1
|
Qin Q, Zhang H, Li X, Ruan H, Liu S, Chen Y, Xu Z, Wang Y, Yan X, Jiang X. MiR-129-5p alleviates depression and anxiety by increasing astrocyte ATP production partly through targeting deubiquitinase Mysm1. PLoS One 2025; 20:e0322715. [PMID: 40344568 PMCID: PMC12064192 DOI: 10.1371/journal.pone.0322715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 03/26/2025] [Indexed: 05/11/2025] Open
Abstract
Major depressive disorder (MDD) is a major global mental concern that severely affects quality of life, yet current pharmacological treatments remain limited in their effectiveness. Long-term chronic stress has been shown to increase the incidence of depression and anxiety. Micro RNAs (miRNAs) have been revealed to participate in the pathological process of depression and represent promising therapeutic targets. In this study, we found that microRNA-129-5p (miR-129-5p) was significantly decreased in the brains of depressive mice. Overexpression of miR-129-5p in the hippocampus effectively alleviated depressive-like behaviors and reduced the activation of microglial cells and astrocytes. In addition, ATP levels in depressive mice were significantly increased following miR-129-5p overexpression. The antidepressant effects of miR-129-5p were reversed when ATP function was blocked with the non-specific P2 receptor antagonist suramin. In vitro experiments revealed that miR-129-5p overexpression enhanced ATP production in astrocytes. Furthermore, using a dual-luciferase reporter assay, we found that miR-129-5p directly targeted Mysm1. When overexpressed in astrocytes, miR-129-5p significantly suppressed Mysm1 expression, promoted phosphorylation of p53 and AMPK, and enhanced the expression of PGC1α, factors previously associated with ATP production. Our findings highlight the crucial role of miR-129-5p in regulating depression, suggesting that miR-129-5p overexpression may serve as an effective strategy for antidepressant treatment.
Collapse
Affiliation(s)
- Qiaozhen Qin
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Heyang Zhang
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Xiaotong Li
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Huaqiang Ruan
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Shuirong Liu
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Yue Chen
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Zhenhua Xu
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Yan Wang
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
| | - Xinlong Yan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences, Haidian, Beijing, P.R. China
- Anhui Medical University, Hefei, Anhui, China
- Jishou University, Jishou, Hunan, China
| |
Collapse
|
2
|
Wei K, Zhou C, Shu Z, Shang X, Zou Y, Zhou W, Xu H, Liang Y, Ma T, Sun X, Xiao J. MYSM1 attenuates osteoarthritis by recruiting PP2A to deubiquitinate and dephosphorylate RIPK2. Bone Res 2025; 13:3. [PMID: 39746943 PMCID: PMC11696715 DOI: 10.1038/s41413-024-00368-y] [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: 12/05/2023] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 01/04/2025] Open
Abstract
Osteoarthritis (OA), the most prevalent degenerative joint disease, is marked by cartilage degradation and pathological alterations in surrounding tissues. Currently, no effective disease-modifying treatments exist. This study aimed to elucidate the critical roles of Myb-like, SWIRM, and MPN domains 1 (MYSM1) and its downstream effector, Receptor-interacting protein kinase 2 (RIPK2), in OA pathogenesis and the underlying mechanisms. Our findings revealed reduced MYSM1 levels in the cartilage of OA patients and mouse models. Genetic or adenovirus-induced MYSM1 knockout exacerbated OA progression in mice, whereas MYSM1 overexpression mitigated it. Mechanistically, MYSM1 inhibited the NF-κB and MAPK signaling pathways. Conversely, downstream RIPK2 significantly increased OA-like phenotypes and activated the NF-κB and MAPK pathways. The Ripk2S176D mutation accelerated OA pathogenesis, while Ripk2 silencing or Ripk2S176A mutation deactivated NF-κB and MAPK pathways, counteracting the role of MYSM1. MYSM1 deubiquitinates and dephosphorylates RIPK2S176 by recruiting protein phosphatase 2 A (PP2A). These results suggest that targeting MYSM1 or downstream RIPK2 offers promising therapeutic potential for OA.
Collapse
Affiliation(s)
- Kang Wei
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chuankun Zhou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zixing Shu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xingru Shang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Zou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Zhou
- Institute of Hepatobiliary Diseases, Transplant Center, Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Huanhuan Xu
- Department of Obstetrics and Gynecology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yulin Liang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuying Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Qin Q, Ruan H, Zhang H, Xu Z, Pan W, Yan X, Jiang X. Deubiquitinase MYSM1: An Important Tissue Development and Function Regulator. Int J Mol Sci 2024; 25:13051. [PMID: 39684760 DOI: 10.3390/ijms252313051] [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: 10/18/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
MYSM1, a deubiquitinating enzyme, plays a pivotal role in diverse biological processes. Both MYSM1 knockout mice and patients with Mysm1 gene mutations exhibit developmental abnormalities across multiple tissues and organs. Serving as a crucial regulator, MYSM1 influences stem cell function, immune responses, and the pathogenesis of diverse diseases. This review comprehensively details MYSM1's deubiquitinating activities in both the nucleus and cytoplasmic compartments, its effects on stem cell proliferation, differentiation, and immune cell function, and its involvement in cancer, aging, and depression. The high sequence homology between murine and human MYSM1, along with similar phenotypes observed in Mysm1-deficient models, provides valuable insights into the etiology of human Mysm1-deficiency syndromes. This review aims to offer a foundation for future comprehensive research on MYSM1.
Collapse
Affiliation(s)
- Qiaozhen Qin
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Huaqiang Ruan
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Heyang Zhang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhenhua Xu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Wenting Pan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Xinlong Yan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| |
Collapse
|
4
|
de Tocqueville S, Martin E, Riller Q, Kermasson L, France B, Magérus A, Rieux‐Laucat F, Delhommeau F, Hirsch P, Touzart A, Echalier A, Fischer A, Moshous D, Revy P. Long-term assessment of haematological recovery following somatic genetic rescue in a MYSM1-deficient patient: Implications for in vivo gene therapy. Br J Haematol 2024; 205:2349-2354. [PMID: 39233474 PMCID: PMC11637720 DOI: 10.1111/bjh.19744] [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: 07/18/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024]
Abstract
MYSM1 deficiency causes inherited bone marrow failure syndrome (IBMFS). We have previously identified an IBMFS patient with a homozygous pathogenic variant in MYSM1 who recovered from cytopenia due to spontaneous correction of one MYSM1 variant in the haematopoietic compartment, an event called somatic genetic rescue (SGR). The study of the genetic and biological aspects of the patient's haematopoietic/lymphopoietic system over a decade after SGR shows that one genetically corrected haematopoietic stem cell (HSC) can restore a healthy and stable haematopoietic system. This supports in vivo gene correction of HSCs as a promising treatment for IBMFS, including MYSM1 deficiency.
Collapse
Affiliation(s)
- Sophie de Tocqueville
- Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue 2023, INSERM UMR 1163Imagine InstituteParisFrance
- Imagine InstituteUniversité Paris Cité, Université Paris SaclayParisFrance
| | - Emmanuel Martin
- Imagine InstituteUniversité Paris Cité, Université Paris SaclayParisFrance
- Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, Equipe Labellisée Ligue 2023, INSERM UMR 1163ParisFrance
| | - Quentin Riller
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, F‐75015, Institut ImagineUniversité Paris CitéParisFrance
| | - Laëtitia Kermasson
- Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue 2023, INSERM UMR 1163Imagine InstituteParisFrance
- Imagine InstituteUniversité Paris Cité, Université Paris SaclayParisFrance
| | - Benoit France
- Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue 2023, INSERM UMR 1163Imagine InstituteParisFrance
- Imagine InstituteUniversité Paris Cité, Université Paris SaclayParisFrance
| | - Aude Magérus
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, F‐75015, Institut ImagineUniversité Paris CitéParisFrance
| | - Frédéric Rieux‐Laucat
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, F‐75015, Institut ImagineUniversité Paris CitéParisFrance
| | - François Delhommeau
- Service d'Hématologie Biologique, Centre de Recherche Saint‐Antoine, CRSA, AP‐HP, SIRIC CURAMUS, INSERM, Hôpital Saint‐AntoineSorbonne UniversitéParisFrance
| | - Pierre Hirsch
- Service d'Hématologie Biologique, Centre de Recherche Saint‐Antoine, CRSA, AP‐HP, SIRIC CURAMUS, INSERM, Hôpital Saint‐AntoineSorbonne UniversitéParisFrance
| | - Aurore Touzart
- Laboratory of Onco‐HematologyHôpital Necker Enfants‐Malades, Assistance Publique‐Hôpitaux de Paris (APHP)ParisFrance
- INSERM U1151, Institut Necker Enfants Malades (INEM)ParisFrance
| | - Aude Echalier
- Leicester Institute of Structural and Chemical Biology, and Department of Molecular and Cell BiologyUniversity of LeicesterLeicesterUK
| | - Alain Fischer
- Department of Pediatric Immunology, Hematology and RheumatologyNecker‐Enfants Malades Hospital, Assistance Publique‐Hôpitaux de Paris (APHP)ParisFrance
- Imagine Institute, INSERM UMR 1163ParisFrance
| | - Despina Moshous
- Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue 2023, INSERM UMR 1163Imagine InstituteParisFrance
- Imagine InstituteUniversité Paris Cité, Université Paris SaclayParisFrance
- Department of Pediatric Immunology, Hematology and RheumatologyNecker‐Enfants Malades Hospital, Assistance Publique‐Hôpitaux de Paris (APHP)ParisFrance
| | - Patrick Revy
- Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue 2023, INSERM UMR 1163Imagine InstituteParisFrance
- Imagine InstituteUniversité Paris Cité, Université Paris SaclayParisFrance
| |
Collapse
|
5
|
Xiao Z, He R, Zhao Z, Chen T, Ying Z. Dysregulation of epigenetic modifications in inborn errors of immunity. Epigenomics 2024; 16:1301-1313. [PMID: 39404224 PMCID: PMC11534118 DOI: 10.1080/17501911.2024.2410695] [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: 08/14/2024] [Accepted: 09/25/2024] [Indexed: 11/01/2024] Open
Abstract
Inborn errors of immunity (IEIs) are a group of typically monogenic disorders characterized by dysfunction in the immune system. Individuals with these disorders experience increased susceptibility to infections, autoimmunity and malignancies due to abnormal immune responses. Epigenetic modifications, including DNA methylation, histone modifications and chromatin remodeling, have been well explored in the regulation of immune cell development and effector function. Aberrant epigenetic modifications can disrupt gene expression profiles crucial for immune responses, resulting in impaired immune cell differentiation and function. Dysregulation of these processes caused by mutations in genes involving in epigenetic modifications has been associated with various IEIs. In this review article, we focus on IEIs that are caused by mutations in 13 genes involved in the regulation of DNA methylation, histone modification and chromatin remodeling.
Collapse
Affiliation(s)
- Zhongyao Xiao
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Rongjing He
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Zihan Zhao
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Taiping Chen
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Zhengzhou Ying
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
6
|
Mousa M, Liang Y, Tung LT, Wang H, Krawczyk C, Langlais D, Nijnik A. Chromatin-binding deubiquitinase MYSM1 acts in haematopoietic progenitors to control dendritic cell development and to program dendritic cell responses to microbial stimulation. Immunology 2024; 172:109-126. [PMID: 38316548 DOI: 10.1111/imm.13758] [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: 07/23/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Dendritic cells (DCs) are the most significant antigen presenting cells of the immune system, critical for the activation of naïve T cells. The pathways controlling DC development, maturation, and effector function therefore require precise regulation to allow for an effective induction of adaptive immune response. MYSM1 is a chromatin binding deubiquitinase (DUB) and an activator of gene expression via its catalytic activity for monoubiquitinated histone H2A (H2A-K119ub), which is a highly abundant repressive epigenetic mark. MYSM1 is an important regulator of haematopoiesis in mouse and human, and a systemic constitutive loss of Mysm1 in mice results in a depletion of many haematopoietic progenitors, including DC precursors, with the downstream loss of most DC lineage cells. However, the roles of MYSM1 at the later checkpoints in DC development, maturation, activation, and effector function at present remain unknown. In the current work, using a range of novel mouse models (Mysm1flCreERT2, Mysm1flCD11c-cre, Mysm1DN), we further the understanding of MYSM1 functions in the DC lineage: assessing the requirement for MYSM1 in DC development independently of other complex developmental phenotypes, exploring its role at the later checkpoints in DC maintenance and activation in response to microbial stimulation, and testing the requirement for the DUB catalytic activity of MYSM1 in these processes. Surprisingly, we demonstrate that MYSM1 expression and catalytic activity in DCs are dispensable for the maintenance of DC numbers in vivo or for DC activation in response to microbial stimulation. In contrast, MYSM1 acts via its DUB catalytic activity specifically in haematopoietic progenitors to allow normal DC lineage development, and its loss results not only in a severe DC depletion but also in the production of functionally altered DCs, with a dysregulation of many housekeeping transcriptional programs and significantly altered responses to microbial stimulation.
Collapse
Affiliation(s)
- Marwah Mousa
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Yue Liang
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Lin Tze Tung
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - HanChen Wang
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Connie Krawczyk
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, Michigan, United States
| | - David Langlais
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University Genome Centre, McGill University, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Mathias B, O'Leary D, Saucier N, Ahmad F, White LS, Russell L, Shinawi M, Smith MJ, Abraham RS, Cooper MA, Kitcharoensakkul M, Green AM, Bednarski JJ. MYSM1 attenuates DNA damage signals triggered by physiologic and genotoxic DNA breaks. J Allergy Clin Immunol 2024; 153:1113-1124.e7. [PMID: 38065233 PMCID: PMC11417613 DOI: 10.1016/j.jaci.2023.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 11/27/2023] [Accepted: 12/01/2023] [Indexed: 12/31/2023]
Abstract
BACKGROUND Patients with deleterious variants in MYSM1 have an immune deficiency characterized by B-cell lymphopenia, hypogammaglobulinemia, and increased radiosensitivity. MYSM1 is a histone deubiquitinase with established activity in regulating gene expression. MYSM1 also localizes to sites of DNA injury but its function in cellular responses to DNA breaks has not been elucidated. OBJECTIVES This study sought to determine the activity of MYSM1 in regulating DNA damage responses (DDRs) to DNA double-stranded breaks (DSBs) generated during immunoglobulin receptor gene (Ig) recombination and by ionizing radiation. METHODS MYSM1-deficient pre- and non-B cells were used to determine the role of MYSM1 in DSB generation, DSB repair, and termination of DDRs. RESULTS Genetic testing in a newborn with abnormal screen for severe combined immune deficiency, T-cell lymphopenia, and near absence of B cells identified a novel splice variant in MYSM1 that results in nearly absent protein expression. Radiosensitivity testing in patient's peripheral blood lymphocytes showed constitutive γH2AX, a marker of DNA damage, in B cells in the absence of irradiation, suggesting a role for MYSM1 in response to DSBs generated during Ig recombination. Suppression of MYSM1 in pre-B cells did not alter generation or repair of Ig DSBs. Rather, loss of MYSM1 resulted in persistent DNA damage foci and prolonged DDR signaling. Loss of MYSM1 also led to protracted DDRs in U2OS cells with irradiation induced DSBs. CONCLUSIONS MYSM1 regulates termination of DNA damage responses but does not function in DNA break generation and repair.
Collapse
Affiliation(s)
- Brendan Mathias
- Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | - David O'Leary
- Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | - Nermina Saucier
- Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | - Faiz Ahmad
- Department of Medicine, Washington University School of Medicine, St Louis, Mo
| | - Lynn S White
- Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | - Le'Mark Russell
- Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | - Marwan Shinawi
- Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | - Matthew J Smith
- Division of Hematology Research, Mayo Clinic, Rochester, Minn
| | - Roshini S Abraham
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Megan A Cooper
- Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | | | - Abby M Green
- Department of Pediatrics, Washington University School of Medicine, St Louis, Mo
| | - Jeffrey J Bednarski
- Department of Pediatrics, Washington University School of Medicine, St Louis, Mo.
| |
Collapse
|
8
|
Xu Z, Qin Q, Wang Y, Zhang H, Liu S, Li X, Chen Y, Wang Y, Ruan H, He W, Zhang T, Yan X, Wang C, Xu D, Jiang X. Deubiquitinase Mysm1 regulates neural stem cell proliferation and differentiation by controlling Id4 expression. Cell Death Dis 2024; 15:129. [PMID: 38342917 PMCID: PMC10859383 DOI: 10.1038/s41419-024-06530-y] [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: 09/26/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
Neural stem cells (NSCs) are critical for brain development and maintenance of neurogenesis. However, the molecular mechanisms that regulate NSC proliferation and differentiation remain unclear. Mysm1 is a deubiquitinase and is essential for the self-renewal and differentiation of several stem cells. It is unknown whether Mysm1 plays an important role in NSCs. Here, we found that Mysm1 was expressed in NSCs and its expression was increased with age in mice. Mice with Mysm1 knockdown by crossing Mysm1 floxed mice with Nestin-Cre mice exhibited abnormal brain development with microcephaly. Mysm1 deletion promoted NSC proliferation and apoptosis, resulting in depletion of the stem cell pool. In addition, Mysm1-deficient NSCs skewed toward neurogenesis instead of astrogliogenesis. Mechanistic investigations with RNA sequencing and genome-wide CUT&Tag analysis revealed that Mysm1 epigenetically regulated Id4 transcription by regulating histone modification at the promoter region. After rescuing the expression of Id4, the hyperproliferation and imbalance differentiation of Mysm1-deficient NSCs was reversed. Additionally, knockdown Mysm1 in aged mice could promote NSC proliferation. Collectively, the present study identified a new factor Mysm1 which is essential for NSC homeostasis and Mysm1-Id4 axis may be an ideal target for proper NSC proliferation and differentiation.
Collapse
Affiliation(s)
- Zhenhua Xu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Qiaozhen Qin
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China
| | - Yan Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
- Anhui Medical University, Hefei, 230032, Anhui, China
| | - Heyang Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Shuirong Liu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Xiaotong Li
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Yue Chen
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Yuqing Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Huaqiang Ruan
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Wenyan He
- China National Clinical Research Center for Neurological Diseases, Jing-Jin Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Tao Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Xinlong Yan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China
| | - Changyong Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China.
| | - Donggang Xu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China.
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China.
- Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
9
|
Sakovich IS, Haapaniemi E, Saarela J, Sharapova SO. Clinical Course and Family History of Adult Patient with Novel MYSM1 Variant. J Clin Immunol 2023; 44:9. [PMID: 38129711 DOI: 10.1007/s10875-023-01609-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 10/19/2023] [Indexed: 12/23/2023]
Affiliation(s)
- Inga S Sakovich
- Research Department, Immunology Laboratory, Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk Region, Belarus
| | - Emma Haapaniemi
- Department of Hematology and Regenerative Medicine, Karolinska Institutet, Huddinge, Sweden
- Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway
- Department of Pediatrics, Oslo University Hospital, Oslo, Norway
| | - Janna Saarela
- Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Svetlana O Sharapova
- Research Department, Immunology Laboratory, Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk Region, Belarus.
| |
Collapse
|
10
|
Imi T, Mizumaki H, Hosomichi K, Nannya Y, Zaimoku Y, Yoroidaka T, Katagiri T, Ishiyama K, Yamazaki H, Ogawa R, Kuroiwa M, Tajima A, Ogawa S, Nakao S. Familial immune-mediated aplastic anaemia in six different families. EJHAEM 2023; 4:714-718. [PMID: 37601868 PMCID: PMC10435714 DOI: 10.1002/jha2.722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/16/2023] [Accepted: 05/15/2023] [Indexed: 08/22/2023]
Abstract
We studied the pathophysiology of aplastic anaemia (AA) in six different pairs of relatives without a family history of hematologic disorders or congenital AA. Five and four of the six pairs shared the HLA-DRB1*15:01 and B*40:02 alleles, respectively. Glycosylphosphatidylinositol-anchored protein-deficient blood cells were detected in eight of the 10 patients evaluated. In a mother-daughter pair from one family, flow cytometry detected leukocytes lacking HLA-A2 due to loss of heterogeneity in chromosome 6p. Whole-exome sequencing of the family pair revealed a missense mutation in MYSM1. These results suggest that genetic inheritance of immune traits might underlie familial AA in some patients.
Collapse
Affiliation(s)
- Tatsuya Imi
- Department of HematologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Hiroki Mizumaki
- Department of HematologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Kazuyoshi Hosomichi
- Department of Bioinformatics and GenomicsGraduate School of Advanced Preventive Medical SciencesKanazawa UniversityKanazawaJapan
| | - Yasuhito Nannya
- Pathology and Tumor BiologyGraduate School of MedicineKyoto UniversityKyotoJapan
- Division of Hematopoietic Disease ControlInstitute of Medical SciecenThe University of TokyoTokyoJapan
| | - Yoshitaka Zaimoku
- Department of HematologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Takeshi Yoroidaka
- Department of HematologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Takamasa Katagiri
- Department of Clinical Laboratory SciencesGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Ken Ishiyama
- Department of HematologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Hirohito Yamazaki
- Department of HematologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Ryosuke Ogawa
- Department of Hematology and OncologyJapan Community Health Care Organization Kyushu HospitalFukuokaJapan
| | - Mika Kuroiwa
- Department of Hematology and OncologyNational Hospital Organization Fukuoka Higashi Medical CenterFukuokaJapan
| | - Atsushi Tajima
- Department of Bioinformatics and GenomicsGraduate School of Advanced Preventive Medical SciencesKanazawa UniversityKanazawaJapan
| | - Seishi Ogawa
- Pathology and Tumor BiologyGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Shinji Nakao
- Department of HematologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| |
Collapse
|
11
|
Wang H, Langlais D, Nijnik A. Histone H2A deubiquitinases in the transcriptional programs of development and hematopoiesis: a consolidated analysis. Int J Biochem Cell Biol 2023; 157:106384. [PMID: 36738766 DOI: 10.1016/j.biocel.2023.106384] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Monoubiquitinated lysine 119 of histone H2A (H2AK119ub) is a highly abundant epigenetic mark, associated with gene repression and deposited on chromatin by the polycomb repressor complex 1 (PRC1), which is an essential regulator of diverse transcriptional programs in mammalian development and tissue homeostasis. While multiple deubiquitinases (DUBs) with catalytic activity for H2AK119ub (H2A-DUBs) have been identified, we lack systematic analyses of their roles and cross-talk in transcriptional regulation. Here, we address H2A-DUB functions in epigenetic regulation of mammalian development and tissue maintenance by conducting a meta-analysis of 248 genomics datasets from 32 independent studies, focusing on the mouse model and covering embryonic stem cells (ESCs), hematopoietic, and immune cell lineages. This covers all the publicly available datasets that map genomic H2A-DUB binding and H2AK119ub distributions (ChIP-Seq), and all datasets assessing dysregulation in gene expression in the relevant H2A-DUB knockout models (RNA-Seq). Many accessory datasets for PRC1-2 and DUB-interacting proteins are also analyzed and interpreted, as well as further data assessing chromatin accessibility (ATAC-Seq) and transcriptional activity (RNA-seq). We report co-localization in the binding of H2A-DUBs BAP1, USP16, and to a lesser extent others that is conserved across different cell-types, and also the enrichment of antagonistic PRC1-2 protein complexes at the same genomic locations. Such conserved sites enriched for the H2A-DUBs and PRC1-2 are proximal to transcriptionally active genes that engage in housekeeping cellular functions. Nevertheless, they exhibit H2AK119ub levels significantly above the genomic average that can undergo further increase with H2A-DUB knockout. This indicates a cooperation between H2A-DUBs and PRC1-2 in the modulation of housekeeping transcriptional programs, conserved across many cell types, likely operating through their antagonistic effects on H2AK119ub and the regulation of local H2AK119ub turnover. Our study further highlights existing knowledge gaps and discusses important directions for future work.
Collapse
Affiliation(s)
- HanChen Wang
- Department of Physiology, McGill University, Montreal, QC, Canada; McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - David Langlais
- McGill University Research Centre on Complex Traits, McGill University, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada; McGill Genome Centre, Montreal, QC, Canada.
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, QC, Canada; McGill University Research Centre on Complex Traits, McGill University, QC, Canada.
| |
Collapse
|
12
|
Zhao J, Jia Y, Mahmut D, Deik AA, Jeanfavre S, Clish CB, Sankaran VG. Human hematopoietic stem cell vulnerability to ferroptosis. Cell 2023; 186:732-747.e16. [PMID: 36803603 PMCID: PMC9978939 DOI: 10.1016/j.cell.2023.01.020] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 11/20/2022] [Accepted: 01/12/2023] [Indexed: 02/18/2023]
Abstract
Hematopoietic stem cells (HSCs) have a number of unique physiologic adaptations that enable lifelong maintenance of blood cell production, including a highly regulated rate of protein synthesis. Yet, the precise vulnerabilities that arise from such adaptations have not been fully characterized. Here, inspired by a bone marrow failure disorder due to the loss of the histone deubiquitinase MYSM1, characterized by selectively disadvantaged HSCs, we show how reduced protein synthesis in HSCs results in increased ferroptosis. HSC maintenance can be fully rescued by blocking ferroptosis, despite no alteration in protein synthesis rates. Importantly, this selective vulnerability to ferroptosis not only underlies HSC loss in MYSM1 deficiency but also characterizes a broader liability of human HSCs. Increasing protein synthesis rates via MYSM1 overexpression makes HSCs less susceptible to ferroptosis, more broadly illustrating the selective vulnerabilities that arise in somatic stem cell populations as a result of physiologic adaptations.
Collapse
Affiliation(s)
- Jiawei Zhao
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yuemeng Jia
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Dilnar Mahmut
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Amy A Deik
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sarah Jeanfavre
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
13
|
Revy P, Kannengiesser C, Bertuch AA. Genetics of human telomere biology disorders. Nat Rev Genet 2023; 24:86-108. [PMID: 36151328 DOI: 10.1038/s41576-022-00527-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2022] [Indexed: 01/24/2023]
Abstract
Telomeres are specialized nucleoprotein structures at the ends of linear chromosomes that prevent the activation of DNA damage response and repair pathways. Numerous factors localize at telomeres to regulate their length, structure and function, to avert replicative senescence or genome instability and cell death. In humans, Mendelian defects in several of these factors can result in abnormally short or dysfunctional telomeres, causing a group of rare heterogeneous premature-ageing diseases, termed telomeropathies, short-telomere syndromes or telomere biology disorders (TBDs). Here, we review the TBD-causing genes identified so far and describe their main functions associated with telomere biology. We present molecular aspects of TBDs, including genetic anticipation, phenocopy, incomplete penetrance and somatic genetic rescue, which underlie the complexity of these diseases. We also discuss the implications of phenotypic and genetic features of TBDs on fundamental aspects related to human telomere biology, ageing and cancer, as well as on diagnostic, therapeutic and clinical approaches.
Collapse
Affiliation(s)
- Patrick Revy
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Nationale contre le Cancer, Paris, France.
- Université Paris Cité, Imagine Institute, Paris, France.
| | - Caroline Kannengiesser
- APHP Service de Génétique, Hôpital Bichat, Paris, France
- Inserm U1152, Université Paris Cité, Paris, France
| | - Alison A Bertuch
- Departments of Paediatrics and Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
14
|
Liang Y, Bhatt G, Tung LT, Wang H, Kim JE, Mousa M, Plackoska V, Illes K, Georges AA, Gros P, Henneman L, Huijbers IJ, Nagar B, Nijnik A. Deubiquitinase catalytic activity of MYSM1 is essential in vivo for hematopoiesis and immune cell development. Sci Rep 2023; 13:338. [PMID: 36611064 PMCID: PMC9825392 DOI: 10.1038/s41598-023-27486-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Myb-like SWIRM and MPN domains 1 (MYSM1) is a chromatin binding protein with deubiquitinase (DUB) catalytic activity. Rare MYSM1 mutations in human patients result in an inherited bone marrow failure syndrome, highlighting the biomedical significance of MYSM1 in the hematopoietic system. We and others characterized Mysm1-knockout mice as a model of this disorder and established that MYSM1 regulates hematopoietic function and leukocyte development in such models through different mechanisms. It is, however, unknown whether the DUB catalytic activity of MYSM1 is universally required for its many functions and for the maintenance of hematopoiesis in vivo. To test this, here we generated a new mouse strain carrying a Mysm1D660N point mutation (Mysm1DN) and demonstrated that the mutation renders MYSM1 protein catalytically inactive. We characterized Mysm1DN/DN and Mysm1fl/DN CreERT2 mice, against appropriate controls, for constitutive and inducible loss of MYSM1 catalytic function. We report a profound similarity in the developmental, hematopoietic, and immune phenotypes resulting from the loss of MYSM1 catalytic function and the full loss of MYSM1 protein. Overall, our work for the first time establishes the critical role of MYSM1 DUB catalytic activity in vivo in hematopoiesis, leukocyte development, and other aspects of mammalian physiology.
Collapse
Affiliation(s)
- Yue Liang
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1 Canada ,grid.14709.3b0000 0004 1936 8649McGill University Research Centre on Complex Traits, McGill University, Montreal, QC Canada
| | - Garvit Bhatt
- grid.14709.3b0000 0004 1936 8649Department of Pharmacology, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Centre de Recherche en Biologie Structurale (CRBS), McGill University, Montreal, QC Canada
| | - Lin Tze Tung
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1 Canada ,grid.14709.3b0000 0004 1936 8649McGill University Research Centre on Complex Traits, McGill University, Montreal, QC Canada
| | - HanChen Wang
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1 Canada ,grid.14709.3b0000 0004 1936 8649McGill University Research Centre on Complex Traits, McGill University, Montreal, QC Canada
| | - Joo Eun Kim
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1 Canada ,grid.14709.3b0000 0004 1936 8649McGill University Research Centre on Complex Traits, McGill University, Montreal, QC Canada
| | - Marwah Mousa
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1 Canada ,grid.14709.3b0000 0004 1936 8649McGill University Research Centre on Complex Traits, McGill University, Montreal, QC Canada
| | - Viktoria Plackoska
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC H3G 0B1 Canada ,grid.14709.3b0000 0004 1936 8649McGill University Research Centre on Complex Traits, McGill University, Montreal, QC Canada
| | - Katalin Illes
- grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Centre de Recherche en Biologie Structurale (CRBS), McGill University, Montreal, QC Canada
| | - Anna A. Georges
- grid.14709.3b0000 0004 1936 8649McGill University Research Centre on Complex Traits, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, Montreal, QC Canada
| | - Philippe Gros
- grid.14709.3b0000 0004 1936 8649McGill University Research Centre on Complex Traits, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, Montreal, QC Canada
| | - Linda Henneman
- grid.430814.a0000 0001 0674 1393Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Antoni Van Leeuwenhoek Ziekenhuis, Amsterdam, The Netherlands
| | - Ivo J. Huijbers
- grid.430814.a0000 0001 0674 1393Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Antoni Van Leeuwenhoek Ziekenhuis, Amsterdam, The Netherlands
| | - Bhushan Nagar
- grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Centre de Recherche en Biologie Structurale (CRBS), McGill University, Montreal, QC Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC, H3G 0B1, Canada. .,McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada.
| |
Collapse
|
15
|
Zhang H, Liu S, Qin Q, Xu Z, Qu Y, Wang Y, Wang J, Du Z, Yuan S, Hong S, Chang Z, He W, Yan X, Lang Y, Tang R, Wang Y, Zhu L, Jiang X. Genetic and Pharmacological Inhibition of Astrocytic Mysm1 Alleviates Depressive-Like Disorders by Promoting ATP Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2204463. [PMID: 36414403 PMCID: PMC9811473 DOI: 10.1002/advs.202204463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/16/2022] [Indexed: 05/03/2023]
Abstract
Major depressive disorder (MDD) is a leading cause of disability worldwide. A comprehensive understanding of the molecular mechanisms of this disorder is critical for the therapy of MDD. In this study, it is observed that deubiquitinase Mysm1 is induced in the brain tissues from patients with major depression and from mice with depressive behaviors. The genetic silencing of astrocytic Mysm1 induced an antidepressant-like effect and alleviated the osteoporosis of depressive mice. Furthermore, it is found that Mysm1 knockdown led to increased ATP production and the activation of p53 and AMP-activated protein kinase (AMPK). Pifithrin α (PFT α) and Compound C, antagonists of p53 and AMPK, respectively, repressed ATP production and reversed the antidepressant effect of Mysm1 knockdown. Moreover, the pharmacological inhibition of astrocytic Mysm1 by aspirin relieved depressive-like behaviors in mice. The study reveals, for the first time, the important function of Mysm1 in the brain, highlighting astrocytic Mysm1 as a potential risk factor for depression and as a valuable target for drug discovery to treat depression.
Collapse
Affiliation(s)
- Heyang Zhang
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
| | - Shuirong Liu
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
| | - Qiaozhen Qin
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
- Faculty of Environmental and Life SciencesBeijing University of TechnologyBeijing100124China
| | - Zhenhua Xu
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
| | - Yannv Qu
- Department of GeriatricsPeking University Shenzhen HospitalShenzhenGuangzhou518036China
| | - Yadi Wang
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
| | - Jianing Wang
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
| | - Zhangzhen Du
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
| | - Shanshan Yuan
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
| | - Shunming Hong
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
| | - Zhilin Chang
- China National Clinical Research Center for Neurological DiseasesJing‐Jin Center for NeuroinflammationBeijing Tiantan HospitalCapital Medical UniversityBeijing100050China
| | - Wenyan He
- China National Clinical Research Center for Neurological DiseasesJing‐Jin Center for NeuroinflammationBeijing Tiantan HospitalCapital Medical UniversityBeijing100050China
| | - Xinlong Yan
- Faculty of Environmental and Life SciencesBeijing University of TechnologyBeijing100124China
| | - Yiran Lang
- Beijing Innovation Center for Intelligent Robots and SystemsBeijing Institute of TechnologyBeijing100081China
| | - Rongyu Tang
- Beijing Innovation Center for Intelligent Robots and SystemsBeijing Institute of TechnologyBeijing100081China
| | - Yan Wang
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
- Anhui Medical UniversityHefeiAnhui230032China
| | - Lingling Zhu
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
- Anhui Medical UniversityHefeiAnhui230032China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantong226019China
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences27 Taiping Road, Haidian DistrictBeijing100850China
- Anhui Medical UniversityHefeiAnhui230032China
| |
Collapse
|
16
|
Hashem H. Hematopoietic cell transplantation for MYSM1 deficiency: not so much an easy task. Ann Hematol 2022; 101:2543-2544. [PMID: 35902396 DOI: 10.1007/s00277-022-04937-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 07/25/2022] [Indexed: 01/26/2023]
Affiliation(s)
- Hasan Hashem
- Department of Pediatrics, Division of Pediatric Hematology Oncology and Bone Marrow Transplantation, King Hussein Cancer Center, 202 Queen Rania street, Amman, 11941, Jordan.
| |
Collapse
|
17
|
Bleesing J. Gain-of-function defects in toll-like receptor 8 shed light on the interface between immune system and bone marrow failure disorders. Front Immunol 2022; 13:935321. [PMID: 36119097 PMCID: PMC9479092 DOI: 10.3389/fimmu.2022.935321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/08/2022] [Indexed: 11/30/2022] Open
Abstract
In this article, we will share lessons that patients with gain-of-function defects in Toll-like receptor 8 (TLR8-GOF) can teach us about the interface between bone marrow failure (BMF) disorders and inborn errors of immunity (IEI), subsequently referred to as “Interface Disorders”. TLR8-GOF is a relatively young entity (from a discovery standpoint) that—through both similar and dissimilar disease characteristics—can increase our understanding of interface disorders, for example, as it pertains to pathophysiology, the genetic mechanism of disease, and related diagnostics and therapeutics. From a genetics point of view, TLR8-GOF joins a growing list of (interface) disorders that can cause disease both with germline and somatic (mosaic) genetic variants. This not only has repercussions for the diagnostic workup of these disorders, inasmuch that routine genetic testing may miss somatic variants, but has therapeutic implications as well, for example, with the approach to curative treatment, such as hematopoietic stem cell transplantation. Following an introduction and schematic rendering of the interface, we will review the salient features of TLR8-GOF, with the understanding that the phenotype of this new disorder is likely not written in stone yet. In keeping with the principle of “Form Follows Function”, we will discuss specific immunological biomarkers that can be measured in clinical laboratories and highlight key disease features that pertain to TLR8-GOF, and can be found in several interface disorders. As can be seen from a schematic representation, the interface provides not only opportunities for learning and collaboration with respect to shared diagnostics but also the potential for drug repurposing and precision therapeutics. Ideally, collaboration also focuses on education and teaching, such that cross-fertilization and collaboration across these disciplines can create a framework for complementary research.
Collapse
Affiliation(s)
- Jack Bleesing
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- *Correspondence: Jack Bleesing,
| |
Collapse
|
18
|
Revertant Mosaicism in Genodermatoses: Natural Gene Therapy Right before Your Eyes. Biomedicines 2022; 10:biomedicines10092118. [PMID: 36140224 PMCID: PMC9495737 DOI: 10.3390/biomedicines10092118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 11/19/2022] Open
Abstract
Revertant mosaicism (RM) is the intriguing phenomenon in which nature itself has successfully done what medical science is so eagerly trying to achieve: correcting the effect of disease-causing germline variants and thereby reversing the disease phenotype back to normal. RM was molecularly confirmed for the first time in a genodermatosis in 1997, the genetic skin condition junctional epidermolysis bullosa (EB). At that time, RM was considered an extraordinary phenomenon. However, several important discoveries have changed this conception in the past few decades. First, RM has now been identified in all major subtypes of EB. Second, RM has also been identified in many other genodermatoses. Third, a theoretical mathematical exercise concluded that reverse mutations should be expected in all patients with a recessive subtype of EB or any other genodermatosis. This has shifted the paradigm from RM being an extraordinary phenomenon to it being something that every physician working in the field of genodermatoses should be looking for in every patient. It has also raised hope for new treatment options in patients with genodermatoses. In this review, we summarize the current knowledge on RM and discuss the perspectives of RM for the future treatment of patients with genodermatoses.
Collapse
|
19
|
S L, M K, U WK, M M. Somatic compensation of inherited bone marrow failure. Semin Hematol 2022; 59:167-173. [DOI: 10.1053/j.seminhematol.2022.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 02/06/2023]
|
20
|
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
|
21
|
Miyazawa H, Wada T. Reversion Mosaicism in Primary Immunodeficiency Diseases. Front Immunol 2021; 12:783022. [PMID: 34868061 PMCID: PMC8635092 DOI: 10.3389/fimmu.2021.783022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 10/28/2021] [Indexed: 11/13/2022] Open
Abstract
Reversion mosaicism has been reported in an increasing number of genetic disorders including primary immunodeficiency diseases. Several mechanisms can mediate somatic reversion of inherited mutations. Back mutations restore wild-type sequences, whereas second-site mutations result in compensatory changes. In addition, intragenic recombination, chromosomal deletions, and copy-neutral loss of heterozygosity have been demonstrated in mosaic individuals. Revertant cells that have regained wild-type function may be associated with milder disease phenotypes in some immunodeficient patients with reversion mosaicism. Revertant cells can also be responsible for immune dysregulation. Studies identifying a large variety of genetic changes in the same individual further support a frequent occurrence of reversion mosaicism in primary immunodeficiency diseases. This phenomenon also provides unique opportunities to evaluate the biological effects of restored gene expression in different cell lineages. In this paper, we review the recent findings of reversion mosaicism in primary immunodeficiency diseases and discuss its clinical implications.
Collapse
Affiliation(s)
- Hanae Miyazawa
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Taizo Wada
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
22
|
Barhoom D, Mohseni R, Behfar M, Hamidieh AA. Successful allogeneic stem cell transplantation with fludarabine-based reduced intensity conditioning in bone marrow failure syndrome 4. Pediatr Transplant 2021; 25:e14089. [PMID: 34302415 DOI: 10.1111/petr.14089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Myb-like, SWIRM, and MPN domains 1 (MYSM1) is a histone H2A deubiquitinase, has been discovered as one of the transcriptional regulators, and regulates the expression of specific transcription factors, which are essential for immunohematology development. Mutation in MYSM1 in humans leads to a rare autosomal recessive disease that has recently been known as inherited bone marrow failure syndrome 4 (BMFS4) associated with congenital bone marrow failure, immunodeficiency, and developmental aberrations. Allogeneic hematopoietic stem cell transplantation (HSCT) is the only curative option for immunohematology defects. METHODS In this paper, we report a pediatric patient with BMFS4 who suffered from pancytopenia and immunodeficiency affecting B cells and was successfully treated with HSCT from an HLA-identical father at 6 years old of age. Fludarabine-based reduced intensity conditioning was used and resulted in full donor chimerism. RESULTS Acute graft versus host disease (GVHD) grade II involving skin and gastrointestinal tract was observed, which was controlled with prednisolone. CONCLUSION She achieved B-cell recovery, and no blood or platelet transfusion was reported 1 year after HSCT.
Collapse
Affiliation(s)
- Dima Barhoom
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Pediatric Hematopoietic Stem Cell Transplant Department, Children's Hospital, Damascus University, Damascus, Syria
| | - Rashin Mohseni
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Behfar
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Chen X, Wang W, Li Y, Huo Y, Zhang H, Feng F, Xi W, Zhang T, Gao J, Yang F, Chen S, Yang A, Wang T. MYSM1 inhibits human colorectal cancer tumorigenesis by activating miR-200 family members/CDH1 and blocking PI3K/AKT signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:341. [PMID: 34706761 PMCID: PMC8549173 DOI: 10.1186/s13046-021-02106-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 09/16/2021] [Indexed: 01/17/2023]
Abstract
Background Histone epigenetic modification disorder is an important predisposing factor for the occurrence and development of many cancers, including colorectal cancer (CRC). The role of MYSM1, a metalloprotease that deubiquitinates monoubiquitinated histone H2A, in colorectal cancer was identified to evaluate its potential clinical application value. Methods MYSM1 expression levels in CRC cell lines and tumor tissues were detected, and their associations with patient survival rate and clinical stage were analyzed using databases and tissue microarrays. Gain- and loss-of-function studies were performed to identify the roles of MYSM1 in CRC cell proliferation, apoptosis, cell cycle progression, epithelial-mesenchymal transition (EMT) and metastasis in vitro and in vivo. ChIP, rescue assays and signal pathway verification were conducted for mechanistic study. Immunohistochemistry (IHC) was used to further assess the relationship of MYSM1 with CRC diagnosis and prognosis. Results MYSM1 was significantly downregulated and was related to the overall survival (OS) of CRC patients. MYSM1 served as a CRC suppressor by inducing apoptosis and inhibiting cell proliferation, EMT, tumorigenic potential and metastasis. Mechanistically, MYSM1 directly bound to the promoter region of miR-200/CDH1, impaired the enrichment of repressive H2AK119ub1 modification and epigenetically enhanced miR-200/CDH1 expression. Testing of paired CRC patient samples confirmed the positive regulatory relationship between MYSM1 and miR-200/CDH1. Furthermore, silencing MYSM1 stimulated PI3K/AKT signaling and promoted EMT in CRC cells. More importantly, a positive association existed between MYSM1 expression and a favorable CRC prognosis. Conclusions MYSM1 plays essential suppressive roles in CRC tumorigenesis and is a potential target for reducing CRC progression and distant metastasis. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02106-2.
Collapse
Affiliation(s)
- Xu Chen
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China.,Air Force Health Care Center for Special Services, Hangzhou, Zhejiang, 310007, P.R. China
| | - Wei Wang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China
| | - Yufang Li
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China.,Nuclear Medicine Diagnostic Center, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710032, P.R. China
| | - Yi Huo
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China
| | - Han Zhang
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China
| | - Fan Feng
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China
| | - Wenjin Xi
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China
| | - Tianze Zhang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China
| | - Jinjian Gao
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China
| | - Fan Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China
| | - Siyi Chen
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Angang Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China.
| | - Tao Wang
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, P.R. China.
| |
Collapse
|
24
|
Mantravadi V, Bednarski JJ, Ritter MA, Gu H, Kolicheski AL, Horner C, Cooper MA, Kitcharoensakkul M. Immunological Findings and Clinical Outcomes of Infants With Positive Newborn Screening for Severe Combined Immunodeficiency From a Tertiary Care Center in the U.S. Front Immunol 2021; 12:734096. [PMID: 34539671 PMCID: PMC8446381 DOI: 10.3389/fimmu.2021.734096] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/19/2021] [Indexed: 02/03/2023] Open
Abstract
The implementation of severe combined immunodeficiency (SCID) newborn screening has played a pivotal role in identifying these patients early in life as well as detecting various milder forms of T cell lymphopenia (TCL). In this study we reviewed the diagnostic and clinical outcomes, and interesting immunology findings of term infants referred to a tertiary care center with abnormal newborn SCID screens over a 6-year period. Key findings included a 33% incidence of non-SCID TCL including infants with novel variants in FOXN1, TBX1, MYSM1, POLD1, and CD3E; 57% positivity rate of newborn SCID screening among infants with DiGeorge syndrome; and earlier diagnosis and improved transplant outcomes for SCID in infants diagnosed after compared to before implementation of routine screening. Our study is unique in terms of the extensive laboratory workup of abnormal SCID screens including lymphocyte subsets, measurement of thymic output (TREC and CD4TE), and lymphocyte proliferation to mitogens in nearly all infants. These data allowed us to observe a stronger positive correlation of the absolute CD3 count with CD4RTE than with TREC copies, and a weak positive correlation between CD4RTE and TREC copies. Finally, we did not observe a correlation between risk of TCL and history of prenatal or perinatal complications or low birth weight. Our study demonstrated SCID newborn screening improves disease outcomes, particularly in typical SCID, and allows early detection and discovery of novel variants of certain TCL-associated genetic conditions.
Collapse
Affiliation(s)
- Vasudha Mantravadi
- The Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Jeffrey J Bednarski
- The Division of Hematology and Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Michelle A Ritter
- The Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Hongjie Gu
- The Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - Ana L Kolicheski
- The Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Caroline Horner
- The Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Megan A Cooper
- The Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Maleewan Kitcharoensakkul
- The Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States.,The Division of Pediatric Rheumatology/Immunology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
25
|
Abstract
Indirect somatic genetic rescue (SGR) of a germline mutation is thought to be rare in inherited Mendelian disorders. Here, we establish that acquired mutations in the EIF6 gene are a frequent mechanism of SGR in Shwachman-Diamond syndrome (SDS), a leukemia predisposition disorder caused by a germline defect in ribosome assembly. Biallelic mutations in the SBDS or EFL1 genes in SDS impair release of the anti-association factor eIF6 from the 60S ribosomal subunit, a key step in the translational activation of ribosomes. Here, we identify diverse mosaic somatic genetic events (point mutations, interstitial deletion, reciprocal chromosomal translocation) in SDS hematopoietic cells that reduce eIF6 expression or disrupt its interaction with the 60S subunit, thereby conferring a selective advantage over non-modified cells. SDS-related somatic EIF6 missense mutations that reduce eIF6 dosage or eIF6 binding to the 60S subunit suppress the defects in ribosome assembly and protein synthesis across multiple SBDS-deficient species including yeast, Dictyostelium and Drosophila. Our data suggest that SGR is a universal phenomenon that may influence the clinical evolution of diverse Mendelian disorders and support eIF6 suppressor mimics as a therapeutic strategy in SDS.
Collapse
|
26
|
Gutierrez-Rodrigues F, Sahoo SS, Wlodarski MW, Young NS. Somatic mosaicism in inherited bone marrow failure syndromes. Best Pract Res Clin Haematol 2021; 34:101279. [PMID: 34404533 DOI: 10.1016/j.beha.2021.101279] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/01/2021] [Accepted: 06/12/2021] [Indexed: 12/20/2022]
Abstract
Inherited bone marrow failure syndromes (IBMFS) are a heterogenous group of diseases caused by pathogenic germline variants in key pathways associated with haematopoiesis and genomic stability. Germline variants in IBMFS-related genes are known to reduce the fitness of hematopoietic stem and progenitor cells (HSPC), which has been hypothesized to drive clonal selection in these diseases. In many IBMFS, somatic mosaicism predominantly impacts cells by two distinct mechanisms, with contrasting effects. An acquired variation can improve cell fitness towards baseline levels, providing rescue of a deleterious phenotype. Alternatively, somatic mosaicism may result in a fitness advantage that results in malignant transformation. This review will describe these phenomena in IBMFS and delineate their relevance for diagnosis and clinical management. In addition, we will discuss which samples and methods can be used for detection of mosaicism according to clinical phenotype, type of mosaicism, and sample availability.
Collapse
Affiliation(s)
| | - Sushree S Sahoo
- Department of Hematology, St. Jude Children's Research Hospital, TN, USA
| | - Marcin W Wlodarski
- Department of Hematology, St. Jude Children's Research Hospital, TN, USA; Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| |
Collapse
|
27
|
Benyelles M, O'Donohue MF, Kermasson L, Lainey E, Borie R, Lagresle-Peyrou C, Nunes H, Cazelles C, Fourrage C, Ollivier E, Marcais A, Gamez AS, Morice-Picard F, Caillaud D, Pottier N, Ménard C, Ba I, Fernandes A, Crestani B, de Villartay JP, Gleizes PE, Callebaut I, Kannengiesser C, Revy P. NHP2 deficiency impairs rRNA biogenesis and causes pulmonary fibrosis and Høyeraal-Hreidarsson syndrome. Hum Mol Genet 2021; 29:907-922. [PMID: 31985013 DOI: 10.1093/hmg/ddaa011] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 01/14/2020] [Accepted: 01/17/2020] [Indexed: 12/18/2022] Open
Abstract
Telomeres are nucleoprotein structures at the end of chromosomes. The telomerase complex, constituted of the catalytic subunit TERT, the RNA matrix hTR and several cofactors, including the H/ACA box ribonucleoproteins Dyskerin, NOP10, GAR1, NAF1 and NHP2, regulates telomere length. In humans, inherited defects in telomere length maintenance are responsible for a wide spectrum of clinical premature aging manifestations including pulmonary fibrosis (PF), dyskeratosis congenita (DC), bone marrow failure and predisposition to cancer. NHP2 mutations have been so far reported only in two patients with DC. Here, we report the first case of Høyeraal-Hreidarsson syndrome, the severe form of DC, caused by biallelic missense mutations in NHP2. Additionally, we identified three unrelated patients with PF carrying NHP2 heterozygous mutations. Strikingly, one of these patients acquired a somatic mutation in the promoter of TERT that likely conferred a selective advantage in a subset of blood cells. Finally, we demonstrate that a functional deficit of human NHP2 affects ribosomal RNA biogenesis. Together, our results broaden the functional consequences and clinical spectrum of NHP2 deficiency.
Collapse
Affiliation(s)
- Maname Benyelles
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée La Ligue contre le Cancer, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Marie-Françoise O'Donohue
- Laboratoire de Biologie Moléculaire Eucaryote, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Laëtitia Kermasson
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée La Ligue contre le Cancer, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Elodie Lainey
- Hematology Laboratory, Robert DEBRE Hospital-APHP and INSERM UMR 1131-Hematology University Institute-Denis Diderot School of Medicine, Paris, France
| | - Raphael Borie
- APHP, Hôpital Bichat, Service de Pneumologie A, DHU FIRE, Paris, France.,INSERM, Unité 1152, Paris, France.,Université Paris Diderot, Paris, France
| | - Chantal Lagresle-Peyrou
- Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,University of Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Hilario Nunes
- Service de Pneumologie, Centre de Référence des Maladies Pulmonaires rares, Hôpital Avicenne, AP-HP, INSERM 1272, Université Paris 13, Bobigny, France
| | - Clarisse Cazelles
- Service d'hématologie adulte, Hôpital Necker- Enfants malades, Paris, France
| | - Cécile Fourrage
- INSERM UMR 1163, Genomics platform, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Genomic Core Facility, Imagine Institute-Structure Fédérative de Recherche Necker, INSERM U1163, Paris, France
| | - Emmanuelle Ollivier
- INSERM UMR 1163, Genomics platform, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Genomic Core Facility, Imagine Institute-Structure Fédérative de Recherche Necker, INSERM U1163, Paris, France
| | - Ambroise Marcais
- Service d'hématologie Adultes, Hôpital Necker-Enfants Malades, Assistance publique hôpitaux de Paris, Paris, France, Laboratoire d'onco-hématologie, Institut Necker-Enfants Malades, INSERM U1151, Université Paris Descartes, Paris, France
| | | | - Fanny Morice-Picard
- Service de Dermatologie Pédiatrique, Centre de Reference des Maladies Rares de la Peau, CHU de Bordeaux, Bordeaux F-33076, France
| | - Denis Caillaud
- Service de Pneumologie-Allergologie, Hôpital Gabriel Montpied, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Nicolas Pottier
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, EA4483-IMPECS, Lille, France
| | - Christelle Ménard
- APHP Service de Génétique, Hôpital Bichat, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Ibrahima Ba
- APHP Service de Génétique, Hôpital Bichat, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Alicia Fernandes
- Biological Resources Center, Structure Fédérative de Recherche Necker, INSERM US24, CNRS UMS3633, Assistance Publique des Hôpitaux de Paris and Institut Imagine, Paris, France
| | - Bruno Crestani
- APHP, Hôpital Bichat, Service de Pneumologie A, DHU FIRE, Paris, France
| | - Jean-Pierre de Villartay
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée La Ligue contre le Cancer, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Pierre-Emmanuel Gleizes
- Laboratoire de Biologie Moléculaire Eucaryote, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Isabelle Callebaut
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France
| | - Caroline Kannengiesser
- APHP Service de Génétique, Hôpital Bichat, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Patrick Revy
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée La Ligue contre le Cancer, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| |
Collapse
|
28
|
Zhan X, Zhao A, Wu B, Yang Y, Wan L, Tan P, Huang J, Lu Y. A novel compound heterozygous mutation of MYSM1 gene in a patient with bone marrow failure syndrome 4. Br J Biomed Sci 2021; 78:239-243. [PMID: 33618624 DOI: 10.1080/09674845.2021.1894706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- X Zhan
- Department of Childhood Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - A Zhao
- Department of Childhood Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - B Wu
- Department of Childhood Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Yang
- Department of Childhood Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - L Wan
- Department of Childhood Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - P Tan
- Department of Childhood Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J Huang
- Department of Childhood Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Lu
- Department of Childhood Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Huang J, Zhan XY, Zhao AL, Wu B, Yang Y, Tan P, Wan LJ, Lu YH. [A novel compound heterozygous mutation in MYSM1 gene in a 1-month-old girl: a bone marrow failure syndrome 4 family survey and literature review]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:129-134. [PMID: 33858043 PMCID: PMC8071664 DOI: 10.3760/cma.j.issn.0253-2727.2021.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 11/18/2022]
Abstract
Objective: To report the clinical manifestations and total exon detection results of one case of MYSM1 gene complex heterozygosity mutation of bone marrow failure syndrome 4 and the results of total exon detection of her family to provide a case phenotype for the early diagnosis of bone marrow failure syndrome 4. Methods: A 1-month-old girl with severe anemia was sequenced with trio-WES. Similarly, the family was also sequenced with tribe-WES to confirm the molecular diagnosis. BWA, GATK, and other software were used for annotation analysis of sequencing results. After polymerase chain reaction, Sanger sequencing was performed by ABI3730 sequencer to verify the target sequence. Moreover, the verification results were obtained by the sequence analysis software. The clinical diagnosis of this girl was reported and the relevant pieces of literature were reviewed. Results: The girl presented with pancytopenia, polydactylism, nonspecific white matter changes, and cysts. However, CD3(-)CD19(+) B decreased. The child was identified with MYSM1 complex heterozygous mutation by whole-exome sequencing, NM_001085487.2:c.1607_c.1611delAAGAG and c.1432C>T, which was respectively inherited from his parents. Genealogy verification confirmed that the c.1432C>T mutation carried by the father was from the grandfather (father's father) , whereas the c.1607_c.1611delAAGAG mutation carried by the mother was from the grandfather (mother's father) , whereas the grandmothers, aunts, and uncle did not carry the mutation. The child was diagnosed with BMFS4 combined with clinical phenotypic and molecular genetic findings. Conclusion: This case provides a case phenotype for the early diagnosis of BMFS4 and extends the pathogenicity variation and phenotype spectrum of the MYSM1 gene. The newly discovered pathogenic variant of MYSM1 c. 1607_c.1611delAAGAG has not been reported at home or abroad.
Collapse
Affiliation(s)
- J Huang
- Department of Hematology, Maternal and Child Hospital of Tongji Medical College of HUST, Maternal and Child Hospital of Hubei Province, Wuhan 430030, China
| | - X Y Zhan
- Department of Hematology, Maternal and Child Hospital of Tongji Medical College of HUST, Maternal and Child Hospital of Hubei Province, Wuhan 430030, China
| | - A L Zhao
- Department of Hematology, Maternal and Child Hospital of Tongji Medical College of HUST, Maternal and Child Hospital of Hubei Province, Wuhan 430030, China
| | - B Wu
- Department of Hematology, Maternal and Child Hospital of Tongji Medical College of HUST, Maternal and Child Hospital of Hubei Province, Wuhan 430030, China
| | - Y Yang
- Department of Hematology, Maternal and Child Hospital of Tongji Medical College of HUST, Maternal and Child Hospital of Hubei Province, Wuhan 430030, China
| | - P Tan
- Department of Hematology, Maternal and Child Hospital of Tongji Medical College of HUST, Maternal and Child Hospital of Hubei Province, Wuhan 430030, China
| | - L J Wan
- Department of Hematology, Maternal and Child Hospital of Tongji Medical College of HUST, Maternal and Child Hospital of Hubei Province, Wuhan 430030, China
| | - Y H Lu
- Department of Hematology, Maternal and Child Hospital of Tongji Medical College of HUST, Maternal and Child Hospital of Hubei Province, Wuhan 430030, China
| |
Collapse
|
30
|
Kelemen K, Saft L, Craig FE, Orazi A, Nakashima M, Wertheim GB, George TI, Horny HP, King RL, Quintanilla-Martinez L, Wang SA, Rimsza LM, Reichard KK. Eosinophilia/Hypereosinophilia in the Setting of Reactive and Idiopathic Causes, Well-Defined Myeloid or Lymphoid Leukemias, or Germline Disorders. Am J Clin Pathol 2021; 155:179-210. [PMID: 33367563 DOI: 10.1093/ajcp/aqaa244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES To report the findings of the 2019 Society for Hematopathology/European Association for Haematopathology Workshop within the categories of reactive eosinophilia, hypereosinophilic syndrome (HES), germline disorders with eosinophilia (GDE), and myeloid and lymphoid neoplasms associated with eosinophilia (excluding entities covered by other studies in this series). METHODS The workshop panel reviewed 109 cases, assigned consensus diagnosis, and created diagnosis-specific sessions. RESULTS The most frequent diagnosis was reactive eosinophilia (35), followed by acute leukemia (24). Myeloproliferative neoplasms (MPNs) received 17 submissions, including chronic eosinophilic leukemia, not otherwise specified (CEL, NOS). Myelodysplastic syndrome (MDS), MDS/MPN, and therapy-related myeloid neoplasms received 11, while GDE and HES received 12 and 11 submissions, respectively. CONCLUSIONS Hypereosinophilia and HES are defined by specific clinical and laboratory criteria. Eosinophilia is commonly reactive. An acute leukemic onset with eosinophilia may suggest core-binding factor acute myeloid leukemia, blast phase of chronic myeloid leukemia, BCR-ABL1-positive leukemia, or t(5;14) B-lymphoblastic leukemia. Eosinophilia is rare in MDS but common in MDS/MPN. CEL, NOS is a clinically aggressive MPN with eosinophilia as the dominant feature. Bone marrow morphology and cytogenetic and/or molecular clonality may distinguish CEL from HES. Molecular testing helps to better subclassify myeloid neoplasms with eosinophilia and to identify patients for targeted treatments.
Collapse
Affiliation(s)
| | - Leonie Saft
- Department of Pathology, Karolinska University Hospital and Institute, Stockholm, Sweden
| | - Fiona E Craig
- Division of Hematopathology, Mayo Clinic, Phoenix, AZ
| | - Attilio Orazi
- Department of Pathology, Texas Tech University Health Sciences Center, El Paso
| | - Megan Nakashima
- Department of Laboratory Medicine, Cleveland Clinic, Cleveland, OH
| | - Gerald B Wertheim
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Tracy I George
- Department of Pathology, University of Utah School of Medicine, Salt Lake City
| | - Hans-Peter Horny
- Institute of Pathology, University of Munich (LMU), Munich, Germany
| | | | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, Tübingen University Hospital, Tübingen, Germany
| | - Sa A Wang
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston
| | - Lisa M Rimsza
- Division of Hematopathology, Mayo Clinic, Phoenix, AZ
| | | |
Collapse
|
31
|
Wirth M, Schick M, Keller U, Krönke J. Ubiquitination and Ubiquitin-Like Modifications in Multiple Myeloma: Biology and Therapy. Cancers (Basel) 2020; 12:cancers12123764. [PMID: 33327527 PMCID: PMC7764993 DOI: 10.3390/cancers12123764] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Multiple myeloma is a cancer of plasma cells causing bone fractures, anemia, renal insufficiency and hypercalcemia. Despite the introduction of new drugs in the past years, it still remains incurable and most patients die from the disease. Multiple myeloma cells are characterized by the production of high amounts of monoclonal antibodies. Therefore, maintaining protein homeostasis from synthesis through folding to degradation is crucial for multiple myeloma cells. While protein ubiquitination and organized degradation are typically considered critical for cellular health, an emerging strategy is to block these processes to induce cell death in disease-state cells characterized by protein over-production. Recent development of compounds that alter the ubiquitin proteasome pathway and drugs that affect ubiquitin-like modifications appear promising in both preclinically and in clinical trials. This review summarizes the impact of protein modifications such as ubiquitination and ubiquitin-like modifications in the biology of multiple myeloma and how it can be exploited to develop new effective therapies for multiple myeloma. Abstract Multiple myeloma is a genetically heterogeneous plasma cell malignancy characterized by organ damage and a massive production of (in-)complete monoclonal antibodies. Coping with protein homeostasis and post-translational regulation is therefore essential for multiple myeloma cells to survive. Furthermore, post-translational modifications such as ubiquitination and SUMOylation play key roles in essential pathways in multiple myeloma, including NFκB signaling, epigenetic regulation, as well as DNA damage repair. Drugs modulating the ubiquitin–proteasome system, such as proteasome inhibitors and thalidomide analogs, are approved and highly effective drugs in multiple myeloma. In this review, we focus on ubiquitin and ubiquitin-like modifications in the biology and current developments of new treatments for multiple myeloma.
Collapse
Affiliation(s)
- Matthias Wirth
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany; (M.W.); (M.S.); (U.K.)
| | - Markus Schick
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany; (M.W.); (M.S.); (U.K.)
| | - Ulrich Keller
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany; (M.W.); (M.S.); (U.K.)
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Max-Delbrück Center for Molecular Medicine, 13092 Berlin, Germany
| | - Jan Krönke
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany; (M.W.); (M.S.); (U.K.)
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-30-450-513-538
| |
Collapse
|
32
|
Tian M, Huang Y, Song Y, Li W, Zhao P, Liu W, Wu K, Wu J. MYSM1 Suppresses Cellular Senescence and the Aging Process to Prolong Lifespan. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001950. [PMID: 33240758 PMCID: PMC7675055 DOI: 10.1002/advs.202001950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/31/2020] [Indexed: 05/19/2023]
Abstract
Aging is a universal feature of life that is a major focus of scientific research and a risk factor in many diseases. A comprehensive understanding of the cellular and molecular mechanisms of aging are critical to the prevention of diseases associated with the aging process. Here, it is shown that MYSM1 is a key suppressor of aging and aging-related pathologies. MYSM1 functionally represses cellular senescence and the aging process in human and mice primary cells and in mice organs. MYSM1 mechanistically attenuates the aging process by promoting DNA repair processes. Remarkably, MYSM1 deficiency facilitates the aging process and reduces lifespan, whereas MYSM1 over-expression attenuates the aging process and increases lifespan in mice. The functional role of MYSM1 is demonstrated in suppressing the aging process and prolonging lifespan. MYSM1 is a key suppressor of aging and may act as a potential agent for the prevention of aging and aging-associated diseases.
Collapse
Affiliation(s)
- Mingfu Tian
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Yuqing Huang
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Yunting Song
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Wen Li
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Peiyi Zhao
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Weiyong Liu
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Kailang Wu
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
| | - Jianguo Wu
- State Key Laboratory of VirologyCollege of Life SciencesWuhan UniversityWuhan430072China
- Guangdong Provincial Key Laboratory of VirologyInstitute of Medical MicrobiologyJinan UniversityGuangzhou510632China
| |
Collapse
|
33
|
Li N, Xu Y, Yu T, Yao R, Chen J, Luo C, Wang J. Further delineation of bone marrow failure syndrome caused by novel compound heterozygous variants of MYSM1. Gene 2020; 757:144938. [PMID: 32640305 DOI: 10.1016/j.gene.2020.144938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 12/21/2022]
Abstract
Myb-like SWIRM and MPN domains (MYSM1) is a chromatin-binding transcriptional regulator that mediates histone 2A deubiquitination, which plays a vital role in hematopoiesis and lymphocyte differentiation. Biallelic variants in MYSM1 cause a rare bone marrow failure syndrome (OMIM #618116). To date, only three pathogenic variants (E390*, R478*, and H656R) of MYSM1 have been reported in nine patients, and all variants are homozygous. Here, we describe a Chinese female patient who mainly presented with leukopenia, granulocytopenia, thrombocytopenia, severe anemia, and B-cell and natural killer cell deficiency in the peripheral blood, and was diagnosed with bone marrow failure. Trio whole-exome sequencing revealed a novel compound heterozygous variant in MYSM1 (c.399G > A, p.L133L, and c.1467C > G, p.Y489*). The c.399G > A synonymous variant is located at the 3'-end of exon 6, which is predicted to affect MYSM1 mRNA splicing. Analysis of the products obtained from the reverse transcription-polymerase chain reaction revealed that the c.399G > A variant leads to exon 6 skipping, resulting in a premature termination codon (c.321_399 del, p.V108Lfs*13). cDNA sequencing suggested that the c.1467C > G variant triggered nonsense-mediated mRNA degradation. Moreover, we identified a novel transcript of MYSM1 mRNA (missing exons 5 and 6) in human blood cells. Our results expand the mutation spectrum of MYSM1; additionally, this is the first report of a synonymous splicing variant that induces post-transcriptional skipping of exon 6 leading to a bone marrow failure syndrome phenotype.
Collapse
Affiliation(s)
- Niu Li
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Yufei Xu
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Tingting Yu
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China; Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Ruen Yao
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Jing Chen
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Chengjuan Luo
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| | - Jian Wang
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China; Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
34
|
Tian M, Liu W, Zhang Q, Huang Y, Li W, Wang W, Zhao P, Huang S, Song Y, Shereen MA, Qin M, Liu Y, Wu K, Wu J. MYSM1 Represses Innate Immunity and Autoimmunity through Suppressing the cGAS-STING Pathway. Cell Rep 2020; 33:108297. [PMID: 33086059 DOI: 10.1016/j.celrep.2020.108297] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/25/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
The immune system is not only required for preventing threats exerted by pathogens but also essential for developing immune tolerance to avoid tissue damage. This study identifies a distinct mechanism by which MYSM1 suppresses innate immunity and autoimmunity. The expression of MYSM1 is induced upon DNA virus infection and by intracellular DNA stimulation. MYSM1 subsequently interacts with STING and cleaves STING K63-linked ubiquitination to suppress cGAS-STING signaling. Notably, Mysm1-deficient mice exhibit a hyper-inflammatory response, acute tissue damage, and high mortality upon virus infection. Moreover, in the PBMCs of patients with systemic lupus erythematosus (SLE), MYSM1 production decreases, while type I interferons and pro-inflammatory cytokine expressions increase. Importantly, MYSM1 treatment represses the production of IFNs and pro-inflammatory cytokines in the PBMCs of SLE patients. Thus, MYSM1 is a critical repressor of innate immunity and autoimmunity and is thus a potential therapeutic agent for infectious, inflammatory, and autoimmune diseases.
Collapse
Affiliation(s)
- Mingfu Tian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Weiyong Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Qi Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yuqing Huang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Wen Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Wenbiao Wang
- Guangzhou Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Peiyi Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Shanyu Huang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yunting Song
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Muhammad Adnan Shereen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Mengying Qin
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yingle Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; Guangzhou Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
35
|
Zhang T, Sun J, Cheng J, Yin W, Li J, Miller H, Herrada AA, Gu H, Song H, Chen Y, Gong Q, Liu C. The role of ubiquitinase in B cell development and function. J Leukoc Biol 2020; 109:395-405. [PMID: 32816356 DOI: 10.1002/jlb.1mr0720-185rr] [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: 08/01/2019] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 11/10/2022] Open
Abstract
Ubiquitinases are a select group of enzymes that modify target proteins through ubiquitination, which plays a crucial role in the regulation of protein degradation, location, and function. B lymphocytes that originated from bone marrow hematopoietic stem cells (HSC), exert humoral immune functions by differentiating into plasma cells and producing antibodies. Previous studies have shown that ubiquitination is involved in the regulation of the cell cycle and signal transduction important for B lymphocyte development and function. In this review, how ubiquitinases regulate B cell development, activation, apoptosis, and proliferation is discussed, which could help in understanding the physiological processes and diseases related to B cells and also provides potential new targets for further studies.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianxuan Sun
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiali Cheng
- Department of hematology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yin
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingwen Li
- Department of hematology, Wuhan Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Department of Intracellular Pathogens, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Andrés A Herrada
- Lymphatic and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Heng Gu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongmei Song
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 1, Shuaifuyuan, Dongcheng District, Beijing, China
| | - Yan Chen
- The Second Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Belle JI, Wang H, Fiore A, Petrov JC, Lin YH, Feng CH, Nguyen TTM, Tung J, Campeau PM, Behrends U, Brunet T, Leszinski GS, Gros P, Langlais D, Nijnik A. MYSM1 maintains ribosomal protein gene expression in hematopoietic stem cells to prevent hematopoietic dysfunction. JCI Insight 2020; 5:125690. [PMID: 32641579 DOI: 10.1172/jci.insight.125690] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/21/2020] [Indexed: 01/09/2023] Open
Abstract
Ribosomopathies are congenital disorders caused by mutations in the genes encoding ribosomal and other functionally related proteins. They are characterized by anemia, other hematopoietic and developmental abnormalities, and p53 activation. Ribosome assembly requires coordinated expression of many ribosomal protein (RP) genes; however, the regulation of RP gene expression, especially in hematopoietic stem cells (HSCs), remains poorly understood. MYSM1 is a transcriptional regulator essential for HSC function and hematopoiesis. We established that HSC dysfunction in Mysm1 deficiency is driven by p53; however, the mechanisms of p53 activation remained unclear. Here, we describe the transcriptome of Mysm1-deficient mouse HSCs and identify MYSM1 genome-wide DNA binding sites. We establish a direct role for MYSM1 in RP gene expression and show a reduction in protein synthesis in Mysm1-/- HSCs. Loss of p53 in mice fully rescues Mysm1-/- anemia phenotype but not RP gene expression, indicating that RP gene dysregulation is a direct outcome of Mysm1 deficiency and an upstream mediator of Mysm1-/- phenotypes through p53 activation. We characterize a patient with a homozygous nonsense MYSM1 gene variant, and we demonstrate reduced protein synthesis and increased p53 levels in patient hematopoietic cells. Our work provides insights into the specialized mechanisms regulating RP gene expression in HSCs and establishes a common etiology of MYSM1 deficiency and ribosomopathy syndromes.
Collapse
Affiliation(s)
- Jad I Belle
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - HanChen Wang
- Department of Physiology.,McGill University Research Centre on Complex Traits, and.,Department of Human Genetics, McGill University, Quebec, Canada
| | - Amanda Fiore
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - Jessica C Petrov
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - Yun Hsiao Lin
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - Chu-Han Feng
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - Thi Tuyet Mai Nguyen
- Centre Hospitalier Universitaire St. Justine Research Center, University of Montreal, Quebec, Canada
| | - Jacky Tung
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - Philippe M Campeau
- Centre Hospitalier Universitaire St. Justine Research Center, University of Montreal, Quebec, Canada
| | | | - Theresa Brunet
- Institute of Human Genetics, Technische Universität München (TUM), Munich, Germany
| | - Gloria Sarah Leszinski
- Institute of Human Genetics, Technische Universität München (TUM), Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Philippe Gros
- McGill University Research Centre on Complex Traits, and.,Department of Biochemistry and.,The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Quebec, Canada
| | - David Langlais
- McGill University Research Centre on Complex Traits, and.,Department of Human Genetics, McGill University, Quebec, Canada.,McGill University Genome Centre, Montreal, Quebec, Canada
| | - Anastasia Nijnik
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| |
Collapse
|
37
|
Interaction of Deubiquitinase 2A-DUB/MYSM1 with DNA Repair and Replication Factors. Int J Mol Sci 2020; 21:ijms21113762. [PMID: 32466590 PMCID: PMC7312997 DOI: 10.3390/ijms21113762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/07/2020] [Accepted: 05/19/2020] [Indexed: 01/09/2023] Open
Abstract
The deubiquitination of histone H2A on lysine 119 by 2A-DUB/MYSM1, BAP1, USP16, and other enzymes is required for key cellular processes, including transcriptional activation, apoptosis, and cell cycle control, during normal hematopoiesis and tissue development, and in tumor cells. Based on our finding that MYSM1 colocalizes with γH2AX foci in human peripheral blood mononuclear cells, leukemia cells, and melanoma cells upon induction of DNA double-strand breaks with topoisomerase inhibitor etoposide, we applied a mass spectrometry-based proteomics approach to identify novel 2A-DUB/MYSM1 interaction partners in DNA-damage responses. Differential display of MYSM1 binding proteins significantly enriched after exposure of 293T cells to etoposide revealed an interacting network of proteins involved in DNA damage and replication, including factors associated with poor melanoma outcome. In the context of increased DNA-damage in a variety of cell types in Mysm1-deficient mice, in bone marrow cells upon aging and in UV-exposed Mysm1-deficient skin, our current mass spectrometry data provide additional evidence for an interaction between MYSM1 and key DNA replication and repair factors, and indicate a potential function of 2A-DUB/MYSM1 in DNA repair processes.
Collapse
|
38
|
Sarodaya N, Karapurkar J, Kim KS, Hong SH, Ramakrishna S. The Role of Deubiquitinating Enzymes in Hematopoiesis and Hematological Malignancies. Cancers (Basel) 2020; 12:E1103. [PMID: 32354135 PMCID: PMC7281754 DOI: 10.3390/cancers12051103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/11/2020] [Accepted: 04/26/2020] [Indexed: 12/24/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are responsible for the production of blood cells throughout the human lifespan. Single HSCs can give rise to at least eight distinct blood-cell lineages. Together, hematopoiesis, erythropoiesis, and angiogenesis coordinate several biological processes, i.e., cellular interactions during development and proliferation, guided migration, lineage programming, and reprogramming by transcription factors. Any dysregulation of these processes can result in hematological disorders and/or malignancies. Several studies of the molecular mechanisms governing HSC maintenance have demonstrated that protein regulation by the ubiquitin proteasomal pathway is crucial for normal HSC function. Recent studies have shown that reversal of ubiquitination by deubiquitinating enzymes (DUBs) plays an equally important role in hematopoiesis; however, information regarding the biological function of DUBs is limited. In this review, we focus on recent discoveries about the physiological roles of DUBs in hematopoiesis, erythropoiesis, and angiogenesis and discuss the DUBs associated with common hematological disorders and malignancies, which are potential therapeutic drug targets.
Collapse
Affiliation(s)
- Neha Sarodaya
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (N.S.); (J.K.); (K.-S.K.)
| | - Janardhan Karapurkar
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (N.S.); (J.K.); (K.-S.K.)
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (N.S.); (J.K.); (K.-S.K.)
- College of Medicine, Hanyang University, Seoul 04763, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (N.S.); (J.K.); (K.-S.K.)
- College of Medicine, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
39
|
Fiore A, Liang Y, Lin YH, Tung J, Wang H, Langlais D, Nijnik A. Deubiquitinase MYSM1 in the Hematopoietic System and beyond: A Current Review. Int J Mol Sci 2020; 21:ijms21083007. [PMID: 32344625 PMCID: PMC7216186 DOI: 10.3390/ijms21083007] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 01/21/2023] Open
Abstract
MYSM1 has emerged as an important regulator of hematopoietic stem cell function, blood cell production, immune response, and other aspects of mammalian physiology. It is a metalloprotease family protein with deubiquitinase catalytic activity, as well as SANT and SWIRM domains. MYSM1 normally localizes to the nucleus, where it can interact with chromatin and regulate gene expression, through deubiquitination of histone H2A and non-catalytic contacts with other transcriptional regulators. A cytosolic form of MYSM1 protein was also recently described and demonstrated to regulate signal transduction pathways of innate immunity, by promoting the deubiquitination of TRAF3, TRAF6, and RIP2. In this work we review the current knowledge on the molecular mechanisms of action of MYSM1 protein in transcriptional regulation, signal transduction, and potentially other cellular processes. The functions of MYSM1 in different cell types and aspects of mammalian physiology are also reviewed, highlighting the key checkpoints in hematopoiesis, immunity, and beyond regulated by MYSM1. Importantly, mutations in MYSM1 in human were recently linked to a rare hereditary disorder characterized by leukopenia, anemia, and other hematopoietic and developmental abnormalities. Our growing knowledge of MYSM1 functions and mechanisms of actions sheds important insights into its role in mammalian physiology and the etiology of the MYSM1-deficiency disorder in human.
Collapse
Affiliation(s)
- Amanda Fiore
- Department of Physiology, McGill University, Montreal, QC 3655, Canada; (A.F.); (Y.L.); (Y.H.L.); (J.T.); (H.W.)
- Research Centre on Complex Traits, McGill University, Montreal, QC 3649, Canada;
| | - Yue Liang
- Department of Physiology, McGill University, Montreal, QC 3655, Canada; (A.F.); (Y.L.); (Y.H.L.); (J.T.); (H.W.)
- Research Centre on Complex Traits, McGill University, Montreal, QC 3649, Canada;
| | - Yun Hsiao Lin
- Department of Physiology, McGill University, Montreal, QC 3655, Canada; (A.F.); (Y.L.); (Y.H.L.); (J.T.); (H.W.)
- Research Centre on Complex Traits, McGill University, Montreal, QC 3649, Canada;
| | - Jacky Tung
- Department of Physiology, McGill University, Montreal, QC 3655, Canada; (A.F.); (Y.L.); (Y.H.L.); (J.T.); (H.W.)
- Research Centre on Complex Traits, McGill University, Montreal, QC 3649, Canada;
| | - HanChen Wang
- Department of Physiology, McGill University, Montreal, QC 3655, Canada; (A.F.); (Y.L.); (Y.H.L.); (J.T.); (H.W.)
- Research Centre on Complex Traits, McGill University, Montreal, QC 3649, Canada;
- Department of Human Genetics, McGill University, Montreal, QC 3640, Canada
| | - David Langlais
- Research Centre on Complex Traits, McGill University, Montreal, QC 3649, Canada;
- Department of Human Genetics, McGill University, Montreal, QC 3640, Canada
- McGill University Genome Centre, Montreal, QC 740, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, QC 3655, Canada; (A.F.); (Y.L.); (Y.H.L.); (J.T.); (H.W.)
- Research Centre on Complex Traits, McGill University, Montreal, QC 3649, Canada;
- Correspondence: ; Tel.: +1-514-398-5567
| |
Collapse
|
40
|
Nomura T. Recombination-induced revertant mosaicism in ichthyosis with confetti and loricrin keratoderma. J Dermatol Sci 2019; 97:94-100. [PMID: 31928837 DOI: 10.1016/j.jdermsci.2019.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 12/26/2019] [Accepted: 12/26/2019] [Indexed: 01/23/2023]
Abstract
Revertant mosaicism refers to a condition in which a pathogenic germline mutation is spontaneously corrected in somatic cells, resulting in the presence of two or more cell populations with different genotypes in an organism arising from a single fertilized egg. If the revertant cells are clonally expanded due to a survival advantage over the surrounding mutant cells, patients benefit from this self-healing phenomenon which leads to the development of milder-than-expected clinical phenotypes; in genetic skin diseases, patients with revertant mosaicism present with small islands of healthy skin. To date, revertant mosaicism has been reported in ∼50 genetic diseases involving the skin, blood, liver, muscle, and brain. In this review, I briefly summarize current knowledge on revertant mosaicism in two particular skin diseases, ichthyosis with confetti (IWC) and loricrin keratoderma (LK), both of which develop numerous revertant skin patches. Notably, homologous recombination (HR) is the only mechanism underlying the reversion of pathogenic mutations in IWC and LK, and this was identified following the analysis of ∼50 revertant epidermis samples. All the samples showed long-tract loss of heterozygosity (LOH) that originated at regions centromeric to pathogenic mutations and extended to the telomere of the mutation-harboring chromosomes. Elucidating the molecular mechanisms underlying revertant mosaicism in IWC and LK-especially how mutant proteins induce long-tract LOH-would potentially expand the possibility of manipulating HR to induce the reversion of disease-causing mutations and help devising novel therapies not only for IWC and LK but also for other intractable genetic diseases.
Collapse
Affiliation(s)
- Toshifumi Nomura
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
41
|
Benyelles M, Episkopou H, O'Donohue M, Kermasson L, Frange P, Poulain F, Burcu Belen F, Polat M, Bole‐Feysot C, Langa‐Vives F, Gleizes P, de Villartay J, Callebaut I, Decottignies A, Revy P. Impaired telomere integrity and rRNA biogenesis in PARN-deficient patients and knock-out models. EMBO Mol Med 2019; 11:e10201. [PMID: 31273937 PMCID: PMC6609912 DOI: 10.15252/emmm.201810201] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/24/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
PARN, poly(A)-specific ribonuclease, regulates the turnover of mRNAs and the maturation and stabilization of the hTR RNA component of telomerase. Biallelic PARN mutations were associated with Høyeraal-Hreidarsson (HH) syndrome, a rare telomere biology disorder that, because of its severity, is likely not exclusively due to hTR down-regulation. Whether PARN deficiency was affecting the expression of telomere-related genes was still unclear. Using cells from two unrelated HH individuals carrying novel PARN mutations and a human PARN knock-out (KO) cell line with inducible PARN complementation, we found that PARN deficiency affects both telomere length and stability and down-regulates the expression of TRF1, TRF2, TPP1, RAP1, and POT1 shelterin transcripts. Down-regulation of dyskerin-encoding DKC1 mRNA was also observed and found to result from p53 activation in PARN-deficient cells. We further showed that PARN deficiency compromises ribosomal RNA biogenesis in patients' fibroblasts and cells from heterozygous Parn KO mice. Homozygous Parn KO however resulted in early embryonic lethality that was not overcome by p53 KO. Our results refine our knowledge on the pleiotropic cellular consequences of PARN deficiency.
Collapse
Affiliation(s)
- Maname Benyelles
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| | | | - Marie‐Françoise O'Donohue
- Laboratoire de Biologie Moléculaire EucaryoteCentre de Biologie Intégrative (CBI)CNRS, UPSUniversité de ToulouseToulouseFrance
| | - Laëtitia Kermasson
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| | - Pierre Frange
- EA 7327, Université Paris Descartes, Sorbonne Paris‐CitéParisFrance
- Laboratoire de Microbiologie clinique & Unité d'ImmunologieHématologie et Rhumatologie PédiatriquesAP‐HP, Hôpital Necker, Enfants MaladesParisFrance
| | - Florian Poulain
- de Duve InstituteUniversité catholique de LouvainBrusselsBelgium
| | - Fatma Burcu Belen
- Pediatric HematologyFaculty of MedicineBaskent UniversityAnkaraTurkey
| | - Meltem Polat
- Pediatric Infectious DiseasesDepartment of Pediatric Infectious DiseasesPamukkale University Medical FacultyDenizliTurkey
| | - Christine Bole‐Feysot
- INSERM, UMR 1163Genomics platform, Imagine InstituteParis Descartes–Sorbonne Paris Cité UniversityParisFrance
- Genomic Core FacilityImagine Institute‐Structure Fédérative de Recherche NeckerINSERM U1163ParisFrance
| | | | - Pierre‐Emmanuel Gleizes
- Laboratoire de Biologie Moléculaire EucaryoteCentre de Biologie Intégrative (CBI)CNRS, UPSUniversité de ToulouseToulouseFrance
| | - Jean‐Pierre de Villartay
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| | - Isabelle Callebaut
- Muséum National d'Histoire NaturelleUMR CNRS 7590Institut de Minéralogiede Physique des Matériaux et de Cosmochimie, IMPMCSorbonne UniversitéParisFrance
| | | | - Patrick Revy
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| |
Collapse
|
42
|
Revy P, Kannengiesser C, Fischer A. Somatic genetic rescue in Mendelian haematopoietic diseases. Nat Rev Genet 2019; 20:582-598. [DOI: 10.1038/s41576-019-0139-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2019] [Indexed: 12/30/2022]
|
43
|
Wang YH, Huang XH, Yang YM, He Y, Dong XH, Yang HX, Zhang L, Wang Y, Zhou J, Wang C, Jiang XX. Mysm1 epigenetically regulates the immunomodulatory function of adipose-derived stem cells in part by targeting miR-150. J Cell Mol Med 2019; 23:3737-3746. [PMID: 30895711 PMCID: PMC6484305 DOI: 10.1111/jcmm.14281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/31/2019] [Accepted: 02/25/2019] [Indexed: 12/26/2022] Open
Abstract
Adipose‐derived stem cells (ASCs) are highly attractive for cell‐based therapies in tissue repair and regeneration because they have multilineage differentiation capacity and are immunosuppressive. However, the detailed epigenetic mechanisms of their immunoregulatory capacity are not fully defined. In this study, we found that Mysm1 was induced in ASCs treated with inflammatory cytokines. Adipose‐derived stem cells with Mysm1 knockdown exhibited attenuated immunosuppressive capacity, evidenced by less inhibition of T cell proliferation, more pro‐inflammatory factor secretion and less nitric oxide (NO) production in vitro. Mysm1‐deficient ASCs exacerbated inflammatory bowel diseases but inhibited tumour growth in vivo. Mysm1‐deficient ASCs also showed depressed miR‐150 expression. When transduced with Mysm1 overexpression lentivirus, ASCs exhibited enhanced miR‐150 expression. Furthermore, Mysm1‐deficient cells transduced with lentivirus containing miR‐150 mimics produced less pro‐inflammatory factors and more NO. Our study reveals a new role of Mysm1 in regulating the immunomodulatory activities of ASCs by targeting miR‐150. These novel insights into the mechanisms through which ASCs regulate immune reactions may lead to better clinical utility of these cells.
Collapse
Affiliation(s)
- Yu-Han Wang
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, P.R. China
| | - Xiao-Hui Huang
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, P.R. China.,Anhui Medical University, Hefei, Anhui, China
| | - Yan-Mei Yang
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, P.R. China.,Department of Stomatology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Youdi He
- Department of Neurology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiao-Hui Dong
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, P.R. China
| | - Hui-Xin Yang
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, P.R. China.,Department of Stomatology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Lei Zhang
- College of Agroforestry Engineering and Planning, Tongren University, Tongren, Guizhou, China
| | - Yan Wang
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, P.R. China
| | - Jin Zhou
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, P.R. China
| | - Changyong Wang
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, P.R. China
| | - Xiao-Xia Jiang
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, P.R. China.,Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
44
|
Ulirsch JC, Verboon JM, Kazerounian S, Guo MH, Yuan D, Ludwig LS, Handsaker RE, Abdulhay NJ, Fiorini C, Genovese G, Lim ET, Cheng A, Cummings BB, Chao KR, Beggs AH, Genetti CA, Sieff CA, Newburger PE, Niewiadomska E, Matysiak M, Vlachos A, Lipton JM, Atsidaftos E, Glader B, Narla A, Gleizes PE, O'Donohue MF, Montel-Lehry N, Amor DJ, McCarroll SA, O'Donnell-Luria AH, Gupta N, Gabriel SB, MacArthur DG, Lander ES, Lek M, Da Costa L, Nathan DG, Korostelev AA, Do R, Sankaran VG, Gazda HT. The Genetic Landscape of Diamond-Blackfan Anemia. Am J Hum Genet 2018; 103:930-947. [PMID: 30503522 PMCID: PMC6288280 DOI: 10.1016/j.ajhg.2018.10.027] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/29/2018] [Indexed: 01/19/2023] Open
Abstract
Diamond-Blackfan anemia (DBA) is a rare bone marrow failure disorder that affects 7 out of 1,000,000 live births and has been associated with mutations in components of the ribosome. In order to characterize the genetic landscape of this heterogeneous disorder, we recruited a cohort of 472 individuals with a clinical diagnosis of DBA and performed whole-exome sequencing (WES). We identified relevant rare and predicted damaging mutations for 78% of individuals. The majority of mutations were singletons, absent from population databases, predicted to cause loss of function, and located in 1 of 19 previously reported ribosomal protein (RP)-encoding genes. Using exon coverage estimates, we identified and validated 31 deletions in RP genes. We also observed an enrichment for extended splice site mutations and validated their diverse effects using RNA sequencing in cell lines obtained from individuals with DBA. Leveraging the size of our cohort, we observed robust genotype-phenotype associations with congenital abnormalities and treatment outcomes. We further identified rare mutations in seven previously unreported RP genes that may cause DBA, as well as several distinct disorders that appear to phenocopy DBA, including nine individuals with biallelic CECR1 mutations that result in deficiency of ADA2. However, no new genes were identified at exome-wide significance, suggesting that there are no unidentified genes containing mutations readily identified by WES that explain >5% of DBA-affected case subjects. Overall, this report should inform not only clinical practice for DBA-affected individuals, but also the design and analysis of rare variant studies for heterogeneous Mendelian disorders.
Collapse
Affiliation(s)
- Jacob C Ulirsch
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey M Verboon
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shideh Kazerounian
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael H Guo
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel Yuan
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Leif S Ludwig
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Robert E Handsaker
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Nour J Abdulhay
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Claudia Fiorini
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Giulio Genovese
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Elaine T Lim
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aaron Cheng
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Beryl B Cummings
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine R Chao
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alan H Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Casie A Genetti
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Colin A Sieff
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Peter E Newburger
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Edyta Niewiadomska
- Department of Pediatric Hematology/Oncology, Medical University of Warsaw, Warsaw, Poland
| | - Michal Matysiak
- Department of Pediatric Hematology/Oncology, Medical University of Warsaw, Warsaw, Poland
| | - Adrianna Vlachos
- Feinstein Institute for Medical Research, Manhasset, NY; Division of Hematology/Oncology and Stem Cell Transplantation, Cohen Children's Medical Center, New Hyde Park, NY; Hofstra Northwell School of Medicine, Hempstead, NY 11030, USA
| | - Jeffrey M Lipton
- Feinstein Institute for Medical Research, Manhasset, NY; Division of Hematology/Oncology and Stem Cell Transplantation, Cohen Children's Medical Center, New Hyde Park, NY; Hofstra Northwell School of Medicine, Hempstead, NY 11030, USA
| | - Eva Atsidaftos
- Feinstein Institute for Medical Research, Manhasset, NY; Division of Hematology/Oncology and Stem Cell Transplantation, Cohen Children's Medical Center, New Hyde Park, NY; Hofstra Northwell School of Medicine, Hempstead, NY 11030, USA
| | - Bertil Glader
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 02114, USA
| | - Anupama Narla
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 02114, USA
| | - Pierre-Emmanuel Gleizes
- Laboratory of Eukaryotic Molecular Biology, Center for Integrative Biology (CBI), University of Toulouse, CNRS, Toulouse, France
| | - Marie-Françoise O'Donohue
- Laboratory of Eukaryotic Molecular Biology, Center for Integrative Biology (CBI), University of Toulouse, CNRS, Toulouse, France
| | - Nathalie Montel-Lehry
- Laboratory of Eukaryotic Molecular Biology, Center for Integrative Biology (CBI), University of Toulouse, CNRS, Toulouse, France
| | - David J Amor
- Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Steven A McCarroll
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Anne H O'Donnell-Luria
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Namrata Gupta
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Stacey B Gabriel
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel G MacArthur
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Eric S Lander
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Monkol Lek
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lydie Da Costa
- University Paris VII Denis DIDEROT, Faculté de Médecine Xavier Bichat, 75019 Paris, France; Laboratory of Excellence for Red Cell, LABEX GR-Ex, 75015 Paris, France
| | - David G Nathan
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Andrei A Korostelev
- RNA Therapeutics Institute, Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Ron Do
- Department of Genetics and Genomic Sciences and The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vijay G Sankaran
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| | - Hanna T Gazda
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Huo Y, Li BY, Lin ZF, Wang W, Jiang XX, Chen X, Xi WJ, Yang AG, Chen SY, Wang T. MYSM1 Is Essential for Maintaining Hematopoietic Stem Cell (HSC) Quiescence and Survival. Med Sci Monit 2018; 24:2541-2549. [PMID: 29694335 PMCID: PMC5939948 DOI: 10.12659/msm.906876] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background Histone H2A deubiquitinase MYSM1 has recently been shown to be essential for hematopoiesis and hematopoietic stem cell (HSC) function in both mice and humans. However, conventional MYSM1 knockouts cause partial embryonic lethality and growth retardation, and it is difficult to convincingly remove the effects of environmental factors on HSC differentiation and function. Material/Methods MYSM1 conditional knockout (cKO) mice were efficiently induced by using the Vav1-cre transgenic system. The Vav-Cre MYSM1 cKO mice were then analyzed to verify the intrinsic role of MYSM1 in hematopoietic cells. Results MYSM1 cKO mice were viable and were born at normal litter sizes. At steady state, we observed a defect in hematopoiesis, including reduced bone marrow cellularity and abnormal HSC function. MYSM1 deletion drives HSCs from quiescence into rapid cycling, and MYSM1-deficient HSCs display impaired engraftment. In particular, the immature cycling cKO HSCs have elevated reactive oxygen species (ROS) levels and are prone to apoptosis, resulting in the exhaustion of the stem cell pool during stress response to 5-FU. Conclusions Our study using MYSM1 cKO mice confirms the important role of MYSM1 in maintaining HSC quiescence and survival.
Collapse
Affiliation(s)
- Yi Huo
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland).,Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Bing-Yi Li
- Undergraduate Student at Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Zhi-Feng Lin
- Undergraduate Student at Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Wei Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Xiao-Xia Jiang
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xu Chen
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Wen-Jin Xi
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - An-Gang Yang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland)
| | - Si-Yi Chen
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Tao Wang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi, China (mainland).,Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
46
|
Abstract
Proper regulation of the immune system is required for protection against pathogens and preventing autoimmune disorders. Inborn errors of the immune system due to inherited or de novo germline mutations can lead to the loss of protective immunity, aberrant immune homeostasis, and the development of autoimmune disease, or combinations of these. Forward genetic screens involving clinical material from patients with primary immunodeficiencies (PIDs) can vary in severity from life-threatening disease affecting multiple cell types and organs to relatively mild disease with susceptibility to a limited range of pathogens or mild autoimmune conditions. As central mediators of innate and adaptive immune responses, T cells are critical orchestrators and effectors of the immune response. As such, several PIDs result from loss of or altered T cell function. PID-associated functional defects range from complete absence of T cell development to uncontrolled effector cell activation. Furthermore, the gene products of known PID causal genes are involved in diverse molecular pathways ranging from T cell receptor signaling to regulators of protein glycosylation. Identification of the molecular and biochemical cause of PIDs can not only guide the course of treatment for patients, but also inform our understanding of the basic biology behind T cell function. In this chapter, we review PIDs with known genetic causes that intrinsically affect T cell function with particular focus on perturbations of biochemical pathways.
Collapse
Affiliation(s)
- William A Comrie
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States; Clinical Genomics Program, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States
| | - Michael J Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States; Clinical Genomics Program, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, United States.
| |
Collapse
|
47
|
Lesport E, Ferster A, Biver A, Roch B, Vasquez N, Jabado N, Vives FL, Revy P, Soulier J, de Villartay JP. Reduced recruitment of 53BP1 during interstrand crosslink repair is associated with genetically inherited attenuation of mitomycin C sensitivity in a family with Fanconi anemia. Oncotarget 2018; 9:3779-3793. [PMID: 29423082 PMCID: PMC5790499 DOI: 10.18632/oncotarget.23375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 11/28/2017] [Indexed: 11/25/2022] Open
Abstract
The Fanconi anemia (FA) pathway is implicated in the repair of DNA interstrand crosslinks (ICL). In this process, it has been shown that FA factors regulate the choice for DNA double strand break repair towards homologous recombination (HR). As this mechanism is impaired in FA deficient cells exposed to crosslinking agents, an inappropriate usage of non-homologous end joining (NHEJ) leads to the accumulation of toxic chromosomal abnormalities. We studied a family with two FANCG patients and found a genetically inherited attenuation of mitomycin C sensitivity resulting in-vitro in an attenuated phenotype for one patient or in increased resistance for two healthy relatives. A heterozygous mutation in ATM was identified in these 3 subjects but was not directly linked to the observed phenotype. However, the attenuation of ICL sensitivity was associated with a reduced recruitment of 53BP1 during the course of ICL repair, and increased HR levels. These results further demonstrate the importance of favoring HR over NHEJ for the survival of cells challenged with ICLs.
Collapse
Affiliation(s)
- Emilie Lesport
- Laboratory “Genome Dynamics in The Immune System”, INSERM UMR1163, Université Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Alina Ferster
- Departement d’Hémato-Oncologie, Hôpital Universitaire des Enfants Reine Fabiola, Bruxelles, Belgium
| | - Armand Biver
- Service de Pédiatrie Générale, Centre Hospitalier De Luxembourg, Luxembourg
| | - Benoit Roch
- Laboratory “Genome Dynamics in The Immune System”, INSERM UMR1163, Université Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Nadia Vasquez
- INSERM U944, Institut Universitaire d’Hématologie, Paris, France
| | - Nada Jabado
- Department of Human Genetics and Department of Experimental Medicine, McGill University, Montreal, Canada
| | | | - Patrick Revy
- Laboratory “Genome Dynamics in The Immune System”, INSERM UMR1163, Université Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Jean Soulier
- INSERM U944, Institut Universitaire d’Hématologie, Paris, France
| | - Jean-Pierre de Villartay
- Laboratory “Genome Dynamics in The Immune System”, INSERM UMR1163, Université Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| |
Collapse
|
48
|
Haffner-Luntzer M, Kovtun A, Fischer V, Prystaz K, Hainzl A, Kroeger CM, Krikki I, Brinker TJ, Ignatius A, Gatzka M. Loss of p53 compensates osteopenia in murine Mysm1 deficiency. FASEB J 2018; 32:1957-1968. [PMID: 29203593 DOI: 10.1096/fj.201700871r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Histone modifications critically contribute to the epigenetic orchestration of bone homeostasis-in part, by modifying the access of transcription factors to specific genes involved in the osteogenic differentiation process of bone marrow mesenchymal stem cells (MSCs) and osteoblasts. Based on our previous finding that histone H2A deubiquitinase 2A-DUB/Mysm1 interacts with the p53 axis in hematopoiesis and tissue development, we analyzed the molecular basis of the skeletal phenotype of Mysm1-deficient mice and dissected the underlying p53-dependent and -independent mechanisms. Visible morphologic, skeletal deformations of young Mysm1-deficient mice-including a kinked and truncated tail and shortened long bones-were associated with osteopenia of long bones. On the cellular level, Mysm1-deficient primary osteoblasts displayed reduced potential to differentiate into mature osteoblasts, as indicated by decreased expression of osteogenic markers. Reduced osteogenic differentiation capacity of Mysm1-deficient osteoblasts was accompanied by an impaired induction of osteogenic transcription factor Runx2. Osteogenic differentiation of Mysm1-/- MSCs, however, was not compromised in vitro. In line with defective hematopoietic development of Mysm1-deficient mice, Mysm1-/- osteoclasts had reduced resorption activity and were more prone to apoptosis in TUNEL assays. Skeletal alterations and osteopenia of Mysm1-deficient mice were phenotypically completely rescued by simultaneous ablation of p53 in p53-/-Mysm1-/- double-deficient mice-although p53 deficiency did not restore Runx2 expression in Mysm1-/- osteoblasts on the molecular level but, instead, enhanced proliferation and osteogenic differentiation of MSCs. In summary, our results demonstrate novel roles for Mysm1 in osteoblast differentiation and osteoclast formation, resulting in osteopenia in Mysm1-deficient mice that could be abrogated by the loss of p53 from increased osteogenic differentiation of Mysm1-/-p53-/- MSCs.-Haffner-Luntzer, M., Kovtun, A., Fischer, V., Prystaz, K., Hainzl, A., Kroeger, C. M., Krikki, I., Brinker, T. J., Ignatius, A., Gatzka, M. Loss of p53 compensates osteopenia in murine Mysm1 deficiency.
Collapse
Affiliation(s)
- Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University of Ulm, Ulm, Germany.,Trauma Research Centre, University of Ulm, Ulm, Germany
| | - Anna Kovtun
- Institute of Orthopaedic Research and Biomechanics, University of Ulm, Ulm, Germany.,Trauma Research Centre, University of Ulm, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopaedic Research and Biomechanics, University of Ulm, Ulm, Germany.,Trauma Research Centre, University of Ulm, Ulm, Germany
| | - Katja Prystaz
- Institute of Orthopaedic Research and Biomechanics, University of Ulm, Ulm, Germany.,Trauma Research Centre, University of Ulm, Ulm, Germany
| | - Adelheid Hainzl
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Carsten M Kroeger
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Ioanna Krikki
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| | - Titus J Brinker
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany.,Department of National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University of Ulm, Ulm, Germany.,Trauma Research Centre, University of Ulm, Ulm, Germany
| | - Martina Gatzka
- Department of Dermatology and Allergic Diseases, University of Ulm, Ulm, Germany
| |
Collapse
|
49
|
Lin BD, Carnero Montoro E, Bell JT, Boomsma DI, de Geus EJ, Jansen R, Kluft C, Mangino M, Penninx B, Spector TD, Willemsen G, Hottenga JJ. 2SNP heritability and effects of genetic variants for neutrophil-to-lymphocyte and platelet-to-lymphocyte ratio. J Hum Genet 2017; 62:979-988. [PMID: 29066854 PMCID: PMC5669488 DOI: 10.1038/jhg.2017.76] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/24/2017] [Accepted: 06/13/2017] [Indexed: 01/13/2023]
Abstract
Neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) are important biomarkers for disease development and progression. To gain insight into the genetic causes of variance in NLR and PLR in the general population, we conducted genome-wide association (GWA) analyses and estimated SNP heritability in a sample of 5901 related healthy Dutch individuals. GWA analyses identified a new genome-wide significant locus on the HBS1L-MYB intergenic region for PLR, which replicated in a sample of 2538 British twins. For platelet count, we replicated three known genome-wide significant loci in our cohort (at CCDC71L-PIK3CG, BAK1 and ARHGEF3). For neutrophil count, we replicated the PSMD3 locus. For the identified top SNPs, we found significant cis and trans expression quantitative trait loci effects for several loci involved in hematological and immunological pathways. Linkage Disequilibrium score (LD) regression analyses for PLR and NLR confirmed that both traits are heritable, with a polygenetic SNP heritability for PLR of 14.1%, and for NLR of 2.4%. Genetic correlations were present between ratios and the constituent counts, with the genetic correlation (r=0.45) of PLR with platelet count reaching statistical significance. In conclusion, we established that two important biomarkers have a significant heritable SNP component, and identified the first genome-wide locus for PLR.
Collapse
Affiliation(s)
- Bochao Danae Lin
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Elena Carnero Montoro
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Jordana T. Bell
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eco J. de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- EMGO+ Institute for Health & Care Research, VU Medical Center, Amsterdam, The Netherlands
| | - Rick Jansen
- Department of Psychiatry, VU Medical Center, Amsterdam, The Netherlands
| | | | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
- NIHR Biomedical Research Centre at Guy’s and St Thomas’ Foundation Trust, London SE1 9RT, UK
| | - Brenda Penninx
- Department of Psychiatry, VU Medical Center, Amsterdam, The Netherlands
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- EMGO+ Institute for Health & Care Research, VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
50
|
Wilms C, Kroeger CM, Hainzl AV, Banik I, Bruno C, Krikki I, Farsam V, Wlaschek M, Gatzka MV. MYSM1/2A-DUB is an epigenetic regulator in human melanoma and contributes to tumor cell growth. Oncotarget 2017; 8:67287-67299. [PMID: 28978033 PMCID: PMC5620173 DOI: 10.18632/oncotarget.18617] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 05/31/2017] [Indexed: 01/12/2023] Open
Abstract
Histone modifying enzymes, such as histone deacetylases (HDACs) and polycomb repressive complex (PRC) components, have been implicated in regulating tumor growth, epithelial-mesenchymal transition, tumor stem cell maintenance, or repression of tumor suppressor genes - and may be promising targets for combination therapies of melanoma and other cancers. According to recent findings, the histone H2A deubiquitinase 2A-DUB/Mysm1 interacts with the p53-axis in hematopoiesis and tissue differentiation in mice, in part by modulating DNA-damage responses in stem cell and progenitor compartments. Based on the identification of alterations in skin pigmentation and melanocyte specification in Mysm1-deficient mice, we hypothesized that MYSM1 may be involved in melanoma formation. In human melanoma samples, expression of MYSM1 was increased compared with normal skin melanocytes and nevi and co-localized with melanocyte markers such as Melan-A and c-KIT. Similarly, in melanoma cell lines A375 and SK-MEL-28 and in murine skin, expression of the deubiquitinase was detectable at the mRNA and protein level that was inducible by growth factor signals and UVB exposure, respectively. Upon stable silencing of MYSM1 in A375 and SK-MEL-28 melanoma cells by lentivirally-mediated shRNA expression, survival and proliferation were significantly reduced in five MYSM1 shRNA cell lines analyzed compared with control cells. In addition, MYSM1-silenced melanoma cells proliferated less well in softagar assays. In context with our finding that MYSM1 bound to the c-MET promoter region in close vicinity to PAX3 in melanoma cells, our data indicate that MYSM1 is an epigenetic regulator of melanoma growth and potentially promising new target for tumor therapy.
Collapse
Affiliation(s)
- Christina Wilms
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | - Carsten M Kroeger
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | - Adelheid V Hainzl
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | - Ishani Banik
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany.,ETH, 8092 Zurich, Switzerland
| | - Clara Bruno
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany.,Department of Neurology, Ulm University, 89081 Ulm, Germany
| | - Ioanna Krikki
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | - Vida Farsam
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | - Meinhard Wlaschek
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | - Martina V Gatzka
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| |
Collapse
|