1
|
Dunbar M, Agarwal S, Venkatesan C, Vollmer B, Scelsa B, Pardo AC, Tarui T, Hart AR, Mulkey SB, Lemmon ME, Gano D. Fetal intracerebral hemorrhage: review of the literature and practice considerations. Pediatr Res 2025:10.1038/s41390-025-04000-5. [PMID: 40097829 DOI: 10.1038/s41390-025-04000-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025]
Abstract
Fetal intracerebral hemorrhage is increasingly recognized on prenatal imaging. In this review, we discuss clinically relevant aspects of fetal intracerebral hemorrhage, including germinal matrix-intraventricular hemorrhage, as well as intraparenchymal hemorrhage. We discuss current clinical practice for prenatal counseling and postnatal management of fetal intracerebral hemorrhage, and offer practical recommendations for clinicians. We propose standardized terminology for classification of fetal intracerebral hemorrhage to be used in future research. We also highlight gaps in the literature and priorities for future research, namely the need for prospective large-scale studies to better understand underlying etiologies and neurodevelopmental outcomes in fetal intracerebral hemorrhage. IMPACT STATEMENT: We discuss the diverse etiologies and outcomes of fetal intracerebral hemorrhage, and propose standardized terminology for classification. We outline current practice and offer practical recommendations for management and counseling of fetal intracerebral hemorrhage, recognizing the need for capacity-building in the newly emerging subspecialty of fetal neurology. We highlight gaps in the literature and research priorities in fetal intracerebral hemorrhage to promote collaborative research, and the development of interventions to improve pregnancy and child outcomes.
Collapse
Affiliation(s)
- Mary Dunbar
- Department of Pediatrics, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Sonika Agarwal
- Division of Neurology & Pediatrics, Children's Hospital of Philadelphia; Division of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Charu Venkatesan
- Division of Neurology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brigitte Vollmer
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton; Paediatric and Neonatal Neurology, Southampton Children's Hospital, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Barbara Scelsa
- Department of Pediatric Neurology, Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Andrea C Pardo
- Department of Pediatrics, Division of Neurology, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tomo Tarui
- Division of Pediatric Neurology, Hasbro Children's Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Anthony R Hart
- Department of Paediatric Neurology, King's College Hospital NHS Foundation Trust, London, UK
| | - Sarah B Mulkey
- Departments of Neurology and Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Zickler Family Prenatal Pediatrics Institute, Children's National Hospital, Washington, DC, USA
| | - Monica E Lemmon
- Department of Pediatrics and Population Health Sciences, Duke University School of Medicine, Durham, NC, England
| | - Dawn Gano
- Department of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Chen Y, Wu L, Yang L, Chen J, Gu X, Zhang Y, Lin Q. Decitabine facilitates thrombopoiesis independent of the thrombopoietin receptor in zebrafish. BLOOD SCIENCE 2025; 7:e00216. [PMID: 39949500 PMCID: PMC11822343 DOI: 10.1097/bs9.0000000000000216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/26/2024] [Indexed: 02/16/2025] Open
Affiliation(s)
- Yudong Chen
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Liangliang Wu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Lian Yang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Jiajun Chen
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xiaofan Gu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Yiyue Zhang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Qing Lin
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
- Department of Hematology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong China
| |
Collapse
|
3
|
Gano D, Glenn OA, Rand L, Heraty K, Devine P, Norton ME, Sparks TN. Fetal Intracranial Hemorrhage due to Uniparental Disomy and Unmasked MPL-Related Congenital Amegakaryocytic Thrombocytopenia. Prenat Diagn 2024. [PMID: 39739357 DOI: 10.1002/pd.6737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Affiliation(s)
- Dawn Gano
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Orit A Glenn
- Department of Radiology and Biomedical Imaging, San Francisco, California, USA
| | - Larry Rand
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| | - Kyle Heraty
- Fetal Treatment Center, UCSF Benioff Children's Hospitals, San Francisco, California, USA
| | - Patrick Devine
- Genomic Medicine Laboratory, University of California San Francisco, San Francisco, California, USA
| | - Mary E Norton
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| | - Teresa N Sparks
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
4
|
Hussain MA, Das SP, Kulkarni M, Laha S. A review on the functional characteristics of the c-Myeloproliferative Leukaemia (c-MPL) gene and its isoforms. Cell Oncol (Dordr) 2024; 47:1607-1626. [PMID: 39283476 DOI: 10.1007/s13402-024-00988-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/11/2024] Open
Abstract
The c-MPL-TPO axis regulates hematopoiesis by activating various signalling cascades, including JAK/STAT, MAPK/ERK, and PIK3/AKT. Here, we have summarized how TPO is regulated by c-MPL and, how mutations in the c-MPL regulate hematopoiesis. We also focus on its non-hematological regulatory role in diseases like Unstable Angina and pathways like DNA damage repair, skeletal homeostasis, & apoptotic regulation of neurons/HSCs at the embryonic state. We discuss the therapeutic efficiency of c-MPL and, its potential to be developed as a bio-marker for detecting metastasis and development of chemo-resistance in various cancers, justifying the multifaceted nature of c-MPL. We have also highlighted the importance of c-MPL isoforms and their stoichiometry in controlling the HSC quiescent and proliferative state. The regulation of the ratio of different isoforms through gene-therapy can open future therapeutic avenues. A systematic understanding of c-MPL-isoforms would undoubtedly take one step closer to facilitating c-MPL from basic-research towards translational medicine.
Collapse
Affiliation(s)
- Mohammad Amjad Hussain
- Cell Biology and Molecular Genetics Division, Yenepoya Research Centre, Yenepoya (Deemed to be) University, 3rd Floor, Academic Block, University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - Shankar Prasad Das
- Cell Biology and Molecular Genetics Division, Yenepoya Research Centre, Yenepoya (Deemed to be) University, 3rd Floor, Academic Block, University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - Mithila Kulkarni
- Cell Biology and Molecular Genetics Division, Yenepoya Research Centre, Yenepoya (Deemed to be) University, 3rd Floor, Academic Block, University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - Suparna Laha
- Cell Biology and Molecular Genetics Division, Yenepoya Research Centre, Yenepoya (Deemed to be) University, 3rd Floor, Academic Block, University Road, Deralakatte, Mangalore, Karnataka, 575018, India.
| |
Collapse
|
5
|
Aldebert C, Fahd M, Galimard JE, Ghemlas IA, Zecca M, Silva J, Mohseny A, Kupesiz A, Hamladji RM, Miranda N, Güngör T, Wynn RF, Merli P, Sundin M, Faraci M, Diaz-de-Heredia C, Burkhardt B, Bordon V, Angoso M, Bader P, Ifversen M, Herrera Arroyo C, Maximova N, Riesco S, Stein J, Dalissier A, Locatelli F, Kalwak K, Dalle JH, Corbacioglu S. Outcomes of patients undergoing allogeneic haematopoietic stem cell transplantation for congenital amegakaryocytic thrombocytopenia; a study on behalf of the PDWP of the EBMT. Bone Marrow Transplant 2024:10.1038/s41409-024-02416-x. [PMID: 39289521 DOI: 10.1038/s41409-024-02416-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 07/27/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
Congenital amegakaryocytic thrombocytopenia is a rare, inherited bone marrow failure syndrome. Allogeneic haematopoietic stem cell transplantation (allo-HSCT) is currently the only curative treatment. In this retrospective study, we analysed 66 patients with allo-HSCT, reported in the European Society for Blood and Marrow Transplantation (EBMT) registry. Bone marrow (BM) was the most widely used stem cell source (n = 40; 61%) followed by peripheral blood (PB) (n = 18; 27%), and unrelated umbilical cord blood (UCB) (n = 8; 12%). Most frequently was a HLA-matched graft from related (n = 26; 39%) and unrelated (n = 15; 23%) donors after a myeloablative busulfan-based conditioning regimen. GvHD prophylaxis was mostly cyclosporine and methotrexate (53%). The 6-year cumulative incidence of graft-failure and second transplant were 25% and 17%, respectively. The 6-year disease-free survival (DFS) and overall survival (OS) were 66.9% and 85.6%, respectively. The 6-year transplant-related mortality (TRM) was 8.0%. In conclusion, most patients with CAMT benefit from allo-HSCT, but with many graft failures.
Collapse
Affiliation(s)
- Clémence Aldebert
- Paediatric Haematology and Immunology Department, Robert-Debré Hospital, GHU APHP Nord-Université de Paris, Paris, France.
| | - Mony Fahd
- Paediatric Haematology and Immunology Department, Robert-Debré Hospital, GHU APHP Nord-Université de Paris, Paris, France
| | - Jacques-Emmanuel Galimard
- EBMT Statistical Unit, Sorbonne University, Saint-Antoine Hospital, AP-HP, INSERM UMRs 938, Paris, France
| | - Ibrahim A Ghemlas
- King Faisal Specialist Hospital & Research Centre (Paeds), Riyadh, Saudi Arabia
| | - Marco Zecca
- Fondazione IRCCS Policlinico San Matteo Paediatric Haematology-Oncology, Pavia, Italy
| | - Juliana Silva
- Great Ormond Street Hospital-Bone Marrow Transplant Department, London, UK
| | - Alexander Mohseny
- Willem-Alexander Children's Hospital, Division of Stem Cell Transplantation, Leiden University Medical Center, Leiden, The Netherlands
| | - Alphan Kupesiz
- Paediatric Haematology, Akdeniz University Medical School, Antalya, Turkey
| | - Rose-Marie Hamladji
- Service Hématologie Greffe de Moëlle, Centre Pierre Et Marie Curie, Algiers, Algeria
| | - Nuno Miranda
- BMT, Instituto Portugues de Oncologia, Lisbon, Portugal
| | - Tayfun Güngör
- Division of Stem Cell Transplantation, The Hospital of the Eleonore Foundation, University Children's Hospital, Zürich, Switzerland
| | - Robert F Wynn
- Department of Paediatric Haematology, Bone Marrow Unit, Royal Manchester Children's Hospital, Manchester, UK
| | - Pietro Merli
- Department of Paediatric Haematology and Oncology, IRRCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Mikael Sundin
- Division Paediatric Haematology, Immunology and HCT, Astrid Lindgren Children's Hospital, Karolinska University, Stockholm, Sweden
| | - Maura Faraci
- Department of Haematology and Oncology, HSCT Unit, IRCCS Institute G. Gaslini, Genoa, Italy
| | - Cristina Diaz-de-Heredia
- Department of Paediatric Haematology and Oncology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Birgit Burkhardt
- Paediatric Haematology Oncology and BMT, University Hospital Münster, Münster, Germany
| | - Victoria Bordon
- Department of Paediatric Haematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Marie Angoso
- Department of Paediatric Haematology/Oncology, CHU Bordeaux, Bordeaux, France
| | - Peter Bader
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, Goethe University, University Hospital Frankfurt, Frankfurt, Germany
| | - Marianne Ifversen
- Department of Paediatric and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Concepcion Herrera Arroyo
- Department of Haematology, Reina Sofia University Hospital, IMIBIC, University of Cordoba, Cordoba, Spain
| | - Natalia Maximova
- Bone Marrow Transplant Centre, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Susana Riesco
- Department of Paediatrics, Hospital Universitario de Salamanca - IBSAL, Salamanca, Spain
| | - Jerry Stein
- Department of Haemato-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Arnaud Dalissier
- EBMT Paris Study Unit, Sorbonne University, Saint-Antoine Hospital, AP-HP, INSERM UMRs 938, Paris, France
| | - Franco Locatelli
- Department of Paediatric Haematology and Oncology, IRRCS Ospedale Pediatrico Bambino Gesù, Rome, Catholic University of the Sacred Heart, Rome, Italy
| | - Krzysztof Kalwak
- Department of Paediatric Haematology Oncology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Jean-Hugues Dalle
- Paediatric Haematology and Immunology Department, Robert-Debré Hospital, GHU APHP Nord-Université de Paris, Paris, France
| | - Selim Corbacioglu
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University of Regensburg, Regensburg, Germany
| |
Collapse
|
6
|
Kaushansky K. Thrombopoietin, the Primary Regulator of Platelet Production: From Mythos to Logos, a Thirty-Year Journey. Biomolecules 2024; 14:489. [PMID: 38672505 PMCID: PMC11047867 DOI: 10.3390/biom14040489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Thrombopoietin, the primary regulator of blood platelet production, was postulated to exist in 1958, but was only proven to exist when the cDNA for the hormone was cloned in 1994. Since its initial cloning and characterization, the hormone has revealed many surprises. For example, instead of acting as the postulated differentiation factor for platelet precursors, megakaryocytes, it is the most potent stimulator of megakaryocyte progenitor expansion known. Moreover, it also stimulates the survival, and in combination with stem cell factor leads to the expansion of hematopoietic stem cells. All of these growth-promoting activities have resulted in its clinical use in patients with thrombocytopenia and aplastic anemia, although the clinical development of the native molecule illustrated that "it's not wise to mess with mother nature", as a highly engineered version of the native hormone led to autoantibody formation and severe thrombocytopenia. Finally, another unexpected finding was the role of the thrombopoietin receptor in stem cell biology, including the development of myeloproliferative neoplasms, an important disorder of hematopoietic stem cells. Overall, the past 30 years of clinical and basic research has yielded many important insights, which are reviewed in this paper.
Collapse
Affiliation(s)
- Kenneth Kaushansky
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
7
|
Lee S, Zhan H. Deciphering the differential impact of thrombopoietin/MPL signaling on hematopoietic stem/progenitor cell function in bone marrow and spleen. Stem Cell Reports 2024; 19:211-223. [PMID: 38215758 PMCID: PMC10874852 DOI: 10.1016/j.stemcr.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 01/14/2024] Open
Abstract
Thrombopoietin (TPO) and its receptor MPL play crucial roles in hematopoietic stem cell (HSC) function and platelet production. However, the precise effects of TPO/MPL signaling on HSC regulation in different hematopoietic niches remain unclear. Here, we investigated the effects of TPO/MPL ablation on marrow and splenic hematopoiesis in TPO-/- and MPL-/- mice during aging. Despite severe thrombocytopenia, TPO-/- and MPL-/- mice did not develop marrow failure during a 2-year follow-up. Marrow and splenic HSCs exhibited different responses to TPO/MPL ablation and exogenous TPO treatment. Splenic niche cells compensated for marrow HSC loss in TPO-/- and MPL-/- mice by upregulating CXCL12 levels. These findings provide new insights into the complex regulation of HSCs by TPO/MPL and reveal a previously unknown link between TPO and CXCL12, two key growth factors for HSC maintenance. Understanding the distinct regulatory mechanisms between marrow and spleen hematopoiesis will help to develop novel therapeutic approaches for hematopoietic disorders.
Collapse
Affiliation(s)
- Sandy Lee
- Graduate Program in Molecular & Cellular Pharmacology, Stony Brook University, Stony Brook, NY, USA
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, USA; Medical Service, Northport VA Medical Center, Northport, NY, USA.
| |
Collapse
|
8
|
Bayegi SN, Hamidieh AA, Behfar M, Saghazadeh A, Bozorgmehr M, Tajik N, Delbandi AA, Delavari S, Shekarabi M, Rezaei N. The Reconstitution of T-cells after Allogeneic Hematopoietic Stem Cell Transplant in a Pediatric Patient with Congenital Amegakaryocytic Thrombocytopenia (CAMT). Endocr Metab Immune Disord Drug Targets 2024; 24:265-272. [PMID: 37526450 DOI: 10.2174/1871530323666230801100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/08/2023] [Accepted: 06/27/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND Congenital amegakaryocytic thrombocytopenia (CAMT) is a bone marrow failure syndrome with autosomal recessive inheritance characterized by the lack of megakaryocytes and thrombocytopenia. The cause of the disease is a mutation in the c-Mpl gene, which encodes the thrombopoietin (TPO) receptor. The main treatment for this genetic disorder is an allogeneic hematopoietic stem cell transplant (allo-HSCT). However, transplant-related mortality, development of acute and chronic graft-versushost disease (GvHD), and susceptibility to opportunistic infections are major barriers to transplantation. Delay in the reconstitution of T cells and imbalance in the regeneration of distinct functional CD4 and CD8 T-cell subsets mainly affect post-transplant complications. We report a case of CAMT, who developed acute GvHD but had no signs and symptoms of chronic GvHD following allo-HSCT. CASE PRESENTATION At the age of four, she presented with petechiae and purpura. In laboratory investigations, pancytopenia without organomegaly, and cellularity less than 5% in bone marrow biopsy, were observed. A primary diagnosis of idiopathic aplastic anemia was made, and she was treated with prednisolone, cyclosporine, and anti-thymocyte globulin (ATG), which did not respond. Genetic analysis revealed the mutation c.1481T>G (p. L494W) in exon 10 of the c-Mpl gene, and the diagnosis of CAMT was confirmed. The patient underwent allo-HSCT from a healthy sibling donor. Alloimmunization reactions and immune disorders were present due to long-term treatment with immunosuppressive medications and repeated blood and platelet transfusions. Hence, the regeneration of T-lymphocytes after allo-HSCT was evaluated. CONCLUSION Successful treatment of acute GvHD prevented advancing the condition to chronic GvHD, and this was accompanied by delayed T-cell reconstitution through an increase in Treg:Tcons ratio.
Collapse
Affiliation(s)
- Shideh Namazi Bayegi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Behfar
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amene Saghazadeh
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Systematic Review and Metaanalysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmood Bozorgmehr
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nader Tajik
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Samaneh Delavari
- Systematic Review and Metaanalysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mehdi Shekarabi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
9
|
Collodel L, Chirico M, Cavicchioli P, Gessoni G. A rare case of congenital amegakaryocytic thrombocytopenia associated with possible neonatal alloimmune thrombocytopenia and neutropenia by anti-HLA antibodies. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2024; 22:30-33. [PMID: 36795344 PMCID: PMC10812893 DOI: 10.2450/2023.0240-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/22/2022] [Indexed: 02/17/2023]
Affiliation(s)
- Luca Collodel
- Transfusion Medicine Department, dell’Angelo General Hospital, Mestre, Italy
| | - Michela Chirico
- Pediatrics Ward and Neonatal Critical Care Unit, dell’Angelo General Hospital, Mestre, Italy
| | - Paola Cavicchioli
- Pediatrics Ward and Neonatal Critical Care Unit, dell’Angelo General Hospital, Mestre, Italy
| | - Gianluca Gessoni
- Transfusion Medicine Department, dell’Angelo General Hospital, Mestre, Italy
| |
Collapse
|
10
|
Stefanucci L, Collins J, Sims MC, Barrio-Hernandez I, Sun L, Burren OS, Perfetto L, Bender I, Callahan TJ, Fleming K, Guerrero JA, Hermjakob H, Martin MJ, Stephenson J, Paneerselvam K, Petrovski S, Porras P, Robinson PN, Wang Q, Watkins X, Frontini M, Laskowski RA, Beltrao P, Di Angelantonio E, Gomez K, Laffan M, Ouwehand WH, Mumford AD, Freson K, Carss K, Downes K, Gleadall N, Megy K, Bruford E, Vuckovic D. The effects of pathogenic and likely pathogenic variants for inherited hemostasis disorders in 140 214 UK Biobank participants. Blood 2023; 142:2055-2068. [PMID: 37647632 PMCID: PMC10733830 DOI: 10.1182/blood.2023020118] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023] Open
Abstract
Rare genetic diseases affect millions, and identifying causal DNA variants is essential for patient care. Therefore, it is imperative to estimate the effect of each independent variant and improve their pathogenicity classification. Our study of 140 214 unrelated UK Biobank (UKB) participants found that each of them carries a median of 7 variants previously reported as pathogenic or likely pathogenic. We focused on 967 diagnostic-grade gene (DGG) variants for rare bleeding, thrombotic, and platelet disorders (BTPDs) observed in 12 367 UKB participants. By association analysis, for a subset of these variants, we estimated effect sizes for platelet count and volume, and odds ratios for bleeding and thrombosis. Variants causal of some autosomal recessive platelet disorders revealed phenotypic consequences in carriers. Loss-of-function variants in MPL, which cause chronic amegakaryocytic thrombocytopenia if biallelic, were unexpectedly associated with increased platelet counts in carriers. We also demonstrated that common variants identified by genome-wide association studies (GWAS) for platelet count or thrombosis risk may influence the penetrance of rare variants in BTPD DGGs on their associated hemostasis disorders. Network-propagation analysis applied to an interactome of 18 410 nodes and 571 917 edges showed that GWAS variants with large effect sizes are enriched in DGGs and their first-order interactors. Finally, we illustrate the modifying effect of polygenic scores for platelet count and thrombosis risk on disease severity in participants carrying rare variants in TUBB1 or PROC and PROS1, respectively. Our findings demonstrate the power of association analyses using large population datasets in improving pathogenicity classifications of rare variants.
Collapse
Affiliation(s)
- Luca Stefanucci
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- British Heart Foundation, BHF Centre of Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Janine Collins
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, Barts Health NHS Trust, London, United Kingdom
| | - Matthew C. Sims
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, United Kingdom
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Inigo Barrio-Hernandez
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Luanluan Sun
- Department of Public Health and Primary Care, BHF Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Oliver S. Burren
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Livia Perfetto
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
- Department of Biology and Biotechnology “C.Darwin,” Sapienza University of Rome, Rome, Italy
| | - Isobel Bender
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Tiffany J. Callahan
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY
| | - Kathryn Fleming
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Jose A. Guerrero
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, Barts Health NHS Trust, London, United Kingdom
| | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Maria J. Martin
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - James Stephenson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - NIHR BioResource
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- British Heart Foundation, BHF Centre of Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, Barts Health NHS Trust, London, United Kingdom
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, United Kingdom
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
- Department of Public Health and Primary Care, BHF Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
- Department of Biology and Biotechnology “C.Darwin,” Sapienza University of Rome, Rome, Italy
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- Centre for Genomics Research, Discovery Sciences, AstraZeneca, Cambridge, United Kingdom
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia
- Genomic Medicine, The Jackson Laboratory, Farmington, CT
- Institute for Systems Genomics, University of Connecticut, Farmington, CT
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences RILD Building, University of Exeter Medical School, Exeter, United Kingdom
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
- Heart and Lung Research Institute, University of Cambridge, Cambridge, United Kingdom
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
- Health Data Science Centre, Human Technopole, Milan, Italy
- Haemophilia Centre and Thrombosis Unit, Royal Free London NHS Foundation Trust, London, United Kingdom
- Department of Haematology, Imperial College Healthcare NHS Trust, London, United Kingdom
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, London, United Kingdom
- Department of Haematology, University College London Hospitals NHS Trust, London, United Kingdom
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KULeuven, Leuven, Belgium
- Cambridge Genomics Laboratory, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Kalpana Paneerselvam
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, AstraZeneca, Cambridge, United Kingdom
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia
| | - Pablo Porras
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Peter N. Robinson
- Genomic Medicine, The Jackson Laboratory, Farmington, CT
- Institute for Systems Genomics, University of Connecticut, Farmington, CT
| | - Quanli Wang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Xavier Watkins
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- British Heart Foundation, BHF Centre of Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences RILD Building, University of Exeter Medical School, Exeter, United Kingdom
| | - Roman A. Laskowski
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Pedro Beltrao
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Emanuele Di Angelantonio
- British Heart Foundation, BHF Centre of Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Public Health and Primary Care, BHF Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
- Heart and Lung Research Institute, University of Cambridge, Cambridge, United Kingdom
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, Cambridge, United Kingdom
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, United Kingdom
- Health Data Science Centre, Human Technopole, Milan, Italy
| | - Keith Gomez
- Haemophilia Centre and Thrombosis Unit, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Mike Laffan
- Department of Haematology, Imperial College Healthcare NHS Trust, London, United Kingdom
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, London, United Kingdom
| | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, University College London Hospitals NHS Trust, London, United Kingdom
| | - Andrew D. Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KULeuven, Leuven, Belgium
| | - Keren Carss
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Cambridge Genomics Laboratory, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Nick Gleadall
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Karyn Megy
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Elspeth Bruford
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Dragana Vuckovic
- Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| |
Collapse
|
11
|
Ammeti D, Marzollo A, Gabelli M, Zanchetta ME, Tretti-Parenzan C, Bottega R, Capaci V, Biffi A, Savoia A, Bresolin S, Faleschini M. A novel mutation in MECOM affects MPL regulation in vitro and results in thrombocytopenia and bone marrow failure. Br J Haematol 2023; 203:852-859. [PMID: 37610030 DOI: 10.1111/bjh.19023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/21/2023] [Accepted: 07/21/2023] [Indexed: 08/24/2023]
Abstract
MECOM-associated syndrome (MECOM-AS) is a rare disease characterized by amegakaryocytic thrombocytopenia, progressive bone marrow failure, pancytopenia and radioulnar synostosis with high penetrance. The clinical phenotype may also include finger malformations, cardiac and renal alterations, hearing loss, B-cell deficiency and predisposition to infections. The syndrome, usually diagnosed in the neonatal period because of severe thrombocytopenia, is caused by mutations in the MECOM gene, encoding for the transcription factor EVI1. The mechanism linking the alteration of EVI1 function and thrombocytopenia is poorly understood. In a paediatric patient affected by severe thrombocytopenia, we identified a novel variant of the MECOM gene (p.P634L), whose effect was tested on pAP-1 enhancer element and promoters of targeted genes showing that the mutation impairs the repressive activity of the transcription factor. Moreover, we demonstrated that EVI1 controls the transcriptional regulation of MPL, a gene whose mutations are responsible for congenital amegakaryocytic thrombocytopenia (CAMT), potentially explaining the partial overlap between MECOM-AS and CAMT.
Collapse
Affiliation(s)
- Daniele Ammeti
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Antonio Marzollo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
| | - Maria Gabelli
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
- Maternal and Child Health Department, Padua University, Padua, Italy
| | | | - Caterina Tretti-Parenzan
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
- Maternal and Child Health Department, Padua University, Padua, Italy
| | - Roberta Bottega
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Valeria Capaci
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Alessandra Biffi
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
- Maternal and Child Health Department, Padua University, Padua, Italy
| | - Anna Savoia
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Silvia Bresolin
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
- Maternal and Child Health Department, Padua University, Padua, Italy
| | - Michela Faleschini
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
12
|
Sokkar MF, Hamdy M, Erian PS, Mosaad RM, Elaraby NM, Taher MB, El-Sayed H, Al Komy M, Eid MM, Mohamed AM, Amr KS, El-Kamah GY. Studying the pathogenicity of 26 variants characterized in the first molecular analyses of Egyptian aplastic anemia patients. J Genet Eng Biotechnol 2023; 21:149. [PMID: 38017244 PMCID: PMC10684839 DOI: 10.1186/s43141-023-00585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/31/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Aplastic anemia (AA) is a bone marrow disorder characterized by peripheral pancytopenia and marrow hypoplasia which can lead to life-threatening complications. Our objective was to study the telomerase genes (TERT and TERC) variants, explore their relationship to telomere shortening and TERT gene expression, and to identify variants in the MPL gene within Egyptian AA patients. METHODS Forty AA patients and 40 sex- and age-matched healthy individuals as the control group were studied through sequencing of TERT, TERC, and MPL genes. Quantitative real-time PCR (qRT-PCR) was used for measuring TERT gene expression. Telomere length (TL) was measured using the Quantitative Fluorescence In Situ Hybridization (Q-FISH) technique. In silico analysis was performed for the prediction of the pathogenicity of resultant variants. RESULTS Sequencing of MPL, TERT, and TERC genes identified 26 variants. Eleven variants were identified in the MPL gene. Three of them are pathogenic: two missense [c.305 G>A, c.1589 C>T] and one splice site [g.9130T>G]. TERT gene sequencing showed thirteen variants, among them, four novel [c.484G>A, c.499G>A, c.512G>A, c.3164C>G] and two previously reported [c.835G>A, c.2031C>T] were predicted to be pathogenic. Two variants were characterized within the TERC gene; n.514A>G and n.463 C>T. TERT gene expression was downregulated in 70% of studied patients and the Q-FISH technique detected telomere shortening in 82.5% of patients. CONCLUSIONS Twenty-six pathogenic and benign variants within the TERC, TERT, and MPL genes were identified among the studied AA patients that were in several cases associated with shortened telomeres and/or lower TERT gene expression. Genotype/phenotype correlation in AA patients is of great importance in explaining the disease severity and guiding therapeutic decisions.
Collapse
Affiliation(s)
- Mona F Sokkar
- Molecular Genetics and Enzymology Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt.
| | - Mona Hamdy
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Peter Sf Erian
- Human Cytogenetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Rehab M Mosaad
- Molecular Genetics and Enzymology Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Nesma M Elaraby
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Mohamed B Taher
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Heba El-Sayed
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Mohammed Al Komy
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maha M Eid
- Human Cytogenetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Amal M Mohamed
- Human Cytogenetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Khalda S Amr
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| | - Ghada Y El-Kamah
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre (NRC), Cairo, Egypt
| |
Collapse
|
13
|
Vissers LTW, van der Burg M, Lankester AC, Smiers FJW, Bartels M, Mohseny AB. Pediatric Bone Marrow Failure: A Broad Landscape in Need of Personalized Management. J Clin Med 2023; 12:7185. [PMID: 38002797 PMCID: PMC10672506 DOI: 10.3390/jcm12227185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Irreversible severe bone marrow failure (BMF) is a life-threatening condition in pediatric patients. Most important causes are inherited bone marrow failure syndromes (IBMFSs) and (pre)malignant diseases, such as myelodysplastic syndrome (MDS) and (idiopathic) aplastic anemia (AA). Timely treatment is essential to prevent infections and bleeding complications and increase overall survival (OS). Allogeneic hematopoietic stem cell transplantation (HSCT) provides a cure for most types of BMF but cannot restore non-hematological defects. When using a matched sibling donor (MSD) or a matched unrelated donor (MUD), the OS after HSCT ranges between 60 and 90%. Due to the introduction of post-transplantation cyclophosphamide (PT-Cy) to prevent graft versus host disease (GVHD), alternative donor HSCT can reach similar survival rates. Although HSCT can restore ineffective hematopoiesis, it is not always used as a first-line therapy due to the severe risks associated with HSCT. Therefore, depending on the underlying cause, other treatment options might be preferred. Finally, for IBMFSs with an identified genetic etiology, gene therapy might provide a novel treatment strategy as it could bypass certain limitations of HSCT. However, gene therapy for most IBMFSs is still in its infancy. This review summarizes current clinical practices for pediatric BMF, including HSCT as well as other disease-specific treatment options.
Collapse
Affiliation(s)
- Lotte T. W. Vissers
- Laboratory for Pediatric Immunology, Department of Pediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (L.T.W.V.); (M.v.d.B.)
| | - Mirjam van der Burg
- Laboratory for Pediatric Immunology, Department of Pediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (L.T.W.V.); (M.v.d.B.)
| | - Arjan C. Lankester
- Department of Pediatrics, Hematology and Stem Cell Transplantation, Willem-Alexander Children’s Hospital, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.C.L.); (F.J.W.S.)
| | - Frans J. W. Smiers
- Department of Pediatrics, Hematology and Stem Cell Transplantation, Willem-Alexander Children’s Hospital, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.C.L.); (F.J.W.S.)
| | - Marije Bartels
- Department of Pediatric Hematology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Alexander B. Mohseny
- Department of Pediatrics, Hematology and Stem Cell Transplantation, Willem-Alexander Children’s Hospital, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.C.L.); (F.J.W.S.)
| |
Collapse
|
14
|
Shah A, Kumar C, Shanmukhaiah C, Rajendran A, Mudaliar S, Idicula-Thomas S, Vundinti BR. Genomic and computational analysis of four novel variants of MPL gene in Congenital Amegakaryocytic Thrombocytopenia. Ann Hematol 2023; 102:2683-2693. [PMID: 37438490 DOI: 10.1007/s00277-023-05347-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023]
Abstract
Congenital amegakaryocytic thrombocytopenia (CAMT) is a rare, genetic, autosomal recessive disorder characterized by severe thrombocytopenia, due to inefficient bone marrow megakaryopoiesis eventually leading to aplasia. Majority of the cases are due to homozygous or compound heterozygous mutations in MPL gene encoding for thrombopoietin (THPO) receptor protein. CAMT can be diagnosed at early phase of life, with major complication of transfusion dependency and hematopoietic transplantation as only curative treatment. We have investigated the sequence variations in MPL gene of 7 bone marrow failure (BMF) subjects, who presented with clinically diverse phenotypes, through next generation sequencing (NGS). Plasma THPO levels were estimated using ELISA. Insilico sequence and structure-based analyses were performed to understand the structural and functional implications of mutations, identified through NGS. We studied 7 CAMT subjects suspected of BMF, who presented with severe thrombocytopenia followed by pancytopenia, bleeding manifestation and physical anomalies. The plasma THPO levels were significantly elevated (p<0.05) in all the cases. Molecular analysis by NGS identified 9 genomic mutations in MPL gene. These included 7 non-synonymous substitution, 1 nonsense substitution and 1 in-del mutations, of which 4 are novel mutations. Insilico analysis predicted damaging effects on THPO-R and its reduced affinity for THPO for all the identified mutations. CAMT is a rare disorder with diverse clinical phenotypes and diagnosis is challenging. The elevated plasma THPO levels should be considered for the primary diagnosis and prognosis of the disease. However, molecular analysis of MPL gene is important for the diagnosis and management of the disease through genetic counselling. Though the cytokines, THPO-R agonist are used for the treatment of CAMT, HSCT is the only curative therapy.
Collapse
Affiliation(s)
- Anjali Shah
- Department of Cytogenetics, ICMR-National Institute of Immunohaematology, 13th floor, New Multi-storeyed building, KEM hospital Campus, Parel, Mumbai, Maharashtra, 400012, India
| | - Chandan Kumar
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, 400012, India
| | - Chandrakala Shanmukhaiah
- Department of Haematology, KEM Hospital, 10th Floor, New Multistoried Building, Parel, Mumbai, Maharashtra, 400012, India
| | - Aruna Rajendran
- Department of Pediatric Hematology, Institute of Child Health and Hospital for Children, Chennai, India
| | | | - Susan Idicula-Thomas
- Biomedical Informatics Centre, ICMR-National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, 400012, India
| | - Babu Rao Vundinti
- Department of Cytogenetics, ICMR-National Institute of Immunohaematology, 13th floor, New Multi-storeyed building, KEM hospital Campus, Parel, Mumbai, Maharashtra, 400012, India.
| |
Collapse
|
15
|
Gachard N, Lafage-Pochitaloff M, Quessada J, Auger N, Collonge-Rame MA. Cytogenetics in the management of hematologic neoplasms with germline predisposition: guidelines from the Groupe Francophone de Cytogénétique Hématologique (GFCH). Curr Res Transl Med 2023; 71:103416. [PMID: 37865978 DOI: 10.1016/j.retram.2023.103416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/24/2023]
Abstract
The number of predisposing genes is continuously growing with the widespread availability of DNA sequencing, increasing the prevalence of hematologic malignancies with germline predisposition. Cytogenetic analyses provide an effective approach for the recognition of these malignancies with germline predisposition, which is critical for proper diagnosis, optimal treatment and genetic counseling. Based on the World Health Organization and the international consensus classifications as well as the European LeukemiaNet recommendations, this review first presents an advanced classification of neoplasms with germline predisposition focused on the acquired cytogenetic alterations during leukemogenesis. The various genetic rescue mechanisms and the progression to transformation are then explained. The review also outlines the specific constitutional and somatic cytogenetic aberrations indicative of germline predisposition disorders in B-acute lymphoblastic leukemia (ALL), T-ALL, bone marrow failure syndrome and myeloid neoplasms. An emphasis is made on monosomy 7 in the predisposition field, its frequency and diagnosis impact as well as its various circumstances of occurrence. Lastly, we propose cytogenetic technical recommendations and guidelines for clinical reporting of these specific aberrations.
Collapse
Affiliation(s)
- Nathalie Gachard
- Laboratoire d'hématologie, Centre de Biologie et de Recherche en Santé, CHU de Limoges, Limoges 87042, France; UMR CNRS 7276, INSERM U1262 Université de Limoges, Limoges 87025, France.
| | - Marina Lafage-Pochitaloff
- Laboratoire de Cytogénétique Hématologique, Département d'Hématologie, CHU Timone, APHM, Aix Marseille Université, Marseille 13005, France
| | - Julie Quessada
- Laboratoire de Cytogénétique Hématologique, Département d'Hématologie, CHU Timone, APHM, Aix Marseille Université, Marseille 13005, France
| | - Nathalie Auger
- Laboratoire de Cytogénétique -Génétique des Tumeurs - Gustave Roussy - 144 rue Edouard Vaillant, Villejuif 94805, France
| | - Marie-Agnès Collonge-Rame
- Oncobiologie Génétique Bioinformatique, UF Cytogénétique et Génétique Moléculaire, CHU de Besançon, Besançon 25030, France
| |
Collapse
|
16
|
Capaci V, Adam E, Bar-Joseph I, Faleschini M, Pecci A, Savoia A. Defective binding of ETS1 and STAT4 due to a mutation in the promoter region of THPO as a novel mechanism of congenital amegakaryocytic thrombocytopenia. Haematologica 2023; 108:1385-1393. [PMID: 36226497 PMCID: PMC10153527 DOI: 10.3324/haematol.2022.281392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/30/2022] [Indexed: 11/09/2022] Open
Abstract
Congenital amegakaryocytic thrombocytopenia (CAMT) is a recessive disorder characterized by severe reduction of megakaryocytes and platelets at birth, which evolves toward bone marrow aplasia in childhood. CAMT is mostly caused by mutations in MPL (CAMT-MPL), the gene encoding the receptor of thrombopoietin (THPO), a crucial cytokine regulating hematopoiesis. CAMT can be also due to mutations affecting the THPO coding region (CAMT-THPO). In a child with the clinical picture of CAMT, we identified the homozygous c.-323C>T substitution, affecting a potential regulatory region of THPO. Although mechanisms controlling THPO transcription are not characterized, bioinformatics and in vitro analysis showed that c.-323C>T prevents the binding of transcription factors ETS1 and STAT4 to the putative THPO promoter, impairing THPO expression. Accordingly, in the proband the serum THPO concentration indicates defective THPO production. Based on these findings, the patient was treated with the THPO-mimetic agent eltrombopag, which induced a significant increase in platelet count and stable remission of bleeding symptoms. Herein, we report a novel pathogenic variant responsible for CAMT and provide new insights into the mechanisms regulating transcription of the THPO gene.
Collapse
Affiliation(s)
- Valeria Capaci
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste
| | - Etai Adam
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant-Sheba Medical Center, Hashomer
| | - Ifat Bar-Joseph
- The Center for Cancer Research-Sheba Medical Center, Hashomer
| | | | - Alessandro Pecci
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation; University of Pavia, Pavia
| | - Anna Savoia
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy; Department of Medical Sciences, University of Trieste, Trieste.
| |
Collapse
|
17
|
Vanholder IM, Ecury-Goossen GM, Admiraal J, Porcelijn L, van Spaendonk RM, Gouw SC. Congenital amegakaryocytic thrombocytopenia presenting with a new thrombopoietin receptor (MPL) pathogenic variant: An instructive neonatal case. J Paediatr Child Health 2022; 58:2319-2321. [PMID: 35972311 DOI: 10.1111/jpc.16168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/08/2022] [Accepted: 08/03/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Inez M Vanholder
- Department of Neonatology and Paediatrics, Curaçao Medical Center, J.H.J. Hamelweg, Willemstad, Curaçao.,Department of Paediatrics, University of Antwerp, Edegem, Belgium
| | - Ginette M Ecury-Goossen
- Department of Neonatology and Paediatrics, Curaçao Medical Center, J.H.J. Hamelweg, Willemstad, Curaçao
| | - Jop Admiraal
- Department of Neonatology and Paediatrics, Curaçao Medical Center, J.H.J. Hamelweg, Willemstad, Curaçao.,Department of Paediatrics, University of Amsterdam, Amsterdam, The Netherlands
| | - Leendert Porcelijn
- Immunohematology Diagnostic Services, Sanquin Diagnostic Services, Amsterdam, The Netherlands
| | | | - Samantha C Gouw
- Department of Pediatric Hematology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
18
|
Warren JT, Di Paola J. Genetics of inherited thrombocytopenias. Blood 2022; 139:3264-3277. [PMID: 35167650 PMCID: PMC9164741 DOI: 10.1182/blood.2020009300] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/04/2022] [Indexed: 01/19/2023] Open
Abstract
The inherited thrombocytopenia syndromes are a group of disorders characterized primarily by quantitative defects in platelet number, though with a variety demonstrating qualitative defects and/or extrahematopoietic findings. Through collaborative international efforts applying next-generation sequencing approaches, the list of genetic syndromes that cause thrombocytopenia has expanded significantly in recent years, now with over 40 genes implicated. In this review, we focus on what is known about the genetic etiology of inherited thrombocytopenia syndromes and how the field has worked to validate new genetic discoveries. We highlight the important role for the clinician in identifying a germline genetic diagnosis and strategies for identifying novel causes through research-based endeavors.
Collapse
Affiliation(s)
- Julia T Warren
- Division of Hematology-Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Jorge Di Paola
- Division of Hematology-Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
19
|
Davenport P, Liu ZJ, Sola-Visner M. Fetal vs adult megakaryopoiesis. Blood 2022; 139:3233-3244. [PMID: 35108353 PMCID: PMC9164738 DOI: 10.1182/blood.2020009301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/12/2022] [Indexed: 11/20/2022] Open
Abstract
Fetal and neonatal megakaryocyte progenitors are hyperproliferative compared with adult progenitors and generate a large number of small, low-ploidy megakaryocytes. Historically, these developmental differences have been interpreted as "immaturity." However, more recent studies have demonstrated that the small, low-ploidy fetal and neonatal megakaryocytes have all the characteristics of adult polyploid megakaryocytes, including the presence of granules, a well-developed demarcation membrane system, and proplatelet formation. Thus, rather than immaturity, the features of fetal and neonatal megakaryopoiesis reflect a developmentally unique uncoupling of proliferation, polyploidization, and cytoplasmic maturation, which allows fetuses and neonates to populate their rapidly expanding bone marrow and blood volume. At the molecular level, the features of fetal and neonatal megakaryopoiesis are the result of a complex interplay of developmentally regulated pathways and environmental signals from the different hematopoietic niches. Over the past few years, studies have challenged traditional paradigms about the origin of the megakaryocyte lineage in both fetal and adult life, and the application of single-cell RNA sequencing has led to a better characterization of embryonic, fetal, and adult megakaryocytes. In particular, a growing body of data suggests that at all stages of development, the various functions of megakaryocytes are not fulfilled by the megakaryocyte population as a whole, but rather by distinct megakaryocyte subpopulations with dedicated roles. Finally, recent studies have provided novel insights into the mechanisms underlying developmental disorders of megakaryopoiesis, which either uniquely affect fetuses and neonates or have different clinical presentations in neonatal compared with adult life.
Collapse
Affiliation(s)
- Patricia Davenport
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA; and
- Harvard Medical School, Boston, MA
| | - Zhi-Jian Liu
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA; and
- Harvard Medical School, Boston, MA
| | - Martha Sola-Visner
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA; and
- Harvard Medical School, Boston, MA
| |
Collapse
|
20
|
Chen Y, Xie J, Shen Z, Shi J, Chen S, Wang G. Clinical and molecular characteristics of acute myeloid leukemia with MPL mutation. Hematology 2022; 27:530-534. [PMID: 35544613 DOI: 10.1080/16078454.2022.2066244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES The current study aimed to explore the incidence of MPL mutations and the clinical and molecular characteristics of AML with MPL mutation. METHODS In total, 1509 patients with newly diagnosed AML were retrospectively analyzed between January 2017 and December 2020. MPL mutations were detected via next-generation sequencing. During the same period, we also enrolled 30 patients with other myeloid neoplasms (MNs) with MPL mutation, which included myelodysplastic syndrome (n = 15), myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) (n = 6), and MPN (n = 9). The clinical characteristics of MPL-mutated AML and other types of MNs or MPL-wide type (MPL-wt) AML were compared, and the spectrum of co-mutations and MPL mutation profiles in MPL-mutated AML were analyzed. RESULTS MPL mutations were identified in 19 (1.26%) of 1509 patients with AML. The waterfall diagram showed that the co-mutations were mainly epigenetic modifications (TET2, IDH1, and EZH2), spliceosomes (SRSF2), and transcription factors (RUNX1). The platelet count of the AML group was significantly lower than that of the MPN group (p = 0.001). MPL mutations were commonly observed in the intracellular region in AML but the transmembrane region in MPN (p = 0.013). The MPL-mutated AML group had a lower white blood cell count and a lower rate of complete remission than the MPL wild-type AML group (p = 0.037). CONCLUSION MPL mutations are clinically relevant in patients with AML, and they may be a novel subtype characterized by lower white blood cell counts and poor complete remission rates. However, further studies must be conducted to identify its correlated mechanism.
Collapse
Affiliation(s)
- Yu Chen
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,National Clinical Research Center for Hematologic Diseases, Suzhou, People's Republic of China.,Department of Hematology, the Second Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Jundan Xie
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,National Clinical Research Center for Hematologic Diseases, Suzhou, People's Republic of China
| | - Zhen Shen
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,National Clinical Research Center for Hematologic Diseases, Suzhou, People's Republic of China
| | - Jie Shi
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,National Clinical Research Center for Hematologic Diseases, Suzhou, People's Republic of China
| | - Suning Chen
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,National Clinical Research Center for Hematologic Diseases, Suzhou, People's Republic of China
| | - Gang Wang
- Department of Hematology, the Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| |
Collapse
|
21
|
Coste T, Vincent-Delorme C, Stichelbout M, Devisme L, Gelot A, Deryabin I, Pelluard F, Aloui C, Leutenegger AL, Jouannic JM, Héron D, Gould DB, Tournier-Lasserve E. COL4A1/COL4A2 and inherited platelet disorder gene variants in fetuses showing intracranial hemorrhage. Prenat Diagn 2022; 42:601-610. [PMID: 35150448 PMCID: PMC10434296 DOI: 10.1002/pd.6113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/12/2022] [Accepted: 02/09/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Variants of COL4A1/COL4A2 genes have been reported in fetal intracranial hemorrhage (ICH) cases but their prevalence and characteristics have not been established in a large series of fetuses. Fetal neonatal alloimmune thrombocytopenia is a major acquired ICH factor but the prevalence and characteristics of inherited platelet disorder (IPD) gene variants leading to thrombocytopenia are unknown. Herein, we screened COL4A1/COL4A2 and IPD genes in a large series of ICH fetuses. METHODS A cohort of 194 consecutive ICH fetuses were first screened for COL4A1/COL4A2 variants. We manually curated a list of 64 genes involved in IPD and investigated them in COL4A1/COL4A2 negative fetuses, using exome sequencing data from 101 of these fetuses. RESULT Pathogenic variants of COL4A1/COL4A2 genes were identified in 36 fetuses (19%). They occurred de novo in 70% of the 32 fetuses for whom parental DNA was available. Pathogenic variants in two megakaryopoiesis genes (MPL and MECOM genes) were identified in two families with recurrent and severe fetal ICH, with variable extraneurological pathological features. CONCLUSION Our study emphasizes the genetic heterogeneity of fetal ICH and the need to screen both COL4A1/COL4A2 and IPD genes in the etiological investigation of fetal ICH to allow proper genetic counseling.
Collapse
Affiliation(s)
- Thibault Coste
- AP-HP, Service de génétique moléculaire Neurovasculaire, Hôpital Saint-Louis, Paris, France
- Université de Paris, INSERM UMR-1141 Neurodiderot, Paris, France
| | | | | | | | - Antoinette Gelot
- APHP, Service de fœtopathologie, Hôpital Trousseau, Paris, France
| | - Igor Deryabin
- APHP, Service de fœtopathologie, Hôpital Trousseau, Paris, France
| | - Fanny Pelluard
- University Bordeaux, INSERM, BaRITOn, U1053, Bordeaux, France
| | - Chaker Aloui
- Université de Paris, INSERM UMR-1141 Neurodiderot, Paris, France
| | | | - Jean-Marie Jouannic
- APHP Sorbonne Université, Service de médecine fœtale, Hôpital Trousseau, Paris, France
| | - Delphine Héron
- AP-HP, Service de génétique clinique, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Douglas B Gould
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA
| | - Elisabeth Tournier-Lasserve
- AP-HP, Service de génétique moléculaire Neurovasculaire, Hôpital Saint-Louis, Paris, France
- Université de Paris, INSERM UMR-1141 Neurodiderot, Paris, France
| |
Collapse
|
22
|
Avagyan S, Shimamura A. Lessons From Pediatric MDS: Approaches to Germline Predisposition to Hematologic Malignancies. Front Oncol 2022; 12:813149. [PMID: 35356204 PMCID: PMC8959480 DOI: 10.3389/fonc.2022.813149] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/26/2022] [Indexed: 12/13/2022] Open
Abstract
Pediatric myelodysplastic syndromes (MDS) often raise concern for an underlying germline predisposition to hematologic malignancies, referred to as germline predisposition herein. With the availability of genetic testing, it is now clear that syndromic features may be lacking in patients with germline predisposition. Many genetic lesions underlying germline predisposition may also be mutated somatically in de novo MDS and leukemias, making it critical to distinguish their germline origin. The verification of a suspected germline predisposition informs therapeutic considerations, guides monitoring pre- and post-treatment, and allows for family counseling. Presentation of MDS due to germline predisposition is not limited to children and spans a wide age range. In fact, the risk of MDS may increase with age in many germline predisposition conditions and can present in adults who lack classical stigmata in their childhood. Furthermore, germline predisposition associated with DDX41 mutations presents with older adult-onset MDS. Although a higher proportion of pediatric patients with MDS will have a germline predisposition, the greater number of MDS diagnoses in adult patients may result in a larger overall number of those with an underlying germline predisposition. In this review, we present a framework for the evaluation of germline predisposition to MDS across all ages. We discuss characteristics of personal and family history, clinical exam and laboratory findings, and integration of genetic sequencing results to assist in the diagnostic evaluation. We address the implications of a diagnosis of germline predisposition for the individual, for their care after MDS therapy, and for family members. Studies on MDS with germline predisposition have provided unique insights into the pathogenesis of hematologic malignancies and mechanisms of somatic genetic rescue vs. disease progression. Increasing recognition in adult patients will inform medical management and may provide potential opportunities for the prevention or interception of malignancy.
Collapse
Affiliation(s)
- Serine Avagyan
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, United States
| | - Akiko Shimamura
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
23
|
Oved JH, Shah YB, Venella K, Paessler ME, Olson TS. Non-myeloablative conditioning is sufficient to achieve complete donor myeloid chimerism following matched sibling donor bone marrow transplant for myeloproliferative leukemia virus oncogene ( MPL) mutation-driven congenital amegakaryocytic thrombocytopenia: Case report. Front Pediatr 2022; 10:903872. [PMID: 35967582 PMCID: PMC9366100 DOI: 10.3389/fped.2022.903872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Congenital amegakaryocytic thrombocytopenia (CAMT) is a rare platelet production disorder caused mainly by loss of function biallelic mutations in myeloproliferative leukemia virus oncogene (MPL), the gene encoding the thrombopoietin receptor (TPOR). Patients with MPL-mutant CAMT are not only at risk for life-threatening bleeding events, but many affected individuals will also ultimately develop bone marrow aplasia owing to the absence of thrombopoietin/TPOR signaling required for maintenance of hematopoietic stem cells. Curative allogeneic stem cell transplant for patients with CAMT has historically used myeloablative conditioning; however, given the inherent stem cell defect in MPL-mutant CAMT, a less intensive regimen may prove equally effective with reduced morbidity, particularly in patients with evolving aplasia. METHODS We report the case of a 2-year-old boy with MPL-mutant CAMT and bone marrow hypocellularity who underwent matched sibling donor bone marrow transplant (MSD-BMT) using a non-myeloablative regimen consisting of fludarabine, cyclophosphamide, and antithymocyte globulin (ATG). RESULTS The patient achieved rapid trilinear engraftment and resolution of thrombocytopenia. While initial myeloid donor chimerism was mixed (88% donor), due to the competitive advantage of donor hematopoietic cells, myeloid chimerism increased to 100% by 4 months post-transplant. Donor chimerism and blood counts remained stable through 1-year post-transplant. CONCLUSION This experience suggests that non-myeloablative conditioning is a suitable approach for patients with MPL-mutant CAMT undergoing MSD-BMT and is associated with reduced risks of conditioning-related toxicity compared to traditional myeloablative regimens.
Collapse
Affiliation(s)
- Joseph Hai Oved
- Pediatric Transplantation and Cell Therapy, MSK Kids, New York, NY, United States
| | - Yash B Shah
- Cell Therapy and Transplant Section, Division of Pediatric Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Kimberly Venella
- Cell Therapy and Transplant Section, Division of Pediatric Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Michele E Paessler
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Timothy S Olson
- Cell Therapy and Transplant Section, Division of Pediatric Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
24
|
Congenital amegakaryocytic thrombocytopenia - Not a single disease. Best Pract Res Clin Haematol 2021; 34:101286. [PMID: 34404532 DOI: 10.1016/j.beha.2021.101286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 01/05/2023]
Abstract
Congenital amegakaryocytic thrombocytopenia (CAMT) is a rare inherited bone marrow failure syndrome (IBMFS) that is characterized by severe thrombocytopenia at birth due to ineffective megakaryopoiesis and development towards aplastic anemia during the first years of life. CAMT is not a single monogenetic disorder; rather, many descriptions of CAMT include different entities with different etiologies. CAMT in a narrow sense, which is primarily restricted to the hematopoietic system, is caused mainly by mutations in the gene for the thrombopoietin receptor (MPL), sometimes in the gene for its ligand (THPO). CAMT in association with radio-ulnar synostosis, which is not always clinically apparent, is mostly caused by mutations in MECOM, rarely in HOXA11. Patients affected by other IBMFS - especially Fanconi anemia or dyskeratosis congenita - may be misdiagnosed as having CAMT when they lack typical disease features of these syndromes or have only mild symptoms. This article reviews scientific and clinical aspects of the various disorders associated with the term "CAMT" with a main focus on the disease caused by mutations in the MPL gene.
Collapse
|
25
|
Inherited Platelet Disorders: An Updated Overview. Int J Mol Sci 2021; 22:ijms22094521. [PMID: 33926054 PMCID: PMC8123627 DOI: 10.3390/ijms22094521] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Platelets play a major role in hemostasis as ppwell as in many other physiological and pathological processes. Accordingly, production of about 1011 platelet per day as well as appropriate survival and functions are life essential events. Inherited platelet disorders (IPDs), affecting either platelet count or platelet functions, comprise a heterogenous group of about sixty rare diseases caused by molecular anomalies in many culprit genes. Their clinical relevance is highly variable according to the specific disease and even within the same type, ranging from almost negligible to life-threatening. Mucocutaneous bleeding diathesis (epistaxis, gum bleeding, purpura, menorrhagia), but also multisystemic disorders and/or malignancy comprise the clinical spectrum of IPDs. The early and accurate diagnosis of IPDs and a close patient medical follow-up is of great importance. A genotype-phenotype relationship in many IPDs makes a molecular diagnosis especially relevant to proper clinical management. Genetic diagnosis of IPDs has been greatly facilitated by the introduction of high throughput sequencing (HTS) techniques into mainstream investigation practice in these diseases. However, there are still unsolved ethical concerns on general genetic investigations. Patients should be informed and comprehend the potential implications of their genetic analysis. Unlike the progress in diagnosis, there have been no major advances in the clinical management of IPDs. Educational and preventive measures, few hemostatic drugs, platelet transfusions, thrombopoietin receptor agonists, and in life-threatening IPDs, allogeneic hematopoietic stem cell transplantation are therapeutic possibilities. Gene therapy may be a future option. Regular follow-up by a specialized hematology service with multidisciplinary support especially for syndromic IPDs is mandatory.
Collapse
|