells can attach to Fn?’ which appears to contradict<br />

the results presented in this study. However, reticulocytes<br />

produced in response to hemolysis are not identical to<br />

those produced during steady-state erythropoie~is,”~ and<br />

their adhesion properties might be altered by the underlying<br />

disorder.” Furthermore, the marked susceptibility of cell<br />

adhesion to the conditions in which the functional attachment<br />

assay is performed might explain quantitatively different<br />

results.<br />

Both the anti-a5/31 polyclonal IgG and the anti-d MoAb<br />

inhibited CFU-E and preproerythroblast adhesion to Fn.<br />

This strongly suggests that VLA-5 (a5pl) functions as an Fn<br />

receptor on normal human erythroblastic cells. The Fn<br />

receptor was undetectable from reticulocytes by indirect<br />

immunofluorescence. However, changes in the expression of<br />

the Fn receptor during in vitro erythroblastic differentiation<br />

could not be analyzed in more detail by immunofluorescence:<br />

the polyclonal antibody recognizes not only VLA-5, but all<br />

integrins sharing the /3l subunit, and some of these molecules<br />

may be expressed by erythroblasts, and the B1E5 MoAb is<br />

not suitable for fluorescence labeling (C. Damsky, personal<br />

communication, January 1989).<br />

The above results demonstrate that adhesion to Fn is<br />

precisely regulated during normal human erythroid differentiation<br />

and may influence cell migration in the bone marrow.<br />

A challenging question will be to determine if stage-specific<br />

expression of integrins will influence cell behavior by triggering<br />

early events associated with erythroid cell proliferation<br />

and differentiati~n.~’.~~<br />

ACKNOWLEDGMENT<br />

We gratefully acknowledge the help of the surgeons who provided<br />

marrow samples: Drs Brunet, Missenard, and Lapresle of Clinique<br />

Arago and Drs Bonnet, Judet, and Koechlin of HBpital de la<br />

Croix-Rouge and their nursing staff. We thank Dr A. Eaves (Terry<br />

Fox Laboratory, Vancouver, Canada) for the gift of erythropoietin;<br />

Drs L.C. Anderson, R. Berthier, C. Damsky, L. Edelmann, J.P.<br />

Fermand, and F. Tetteroo for their generous gifts of antibodies; C.<br />

Eaves, D. Baruch, and M. Rosemblatt for helpful discussions and<br />

critical reading of the manuscript, and M. Laroche for excellent<br />

secretarial assistance.


VUILLET-GAUGLER ET AL<br />

1. Edelman GM: Morphoregulatory molecules. Biochem 27:<br />

3533,1988<br />

2. Li ML, Aggeler J, Farson A, Hatier C, Hassel J, Bissell M:<br />

Influence of a reconstituted basement membrane and its components<br />

on casein gene expression and secretion in mouse mammary epithelial<br />

cells. Proc Natl Acad Sci USA 84:136, 1987<br />

3. Dexter TM, Allen TD, Lajtha LG: Conditions controlling the<br />

proliferation of haemopoietic stem cells in vitro. J Cell Physiol<br />

91:335, 1977<br />

4. Lichtman MA: The ultrastructure of the hematopoietic environment<br />

of the marrow. Exp Hematol9:391, 1981<br />

5. Dexter TM, Spncer E, Toksoz D, Lajtha LG: The role of<br />

cells and their products in the regulation of in vitro stem cell<br />

proliferation and granulocyte development. J Supramol Struct<br />

13513, 1980<br />

6. Coulombel L, Eaves C, Eaves A: Enzymatic treatment of long<br />

term human marrow cultures reveals the preferential location of<br />

primitive hemopoietic progenitors in the adherent layer. <strong>Blood</strong><br />

62:291, 1983<br />

7. Tsai S, Emerson SG, Sieff C: Isolation of a human stromal cell<br />

strain secreting hemopoietic growth factors. J Cell Physiol 127:137,<br />

1986<br />

8. Zsebo KM, Yuschenkoff VN, Schiffer S, Chang D, McWall E,<br />

Dinarello C, Brolun MA, Altrock B, Bagby GC: Vascular endothelial<br />

cells and granulopoiesis: Interleukin-1 stimulates release of<br />

G-CSF and GM-CSF. <strong>Blood</strong> 71:99,1988<br />

9. Rettenmeier CN, Roussel MF Differential processing of<br />

colony-stimulating factor 1 precursors encoded by two human<br />

cDNAs. Mol Cell Biol8:5026, 1988<br />

10. Gordon MY, Graham PR, Watt SM, Greaves MF: Compartmentalization<br />

of a haematopoietic growth factor (GM-CSF) by<br />

glycosaminoglycans in the bone marrow microenvironment. Nature<br />

326:403, 1987<br />

11. Ruoslahti E, Pierschbacher M: New perspectives in cell<br />

adhesion: RGD and integrins. Science 238:491, 1987<br />

12. Hemler ME, Huang C, Schwarz L: The VLA protein family.<br />

Characterization of five distinct cell surface heterodimers each with<br />

a common 130,000 molecular weight @ subunit. J Biol Chem<br />

262:3300,1987<br />

13. Campbell A, Wicha MS, Long M: Extracellular matrix<br />

promotes the growth and differentiation of murine hematopoietic<br />

cells in vitro. J Clin Invest 75:2085, 1985<br />

14. Spooncer E, Gallagher JT, Krizsa F, Dexter TM: Regulation<br />

of haemopoiesis in long-term bone marrow cultures. IV: Glycosaminoglycan<br />

synthesis and the stimulation of haematopoiesis by @-<br />

xylosides. J Cell Biol96:510, 1983<br />

15. Coulombel L, Vuillet MH, Leroy C, Tchernia G: Lineageand<br />

stage-specific adhesion of human hematopoietic cells to extracellular<br />

matrices from marrow fibroblasts. <strong>Blood</strong> 71:329, 1988<br />

16. Tsai S, Patel V, Beaumont E, Lodish HF, Nathan DG, Sieff<br />

CA: Differential binding of erythroid and myeloid progenitors to<br />

fibroblasts and fibronectin. <strong>Blood</strong> 69:1587, 1987<br />

17. Campbell A, Long MW, Wicha MS: Haemonectin, a bone<br />

marrow adhesion protein specific for cells of the granulocytic<br />

lineage. Nature 329:744, 1987<br />

18. Patel VP, Lodish HF: Loss of adhesion of murine erythroleukemia<br />

cells to fibronectin during erythroid differentiation. Science<br />

224:996, 1984<br />

19. Virtanen I, Ylanne J, Vartio T: Human erythroleukemia cells<br />

adhere to fibronectin: Evidence for a Mr 190000 receptor protein.<br />

<strong>Blood</strong> 69:578,1987<br />

20. Mouchiroud G, Berthier R, Newton IA, Chapel A, Hollard<br />

D: Monoclonal antibodies against human hematopoietic cells and<br />

REFERENCES<br />

the separation of progenitor cells from bone marrow. Exp Hematol<br />

13:566, 1985<br />

21. Edelman P, Vinci G, Villeval JL, Vainchenker W, Henri A,<br />

Miglierina R, Rouger R, Reviron J, Breton-Gorius J, Sureau C,<br />

Edelman L: A monoclonal antibody against an erythrocyte ontogenic<br />

antigen identifies fetal and adult erythroid progenitors. <strong>Blood</strong><br />

67:56, 1986<br />

22. Tetteroo PAT, Lansdorp PM, Leeksma OC, Von Dem Borne<br />

AEG: Monoclonal antibodies against human platelet glycoprotein<br />

IIIa. Br J Haematol55:509, 1983<br />

23. Anderson LC, Gahmberg CG, Teerenhovi L, Vuopio P<br />

Glycophorin A as a cell surface marker of early erythroid differentiation<br />

in acute leukemia. Int J Cancer 23:717, 1979<br />

24. Argraves WS, Suzuki S, Arai H, Thompson K, Pierschbacher<br />

M, Ruoslahti E: Amino acid sequence of the human fibronectin<br />

receptor. J Cell Biol 105:1183, 1987<br />

25. Werb Z, Tremble P, Behrendtsen 0, Crowley E, Damsky C:<br />

Signal transduction through the fibronectin receptor induces collagenase<br />

and stromelysin gene expression. J Cell Biol 109377, 1989<br />

26. Furfang J, Thierfelder S: Effective avidin coating of erythrocytes<br />

by disulfide bound formation. J Immunol Methods 91:123,<br />

1986<br />

27. Breton-Gorius J, Van Haeke D, Pryzwansky KB, Guichard J,<br />

Tabilio A, Vainchenker W, Carmel R: Simultaneous detection of<br />

membrane markers with monoclonal antibodies and peroxidatic<br />

activities in leukaemia: Ultrastructural analysis using a new method<br />

of fixation preserving the platelet peroxidase. Br J Haematol58:447,<br />

1984<br />

28. Graham RC, Karnovsky MJ: The early stages of absorption<br />

of injected horseradish peroxidase in proximal tubules of mouse<br />

kidney. Ultrastructural cytochemistry by a new technique. J Histochem<br />

Cytochem 14:291, 1966<br />

29. Kieffer N, Bettaieb A, Legrand C, Vainchenker W, Breton-<br />

Gorius J, Edelman L: Immunochemical identification of the 88 Kd<br />

platelet glycoprotein IV on human erythroid progenitor cells. <strong>Blood</strong><br />

70:156a, 1987 (abstr)<br />

30. Greenberg PL, Baker S, Link M, Minowada J: Immunologic<br />

selection of hematopoietic precursor cells utilizing antibodymediated<br />

plate binding (“panning”). <strong>Blood</strong> 65:190, 1985<br />

31. Breton-Gorius J, Villeval JL, Mitjavila MT, Vinci G, Guichard<br />

J, Rochant H, Flandrin G, Vainchenker W: Ultrastructural<br />

and cytochemical characterization of blasts from early erythroblastic<br />

leukemias. Leukemia 1:173, 1987<br />

32. Loken MR, Shah VO, Dattilio KL, Civin CI: Flow cytometry<br />

analysis of human bone marrow: I. Normal erythroid development.<br />

<strong>Blood</strong> 69:255,1987<br />

33. Breton-Gorius J, Reyes F Ultrastructure of human bone<br />

marrow cell maturation. Int Rev Cytol46:251, 1976<br />

34. Fibach E, Manor D, Oppenheim A, Rachilewitch EA: Proliferation<br />

and maturation of human erythroid progenitors in liquid<br />

culture. <strong>Blood</strong> 73:100, 1989<br />

35. Sawada K, Krantz SB, Kans JS, Dessypris EN, Sawyer S,<br />

Click AD, Civin CI: Purification of human erythroid colony-forming<br />

units and demonstration of specific binding of erythropoietin. J Clin<br />

Invest 80357,1987<br />

36. Nijhof W, Wierenga PK Isolation and characterization of<br />

the erythroid progenitor cells CFU-E. J Cell Biol96:386, 1983<br />

37. Koury MJ, Bondurant MC, Rana SS: Changes in erythroid<br />

membrane proteins during erythropoietin-mediated terminal differentiation.<br />

J Cell Physiol 133:438, 1987<br />

38. Eaves AC, Eaves CJ: Erythropoiesis in culture, in McCulloch<br />

EA (ed): Cell Culture Techniques. Clinics in Haematology, vol 13.<br />

Philadelphia, PA, WB Saunders, 1981, p 371


ERYTHROBLAST ADHESION TO FIBRONECTIN<br />

a73<br />

39. Asch AS, Barnwell J, Silverstein RL, Nachman RL: Isolation<br />

of the thrombospondin membrane receptor. J Clin Invest 79:1054,<br />

1987<br />

40. Takada Y, Huang C, Hemler M: Fibronectin receptor structures<br />

in the VLA family of heterodimers. Nature 326:607, 1987<br />

41. Patel VP, Lodish H F The fibronectin receptor on mammalian<br />

erythroid precursor cells: Characterization and developmental regulation.<br />

J Cell Biol 102:449, 1986<br />

42. Patel VP, Ciechanover A, Platt 0, Lodish HF: Mammalian<br />

reticulocytes lose adhesion to fibronectin during maturation to<br />

erythrocytes. Proc Natl Acad Sci USA 82:440, 1985<br />

43. Coulombel L, Tchernia G, Mohandas N: Human reticulocyte<br />

maturation and its relevance to erythropoietic stress. J Lab Clin Med<br />

94:467, 1979<br />

44. Mohandas N, Evans E: Sickle erythrocyte adherence to<br />

vascular endothelium: Morphologic correlates and the requirement<br />

for divalent cations and collagen binding plasma proteins. J Clin<br />

Invest 76:1605, 1985<br />

45. Dike LE, Farmer SR: Cell adhesion induces expression of<br />

growth-associated genes in suspension-arrested fibroblasts. Proc<br />

Natl Acad Sci USA 85:6792,1988<br />

46. Patel VP, Lodish HF A fibronectin matrix is required for<br />

differentiation of murine erythroleukemia cells into reticulocytes. J<br />

Cell Biol 105:3105, 1987

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!