Primary familial erythrocytosis (familial polycythemia) is a rare myeloproliferative disorder with an autosomal dominant mode of inheritance. We studied a new kindred with autosomal dominantly inherited familial erythrocytosis. The molecular basis for the observed phenotype of isolated erythrocytosis is heterozygosity for a novel nonsense mutation affecting codon 399 in exon 8 of the erythropoietin receptor (EPOR) gene, encoding an EpoR peptide that is truncated by 110 amino acids at its C-terminus. The newEPOR gene mutation 5881G>T was found to segregate with isolated erythrocytosis in the affected family and this mutation represents the most extensive EpoR truncation reported to date, associated with familial erythrocytosis. Erythroid progenitors from an affected individual displayed Epo hypersensitivity in in vitro methylcellulose cultures, as indicated by more numerous erythroid burst-forming unit-derived colonies in low Epo concentrations compared to normal controls. Expression of mutant EpoR in interleukin 3–dependent hematopoietic cells was associated with Epo hyperresponsiveness compared to cells expressing wild-type EpoR.

Familial erythrocytosis, also known as primary familial and congenital polycythemia (PFCP), is a rare disorder involving isolated proliferation of bone marrow progenitors of the erythroid lineage.1,2 This disorder is typically characterized by an autosomal dominant mode of inheritance, and less frequently, by the occurrence of sporadic cases.3,4The clinical features include the presence of isolated erythrocytosis without evolution into leukemia or other myeloproliferative disorders, absence of splenomegaly, normal white blood cell and platelet counts, low plasma erythropoietin (Epo) levels, normal hemoglobin-oxygen dissociation curve indicated by a normal P50, and hypersensitivity of erythroid progenitors to exogenous erythropoietin in vitro.5-7 Mutations in the gene encoding the erythropoietin receptor (EPOR) have been described in several families with isolated familial erythrocytosis.4 8-14 We report a new kindred with dominantly inherited familial erythrocytosis associated with heterozygosity for a novel point mutation in theEPOR gene.

Patients, erythroid colony formation assays, DNA and RNA analyses

Peripheral blood samples from affected and unaffected individuals were obtained under a protocol approved by the Institutional Review Board at Yale University School of Medicine. In vitro erythroid colony formation assays were performed as described.10 The genomic DNA structure of theEPOR gene and RNA analyses for cloning of mutant EpoR complementary DNA (cDNA) were performed as described.10Identity of subcloned polymerase chain reaction (PCR) products containing amplified EPOR genomic DNA and cDNA fragments were confirmed by nucleotide sequencing.

Cloning and expression of mutant EpoR and Epo dose-response assays

Wild-type (WT) and mutant human EPOR cDNAs were cloned in pBabe-puro retroviral vector and plasmids were stably transfected into amphotropic producer cell line PA317 using Lipofectamine reagent (Invitrogen, Carlsbad, CA).15Puromycin-resistant (3.5 μg/mL) PA317 cells were cocultured with murine interleukin 3 (IL-3)–dependent 32D cells in the presence of 4 μg/mL polybrene (Sigma, St Louis, MO) for 24 hours. 32D cells infected with retrovirus were selected in medium containing 1.0 μg/mL puromycin for 10 days and cultured in Epo 2 U/mL (Amgen, Thousand Oaks, CA) instead of IL-3. As negative control, 32D cells transduced with empty pBabe-puro vector were generated. Single-cell clones of 32D cells were isolated by limiting dilution. Expression of EpoR protein in single-cell clones was demonstrated by immunoprecipitation and Western analysis as described15 and surface expression of EpoR was confirmed by a flow cytometry assay (data not shown) using recombinant human Epo (Amgen) that was biotinylated as described.16Negative control 32D cells transduced with vector only do not express EpoR protein, do not proliferate in Epo, and do not demonstrate surface binding of biotin-Epo by flow cytometry assay (data not shown). Epo dose-response assays were performed using MTT reagent (dimethylthiazol-2-yl-2,5-diphenyltetrazolium) as described.17 

We studied a 4-generation white family from Maine (Figure1). The propositus (patient III:24 in Figure 1) was first evaluated at the age of 15 years because of headaches associated with a hemoglobin of 20.7 g/dL and a hematocrit of 62%. The white blood cell and platelet counts and morphology of the red blood cells in peripheral blood smear were normal. The plasma Epo level was less than 10 mU/mL and hemoglobin electrophoresis and P50 were normal. The persistent headaches were relieved by phlebotomy. Many affected individuals in this family underwent periodic phlebotomy to control hematocrit levels.

Fig. 1.

Pedigree of affected family.

The dark filled symbols represent affected individuals with erythrocytosis or history of periodic phlebotomy for polycythemia. The symbols with patterned fill represent individuals for whom clinical information was not available. The presence or absence of the 5881G>T mutation in tested individuals is indicated by + or − symbols, respectively. The propositus is indicated by the arrow (III:24).

Fig. 1.

Pedigree of affected family.

The dark filled symbols represent affected individuals with erythrocytosis or history of periodic phlebotomy for polycythemia. The symbols with patterned fill represent individuals for whom clinical information was not available. The presence or absence of the 5881G>T mutation in tested individuals is indicated by + or − symbols, respectively. The propositus is indicated by the arrow (III:24).

Close modal

The phenotype of the propositus and other evaluated affected individuals in the pedigree were consistent with clinical features of PFCP.18 To determine whether erythrocytosis in this family was associated with Epo hypersensitivity of erythroid progenitors, we performed in vitro semisolid medium cultures using peripheral blood mononuclear cells. The results of scoring of day 14 erythroid burst-forming unit (BFU-E)–derived colonies in semisolid methylcellulose medium containing serum and various added Epo concentrations are illustrated in Figure2A. In the absence of added Epo in the culture medium, 0 to 1 BFU-E–derived colonies were detected in the samples from the affected individual and no erythroid colonies were present in control cultures. At highest added Epo concentration of 1 U/mL, the numbers of BFU-E–derived colonies were similar in cultures of the affected individual and those of the unaffected family member (30 ± 3.7 versus 33 ± 7, respectively). Notably, erythroid progenitors of the affected individual displayed hypersensitivity to Epo when compared to those of an unaffected family member in the presence of relatively low and intermediate range of Epo concentrations (25-100 mU/mL). A significant difference in the number of erythroid colonies was observed in 25 mU/mL, with 1.3 ± 0.5 colonies in the control plates compared to 8 ± 0.5 colonies in the patient cultures (P < .0002 by Student t test). In 50 mU/mL Epo, 20 ± 5.8 colonies were present in the cultures from the individual with erythrocytosis compared to 11 ± 1.5 in the unaffected family member (not significant). In 100 mU/mL Epo, the difference in colony numbers remained significant with 28 ± 0.5 in the patient versus 17 ± 2.8 in controls (P < .03). The differences in the total numbers of erythroid colonies was accompanied by a visible difference in the size of individual colonies when culture plates from the affected individual and control were examined (data not shown).

Fig. 2.

Erythroid colony assays, mutation analysis, and Epo-dose response.

(A) Erythroid colony formation assays in 2 family members. The vertical axis indicates the numbers of BFU-E–derived colonies per 2.5 × 105 peripheral blood mononuclear cells, expressed as the mean ± SD of assays performed in triplicate. The final Epo concentration added to the cultures is indicated on the horizontal axis. (B) DNA sequence of subcloned genomic PCR products from the EpoR gene of the proband. Analysis of individual clones shows the sequences for the normal allele (left) and the mutant allele (right). The bold letter T indicates 5881G>T substitution resulting in introduction of a STOP codon. (C) Upper panel shows diagram of exon 8 of EPORgene. The coding sequence is shown as solid box and the 3′ untranslated region as an open box. The position and size of genomic PCR amplification products and a Tru9I (T) restriction map of the 493-base pair (bp) PCR amplification product for the mutant allele are shown. Lower panel shows detection of the mutation by restriction endonuclease digestion of PCR-amplified genomic DNA. Part of the pedigree is shown at the top of the figure. Genomic DNA amplification products were digested with Tru9I and fractionated by electrophoresis in a 2% agarose gel. The mutation creates a uniqueTru9I site in mutant allele and yields fragments of 369 bp and 124 bp in addition to the 493-bp fragment from the normal allele in individuals heterozygous for the new mutation. None of the unaffected individuals (open symbols) have Tru9I site in their genomic DNA, whereas all affected individuals in the family (filled symbols) are heterozygous for the mutation. (D) Epo dose-response of 32D cells transfected with wild type (WT) or mutant (ME) EpoRs. The cells were cultured in the indicated concentrations of Epo (U/mL) and the results are expressed as a percentage of maximal proliferation in 10 U/mL Epo as determined by MTT assay. Each data point represents the mean of 4 determinations with SE bars shown. Similar results were obtained in several experiments using multiple independent single cell clones of transfected cells.

Fig. 2.

Erythroid colony assays, mutation analysis, and Epo-dose response.

(A) Erythroid colony formation assays in 2 family members. The vertical axis indicates the numbers of BFU-E–derived colonies per 2.5 × 105 peripheral blood mononuclear cells, expressed as the mean ± SD of assays performed in triplicate. The final Epo concentration added to the cultures is indicated on the horizontal axis. (B) DNA sequence of subcloned genomic PCR products from the EpoR gene of the proband. Analysis of individual clones shows the sequences for the normal allele (left) and the mutant allele (right). The bold letter T indicates 5881G>T substitution resulting in introduction of a STOP codon. (C) Upper panel shows diagram of exon 8 of EPORgene. The coding sequence is shown as solid box and the 3′ untranslated region as an open box. The position and size of genomic PCR amplification products and a Tru9I (T) restriction map of the 493-base pair (bp) PCR amplification product for the mutant allele are shown. Lower panel shows detection of the mutation by restriction endonuclease digestion of PCR-amplified genomic DNA. Part of the pedigree is shown at the top of the figure. Genomic DNA amplification products were digested with Tru9I and fractionated by electrophoresis in a 2% agarose gel. The mutation creates a uniqueTru9I site in mutant allele and yields fragments of 369 bp and 124 bp in addition to the 493-bp fragment from the normal allele in individuals heterozygous for the new mutation. None of the unaffected individuals (open symbols) have Tru9I site in their genomic DNA, whereas all affected individuals in the family (filled symbols) are heterozygous for the mutation. (D) Epo dose-response of 32D cells transfected with wild type (WT) or mutant (ME) EpoRs. The cells were cultured in the indicated concentrations of Epo (U/mL) and the results are expressed as a percentage of maximal proliferation in 10 U/mL Epo as determined by MTT assay. Each data point represents the mean of 4 determinations with SE bars shown. Similar results were obtained in several experiments using multiple independent single cell clones of transfected cells.

Close modal

Mutations in EPOR have been found in about 12% of cases in a series of individuals with PFCP and it appears that mutation in a gene(s) other than EPOR may account for the polycythemic phenotype in the majority of PFCP cases.4 To determine whether the PFCP phenotype in the new family was associated with a mutation in the EPOR gene, we evaluated the structure of the gene with particular focus on the region encoding the C-terminal domain of the receptor that exerts a negative effect on receptor mitogenic function.19-21 Direct nucleotide sequencing as well as sequencing of subcloned genomic PCR amplification products from the propositus revealed heterozygosity for presence of G>T substitution at position 5881 of EpoR (Figure 2B), introducing a premature termination codon at position 399 that would result in deletion of 110 amino acid residues at the C-terminus of the receptor. This novel mutation introduces a new restriction endonuclease site (Tru9I) in exon 8 of EPOR gene, which allowed rapid, PCR-based detection of the mutation in each family member (Figure 2C). Restriction endonuclease digestion of genomic PCR amplification products with Tru9I was observed only in affected individuals all of whom were heterozygous for the 5881G>T mutation. The mutation was not present in any of the unaffected individuals tested.

The EpoR mutations associated with PFCP phenotype described to date are all located within exon 8 of the gene and result in truncation of 59 to 84 amino acids from the C-terminal region.4 The function of the new EpoR mutation (5881G>T) was evaluated in vitro in 32D cells engineered to express either wild-type or mutant EpoR cDNA isolated from the proband. Figure 2D illustrates an assay measuring the Epo dose-response of 32D cells transfected with wild-type EpoR or mutant EpoR. A significant percentage of mutant EpoR-expressing cells proliferate in relatively low concentrations of Epo and thus display increased sensitivity to Epo compared to wild-type EpoR-expressing cells within an Epo concentration range of 0.01 to 0.1 U/mL. This result is consistent with previous findings from our laboratory as well as others demonstrating increased Epo sensitivity of transfected hematopoietic cells expressing truncated human EpoRs compared to cells expressing wild-type EpoR.9,10 14 

The distal cytoplasmic region of EpoR is required for down-regulation of Epo-mediated activation of JAK2, a cytoplasmic tyrosine kinase critical for Epo-induced mitogenesis as well as inhibition of apoptosis.14,17,22,23 Recently, a mouse model for congenital polycythemia has been generated by homologous replacement of murine EPOR gene by a mutant human EPOR gene described in a family with PFCP.24 This mutant human EpoR is truncated by 83 amino acids at its C-terminus13 and mice heterozygous for the mutant EpoR allele developed marked polycythemia, mimicking the dominantly inherited human disorder.24 In contrast, in a study by Zang et al,25 mice heterozygous for a targeted mutation in murine EpoR gene, with a more extensive truncation of 108 distal cytoplasmic amino acids, did not display increased hematocrits. The differences between the polycythemic phenotype obtained with in vivo expression of mutant Epo receptors with different cytoplasmic truncations suggested that the extent of the truncations may be important for generation of the PFCP phenotype.24,25 The new human EpoR mutant described in our study represents the most extensive human EpoR truncation reported to date that is associated with PFCP in heterozygous individuals and confers Epo hypersensitivity in erythroid progenitors and transfected hematopoietic cells. This human EpoR mutant lacks 110 distal amino acid residues, comparable with regard to the extent of the cytoplasmic truncation, to the truncated murine EpoR studied by Zang et al.25 It is possible that specific structural differences between truncated human and murine EpoRs other than the extent of the cytoplasmic truncation play a role in generation of the polycythemic phenotype. For instance, compared to the murine EpoR, a notable difference in the structure of truncated human EpoR mutants is the presence of an additional tyrosine residue in the membrane proximal region of human EpoR that could potentially contribute to the dominant, gain-of-function effect of truncated human receptors resulting in generation of PFCP phenotype in vivo.

The authors thank Amgen for providing recombinant human erythropoietin.

Supported in part by a grant from the National Institutes of Health DK-02566 (to M.O.A.).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Prchal
JT
Sokol
L
“Benign erythrocytosis” and other familial and congenital polycythemias.
Eur J Haematol.
57
1996
263
268
2
Forget
BG
Degar
BA
Arcasoy
MO
Familial polycythemia due to truncations of the erythropoietin receptor.
Trans Am Clin Climatol Assoc.
111
2000
38
44
3
Percy
MJ
McMullin
MF
Roques
AW
et al
Erythrocytosis due to a mutation in the erythropoietin receptor gene.
Br J Haematol.
100
1998
407
410
4
Kralovics
R
Prchal
JT
Genetic heterogeneity of primary familial and congenital polycythemia.
Am J Hematol.
68
2001
115
121
5
Prchal
JT
Crist
WM
Goldwasser
E
Perrine
G
Prchal
JF
Autosomal dominant polycythemia.
Blood.
66
1985
1208
1214
6
Juvonen
E
Ikkala
E
Fyhrquist
F
Ruutu
T
Autosomal dominant erythrocytosis caused by increased sensitivity to erythropoietin.
Blood.
78
1991
3066
3069
7
Emanuel
PD
Eaves
CJ
Broudy
VC
et al
Familial and congenital polycythemia in three unrelated families.
Blood.
79
1992
3019
3030
8
de la Chapelle
A
Träskelin
AL
Juvonen
E
Truncated erythropoietin receptor causes dominantly inherited benign human erythrocytosis.
Proc Natl Acad Sci U S A.
90
1993
4495
4499
9
Sokol
L
Luhovy
M
Guan
Y
Prchal
JF
Semenza
GL
Prchal
JT
Primary familial polycythemia: a frameshift mutation in the erythropoietin receptor gene and increased sensitivity of erythroid progenitors to erythropoietin.
Blood.
86
1995
15
22
10
Arcasoy
MO
Degar
BA
Harris
KW
Forget
BG
Familial erythrocytosis associated with a short deletion in the erythropoietin receptor gene.
Blood.
89
1997
4628
4635
11
Kralovics
R
Indrak
K
Stopka
T
Berman
BW
Prchal
JF
Prchal
JT
Two new EPO receptor mutations: truncated EPO receptors are most frequently associated with primary familial and congenital polycythemias.
Blood.
90
1997
2057
2061
12
Furukawa
T
Narita
M
Sakaue
M
et al
Primary familial polycythaemia associated with a novel point mutation in the erythropoietin receptor.
Br J Haematol.
99
1997
222
227
13
Kralovics
R
Sokol
L
Prchal
JT
Absence of polycythemia in a child with a unique erythropoietin receptor mutation in a family with autosomal dominant primary polycythemia.
J Clin Invest.
102
1998
124
129
14
Watowich
SS
Xie
X
Klingmüller
U
et al
Erythropoietin receptor mutations associated with familial erythrocytosis cause hypersensitivity to erythropoietin in the heterozygous state.
Blood.
94
1999
2530
2532
15
Arcasoy
MO
Maun
NA
Perez
L
Forget
BG
Berliner
N
Erythropoietin mediates terminal granulocytic differentiation of committed myeloid cells with ectopic erythropoietin receptor expression.
Eur J Haematol.
67
2001
77
87
16
Shinjo
K
Takeshita
A
Higuchi
M
Ohnishi
K
Ohno
R
Erythropoietin receptor expression on human bone marrow erythroid precursor cells by a newly-devised quantitative flow-cytometric assay.
Br J Haematol.
96
1997
551
558
17
Arcasoy
MO
Harris
KW
Forget
BG
A human erythropoietin receptor gene mutant causing familial erythrocytosis is associated with deregulation of the rates of Jak2 and Stat5 inactivation.
Exp Hematol.
27
1999
63
74
18
Prchal
JF
Prchal
JT
Molecular basis for polycythemia.
Curr Opin Hematol.
6
1999
100
109
19
Yoshimura
A
Longmore
G
Lodish
HF
Point mutation in the exoplasmic domain of the erythropoietin receptor resulting in hormone-independent activation and tumorigenicity.
Nature.
348
1990
647
649
20
D'Andrea
AD
Yoshimura
A
Youssoufian
H
Zon
LI
Koo
JW
Lodish
HF
The cytoplasmic region of the erythropoietin receptor contains nonoverlapping positive and negative growth-regulatory domains.
Mol Cell Biol.
11
1991
1980
1987
21
Youssoufian
H
Longmore
G
Neumann
D
Yoshimura
A
Lodish
HF
Structure, function, and activation of the erythropoietin receptor.
Blood.
81
1993
2223
2236
22
Klingmüller
U
Lorenz
U
Cantley
LC
Neel
BG
Lodish
HF
Specific recruitment of SH-PTP1 to the erythropoietin receptor causes inactivation of JAK2 and termination of proliferative signals.
Cell.
80
1995
729
738
23
Witthuhn
BA
Quelle
FW
Silvennoinen
O
et al
JAK2 associates with the erythropoietin receptor and is tyrosine phosphorylated and activated following stimulation with erythropoietin.
Cell.
74
1993
227
236
24
Divoky
V
Liu
Z
Ryan
TM
Prchal
JF
Townes
TM
Prchal
JT
Mouse model of congenital polycythemia: homologous replacement of murine gene by mutant human erythropoietin receptor gene.
Proc Natl Acad Sci U S A.
98
2001
986
991
25
Zang
H
Sato
K
Nakajima
H
McKay
C
Ney
PA
Ihle
JN
The distal region and receptor tyrosines of the Epo receptor are non-essential for in vivo erythropoiesis.
EMBO J.
20
2001
3156
3166

Author notes

Murat O. Arcasoy, Division of Hematology-Oncology, Duke University Medical Center, DUMC Box 3912, Durham, NC 27710; e-mail: arcas001@mc.duke.edu.

Sign in via your Institution