Next Article in Journal
Missense Variants in GFRA1 and NPNT Are Associated with Congenital Anomalies of the Kidney and Urinary Tract
Previous Article in Journal
Paleogenomes Reveal a Complex Evolutionary History of Late Pleistocene Bison in Northeastern China
Previous Article in Special Issue
Genetic Background of Polycythemia Vera
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

Identification of Two Novel EPOR Gene Variants in Primary Familial Polycythemia: Case Report and Literature Review

by
Laura Lo Riso
1,†,
Gardenia Vargas-Parra
2,3,†,
Gemma Navarro
2,
Leonor Arenillas
3,4,
Lierni Fernández-Ibarrondo
3,
Beatriz Robredo
5,
Carmen Ballester
1,
Bernardo López
1,
Albert Perez-Montaña
1,
Antonia Sampol
1,
Lourdes Florensa
3,4,
Carles Besses
3,6,
María Antonia Duran
1 and
Beatriz Bellosillo
2,3,*
1
Hematology and Hemotherapy Department, Hospital Universitario Son Espases, IDISBA, 07120 Palma de Mallorca, Spain
2
Molecular Biology Laboratory, Pathology Department, Hospital del Mar, 08003 Barcelona, Spain
3
Hospital del Mar Medical Research Institute, IMIM, 08003 Barcelona, Spain
4
Hematological Cytology Laboratory, Pathology Department, Hospital del Mar, 08003 Barcelona, Spain
5
Hematology Department, Hospital Punta Europa, 11207 Algeciras, Spain
6
Hematology Department, Hospital del Mar, 08003 Barcelona, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 10 August 2022 / Revised: 14 September 2022 / Accepted: 16 September 2022 / Published: 20 September 2022
(This article belongs to the Special Issue Genetics and Genomics of Erythrocytosis)

Abstract

:

Simple Summary

Erythrocytosis can be caused by a wide variety of diseases. Some forms of erythrocytosis have an obvious cause, such as a kidney injury, or it may have an oncological cause, but in some patients, the origin of the disease is not entirely clear, and since the symptoms of an isolated erythrocytosis are not usually cumbersome, sometimes the diagnosis takes several months or years. In the present work, we report a couple of cases of familial erythrocytosis associated with novel variants in the erythropoietin receptor gene. This study serves as a reminder of the clinical and molecular study of this rare disease and expands the list of mutations associated with primary familial polycythemia.

Abstract

Primary familial and congenital polycythemia is a rare disease characterized by an increase in red cell mass that may be due to pathogenic variants in the EPO receptor (EPOR) gene. To date, 33 genetic variants have been reported to be associated. We analyzed the presence of EPOR variants in two patients with polycythemia in whom JAK2 pathogenic variants had been previously discarded. Molecular analysis of the EPOR gene was performed by Sanger sequencing of the coding regions and exon/intron boundaries of exon 8. We performed in vitro culture of erythroid progenitor cells. Segregation studies were done whenever possible. The two patients studied showed hypersensitivity to EPO in in vitro cultures. Analysis of the EPOR gene unveiled two novel pathogenic variants. Genetic testing of asymptomatic relatives could guarantee surveillance and proper management.

1. Introduction

Erythrocytosis is the increase in the number of erythrocytes in peripheral blood. It can be either primary due to an intrinsic defect of the erythroid compartment that is associated with low erythropoietin (EPO) levels or secondary, which is extrinsic to the red cells and associated with normal or high EPO levels. Both primary and secondary erythrocytosis can be acquired or arise from hereditary alterations [1]. Among all types of erythrocytosis, the most common type is polycythemia vera (PV), which is an acquired primary erythrocytosis due to somatic mutations in the Janus kinase gene (JAK2). For this reason, apart from excluding secondary causes, the workup of erythrocytosis in the clinical practice includes in the initial testing the screening for JAK2 mutations [2].
Regarding hereditary alterations causing erythrocytosis, known as primary congenital erythrocytosis (CE), pathogenic variants in the EPOR gene have been described in 12-15% of cases, producing defects in the erythropoietin (EPO) receptor (EPOR) [3,4,5]. This entity encompassing alterations in the EPOR gene is designated as Primary Familial and Congenital Polycythemia (PFCP) or ECYT1 [6].
The EPOR gene codifies for the EPO receptor of 508 amino acids, which belongs to the type I cytokine receptors and consists of an extracellular domain that binds to the EPO ligand, a transmembrane domain and an intracellular domain. Upon EPO binding, the EPOR dimerizes enabling two JAK2 tyrosine kinases, which are pre-attached to the receptor, to become close enough for their transphosphorylation and activation [7]. Activated JAK2 proteins phosphorylate the tyrosine residues located in the intracellular domain of the EPOR. The C-terminal domain contains eight tyrosines that act as binding sites for regulatory proteins such as SHP-1 or SOCS family proteins that dephosphorylate the tyrosine residues of both EPOR and JAK2 [5]. Phosphorylated tyrosine residues become docking sites for other signaling proteins containing SH2 residues, such as STAT5 and PI3K, which are in turn activated by phosphorylation. Activated STAT proteins translocate to the nucleus and activate transcription of genes involved in cell differentiation, division, and apoptosis inhibition [8].
Here, we report two cases of erythrocytosis in which the diagnosis of PV and acquired secondary polycythemia had been ruled out that were further studied for mutations in the EPOR gene.

2. Materials and Methods

Samples: Two patients presenting sustained erythrocytosis were referred to the Pathology Department of Hospital del Mar. Common causes of secondary erythrocytosis and JAK2 pathogenic variants in exons 12 and 14 had been previously discarded in both cases. Cases were referred for performing cultures of hematopoietic progenitors and molecular analysis of EPOR gene. A total of 20 mL of peripheral blood was collected after informed consent of the patients.
EPOR sequencing: The complete coding region and the exon–intron boundaries of EPOR gene were sequenced in peripheral blood granulocytes DNA. Sanger sequencing was performed in an ABI3500 instrument after amplification using intronic primers (conditions available upon request). To ensure whether the resulting variants were germinal, variants were confirmed in isolated CD3+ lymphocytes. Variant annotation was performed using the RefSeq accession number NM_000121 for the EPOR gene. Segregation studies of EPOR variants in family relatives were conducted when possible, using the same EPOR sequencing methods.
Next-generation sequencing (NGS): ASXL1, BCOR, BCORL1, BPGM, CALR, CBL, CEBPA, CHEK2, CSF3R, CSNK1A1, CUX1, DDX41, DLEU7, DNMT3A, EGR1, EGLN1, EPAS1, EPO, EPOR, ETV6, EZH2, FLT3, GATA2, HBA1, HBA2, HBB, IDH1, IDH2, JAK2, KIT, KMT2A, KRAS, MPL, NF1, NPM1, NRAS, PHF6, PPM1D, PRPF8, PTPN11, RAD21, RUNX1, SETBP1, SF3B1, SH2B3, SRSF2, STAG2, THPO, TET2, TNFSF11, TP53, TP53RK, TP53TG5, U2AF1, VHL, WT1, and ZRSR2 coding sequences were amplified using two custom GeneRead™ DNAseq Targeted panels (Qiagen). The resulting libraries were sequenced on a NextSeq® platform (Illumina) and analyzed with the QIASeq DNA pipeline (Qiagen). Variants obtained were filtered and annotated with the Illumina VariantStudio software and visualized with the Integrative Genomics Viewer.
Cell culture of hematopoietic progenitors: Endogenous erythroid colony (EEC) assay was conducted and scored precisely as previously described [9]. To assess erythropoietin sensitivity, peripheral blood mononuclear cells from patients and controls were plated in methylcellulose medium (2 × 105/mL; Methocult H-4533, Stem Cell Technology). Epo was added at the following concentrations: 0.0, 0.03, 0.06, 0.125, 0.25, 0.5, 1, 3, and 4 IU/mL. Cultures were maintained in a humidified atmosphere with 5% carbon dioxide at 37 °C. Large erythroid colonies (BFU-E) were scored at day 14.
Literature review and variant classification. We searched for EPOR variants using the terms (EPOR)+(variants) or (EPOR)+(mutations) in PubMed and selected the variants that were reported to be directly associated with PFCP. We classified the variants following the ACMG/AMP general guidelines [10].

3. Results

We have identified two novel EPOR pathogenic variants in two cases referred to the Molecular Diagnostics Laboratory in Hospital del Mar (Barcelona).
Case 1 was a 17-year-old woman of Spanish origin with hemoglobin (Hb) levels of 18.6 g/dL and hematocrit of 62%, who had severe headaches. The only family history related was a 32-year-old brother who resides in Germany, who also had been treated with phlebotomies. Case 2 was a 55-year-old male from Syria who presented with Hb levels of 17.4 g/dL and hematocrit of 52.6%, severe iron deficiency, and severe headaches, mental dullness, and asthenia; his father presented a history of phlebotomies. He had three children of 18, 16, and 12 years old with no clinical/analytical alterations.
Both patients had low levels of serum EPO (<1.5 mU/mL) excluding hypoxia-driven polycythemia. At physical examination, no splenomegaly was reported in any of them, leucocyte and platelet counts were normal, and bone marrow biopsies showed no panmyelosis.
Cell culture of hematopoietic progenitors showed a slight endogenous growth of erythroid cell colonies in the absence of EPO in case 1. Hypersensitivity to EPO was observed in both cases.
Sanger sequencing of the exon 8 of EPOR showed the presence of two novel mutations: in the first case, a duplication of 16 nucleotides leading to a frameshift at proline 431 and a subsequent termination codon after 19 amino acids (c.1275_1290dup, p.Pro431Valfs*19) and in the second case, a deletion of 1 cytosine also causing a frameshift, with the substitution of the proline 449 for a histidine and the appearance of a stop codon after four amino acids (c.1346del, p.Pro449Hisfs*4) (Figure 1). The two variants produced a shorter protein with loss of six of the eight tyrosine residues involved in phosphorylation signaling and were therefore considered as pathogenic.
These findings were corroborated in the NGS analyses, and no further pathogenic variants were found in genes related to somatic or germline erythrocytosis.
Segregation studies were attainable in the brother of case 1 and in all three healthy children of case 2. The 32-year-old brother, who also had erythrocytosis and low EPO levels, resulted as a carrier of the same c.1275_1290dup variant. EPOR sequencing of case 2′s healthy children revealed that his 18-year-old son was the only of the three inheriting the c.1346del variant.
All 33 EPOR variants reported to be associated with PFCP to date, and their classification are listed in Table 1. According to ACMG/AMP guidelines, 26 of them are classified as pathogenic or likely pathogenic, 3 are variants of uncertain significance, and 4 are benign or likely benign.

4. Discussion

PFCP is an autosomal dominant disease with incomplete penetrance, characterized by an isolated primary polycythemia associated with increased red cell mass and subnormal EPO levels. It mimics the clinical presentation of PV, a neoplastic (clonal) disorder, whereas in PFCP, the hematopoiesis is polyclonal, benign, and usually shows good response to phlebotomies if needed [34]. It is important to correctly diagnose these patients in order to avoid labeling them as “neoplastic”.
EPOR gain of function variants account for around 12% of the ECYT1 suspected patients [4,35]. In our experience, EPOR pathogenic variants were found in 6% (2/34) of all studied patients, presenting polycythemia of unknown origin and no JAK2 pathogenic variants.
Congenital predisposition to erythrocytosis can be caused by mutations in a wide range of genes such as EPOR, VHL, EGLN1, EPAS1, HBB, HBA1, HBA2, and BPGM. Until 2008, there were 11 EPOR pathogenic variants reported as causative of familiar polycythemia. Nevertheless, some of the previously reported variants are classified as benign or likely benign according to the current ACMG/AMP guidelines [10]. In Table 1, we classified all EPOR variants reported to date following these criteria. Our cases expand the list of EPOR described mutations so far, and we can expect this number to rise soon with the inclusion of EPOR in NGS panels [36].
In a small proportion of patients with idiopathic erythrocytosis (IE), the presence of probably pathogenic variants in the SH2B3 gene has been reported. This gene is also associated with myeloproliferative neoplasms, especially when they are associated with variants in LNK or other genes of the JAK2 pathway [37]. Therefore, it would be recommended that in cases with erythrocytosis and normal levels of EPO, all these genes be studied at the same time whenever possible, betting on the use of NGS approaches. In the case of acquired mutations, we would be able to assess clonality and in IE possible multilocus inherited alleles.
Since there is a great number of already-existing variants of uncertain significance (VUS), and more is to come as NGS is more widely used, standardized functional studies are a must. Here, we performed cell culture proliferation assays to demonstrate hypersensitivity of hematopoietic progenitors containing pathogenic variants in EPOR to low concentrations of EPO as a demonstration of pathogenicity of these variants. These studies have been previously used as a functional analysis for mutations in this gene [28,38].
Previously reported pathogenic mutations in the EPOR gene produce a shorter EPOR that lacks the docking sites for the negative regulators in the EPO signaling [39]. SHP-1 is a protein tyrosine phosphatase mainly expressed in hematopoietic cells, which inactivates EPOR upon binding to a tyrosine residue located at the C-terminal tail [40]; presumably, loss of these residues, due to truncation mutations, results in an EPO receptor lacking a SHP-1 binding site at Y454, maintaining the observed activation of the EPO receptor. The two novel mutations identified here also cause a shorter protein in which six of the eight conserved tyrosine residues, including Y454, are lost. It has been widely reported that truncations of the EPOR that lack all negative regulatory sites are sufficient to produce erythrocytosis [6]. Pasquier et al. demonstrated that frameshift mutations leading to a new C-terminal cytoplasmic tail including a MDTVP motif increase EPO dimerization and stability [28] compared with wild type and even when compared with its nonsense equivalent lacking the MDTVP motif. Case 1, showing a duplication of 16 base pairs, results in the formation of a new cytoplasmic tail containing this MDTVP motif.

5. Conclusions

We have reported two new mutations in the EPOR gene (c.1275_1290dup and c.1346del). The presence of mutations in EPOR establishes the diagnosis of PFCP, an underdiagnosed disease of benign origin that can cause a severe and disabling disease and that, once diagnosed, is usually easy to treat.

Author Contributions

Conceptualization, L.L.R. and B.B.; data curation, G.V.-P.; formal analysis, L.F., C.B. (Carles Besses), M.A.D. and B.B.; funding acquisition, B.B.; investigation, G.N. and L.F.-I.; methodology, G.N., L.A., B.R., C.B. (Carmen Ballester) and B.L.; project administration, B.B.; supervision, L.F., C.B. (Carles Besses) and B.B.; validation, A.P.-M. and A.S.; writing—original draft, G.V.-P. and L.L.R.; writing—review and editing, B.B. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported in part by the Government of Catalonia, project ID: 2017SGR205, and Xarxa de Banc de Tumors de Catalunya.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Ethics Committee of Hospital del Mar (Approval ID: 2020/9021/I).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mcmullin, M.F. Genetic background of congenital erythrocytosis. Genes 2021, 12, 1151. [Google Scholar] [CrossRef]
  2. Barbui, T.; Thiele, J.; Ferrari, A.; Vannucchi, A.M.; Tefferi, A. The new WHO classification for essential thrombocythemia calls for revision of available evidences. Blood Cancer J. 2020, 10, 22. [Google Scholar] [CrossRef] [PubMed]
  3. Kralovics, R.; Prchal, J.T. Genetic heterogeneity of primary familial and congenital polycythemia. Am. J. Hematol. 2001, 68, 115–121. [Google Scholar] [CrossRef] [PubMed]
  4. Bento, C.; McMullin, M.F.; Gardie, B.; Percy, M.; Cario, H.; Kučerová, J.; Girodon, F.; Rossi, C.; Aström, M.; Diaz-Aguado, A.; et al. Congenital Erythrocytosis and Hereditary Thrombocytosis; COST (European Cooperation in Science and Technology): Brussels, Belgium, 2015; ISBN 978-989-20-5646-3. [Google Scholar]
  5. Bento, C.; Percy, M.J.; Gardie, B.; Maia, T.M.; van Wijk, R.; Perrotta, S.; Della Ragione, F.; Almeida, H.; Rossi, C.; Girodon, F.; et al. Genetic basis of congenital erythrocytosis: Mutation update and online databases. Hum. Mutat. 2014, 35, 15–26. [Google Scholar] [CrossRef] [PubMed]
  6. Huang, L.J.-S.; Shen, Y.-M.; Bulut, G.B. Advances in understanding the pathogenesis of primary familial and congenital polycythaemia. Br. J. Haematol. 2010, 101, 1306–1318. [Google Scholar] [CrossRef]
  7. Remy, I.; Wilson, I.A.; Michnick, S.W. Erythropoietin receptor activation by a ligand-induced conformation change. Science 1999, 283, 990–993. [Google Scholar] [CrossRef]
  8. Morris, R.; Kershaw, N.J.; Babon, J.J. The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci. 2018, 27, 1984–2009. [Google Scholar] [CrossRef]
  9. Florensa, L.; Besses, C.; Woessner, S.; Solé, F.; Acín, P.; Pedro, C.; Sans-Sabrafen, J. Endogenous megakaryocyte and erythroid colony formation from blood in essential thrombocythaemia. Leukemia 1995, 9, 271–273. [Google Scholar]
  10. Richards, S.; Aziz, N.; Bale, S.; Bick, D.; Das, S.; Gastier-Foster, J.; Grody, W.W.; Hegde, M.; Lyon, E.; Spector, E.; et al. Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med. 2015, 17, 405–424. [Google Scholar] [CrossRef]
  11. Peroni, E.; Bertozzi, I.; Gherlinzoni, F.; Stefani, P.M.; Lombardi, A.; Biagetti, G.; Fabris, F.; Randi, M.L. Two novel missense mutations in EPOR gene causes erythrocytosis in two unrelated patients. Br. J. Haematol. 2018, 180, 908–911. [Google Scholar] [CrossRef]
  12. Bento, C.; Almeida, H.; Fernandez-Lago, C.; Ribeiro, M.L. Primary familial congenital erythrocytosis: Two novel EPOR mutations found in Spain. Int. J. Lab. Hematol. 2013, 35, e27–e28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Loganathan, S.E.; Kattaru, S.; Chandrasekhar, C.; Vengamma, B.; Sarma, P.V.G.K. Novel mutations in EPO-R and oxygen-dependent degradation (ODD) domain of EPAS1 genes-a causative reason for Congenital Erythrocytosis. Eur. J. Med. Genet. 2022, 65, 1–20. [Google Scholar] [CrossRef]
  14. Al-Sheikh, M.; Mazurier, E.; Gardie, B.; Casadevall, N.; Galactéros, F.; Goossens, M.; Wajcman, H.; Préhu, C.; Ugo, V. A study of 36 unrelated cases with pure erythrocytosis revealed three new mutations in the erythropoietin receptor gene. Haematologica 2008, 93, 1072–1075. [Google Scholar] [CrossRef] [PubMed]
  15. Arcasoy, M.; Karayal, A.; Segal, H.; Sinning, J.; Forget, B. A novel mutation in the erythropoietin receptor gene is associated with familial erythrocytosis. Blood 2002, 99, 3066–3069. [Google Scholar] [CrossRef] [PubMed]
  16. Toriumi, N.; Kaneda, M.; Hatakeyama, N.; Manabe, H.; Okajima, K.; Sakurai, Y.; Yamamoto, M.; Sarashina, T.; Ikuta, K.; Azuma, H. A case of primary familial congenital polycythemia with a novel EPOR mutation: Possible spontaneous remission/alleviation by menstrual bleeding. Int. J. Hematol. 2018, 108, 339–343. [Google Scholar] [CrossRef]
  17. O’Rourke, K.; Fairbairn, D.J.; Jackson, K.A.; Morris, K.L.; Tey, S.K.; Kennedy, G.A. A novel mutation of the erythropoietin receptor gene associated with primary familial and congenital polycythaemia. Int. J. Hematol. 2011, 93, 542–544. [Google Scholar] [CrossRef]
  18. Gross, M.; Ben-Califa, N.; McMullin, M.F.; Percy, M.J.; Bento, C.; Cario, H.; Minkov, M.; Neumann, D. Polycythaemia-inducing mutations in the erythropoietin receptor (EPOR): Mechanism and function as elucidated by epidermal growth factor receptor-EPOR chimeras. Br. J. Haematol. 2014, 165, 519–528. [Google Scholar] [CrossRef]
  19. Perrotta, S.; Cucciolla, V.; Ferraro, M.; Ronzoni, L.; Tramontano, A.; Rossi, F.; Scudieri, A.C.; Borriello, A.; Roberti, D.; Nobili, B.; et al. EPO receptor gain-of-function causes hereditary polycythemia, alters CD34+ cell differentiation and increases circulating endothelial precursors. PLoS ONE 2010, 5, e12015. [Google Scholar] [CrossRef]
  20. Petersen, K.B.; Hokland, P.; Petersen, G.B.; Nyvold, C.G. Erythropoietin receptor defect: A cause of primary polycythaemia. Br. J. Haematol. 2004, 125, 537–538. [Google Scholar] [CrossRef]
  21. Kralovics, R.; Sokol, L.; Prchal, J.T. Absence of polycythemia in a child with a unique erythropoietin receptor mutation in a family with autosomal dominant primary polycythemia. J. Clin. Investig. 1998, 102, 124–129. [Google Scholar] [CrossRef]
  22. Rives, S.; Pahl, H.L.; Florensa, L.; Bellosillo, B.; Neusuess, A.; Estella, J.; Debatin, K.M.; Kohne, E.; Schwarz, K.; Cario, H. Molecular genetic analyses in familial and sporadic congenitalprimary erythrocytosis. Haematologica 2007, 92, 674–677. [Google Scholar] [CrossRef] [PubMed]
  23. Kralovics, R.; Indrak, K.; Stopka, T.; Berman, B.W.; Prchal, J.F.; Prchal, J.T. Two new EPO receptor mutations: Truncated EPO receptors are most frequently associated with primary familial and congenital polycythemias. Blood 1997, 90, 2057–2061. [Google Scholar] [CrossRef] [PubMed]
  24. Watowich, S.; Xie, X.; Klingmüller, U.; Kere, J.; Lindlof, M.; Berglund, S.; De La Chapelle, A. Erythropoietin receptor mutations associated with familial erythrocytosis cause hypersensitivity to erythropoietin in the heterozygous state. Blood 1999, 94, 2530–2532. [Google Scholar] [CrossRef] [PubMed]
  25. Sokol, L.; Luhovy, M.; Guan, Y.; Prchal, J.F.; Semenza, G.L.; Prchal, J.T. Primary familial polycythemia: A frameshift mutation in the erythropoietin receptor gene and increased sensitivity of erythroid progenitors to erythropoietin. Blood 1995, 86, 15–22. [Google Scholar] [CrossRef] [PubMed]
  26. Filser, M.; Aral, B.; Airaud, F.; Chauveau, A.; Bruce, A.; Polfrit, Y.; Thiebaut, A.; Gauthier, M.; le Marechal, C.; Lippert, E.; et al. Low incidence of EPOR mutations in idiopathic erythrocytosis. Haematologica 2021, 106, 299–301. [Google Scholar] [CrossRef]
  27. Furukawa, T.; Narita, M.; Sakaue, M.; Otsuka, T.; Kuroha, T.; Masuko, M.; Azegami, T.; Kishi, K.; Takahashi, M.; Utsumi, J.; et al. Primary familial polycythaemia associated with a novel point mutation in the erythropoietin receptor. Br. J. Haematol. 1997, 99, 222–227. [Google Scholar] [CrossRef]
  28. Pasquier, F.; Marty, C.; Balligand, T.; Verdier, F.; Grosjean, S.; Gryshkova, V.; Raslova, H.; Constantinescu, S.N.; Casadevall, N.; Vainchenker, W.; et al. New pathogenic mechanisms induced by germline erythropoietin receptor mutations in primary erythrocytosis. Haematologica 2017, 103, 575–586. [Google Scholar] [CrossRef]
  29. Chauveau, A.; Luque, D.; Lecucq, L.; Le Gac, G.; Le Maréchal, C.; Gueguen, P.; Berthou, C.; Ugo, V. A new point mutation in EPOR inducing a short deletion incongenital erythrocytosis. Br. J. Haematol. 2015, 172, 461–486. [Google Scholar] [CrossRef]
  30. Bento, C.; Almeida, H.; Maia, T.M.; Relvas, L.; Oliveira, A.C.; Rossi, C.; Girodon, F.; Fernandez-Lago, C.; Aguado-Diaz, A.; Fraga, C.; et al. Molecular study of congenital erythrocytosis in 70 unrelated patients revealed a potential causal mutation in less than half of the cases (Where is/are the missing gene(s)?). Eur. J. Haematol. 2013, 91, 361–368. [Google Scholar] [CrossRef]
  31. De la Chapelle, A.; Traskelin, A.L.; Juvonen, E. Truncated erythropoietin receptor causes dominantly inherited benign human erythrocytosis. Proc. Natl. Acad. Sci. USA 1993, 90, 4495–4499. [Google Scholar] [CrossRef]
  32. Percy, M.J.; McMullin, M.F.; Roques, A.W.; Westwood, N.B.; Acharya, J.; Hughes, A.E.; Lappin, T.R.J.; Pearson, T.C. Erythrocytosis due to a mutation in the erythropoietin receptor gene. Br. J. Haematol. 1998, 100, 407–410. [Google Scholar] [CrossRef] [PubMed]
  33. Le Couedic, J.; Mitjavila, M.; Villeval, J.-L.; Feger, F.; Gobert, S.; Mayeux, P.; Casadevall, N.; Vainchenker, W. Missense Mutation of the Erythropoietin Receptor Is a Rare Event in Human Erythroid Malignancies. Blood 1996, 87, 1502–1511. [Google Scholar] [CrossRef] [PubMed]
  34. Aljabry, M. Primary familial and congenital polycythemia; The forgotten entity. J. Appl. Hematol. 2018, 9, 39. [Google Scholar] [CrossRef]
  35. Bento, C.; McMullin, M.F.; Percy, M.; Cario, H.; Adam, M.; Everman, D.; Mirzaa, G.; Pagon, R.; Wallace, S.; Bean, L.; et al. Primary Familial and Congenital Polycythemia. In GeneReviews [Internet]; University of Washington: Seattle WA, USA, 2016. [Google Scholar] [PubMed]
  36. Camps, C.; Petousi, N.; Bento, C.; Cario, H.; Copley, R.R.; McMullin, M.F.; Van Wijk, R.; Ratcliffe, P.; Robbins, P.A.; Taylor, J.C.; et al. Gene panel sequencing improves the diagnostic work-up of patients with idiopathic erythrocytosis and identifies new mutations. Haematologica 2016, 101, 1306–1318. [Google Scholar] [CrossRef] [PubMed]
  37. Maslah, N.; Cassinat, B.; Verger, E.; Kiladjian, J.J.; Velazquez, L. The role of LNK/SH2B3 genetic alterations in myeloproliferative neoplasms and other hematological disorders. Leukemia 2017, 31, 1661–1670. [Google Scholar] [CrossRef]
  38. Juvonen, E.; Ikkala, E.; Fyhrquist, F.; Ruutu, T. Autosomal Dominant Erythrocytosis Caused by Increased Sensitivity to Erythropoietin. Blood 1991, 78, 3066–3069. [Google Scholar] [CrossRef] [PubMed]
  39. Lacombe, C.; Mayeux, P. Erythropoietin (Epo) receptor and Epo mimetics. Adv. Nephrol. Necker. Hosp. 1999, 29, 177–189. [Google Scholar]
  40. Bittorf, T.; Seiler, J.; Zhang, Z.; Jaster, R.; Brock, J. SHP1 protein tyrosine phosphatase negatively modulates erythroid differentiation and suppression of apoptosis in J2E erythroleukemic cells. Biol. Chem. 1999, 380, 1201–1209. [Google Scholar] [CrossRef]
Figure 1. EPOR pathogenic variants identified in this study.
Figure 1. EPOR pathogenic variants identified in this study.
Genes 13 01686 g001
Table 1. PFCP-associated EPOR variants reported to date.
Table 1. PFCP-associated EPOR variants reported to date.
Exon Nucleotide Change Protein Effect Variant TypeReferences Class
8c.1013G>Ap.(Cys338Tyr)Missense[11]VUS
8c.1022C>Tp.(Thr341Met)Missense[11]VUS
8c.1138C>G p.(Pro380Ala) Missense[12]B
8c.1183G>Cp.(Val395Leu)Missense[13]LB
8c.1142_1143delp.(Pro381Glnfs∗2)Frameshift[14]PAT
8c.1195G>Tp.(Glu399∗)Nonsense[15]PAT
8c.1220C>Ap.(Ser407∗)Nonsense[16]LPAT
8c.1228A>Cp.(Gln343Pro)Missense[13]VUS
8c.1234delp.(Ser412Argfs∗41)Frameshift[17]PAT
8c.1235C>Ap.(Ser412∗)Nonsense[12,18]PAT
8c.1242_1276del p.(Ser415Hisfs∗18)Frameshift[12,18]PAT
8c.1249G>Tp.(Glu417∗)Nonsense[19]PAT
8c.1252 1255del p.(Gly418Profs∗34)Frameshift[20]PAT
8c.1271_1272delp.(Phe424∗)Nonsense[14]PAT
8c.1273G>T p.(Glu425∗)Nonsense[3]LPAT
8c.1275_1290dupp.(Pro431Valfs*19)FrameshiftPresent studyPAT
8c.1278C>Gp.(Tyr426∗)Nonsense[21,22] PAT
8c.1281dupp.(Ile428Tyrfs∗17)Frameshift[23]PAT
8c.1283_1289dupp.(Ser432Glyfs*15)Frameshift[24]PAT
8c.1285delp.(Leu429Trpfs∗24)Frameshift[14]PAT
8c.1288dupp.(Asp430Glyfs∗15)Frameshift[25] PAT
8c.1293delp.(Ser432Alafs∗21)Frameshift[26]PAT
8c.1299_1305delp.(Gln434Cysfs∗17)Frameshift[15,23] PAT
8c.1300C>Tp.(Gln434∗)Nonsense[27]PAT
8c.1300dupp.(Gln434Profs*11)Frameshift[28]PAT
8c.1310G>Ap.(Arg437His)Missense[12,18,29] LB
8c.1311_1312delp.(Pro438Metfs∗6)Frameshift[30]PAT
8c.1316G>Ap.(Trp439∗)Nonsense[31,32] PAT
8c.1317G>Ap.(Trp439∗)Nonsense[22]PAT
8c.1346delp.(Pro449Hisfs∗4)FrameshiftPresent study PAT
8c.1362C>Gp.(Tyr454∗)Nonsense[29]LPAT
8c.1460A>Gp.(Asn487Ser)Missense[14,33] LB
8c.1462C>Tp.(Pro488Ser)Missense[23,25]LPAT
Abbreviations: B, benign; LB, likely benign; VUS, variant of uncertain significance; LPAT, likely pathogenic; PAT, pathogenic.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lo Riso, L.; Vargas-Parra, G.; Navarro, G.; Arenillas, L.; Fernández-Ibarrondo, L.; Robredo, B.; Ballester, C.; López, B.; Perez-Montaña, A.; Sampol, A.; et al. Identification of Two Novel EPOR Gene Variants in Primary Familial Polycythemia: Case Report and Literature Review. Genes 2022, 13, 1686. https://0-doi-org.brum.beds.ac.uk/10.3390/genes13101686

AMA Style

Lo Riso L, Vargas-Parra G, Navarro G, Arenillas L, Fernández-Ibarrondo L, Robredo B, Ballester C, López B, Perez-Montaña A, Sampol A, et al. Identification of Two Novel EPOR Gene Variants in Primary Familial Polycythemia: Case Report and Literature Review. Genes. 2022; 13(10):1686. https://0-doi-org.brum.beds.ac.uk/10.3390/genes13101686

Chicago/Turabian Style

Lo Riso, Laura, Gardenia Vargas-Parra, Gemma Navarro, Leonor Arenillas, Lierni Fernández-Ibarrondo, Beatriz Robredo, Carmen Ballester, Bernardo López, Albert Perez-Montaña, Antonia Sampol, and et al. 2022. "Identification of Two Novel EPOR Gene Variants in Primary Familial Polycythemia: Case Report and Literature Review" Genes 13, no. 10: 1686. https://0-doi-org.brum.beds.ac.uk/10.3390/genes13101686

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop