Go to JCI Insight
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
  • Clinical Research and Public Health
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All ...
  • Videos
    • Conversations with Giants in Medicine
    • Video Abstracts
  • Reviews
    • View all reviews ...
    • Complement Biology and Therapeutics (May 2025)
    • Evolving insights into MASLD and MASH pathogenesis and treatment (Apr 2025)
    • Microbiome in Health and Disease (Feb 2025)
    • Substance Use Disorders (Oct 2024)
    • Clonal Hematopoiesis (Oct 2024)
    • Sex Differences in Medicine (Sep 2024)
    • Vascular Malformations (Apr 2024)
    • View all review series ...
  • Viewpoint
  • Collections
    • In-Press Preview
    • Clinical Research and Public Health
    • Research Letters
    • Letters to the Editor
    • Editorials
    • Commentaries
    • Editor's notes
    • Reviews
    • Viewpoints
    • 100th anniversary
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • Reviews
  • Review series
  • Conversations with Giants in Medicine
  • Video Abstracts
  • In-Press Preview
  • Clinical Research and Public Health
  • Research Letters
  • Letters to the Editor
  • Editorials
  • Commentaries
  • Editor's notes
  • Reviews
  • Viewpoints
  • 100th anniversary
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
Top
  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal
  • Top
  • Abstract
  • Beyond stem cell intrinsic defects in IBMFS
  • The value of exploring rare pathways in IBMFS
  • The mysteries of OSM unraveled
  • The quest for new genetic causes of IBMFS
  • Discovering the unwritten chapters of IBMFS
  • Acknowledgments
  • Footnotes
  • References
  • Version history
Article has an altmetric score of 4

See more details

Posted by 6 X users
Referenced by 1 Bluesky users
  • Article usage
  • Citations to this article (0)

Advertisement

Commentary Open Access | 10.1172/JCI190955

Oncostatin M silence and neopeptide: the value of exploring patients with rare inherited bone marrow failure

Selket Delafontaine1,2 and Isabelle Meyts1,2

1Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.

2Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium.

Address correspondence to: Isabelle Meyts, Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49, 3000 Leuven, Belgium. Phone: 321634384; Email: isabelle.meyts@uzleuven.be.

Find articles by Delafontaine, S. in: JCI | PubMed | Google Scholar |

1Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.

2Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium.

Address correspondence to: Isabelle Meyts, Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49, 3000 Leuven, Belgium. Phone: 321634384; Email: isabelle.meyts@uzleuven.be.

Find articles by Meyts, I. in: JCI | PubMed | Google Scholar |

Published March 17, 2025 - More info

Published in Volume 135, Issue 6 on March 17, 2025
J Clin Invest. 2025;135(6):e190955. https://doi.org/10.1172/JCI190955.
© 2025 Delafontaine, et al. This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
Published March 17, 2025 - Version history
View PDF

Related article:

Human oncostatin M deficiency underlies an inherited severe bone marrow failure syndrome
Alexandrine Garrigue, … , Chantal Lagresle-Peyrou, Patrick Revy
Alexandrine Garrigue, … , Chantal Lagresle-Peyrou, Patrick Revy
Human OSM homozygous loss-of-function variants cause profound anemia, thrombocytopenia and neutropenia Human OSM-deficient hematopoietic stem and progenitor cells can differentiate into erythroblasts and megakaryocytes
Research Article Genetics Hematology Article has an altmetric score of 14

Human oncostatin M deficiency underlies an inherited severe bone marrow failure syndrome

  • Text
  • PDF
Abstract

Oncostatin M (OSM) is a cytokine with the unique ability to interact with both the OSM receptor (OSMR) and the leukemia inhibitory factor receptor (LIFR). On the other hand, OSMR interacts with IL31RA to form the interleukin-31 receptor. This intricate network of cytokines and receptors makes it difficult to understand the specific function of OSM. While monoallelic loss-of-function (LoF) mutations in OSMR underlie autosomal dominant familial primary localized cutaneous amyloidosis, the in vivo consequences of human OSM deficiency have never been reported so far. Here, we identified 3 young individuals from a consanguineous family presenting with inherited severe bone marrow failure syndromes (IBMFS) characterized by profound anemia, thrombocytopenia, and neutropenia. Genetic analysis revealed a homozygous 1 base-pair insertion in the sequence of OSM associated with the disease. Structural and functional analyses showed that this variant causes a frameshift that replaces the C-terminal portion of OSM, which contains the FxxK motif that interacts with both OSMR and LIFR, with a neopeptide. The lack of detection and signaling of the mutant OSM suggests a LoF mutation. Analysis of zebrafish models further supported the role of the OSM/OSMR signaling in erythroid progenitor proliferation and neutrophil differentiation. Our study provides the previously uncharacterized and unexpectedly limited in vivo consequence of OSM deficiency in humans.

Authors

Alexandrine Garrigue, Laëtitia Kermasson, Sandrine Susini, Ingrid Fert, Christopher B. Mahony, Hanem Sadek, Sonia Luce, Myriam Chouteau, Marina Cavazzana, Emmanuelle Six, Marie-Caroline Le Bousse-Kerdilès, Adrienne Anginot, Jean-Baptiste Souraud, Valérie Cormier-Daire, Marjolaine Willems, Anne Sirvent, Jennifer Russello, Isabelle Callebaut, Isabelle André, Julien Y. Bertrand, Chantal Lagresle-Peyrou, Patrick Revy

×

Abstract

Inherited bone marrow failure syndromes (IBMFSs) encompass a diverse group of hematological disorders characterized by a progressive single-lineage cytopenia or pancytopenia. Despite their heterogeneity, these syndromes often result from genetic errors affecting key biological mechanisms, including telomere maintenance, DNA repair and chromosomal stability, and ribosome assembly, generally leading to accelerated apoptosis of hematopoietic cells. Nevertheless, a genetic diagnosis remains elusive in more than half of the cases. The increased risk of myelodysplastic syndrome (MDS), acute leukemia, and solid tumors associated with IBMFS frequently prompts early hematopoietic stem cell transplantation (HSCT). In this issue of the JCI, Garrigue, Kermasson, and colleagues identified a homozygous variant in Oncostatin M (OSM) in 3 children from a consanguineous family presenting with IBMFS characterized by profound anemia, thrombocytopenia, and neutropenia. The findings suggest that the loss-of-function OSM variant affected hematopoietic stem cell function through changes to the bone marrow microenvironment (BMM).

Beyond stem cell intrinsic defects in IBMFS

In this issue of the JCI, Garrigue, Kermasson, and colleagues demonstrate that human oncostatin M (OSM) deficiency underlies profound anemia, thrombocytopenia, and neutropenia, unraveling another inherited bone marrow failure syndrome (IBMFS) (1). Using a combination of whole-genome homozygosity mapping (WGHM) and whole-exome sequencing (WES), the group identified a homozygous loss-of-function (LoF) variant in three children from a consanguineous family presenting with a particular phenotype of isolated pancytopenia (1) (Figure 1). IBMFSs represent a heterogenous group of rare monogenic disorders characterized by a progressive mono-, bi-, or trilineage cytopenia, often associated with nonhematological manifestations (2, 3). Known genetic causes of IBMFS comprise deleterious germline variants affecting DNA repair, telomere maintenance, and ribosome biogenesis and function, processes which are essential for the self-renewal of hematopoietic stem and progenitor cells (HSPCs) (4). Finally, defects in erythromyeloid differentiation, metabolism, and HSPC homeostasis can also underlie IBFMS. In IBMFSs such as Fanconi anemia, Diamond-Blackfan syndrome, dyskeratosis congenita, constitutional thrombocytopenia, and congenital neutropenia, different stresses elicit a p53-dependent growth arrest, resulting in an accelerated apoptosis of HSPCs (5).

Autosomal recessive OSM deficiency causes an isolated phenotype of IBMFS thFigure 1

Autosomal recessive OSM deficiency causes an isolated phenotype of IBMFS through an alteration of the BMM. OSM is a pleiotropic cytokine, secreted by activated immune cells such as T cells, monocytes, macrophages, and neutrophils. Upon binding to its receptors, composed of gp130 and LIFR and/or gp130 and OSMRb, which are widely expressed in hematopoietic and nonhematopoietic tissue, OSM activates the JAK/STAT, the RAS/MAPK, and the PI3K/AKT signaling pathways. Garrigue, Kermasson, and colleagues (1) demonstrated how a homozygous LoF mutation, found in three children from a consanguineous family, leads to the production of a neopeptide, impeding the interaction with both OSM receptors on HSPCs, which ultimately underlies an IBMFS characterized by profound anemia, neutropenia, and thrombocytopenia. This discovery demonstrates that genetic causes of IBMFS extend beyond HSPC intrinsic defects, as alterations of the BMM and inflammatory cytokines can play a pathogenic role in the development of IBMFS.

Garrigue, Kermasson, and colleagues expand the etiology of IBMFS, demonstrating that next to intrinsic genetic defects affecting HSPCs, genetically defined alterations of the bone marrow microenvironment (BMM) may cause an IBMFS (1). Indeed, patients’ HSPCs differentiated normally into erythroblasts and thrombocytes in vitro, suggesting that OSM deficiency hampers maturation after erythroblast and megakaryocyte differentiation in an HSPC-intrinsic way, or that it alters the BMM. This hypothesis was supported by the normal colony-forming assay results and mega-cult assays using patient cells and previously by decreases in erythroid and megakaryocytic cells following transfer of bone marrow cells from WT mice to OSM receptor-deficient (OSMR-deficient) mice (1, 6). Furthermore, the Garrigue et al. findings support the emerging concept that sustained or aberrant exposure to inflammatory signals, force repeated HSC cycling, which can result in HSC exhaustion and hematopoietic failure (4).

The value of exploring rare pathways in IBMFS

The three patients described by Garrigue, Kermasson, and colleagues were affected by an isolated IBMFS. The disease initially manifested between the ages of three months and six months. One sibling presented with anemia, thrombocytopenia, and neutropenia, whereas the two others presented with anemia and gradually developed thrombocytopenia and/or neutropenia over the following years. Dyserythropoiesis and dysgranulopoiesis became apparent upon cytological bone marrow analysis of two siblings by the age of seven to nine-and-a-half years, and reduced cellular density in one patient by the age of 18 years (1). Using WGHM and WES, the authors identified a homozygous one base-pair insertion in OSM, which segregated with the phenotype. OSM is a pleiotropic cytokine, belonging to the IL-6 subfamily, which has the particularity of signaling through two types of OSM receptors (OSMRs) that are widely expressed in hematopoietic and nonhematopoietic cells (7). OSM is secreted by activated immune cells, such as T cells, monocytes, macrophages, dendritic cells. and neutrophils. Upon binding to its receptors, OSM activates the JAK/STAT, the RAS/MAPK, and the PI3K/AKT signaling pathways. OSM was initially discovered due to its growth-inhibitory effect on tumor cell lines, which led to its naming (7). Meanwhile, it has become evident that OSM has pleiotropic effects, not only on cell proliferation but also on the regulation of inflammation, hematopoiesis, bone remodeling, and neuroprotection (8). Garrigue, Kermasson, and colleagues demonstrate that the homozygous OSM variant, detected in their patients, leads to a frameshift in the C-terminal part of OSM, resulting in a neopeptide. Notably, this neopeptide impeded interaction with the leukemia inhibitory factor receptor (LIFR) and the OSMR subunits, as confirmed in patient cells and CRISPR/Cas9 engineered UT7 cells. Modeling this loss of OSM signaling in splice-morpholino–treated zebrafish unveiled defective erythrocyte and neutrophil production (1). However, the morpholino zebrafish were not thrombocytopenic, pointing to the different thrombocyte development pathways in mammals versus zebrafish. This genetic discovery supports the use of next-generation sequencing to discover germline variants in patients with undefined and isolated IBMFS (5). Since IBMFS predispose to malignant transformation, they may also be discovered in patients affected by aplastic anemia, myelodysplastic syndrome (MDS), or malignancy over a range of ages (9).

The mysteries of OSM unraveled

Abnormal levels of OSM play a crucial role in a multitude of inflammatory diseases. However, since OSM exerts various regulatory effects on inflammation, it may have opposing effects in different diseases. In humans, excessive OSM has been shown to drive inflammation in conditions such as inflammatory bowel disease, multiple sclerosis, rheumatoid arthritis, systemic sclerosis, and chronic inflammatory lung disease (7, 10–12). For a subset of these diseases, clinical trials with monoclonal antibodies directed against OSM are ongoing and small molecule drugs targeting OSM and OSMR are being developed (7, 8). On the other hand, OSM-deficient mice are affected by a reduced hematopoietic progenitor activity in the bone marrow and have a massive accumulation of apoptotic thymocytes with the production of autoantibodies (13).

Human disease linked to genetic OSM deficiency was not previously described. Biallelic LoF mutations in LIFR cause Stüve-Wiedemann syndrome (OMIM #601559), characterized by neonatal skeletal dysplasia, feeding difficulties, respiratory distress, and hyperthermia; extended Stüve-Wiedemann syndrome (OMIM #619751) is caused by biallelic deleterious variants in the gene encoding gp130, showing that gp130 deficiency is mostly due to LIFR signaling deficiency. Cytopenia has been very rarely reported in this condition. Also, IL31RA deficiency and OSMRb deficiency both result in primary localized cutaneous amyloidosis (OMIM #613955 and #105250) (1). Thus, the careful dissection of OSM deficiency as an inborn error unveils a rather limited effect, at least of this single variant causing a LoF in OSM. Nevertheless, the description of OSM deficiency suggests that therapeutic strategies addressing inflammatory disease with complete OSM blockage could result in adverse outcomes. Indeed, the human model of OSM deficiency, as described by Garrigue et al., is that of a progressive IBMFS. Although the patients had almost no extrahematopoietic manifestations, surprisingly, these symptoms may appear with age, and it is possible that the description of additional patients with other or identical genetic causes of OSM deficiency will broaden the phenotype over time.

The quest for new genetic causes of IBMFS

The discovery of new causes of IBMFS, such as the OSM deficiency described in this report by Garrigue, Kermasson, and colleagues, has demonstrated that these conditions might not be as rare as previously thought (1). Next-generation sequencing enables broadening the spectrum of known germline variants causing IBMFS, and functional validation is needed to fully grasp the pathophysiology, to comprehend the natural evolution of these disorders, and ultimately to guide precision medicine. In fact, bone marrow failure has been classified as one of the presenting phenotypes of inborn errors of immunity (IEI) (14, 15). Recent discoveries of genetic causes of IBMFS with associated immune and/or syndromic features include deleterious variants in SNM1B, DUT, and RAD50 (16–18). Recent additions also include (a) LoF mutations in MECOM underlying severe neonatal aplastic anemia without bone marrow dysplasia, radioulnar synostosis, and cardiac abnormalities, necessitating HSCT within the first year of life, (b) homozygous variants in ERCC6L2 causing a mild pancytopenia with possible MDS, and (c) IBMFS due to variants in SAMD9 and SAMD9L, in which hematopoietic somatic mosaicism can lead to attenuated bone marrow failure, denoting the need for HSCT (5, 19). On the other hand, for some less well-known causes of IBMFS, such as ADA2 deficiency, the precise pathological mechanism of bone marrow failure remains elusive and HSCT remains the only curative option until now (20, 21). Indeed, whereas TNF inhibition is mostly effective in treating the vasculitis in these patients, the bone marrow failure does not respond to it (22). The discovery of human autosomal recessive (AR) OSM deficiency underscores the importance of the BMM in the development of bone marrow failure and opens new avenues in the study of the pathophysiology of this and other ill-explained IBMFS.

Discovering the unwritten chapters of IBMFS

The discovery of OSM deficiency as an underlying cause of IBMFS by Garrigue, Kermasson, and colleagues demonstrates that genetic causes of IBMFS extend beyond HSPC intrinsic defects, as alterations of the BMM can also result in IBMFS, and highlights inflammatory cytokines as playing a pathogenic role in the development of IBMFS. Unravelling the underlying pathophysiology of IBMFS is primordial to guide precision medicine and treatment. Although allogeneic hematopoietic stem cell transplantation (HSCT) remains a curative option for the hematological manifestations of IBMFS and provides malignancy prevention, its extrahematopoietic manifestations of IBMFS necessitate a careful risk-benefit evaluation. Novel therapeutic options might target the BMM or explore treatment alternatives beyond transplantation (23). Finally, the insights gained from OSM deficiency should prompt caution regarding the use of therapies inhibiting OSM or OSMR in common inflammatory diseases.

Acknowledgments

IM is a Senior Clinical Investigator at the Research Foundation–Flanders (FWO), and is supported by the CSL Behring Chair of Primary Immunodeficiencies, by KU Leuven C1 Grant C16/18/007, by FWO grants G0B5120N and G0E8420N, and by the Jeffrey Modell Foundation. This project has received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 Research and Innovation Programme (grant agreement No. 948959). This work is supported by the European Reference Network on Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (ERN-RITA).

Address correspondence to: Isabelle Meyts, Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49, 3000 Leuven, Belgium. Phone: 321634384; Email: isabelle.meyts@uzleuven.be.

Footnotes

Conflict of interest: KU Leuven LRD has received advisory honorary for IM by Boehringer-Ingelheim and Takeda. IM has received research funding from CSL-Behring. IM is an inventor of the patent EP23201683.2 pending at KU Leuven.

Copyright: © 2025, Delafontaine et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.

Reference information: J Clin Invest. 2025;135(6):e190955. https://doi.org/10.1172/JCI190955.

See the related article at Human oncostatin M deficiency underlies an inherited severe bone marrow failure syndrome.

References
  1. Garrigue A, et al. Human oncostatin M deficiency underlies an inherited severe bone marrow failure syndrome. J Clin Invest. 2025;135(6):e180981.
    View this article via: JCI PubMed Google Scholar
  2. Dokal I, et al. Inherited bone marrow failure in the pediatric patient. Blood. 2022;140(6):556–570.
    View this article via: CrossRef PubMed Google Scholar
  3. Narla A. When to worry about inherited bone marrow failure and myeloid malignancy predisposition syndromes in the setting of a hypocellular marrow. Hematology Am Soc Hematol Educ Program. 2021;2021(1):153–156.
    View this article via: CrossRef PubMed Google Scholar
  4. Wang J, et al. The role of inflammation in hematopoiesis and bone marrow failure: What can we learn from mouse models? Front Immunol. 2022;13:951937.
    View this article via: CrossRef PubMed Google Scholar
  5. Bluteau O, et al. A landscape of germ line mutations in a cohort of inherited bone marrow failure patients. Blood. 2018;131(7):717–732.
    View this article via: CrossRef PubMed Google Scholar
  6. Sato F, et al. Oncostatin M maintains the hematopoietic microenvironment in the bone marrow by modulating adipogenesis and osteogenesis. PLoS One. 2014;9(12):e116209.
    View this article via: CrossRef PubMed Google Scholar
  7. Han L, et al. Multifaceted oncostatin M: novel roles and therapeutic potential of the oncostatin M signaling in rheumatoid arthritis. Front Immunol. 2023;14:1258765.
    View this article via: CrossRef PubMed Google Scholar
  8. Wolf CL, et al. The clinical relevance of OSM in inflammatory diseases: a comprehensive review. Front Immunol. 2023;14:1239732.
    View this article via: CrossRef PubMed Google Scholar
  9. Kawashima N, et al. The molecular and genetic mechanisms of inherited bone marrow failure syndromes: the role of inflammatory cytokines in their pathogenesis. Biomolecules. 2023;13(8):1249.
    View this article via: CrossRef PubMed Google Scholar
  10. West NR, et al. Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor-neutralizing therapy in patients with inflammatory bowel disease. Nat Med. 2017;23(5):579–589.
    View this article via: CrossRef PubMed Google Scholar
  11. Richards CD, Botelho F. Oncostatin M in the regulation of connective tissue cells and macrophages in pulmonary disease. Biomedicines. 2019;7(4):95.
    View this article via: CrossRef PubMed Google Scholar
  12. Hermans D, et al. Oncostatin M triggers brain inflammation by compromising blood-brain barrier integrity. Acta Neuropathol. 2022;144(2):259–281.
    View this article via: CrossRef PubMed Google Scholar
  13. Esashi E, et al. Oncostatin M deficiency leads to thymic hypoplasia, accumulation of apoptotic thymocytes and glomerulonephritis. Eur J Immunol. 2009;39(6):1664–1670.
    View this article via: CrossRef PubMed Google Scholar
  14. Tangye SG, et al. Human inborn errors of immunity: 2022 update on the classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol. 2022;42(7):1473–507.
    View this article via: CrossRef PubMed Google Scholar
  15. Bousfiha A, et al. The 2022 Update of IUIS Phenotypical Classification for Human Inborn Errors of Immunity. J Clin Immunol. 2022;42(7):1508–1520.
    View this article via: CrossRef PubMed Google Scholar
  16. Kermasson L, et al. Inherited human Apollo deficiency causes severe bone marrow failure and developmental defects. Blood. 2022;139(16):2427–2440.
    View this article via: CrossRef PubMed Google Scholar
  17. Dos Santos RS, et al. dUTPase (DUT) is mutated in a novel monogenic syndrome with diabetes and bone marrow failure. Diabetes. 2017;66(4):1086–1096.
    View this article via: CrossRef PubMed Google Scholar
  18. Takagi M, et al. Bone marrow failure and immunodeficiency associated with human RAD50 variants. J Clin Immunol. 2023;43(8):2136–2145.
    View this article via: CrossRef PubMed Google Scholar
  19. Sahoo SS, et al. Clinical evolution, genetic landscape and trajectories of clonal hematopoiesis in SAMD9/SAMD9L syndromes. Nat Med. 2021;27(10):1806–1817.
    View this article via: CrossRef PubMed Google Scholar
  20. Hashem H, et al. Hematopoietic cell transplantation cures adenosine deaminase 2 deficiency: report on 30 patients. J Clin Immunol. 2021;41(7):1633–1647.
    View this article via: CrossRef PubMed Google Scholar
  21. Van Eyck L, et al. Hematopoietic stem cell transplantation rescues the immunologic phenotype and prevents vasculopathy in patients with adenosine deaminase 2 deficiency. J Allergy Clin Immunol. 2015;135(1):283–287.
    View this article via: CrossRef PubMed Google Scholar
  22. Lee PY, et al. Evaluation and management of deficiency of adenosine deaminase 2: an international consensus statement. JAMA Netw Open. 2023;6(5):e2315894.
    View this article via: CrossRef PubMed Google Scholar
  23. Calado RT, Clé DV. Treatment of inherited bone marrow failure syndromes beyond transplantation. Hematology Am Soc Hematol Educ Program. 2017;2017(1):96–101.
    View this article via: CrossRef PubMed Google Scholar
Version history
  • Version 1 (March 17, 2025): Electronic publication

Article tools

  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal

Metrics

Article has an altmetric score of 4
  • Article usage
  • Citations to this article (0)

Go to

  • Top
  • Abstract
  • Beyond stem cell intrinsic defects in IBMFS
  • The value of exploring rare pathways in IBMFS
  • The mysteries of OSM unraveled
  • The quest for new genetic causes of IBMFS
  • Discovering the unwritten chapters of IBMFS
  • Acknowledgments
  • Footnotes
  • References
  • Version history
Advertisement
Advertisement

Copyright © 2025 American Society for Clinical Investigation
ISSN: 0021-9738 (print), 1558-8238 (online)

Sign up for email alerts

Blogged by 1
Posted by 11 X users
Referenced by 3 Bluesky users
2 readers on Mendeley
See more details
Posted by 6 X users
Referenced by 1 Bluesky users
See more details