1
|
Chen Y, Gong S, Tang J, Wang X, Gao Y, Yang H, Chen W, Hu H, Tong W, Lv K. LNK/SH2B3 Loss Exacerbates the Development of Myeloproliferative Neoplasms in CBL-deficient Mice. Stem Cell Rev Rep 2025; 21:509-519. [PMID: 39560864 PMCID: PMC11976319 DOI: 10.1007/s12015-024-10825-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
Genetic variations of signaling modulator protein LNK (also called SH2B3) are associated with relatively mild myeloproliferative phenotypes in patients with myeloproliferative neoplasms (MPN). However, these variations can induce more severe MPN disease and even leukemic transformation when co-existing with other driver mutations. In addition to the most prevalent driver mutation JAK2V617F, LNK mutations have been clinically identified in patients harboring CBL inactivation mutations, but its significance remains unclear. Here, using a transgenic mouse model, we demonstrated that mice with the loss of both Lnk and Cbl exhibited severe splenomegaly, extramedullary hematopoiesis and exacerbated myeloproliferative characteristics. Moreover, a population of Mac1+ myeloid cells expressed c-Kit in aged mice. Mechanistically, we discovered that LNK could pull down multiple regulatory subunits of the proteosome. Further analysis confirmed a positive role of LNK in regulating proteasome activity, independent of its well-established function in signaling transduction. Thus, our work reveals a novel function of LNK in coordinating with the E3 ligase CBL to regulate myeloid malignancies.
Collapse
Affiliation(s)
- Yafei Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, 518071, China
| | - Shangyu Gong
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, 518071, China
| | - Juan Tang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, 518071, China
| | - Xinying Wang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, 518071, China
| | - Yudan Gao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, 518071, China
| | - Hanying Yang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, 518071, China
| | - Wanze Chen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hailiang Hu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, 518071, China.
| | - Wei Tong
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
| | - Kaosheng Lv
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, School of Biomedical Sciences, Hunan University, Changsha, 410013, China.
| |
Collapse
|
2
|
Wintering A, Hecht A, Meyer J, Wong EB, Hübner J, Abelson S, Feldman K, Kennedy VE, Peretz CAC, French DL, Maguire JA, Jobaliya C, Vasquez MR, Desai S, Dulman R, Nemecek E, Haines H, Hammad M, El Haddad A, Kogan SC, Abdullaev Z, Chehab FF, Tasian SK, Smith CC, Loh ML, Stieglitz E. LNK/ SH2B3 as a novel driver in juvenile myelomonocytic leukemia. Haematologica 2024; 109:2533-2541. [PMID: 38152053 PMCID: PMC11290546 DOI: 10.3324/haematol.2023.283776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 12/19/2023] [Indexed: 12/29/2023] Open
Abstract
Mutations in five canonical Ras pathway genes (NF1, NRAS, KRAS, PTPN11 and CBL) are detected in nearly 90% of patients with juvenile myelomonocytic leukemia (JMML), a frequently fatal malignant neoplasm of early childhood. In this report, we describe seven patients diagnosed with SH2B3-mutated JMML, including five patients who were found to have initiating, loss-of-function mutations in the gene. SH2B3 encodes the adaptor protein LNK, a negative regulator of normal hematopoiesis upstream of the Ras pathway. These mutations were identified to be germline, somatic or a combination of both. Loss of function of LNK, which has been observed in other myeloid malignancies, results in abnormal proliferation of hematopoietic cells due to cytokine hypersensitivity and activation of the JAK/STAT signaling pathway. In vitro studies of induced pluripotent stem cell-derived JMML-like hematopoietic progenitor cells also demonstrated sensitivity of SH2B3-mutated hematopoietic progenitor cells to JAK inhibition. Lastly, we describe two patients with JMML and SH2B3 mutations who were treated with the JAK1/2 inhibitor ruxolitinib. This report expands the spectrum of initiating mutations in JMML and raises the possibility of targeting the JAK/STAT pathway in patients with SH2B3 mutations.
Collapse
Affiliation(s)
- Astrid Wintering
- Department of Pediatrics, Benioff Children's Hospitals, University of California San Francisco, San Francisco, CA 94158
| | - Anna Hecht
- Department of Hematology/Oncology, Klinikum Rechts der Isar, Technische Universität München, München
| | - Julia Meyer
- Department of Pediatrics, Benioff Children's Hospitals, University of California San Francisco, San Francisco, CA 94158
| | - Eric B Wong
- Department of Pediatrics, Benioff Children's Hospitals, University of California San Francisco, San Francisco, CA 94158
| | - Juwita Hübner
- Department of Pediatrics, Benioff Children's Hospitals, University of California San Francisco, San Francisco, CA 94158
| | - Sydney Abelson
- Department of Pediatrics, Benioff Children's Hospitals, University of California San Francisco, San Francisco, CA 94158
| | - Kira Feldman
- Department of Pediatrics, Benioff Children's Hospitals, University of California San Francisco, San Francisco, CA 94158
| | - Vanessa E Kennedy
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA 94158
| | - Cheryl A C Peretz
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158
| | - Deborah L French
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Jean Ann Maguire
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Chintan Jobaliya
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Marta Rojas Vasquez
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Sunil Desai
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada
| | - Robin Dulman
- Pediatric Hematology and Oncology, Pediatric Specialists of Virginia, Fairfax, VA 22031
| | - Eneida Nemecek
- OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239
| | - Hilary Haines
- Children's of Alabama, University of Alabama Hospital, Birmingham, AL 35233
| | - Mahmoud Hammad
- National Cancer Institute, Cairo University, Cairo, Egypt
| | - Alaa El Haddad
- National Cancer Institute, Cairo University, Cairo, Egypt
| | - Scott C Kogan
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94158
| | - Zied Abdullaev
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814
| | - Farid F Chehab
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Philadelphia, PA 19104, USA; Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA 19104
| | - Catherine C Smith
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA; Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158
| | - Mignon L Loh
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, and the Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, WA, 98105.
| | - Elliot Stieglitz
- Department of Pediatrics, Benioff Children's Hospitals, University of California San Francisco, San Francisco, CA 94158, USA; Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158.
| |
Collapse
|
3
|
Arfeuille C, Vial Y, Cadenet M, Caye-Eude A, Fenneteau O, Neven Q, Bonnard AA, Pizzi S, Carpentieri G, Capri Y, Girardi K, Pedace L, Macchiaiolo M, Boudhar K, Khaled MB, Chahla WA, Lutun A, Fahd M, Drunat S, Flex E, Dalle JH, Strullu M, Locatelli F, Tartaglia M, Cavé H. Germline bi-allelic SH2B3/LNK alteration predisposes to a neonatal juvenile myelomonocytic leukemia-like disorder. Haematologica 2024; 109:2542-2554. [PMID: 37981895 PMCID: PMC11290538 DOI: 10.3324/haematol.2023.283917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare, generally aggressive myeloproliferative neoplasm affecting young children. It is characterized by granulomonocytic expansion, with monocytosis infiltrating peripheral tissues. JMML is initiated by mutations upregulating RAS signaling. Approximately 10% of cases remain without an identified driver event. Exome sequencing of two unrelated cases of familial JMML of unknown genetics and analysis of the French JMML cohort identified 11 patients with variants in SH2B3, encoding LNK, a negative regulator of the JAK-STAT pathway. All variants were absent from healthy population databases, and the mutation spectrum was consistent with a loss of function of the LNK protein. A stoploss variant was shown to affect both protein synthesis and stability. The other variants were either truncating or missense, the latter affecting the SH2 domain that interacts with activated JAK. Of the 11 patients, eight from five families inherited pathogenic bi-allelic SH2B3 germline variants from their unaffected heterozygous parents. These children represent half of the cases with no identified causal mutation in the French cohort. They displayed typical clinical and hematologic features of JMML with neonatal onset and marked thrombocytopenia. They had a hypomethylated DNA profile with fetal characteristics and did not have additional genetic alterations. All patients showed partial or complete spontaneous clinical resolution. However, progression to thrombocythemia and immunity-related pathologies may be of concern later in life. Bi-allelic SH2B3 germline mutations thus define a new condition predisposing to a JMML-like disorder, suggesting that JAK pathway deregulation is capable of initiating JMML, and opening new therapeutic options.
Collapse
Affiliation(s)
- Chloé Arfeuille
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Yoann Vial
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Margaux Cadenet
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Aurélie Caye-Eude
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Odile Fenneteau
- Service d'Hématologie Biologique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Quentin Neven
- Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Adeline A Bonnard
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Simone Pizzi
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Giovanna Carpentieri
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Yline Capri
- Département de Génétique, Unité de Génétique clinique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Katia Girardi
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Lucia Pedace
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Marina Macchiaiolo
- Rare Diseases and Medical Genetics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Kamel Boudhar
- Service de réanimation néonatale, Hôpital Central de l'Armée, Alger, Algérie
| | - Monia Ben Khaled
- University of Tunis El Manar, Faculty of Medicine of Tunis, 1007, Tunisia. Pediatric Immuno- Hematology Unit, Bone Marrow Transplantation Center Tunis, Tunis, Tunisia
| | - Wadih Abou Chahla
- Service d'Hématologie Pédiatrique, Centre Hospitalier Universitaire de Lille, Lille
| | - Anne Lutun
- Service d'Hématologie Pédiatrique, Centre Hospitalier Universitaire d'Amiens, Amiens
| | - Mony Fahd
- Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Séverine Drunat
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome
| | - Jean-Hugues Dalle
- Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Marion Strullu
- INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris, France; Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Franco Locatelli
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy; Department of Pediatrics, Catholic University of the Sacred Hearth, 00168 Rome
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Hélène Cavé
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris.
| |
Collapse
|
4
|
Iseki M, Hidano S, Kudo F, Takaki S. Control of germinal center B cell survival and IgE production by an adaptor molecule containing PH and SH2 domains, Aps/Sh2b2. Sci Rep 2024; 14:17767. [PMID: 39090233 PMCID: PMC11294469 DOI: 10.1038/s41598-024-68739-3] [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: 12/21/2023] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
The germinal centers (GCs) are structure found within secondary lymphoid organs and are important for the antibody-producing response against foreign antigens. In GCs, antigen-specific B cells proliferate intensely, inducing immunoglobulin class switching. Recent studies have shown that GCs are also an important site for class switching to IgE, which is implicated in allergy. However, the mechanisms by which IgE production is regulated in GCs remain unclear. Here, we found impairment in IgE-specific production and a reduction of GC B cells after immunization in mice deficient in the Aps/Sh2b2 gene encoding the Lnk/Sh2b family adaptor protein Aps. GC B cells express higher levels of the Aps gene than non-GC B cells, and cell death of Aps-/- GC B cells is enhanced compared to wild-type GC B cells. An in vitro culture system with purified Aps-/- B cells induced the same level of IgE production and frequencies of IgE+ B cells as wild-type B cells. We found that Aps deficiency in B cells resulted in augmented depletion of IgE+ blasts by B cell receptor crosslinking with anti-CD79b antibodies compared to wild-type IgE+ cells. These results suggest that Aps regulates IgE production by controlling the survival of GC B cells and IgE+ plasma cells and may serve as a potential therapeutic target to control IgE production.
Collapse
Affiliation(s)
- Masanori Iseki
- Department of Immune Regulation, The Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Ichikawa, Chiba, Japan.
- Department of Immunology and Molecular Genetics, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| | - Shinya Hidano
- Department of Immune Regulation, The Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Ichikawa, Chiba, Japan
| | - Fujimi Kudo
- Department of Immune Regulation, The Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Ichikawa, Chiba, Japan
- Department of Systems Medicine, Chiba University Graduate School of Medicine, Inohana, Chuo-Ku, Chiba, Japan
| | - Satoshi Takaki
- Department of Immune Regulation, The Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Ichikawa, Chiba, Japan
| |
Collapse
|
5
|
Dou H, Wang R, Tavallaie M, Xiao T, Olszewska M, Papapetrou EP, Tall AR, Wang N. Hematopoietic and eosinophil-specific LNK(SH2B3) deficiency promotes eosinophilia and arterial thrombosis. Blood 2024; 143:1758-1772. [PMID: 38096361 PMCID: PMC11443577 DOI: 10.1182/blood.2023021055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/28/2023] [Accepted: 12/03/2023] [Indexed: 03/25/2024] Open
Abstract
ABSTRACT Increased eosinophil counts are associated with cardiovascular disease and may be an independent predictor of major cardiovascular events. However, the causality and underlying mechanisms are poorly understood. Genome-wide association studies have shown an association of a common LNK variant (R262W, T allele) with eosinophilia and atherothrombotic disorders. LNK(TT) reduces LNK function, and Lnk-deficient mice display accelerated atherosclerosis and thrombosis. This study was undertaken to assess the role of eosinophils in arterial thrombosis in mice with hematopoietic Lnk deficiency. Hematopoietic Lnk deficiency increased circulating and activated eosinophils, JAK/STAT signaling in eosinophils, and carotid arterial thrombosis with increased eosinophil abundance and extracellular trap formation (EETosis) in thrombi. Depletion of eosinophils by anti-Siglec-F antibody or by the ΔdbIGata1 mutation eliminated eosinophils in thrombi and markedly reduced thrombosis in mice with hematopoietic Lnk deficiency but not in control mice. Eosinophil depletion reduced neutrophil abundance and NETosis in thrombi without altering circulating neutrophil counts. To assess the role of Lnk specifically in eosinophils, we crossed Lnkf/f mice with eoCre mice. LnkΔeos mice displayed isolated eosinophilia, increased eosinophil activation, and accelerated arterial thrombosis associated with increased EETosis and NETosis in thrombi. DNase I infusion abolished EETs and neutrophil extracellular traps (NETs) in thrombi and reversed the accelerated thrombosis. Human induced pluripotent stem cell-derived LNK(TT) eosinophils showed increased activation and EETosis relative to isogenic LNK(CC) eosinophils, demonstrating human relevance. These studies show a direct link between eosinophilia, EETosis, and atherothrombosis in hematopoietic Lnk deficiency and an essential role of eosinophil LNK in suppression of arterial thrombosis.
Collapse
Affiliation(s)
- Huijuan Dou
- Molecular Medicine, Columbia University Medical Center, New York, NY
| | - Ranran Wang
- Molecular Medicine, Columbia University Medical Center, New York, NY
| | - Mojdeh Tavallaie
- Molecular Medicine, Columbia University Medical Center, New York, NY
| | - Tong Xiao
- Molecular Medicine, Columbia University Medical Center, New York, NY
| | - Malgorzata Olszewska
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eirini P. Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alan R. Tall
- Molecular Medicine, Columbia University Medical Center, New York, NY
| | - Nan Wang
- Molecular Medicine, Columbia University Medical Center, New York, NY
| |
Collapse
|
6
|
Zhang Y, Morris R, Brown GJ, Lorenzo AMD, Meng X, Kershaw NJ, Kiridena P, Burgio G, Gross S, Cappello JY, Shen Q, Wang H, Turnbull C, Lea-Henry T, Stanley M, Yu Z, Ballard FD, Chuah A, Lee JC, Hatch AM, Enders A, Masters SL, Headley AP, Trnka P, Mallon D, Fletcher JT, Walters GD, Šestan M, Jelušić M, Cook MC, Athanasopoulos V, Fulcher DA, Babon JJ, Vinuesa CG, Ellyard JI. Rare SH2B3 coding variants in lupus patients impair B cell tolerance and predispose to autoimmunity. J Exp Med 2024; 221:e20221080. [PMID: 38417019 PMCID: PMC10901239 DOI: 10.1084/jem.20221080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 03/14/2023] [Accepted: 01/17/2024] [Indexed: 03/01/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease with a clear genetic component. While most SLE patients carry rare gene variants in lupus risk genes, little is known about their contribution to disease pathogenesis. Amongst them, SH2B3-a negative regulator of cytokine and growth factor receptor signaling-harbors rare coding variants in over 5% of SLE patients. Here, we show that unlike the variant found exclusively in healthy controls, SH2B3 rare variants found in lupus patients are predominantly hypomorphic alleles, failing to suppress IFNGR signaling via JAK2-STAT1. The generation of two mouse lines carrying patients' variants revealed that SH2B3 is important in limiting the number of immature and transitional B cells. Furthermore, hypomorphic SH2B3 was shown to impair the negative selection of immature/transitional self-reactive B cells and accelerate autoimmunity in sensitized mice, at least in part due to increased IL-4R signaling and BAFF-R expression. This work identifies a previously unappreciated role for SH2B3 in human B cell tolerance and lupus risk.
Collapse
Affiliation(s)
- Yaoyuan Zhang
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Rhiannon Morris
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Grant J. Brown
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Ayla May D. Lorenzo
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Xiangpeng Meng
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Nadia J. Kershaw
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Pamudika Kiridena
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Gaétan Burgio
- Division of Genome Sciences and Cancer, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Simon Gross
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Jean Y. Cappello
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Qian Shen
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Francis Crick Institute, London, UK
| | - Hao Wang
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Francis Crick Institute, London, UK
| | - Cynthia Turnbull
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Tom Lea-Henry
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- The Canberra Hospital, Garran, Australia
| | - Maurice Stanley
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Zhijia Yu
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Fiona D. Ballard
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Aaron Chuah
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - James C. Lee
- Francis Crick Institute, London, UK
- Department of Gastroenterology, Division of Medicine, Institute for Liver and Digestive Health, University College London, London, UK
| | - Ann-Maree Hatch
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- The Canberra Hospital, Garran, Australia
| | - Anselm Enders
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Seth L. Masters
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | | | - Peter Trnka
- Queensland Children’s Hospital, South Brisbane, Australia
| | | | | | | | - Mario Šestan
- Department of Pediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Marija Jelušić
- Department of Pediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Matthew C. Cook
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- The Canberra Hospital, Garran, Australia
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
| | - Vicki Athanasopoulos
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - David A. Fulcher
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| | - Jeffrey J. Babon
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Carola G. Vinuesa
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Francis Crick Institute, London, UK
| | - Julia I. Ellyard
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, The Australian National University, Acton, Australia
- Centre for Personalised Immunology, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| |
Collapse
|
7
|
Baig MS, Barmpoutsi S, Bharti S, Weigert A, Hirani N, Atre R, Khabiya R, Sharma R, Sarup S, Savai R. Adaptor molecules mediate negative regulation of macrophage inflammatory pathways: a closer look. Front Immunol 2024; 15:1355012. [PMID: 38482001 PMCID: PMC10933033 DOI: 10.3389/fimmu.2024.1355012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/22/2024] [Indexed: 04/13/2024] Open
Abstract
Macrophages play a central role in initiating, maintaining, and terminating inflammation. For that, macrophages respond to various external stimuli in changing environments through signaling pathways that are tightly regulated and interconnected. This process involves, among others, autoregulatory loops that activate and deactivate macrophages through various cytokines, stimulants, and other chemical mediators. Adaptor proteins play an indispensable role in facilitating various inflammatory signals. These proteins are dynamic and flexible modulators of immune cell signaling and act as molecular bridges between cell surface receptors and intracellular effector molecules. They are involved in regulating physiological inflammation and also contribute significantly to the development of chronic inflammatory processes. This is at least partly due to their involvement in the activation and deactivation of macrophages, leading to changes in the macrophages' activation/phenotype. This review provides a comprehensive overview of the 20 adaptor molecules and proteins that act as negative regulators of inflammation in macrophages and effectively suppress inflammatory signaling pathways. We emphasize the functional role of adaptors in signal transduction in macrophages and their influence on the phenotypic transition of macrophages from pro-inflammatory M1-like states to anti-inflammatory M2-like phenotypes. This endeavor mainly aims at highlighting and orchestrating the intricate dynamics of adaptor molecules by elucidating the associated key roles along with respective domains and opening avenues for therapeutic and investigative purposes in clinical practice.
Collapse
Affiliation(s)
- Mirza S. Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Spyridoula Barmpoutsi
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Shreya Bharti
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| | - Nik Hirani
- MRC Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rajat Atre
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rakhi Khabiya
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rahul Sharma
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Shivmuni Sarup
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rajkumar Savai
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
8
|
Andrews C, Conneally E, Langabeer SE. Molecular diagnostic criteria of myeloproliferative neoplasms. Expert Rev Mol Diagn 2023; 23:1077-1090. [PMID: 37999991 DOI: 10.1080/14737159.2023.2277370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/26/2023] [Indexed: 11/26/2023]
Abstract
INTRODUCTION Myeloproliferative neoplasms (MPN) are a heterogeneous group of clonal hematopoietic stem cell neoplasms characterized by the driver mutations JAK2, CALR, and MPL. These mutations cause constitutive activation of JAK-STAT signaling, which is central to pathogenesis of MPNs. Next-generation sequencing has further expanded the molecular landscape allowing for improved diagnostics, prognostication, and targeted therapy. AREAS COVERED This review aims to address current understanding of the molecular diagnosis of MPN not only through improved awareness of the driver mutations but also the disease modifying mutations. In addition, other genetic factors such as clonal hematopoiesis of indeterminate potential (CHIP), order of mutation, and mutation co-occurrence are discussed and how these factors influence disease initiation and ultimately progression. How this molecular information is incorporated into risk stratification models allowing for earlier intervention and targeted therapy in the future will be addressed further. EXPERT OPINION The genomic landscape of the MPN has evolved in the last 15 years with integration of next-generation sequencing becoming the gold standard of MPN management. Although diagnostics and prognostication have become more personalized, additional studies are required to translate these molecular findings into targeted therapy therefore improving patient outcomes.
Collapse
Affiliation(s)
- Claire Andrews
- Department of Haematology, St. Vincent's University Hospital, Dublin, Ireland
| | | | | |
Collapse
|
9
|
Blombery P, Pazhakh V, Albuquerque AS, Maimaris J, Tu L, Briones Miranda B, Evans F, Thompson ER, Carpenter B, Proctor I, Curtin JA, Lambert J, Burns SO, Lieschke GJ. Biallelic deleterious germline SH2B3 variants cause a novel syndrome of myeloproliferation and multi-organ autoimmunity. EJHAEM 2023; 4:463-469. [PMID: 37206266 PMCID: PMC10188477 DOI: 10.1002/jha2.698] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/01/2023] [Accepted: 04/15/2023] [Indexed: 05/21/2023]
Abstract
SH2B3 is a negative regulator of multiple cytokine receptor signalling pathways in haematopoietic tissue. To date, a single kindred has been described with germline biallelic loss-of-function SH2B3 variants characterized by early onset developmental delay, hepatosplenomegaly and autoimmune thyroiditis/hepatitis. Herein, we described two further unrelated kindreds with germline biallelic loss-of-function SH2B3 variants that show striking phenotypic similarity to each other as well as to the previous kindred of myeloproliferation and multi-organ autoimmunity. One proband also suffered severe thrombotic complications. CRISPR-Cas9 gene editing of zebrafish sh2b3 created assorted deleterious variants in F0 crispants, which manifest significantly increased number of macrophages and thrombocytes, partially replicating the human phenotype. Treatment of the sh2b3 crispant fish with ruxolitinib intercepted this myeloproliferative phenotype. Skin-derived fibroblasts from one patient demonstrated increased phosphorylation of JAK2 and STAT5 after stimulation with IL-3, GH, GM-CSF and EPO compared to healthy controls. In conclusion, these additional probands and functional data in combination with the previous kindred provide sufficient evidence for biallelic homozygous deleterious variants in SH2B3 to be considered a valid gene-disease association for a clinical syndrome of bone marrow myeloproliferation and multi-organ autoimmune manifestations.
Collapse
Affiliation(s)
- Piers Blombery
- Clinical HaematologyPeter MacCallum Cancer Centre/Royal Melbourne HospitalMelbourneVictoriaAustralia
- University of MelbourneMelbourneVictoriaAustralia
| | - Vahid Pazhakh
- Clinical HaematologyPeter MacCallum Cancer Centre/Royal Melbourne HospitalMelbourneVictoriaAustralia
- Australian Regenerative Medicine InstituteMonash UniversityClaytonVictoriaAustralia
| | | | - Jesmeen Maimaris
- Institute of Immunity and TransplantationUniversity College LondonLondonUK
- Department of ImmunologyRoyal Free London NHS Foundation TrustLondonUK
| | - Lingge Tu
- Australian Regenerative Medicine InstituteMonash UniversityClaytonVictoriaAustralia
| | | | - Florence Evans
- Institute of Immunity and TransplantationUniversity College LondonLondonUK
| | - Ella R. Thompson
- Clinical HaematologyPeter MacCallum Cancer Centre/Royal Melbourne HospitalMelbourneVictoriaAustralia
- University of MelbourneMelbourneVictoriaAustralia
| | - Ben Carpenter
- Department of HaematologyUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Ian Proctor
- Department of HaematologyUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Julie A. Curtin
- Haematology DepartmentChildren's Hospital at WestmeadWestmeadNew South WalesAustralia
| | - Jonathan Lambert
- Department of HaematologyUniversity College London Hospitals NHS Foundation TrustLondonUK
- Department of HaematologyUCL Cancer InstituteUniversity College LondonLondonUK
| | - Siobhan O. Burns
- Institute of Immunity and TransplantationUniversity College LondonLondonUK
- Department of ImmunologyRoyal Free London NHS Foundation TrustLondonUK
| | - Graham J. Lieschke
- Clinical HaematologyPeter MacCallum Cancer Centre/Royal Melbourne HospitalMelbourneVictoriaAustralia
- Australian Regenerative Medicine InstituteMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
10
|
Beghini A, Leuzzi L, Abazari N, Bossi LE, Guido V, Trojani A, Cairoli R. A novel start-loss mutation of the SH2B3 gene in a family with myeloproliferative neoplasms. Hematol Oncol 2022; 40:1109-1112. [PMID: 35570682 DOI: 10.1002/hon.3023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The ever-increasing advances in high-throughput sequencing have broadened our understanding of the genetic pathogenesis of Philadelphia-negative myeloproliferative neoplasms (MPNs). Convergent studies have shown that MPN driver mutations associate with additional mutations found in genes coding for negative regulators of the JAK/STAT signaling, including the SH2B3 (SH2B-adaptor protein 3, also known as LNK). Here, we describe a novel heterozygous start-loss mutation of the SH2B3 gene (c.3G>A, SH2B3M? ) in a consanguineous family characterized by recurrent early onset of JAK2V617F -positive MPNs. The model represented by this pedigree suggests that the SH2B3 could be a predisposing mutation that facilitates the acquisition of driver mutations.
Collapse
Affiliation(s)
| | - Livia Leuzzi
- Department of Hematology, Oncology and Molecular Medicine, Niguarda Cancer Center, Milan, Italy
| | - Nazanin Abazari
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Luca E Bossi
- Department of Hematology, Oncology and Molecular Medicine, Niguarda Cancer Center, Milan, Italy
| | - Valentina Guido
- Department of Hematology, Oncology and Molecular Medicine, Pathology Unit ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessandra Trojani
- Department of Hematology, Oncology and Molecular Medicine, Niguarda Cancer Center, Milan, Italy
| | - Roberto Cairoli
- Department of Hematology, Oncology and Molecular Medicine, Niguarda Cancer Center, Milan, Italy
| |
Collapse
|
11
|
Alexander MR, Hank S, Dale BL, Himmel L, Zhong X, Smart CD, Fehrenbach DJ, Chen Y, Prabakaran N, Tirado B, Centrella M, Ao M, Du L, Shyr Y, Levy D, Madhur MS. A Single Nucleotide Polymorphism in SH2B3/LNK Promotes Hypertension Development and Renal Damage. Circ Res 2022; 131:731-747. [PMID: 36169218 PMCID: PMC9588739 DOI: 10.1161/circresaha.121.320625] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 09/15/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND SH2B3 (SH2B adaptor protein 3) is an adaptor protein that negatively regulates cytokine signaling and cell proliferation. A common missense single nucleotide polymorphism in SH2B3 (rs3184504) results in substitution of tryptophan (Trp) for arginine (Arg) at amino acid 262 and is a top association signal for hypertension in human genome-wide association studies. Whether this variant is causal for hypertension, and if so, the mechanism by which it impacts pathogenesis is unknown. METHODS We used CRISPR-Cas9 technology to create mice homozygous for the major (Arg/Arg) and minor (Trp/Trp) alleles of this SH2B3 polymorphism. Mice underwent angiotensin II (Ang II) infusion to evaluate differences in blood pressure (BP) elevation and end-organ damage including albuminuria and renal fibrosis. Cytokine production and Stat4 phosphorylation was also assessed in Arg/Arg and Trp/Trp T cells. RESULTS Trp/Trp mice exhibit 10 mmHg higher systolic BP during chronic Ang II infusion compared to Arg/Arg controls. Renal injury and perivascular fibrosis are exacerbated in Trp/Trp mice compared to Arg/Arg controls following Ang II infusion. Renal and ex vivo stimulated splenic CD8+ T cells from Ang II-infused Trp/Trp mice produce significantly more interferon gamma (IFNg) compared to Arg/Arg controls. Interleukin-12 (IL-12)-induced IFNg production is greater in Trp/Trp compared to Arg/Arg CD8+ T cells. In addition, IL-12 enhances Stat4 phosphorylation to a greater degree in Trp/Trp compared to Arg/Arg CD8+ T cells, suggesting that Trp-encoding SH2B3 exhibits less negative regulation of IL-12 signaling to promote IFNg production. Finally, we demonstrated that a multi-SNP model genetically predicting increased SH2B3 expression in lymphocytes is inversely associated with hypertension and hypertensive chronic kidney disease in humans.. CONCLUSIONS Taken together, these results suggest that the Trp encoding allele of rs3184504 is causal for BP elevation and renal dysfunction, in part through loss of SH2B3-mediated repression of T cell IL-12 signaling leading to enhanced IFNg production.
Collapse
Affiliation(s)
- Matthew R. Alexander
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, VUMC, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Samuel Hank
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Bethany L. Dale
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Lauren Himmel
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Xue Zhong
- Department of Medicine, Division of Genetic Medicine, VUMC, Nashville, TN, USA
| | - Charles D. Smart
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Daniel J. Fehrenbach
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Yuhan Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | | | | | - Megan Centrella
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Mingfang Ao
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Liping Du
- Department of Biostatistics, VUMC, Nashville, TN
| | - Yu Shyr
- Department of Biostatistics, VUMC, Nashville, TN
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA and Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Meena S. Madhur
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, VUMC, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
12
|
Baccelli F, Leardini D, Muratore E, Messelodi D, Bertuccio SN, Chiriaco M, Cancrini C, Conti F, Castagnetti F, Pedace L, Pession A, Yoshimi A, Niemeyer C, Tartaglia M, Locatelli F, Masetti R. Immune dysregulation associated with co-occurring germline CBL and SH2B3 variants. Hum Genomics 2022; 16:40. [PMID: 36123612 PMCID: PMC9484243 DOI: 10.1186/s40246-022-00414-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND CBL syndrome is a RASopathy caused by heterozygous germline mutations of the Casitas B-lineage lymphoma (CBL) gene. It is characterized by heterogeneous clinical phenotype, including developmental delay, facial dysmorphisms, cardiovascular malformations and an increased risk of cancer development, particularly juvenile myelomonocytic leukemia (JMML). Although the clinical phenotype has been progressively defined in recent years, immunological manifestations have not been well elucidated to date. METHODS We studied the genetic, immunological, coagulative, and clinical profile of a family with CBL syndrome that came to our observation after the diagnosis of JMML, with homozygous CBL mutation, in one of the members. RESULTS Variant analysis revealed the co-occurrence of CBL heterozygous mutation (c.1141 T > C) and SH2B3 mutation (c.1697G > A) in two other members. Patients carrying both mutations showed an ALPS-like phenotype characterized by lymphoproliferation, cytopenia, increased double-negative T-cells, impaired Fas-mediated lymphocyte apoptosis, altered cell death in PBMC and low TRECs expression. A coagulative work-up was also performed and showed the presence of subclinical coagulative alterations in patients carrying both mutations. CONCLUSION In the reported family, we described immune dysregulation, as part of the clinical spectrum of CBL mutation with the co-occurrence of SH2B3.
Collapse
Affiliation(s)
- Francesco Baccelli
- Pediatric Oncology and Hematology "Lalla Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138, Bologna, Italy
| | - Davide Leardini
- Pediatric Oncology and Hematology "Lalla Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138, Bologna, Italy.
| | - Edoardo Muratore
- Pediatric Oncology and Hematology "Lalla Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138, Bologna, Italy
| | - Daria Messelodi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138, Bologna, Italy
| | | | - Maria Chiriaco
- Chair of Pediatrics, Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Caterina Cancrini
- Chair of Pediatrics, Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
- Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Academic Department of Pediatrics, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, 00165, Rome, Italy
| | - Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138, Bologna, Italy
| | - Fausto Castagnetti
- Hematology "Lorenzo E Ariosto Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
| | - Lucia Pedace
- Department of Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome, 00165, Rome, Italy
| | - Andrea Pession
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138, Bologna, Italy
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138, Bologna, Italy
| | - Ayami Yoshimi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Centre, Faculty of Medicine, University of Freiburg, 79085, Freiburg, Germany
| | - Charlotte Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Centre, Faculty of Medicine, University of Freiburg, 79085, Freiburg, Germany
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, 00165, Rome, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome, 00165, Rome, Italy
| | - Riccardo Masetti
- Pediatric Oncology and Hematology "Lalla Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138, Bologna, Italy
| |
Collapse
|
13
|
Genetic Background of Polycythemia Vera. Genes (Basel) 2022; 13:genes13040637. [PMID: 35456443 PMCID: PMC9027017 DOI: 10.3390/genes13040637] [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: 03/02/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 02/06/2023] Open
Abstract
Polycythemia vera belongs to myeloproliferative neoplasms, essentially by affecting the erythroblastic lineage. JAK2 alterations have emerged as major driver mutations triggering PV-phenotype with the V617F mutation detected in nearly 98% of cases. That’s why JAK2 targeting therapeutic strategies have rapidly emerged to counter the aggravation of the disease. Over decades of research, to go further in the understanding of the disease and its evolution, a wide panel of genetic alterations affecting multiple genes has been highlighted. These are mainly involved in alternative splicing, epigenetic, miRNA regulation, intracellular signaling, and transcription factors expression. If JAK2 mutation, irrespective of the nature of the alteration, is known to be a crucial event for the disease to initiate, additional mutations seem to be markers of progression and poor prognosis. These discoveries have helped to characterize the complex genomic landscape of PV, resulting in potentially new adapted therapeutic strategies for patients concerning all the genetic interferences.
Collapse
|
14
|
Rajabi F, Abdollahimajd F, Jabalameli N, Nassiri Kashani M, Firooz A. The Immunogenetics of Alopecia areata. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:19-59. [DOI: 10.1007/978-3-030-92616-8_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
15
|
Morris R, Butler L, Perkins A, Kershaw NJ, Babon JJ. The Role of LNK (SH2B3) in the Regulation of JAK-STAT Signalling in Haematopoiesis. Pharmaceuticals (Basel) 2021; 15:ph15010024. [PMID: 35056081 PMCID: PMC8781068 DOI: 10.3390/ph15010024] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 01/05/2023] Open
Abstract
LNK is a member of the SH2B family of adaptor proteins and is a non-redundant regulator of cytokine signalling. Cytokines are secreted intercellular messengers that bind to specific receptors on the surface of target cells to activate the Janus Kinase-Signal Transducer and Activator of Transcription (JAK-STAT) signalling pathway. Activation of the JAK-STAT pathway leads to proliferative and often inflammatory effects, and so the amplitude and duration of signalling are tightly controlled. LNK binds phosphotyrosine residues to signalling proteins downstream of cytokines and constrains JAK-STAT signalling. Mutations in LNK have been identified in a range of haematological and inflammatory diseases due to increased signalling following the loss of LNK function. Here, we review the regulation of JAK-STAT signalling via the adaptor protein LNK and discuss the role of LNK in haematological diseases.
Collapse
Affiliation(s)
- Rhiannon Morris
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (R.M.); (N.J.K.)
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Liesl Butler
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3001, Australia; (L.B.); (A.P.)
- Alfred Health, Melbourne, VIC 3001, Australia
| | - Andrew Perkins
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3001, Australia; (L.B.); (A.P.)
- Alfred Health, Melbourne, VIC 3001, Australia
| | - Nadia J. Kershaw
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (R.M.); (N.J.K.)
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Jeffrey J. Babon
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (R.M.); (N.J.K.)
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
- Correspondence: ; Tel.: +61-3-9345-2960; Fax: +61-3-9347-0852
| |
Collapse
|
16
|
Stavnichuk M, Komarova SV. Megakaryocyte-driven changes in bone health: lessons from mouse models of myelofibrosis and related disorders. Am J Physiol Cell Physiol 2021; 322:C177-C184. [PMID: 34910601 DOI: 10.1152/ajpcell.00328.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Over the years, numerous studies demonstrated reciprocal communications between processes of bone marrow hematopoiesis and bone remodeling. Megakaryocytes, rare bone marrow cells responsible for platelet production, were demonstrated to be involved in bone homeostasis. Myelofibrosis, characterized by an increase in pleomorphic megakaryocytes in the bone marrow, commonly leads to the development of osteosclerosis. In vivo, an increase in megakaryocyte number was shown to result in osteosclerosis in GATA-1low, NF-E2-/-, TPOhigh, Mpllf/f;PF4cre, Lnk-/-, Mpig6b-/-, Mpig6bfl/fl;Gp1ba-Cr+/KI, Pt-vWD mouse models. In vitro, megakaryocytes stimulate osteoblast proliferation and have variable effects on osteoclast proliferation and activity through soluble factors and direct cell-cell communications. Intriguingly, new studies revealed that the ability of megakaryocytes to communicate with bone cells is affected by the age and sex of animals. This mini-review summarises changes seen in bone architecture and bone cell function in mouse models with an elevated number of megakaryocytes and the effects megakaryocytes have on osteoblasts and osteoclasts in vitro, and discusses potential molecular players that can mediate these effects.
Collapse
Affiliation(s)
- Mariya Stavnichuk
- Shriners Hospital for Children - Canada, Montreal, Quebec, Canada.,Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Shriners Hospital for Children - Canada, Montreal, Quebec, Canada.,Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Jiao H, Zhang M, Zhang Y, Wang Y, Li WD. Pathway Association Studies Reveal Gene Loci and Pathway Networks that Associated With Plasma Cystatin C Levels. Front Genet 2021; 12:711155. [PMID: 34899825 PMCID: PMC8656399 DOI: 10.3389/fgene.2021.711155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/09/2021] [Indexed: 01/09/2023] Open
Abstract
As a marker for glomerular filtration, plasma cystatin C level is used to evaluate kidney function. To decipher genetic factors that control the plasma cystatin C level, we performed genome-wide association and pathway association studies using United Kingdom Biobank data. One hundred fifteen loci yielded p values less than 1 × 10−100, three genes (clusters) showed the most significant associations, including the CST8-CST9 cluster on chromosome 20, the SH2B3-ATXN2 gene region on chromosome 12, and the SHROOM3-CCDC158 gene region on chromosome 4. In pathway association studies, forty significant pathways had FDR (false discovery rate) and or FWER (family-wise error rate) ≤ 0.001: spermatogenesis, leukocyte trans-endothelial migration, cell adhesion, glycoprotein, membrane lipid, steroid metabolic process, and insulin signaling pathways were among the most significant pathways that associated with the plasma cystatin C levels. We also performed Genome-wide association studies for eGFR, top associated genes were largely overlapped with those for cystatin C.
Collapse
Affiliation(s)
- Hongxiao Jiao
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Miaomiao Zhang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuan Zhang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,College of Public Health, Tianjin Medical University, Tianjin, China
| | - Yaogang Wang
- College of Public Health, Tianjin Medical University, Tianjin, China
| | - Wei-Dong Li
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
18
|
Allenspach EJ, Shubin NJ, Cerosaletti K, Mikacenic C, Gorman JA, MacQuivey MA, Rosen AB, Timms AE, Wray-Dutra MN, Niino K, Liggitt D, Wurfel MM, Buckner JH, Piliponsky AM, Rawlings DJ. The Autoimmune Risk R262W Variant of the Adaptor SH2B3 Improves Survival in Sepsis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2710-2719. [PMID: 34740959 PMCID: PMC8612972 DOI: 10.4049/jimmunol.2100454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/27/2021] [Indexed: 11/19/2022]
Abstract
The single-nucleotide polymorphism (SNP) rs3184504 is broadly associated with increased risk for multiple autoimmune and cardiovascular diseases. Although the allele is uniquely enriched in European descent, the mechanism for the widespread selective sweep is not clear. In this study, we find the rs3184504*T allele had a strong association with reduced mortality in a human sepsis cohort. The rs3184504*T allele associates with a loss-of-function amino acid change (p.R262W) in the adaptor protein SH2B3, a likely causal variant. To better understand the role of SH2B3 in sepsis, we used mouse modeling and challenged SH2B3-deficient mice with a polymicrobial cecal-ligation puncture (CLP) procedure. We found SH2B3 deficiency improved survival and morbidity with less organ damage and earlier bacterial clearance compared with control mice. The peritoneal infiltrating cells exhibited augmented phagocytosis in Sh2b3 -/- mice with enriched recruitment of Ly6Chi inflammatory monocytes despite equivalent or reduced chemokine expression. Rapid cycling of monocytes and progenitors occurred uniquely in the Sh2b3 -/- mice following CLP, suggesting augmented myelopoiesis. To model the hypomorphic autoimmune risk allele, we created a novel knockin mouse harboring a similar point mutation in the murine pleckstrin homology domain of SH2B3. At baseline, phenotypic changes suggested a hypomorphic allele. In the CLP model, homozygous knockin mice displayed improved mortality and morbidity compared with wild-type or heterozygous mice. Collectively, these data suggest that hypomorphic SH2B3 improves the sepsis response and that balancing selection likely contributed to the relative frequency of the autoimmune risk variant.
Collapse
Affiliation(s)
- Eric J. Allenspach
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA,Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Nicholas J. Shubin
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Karen Cerosaletti
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Carmen Mikacenic
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA,Department of Medicine, Division of Pulmonary and Critical Care, University of Washington, Seattle, Washington, USA
| | - Jacquelyn A Gorman
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Matthew A. MacQuivey
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Aaron B.I. Rosen
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Andrew E. Timms
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Michelle N. Wray-Dutra
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Kerri Niino
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, Washington, USA
| | - Mark M. Wurfel
- Department of Medicine, Division of Pulmonary and Critical Care, University of Washington, Seattle, Washington, USA
| | - Jane H. Buckner
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA,Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Adrian M. Piliponsky
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA,Departments of Pediatrics, Pathology and Global Health, University of Washington School of Medicine, Seattle, Washington, USA
| | - David J. Rawlings
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA,Department of Pediatrics, University of Washington, Seattle, Washington, USA,Department of Immunology, University of Washington, Seattle, Washington, USA,Correspondence should be addressed to D.J.R. () and E.J.A. ()
| |
Collapse
|
19
|
LNK (SH2B3) Inhibition Expands Healthy and Fanconi Anemia Human Hematopoietic Stem and Progenitor Cells. Blood Adv 2021; 6:731-745. [PMID: 34844262 PMCID: PMC8945310 DOI: 10.1182/bloodadvances.2021004205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 11/07/2021] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) remains the only curative treatment for a variety of hematological diseases. Allogenic HSCT requires hematopoietic stem cells (HSCs) from matched donors and comes with cytotoxicity and mortality. Recent advances in genome modification of HSCs have demonstrated the possibility of using autologous HSCT-based gene therapy to cure monogenic diseases, such as the inherited bone marrow failure (BMF) syndrome Fanconi Anemia (FA). However, for FA and other BMF syndromes insufficient HSC numbers with functional defects results in delayed hematopoietic recovery and increased risk of graft failure. We and others previously identified the adaptor protein Lnk (Sh2b3) as a critical negative regulator of murine HSC homeostasis. However, whether LNK (SH2B3) controls human HSCs has not been studied. Here, we demonstrate that depletion of LNK via lentiviral expression of miR30-based short hairpin RNAs (shRNAs) resulted in robust expansion of transplantable human HSCs that provided balanced multilineage reconstitution in primary and secondary mouse recipients. Importantly, LNK depletion enhanced cytokine mediated JAK/STAT activation in CD34+ hematopoietic stem and progenitor cells (HSPCs). Moreover, we demonstrate that LNK depletion expands primary HSPCs associated with FA. In xenotransplant, engraftment defects of FANCD2-depleted FA-like HSCs were markedly improved by LNK inhibition. Finally, targeting LNK in primary bone marrow HSPCs from FA patients enhanced their colony forming potential in vitro. Together, these results demonstrate the potential of targeting LNK to expand HSCs to improve HSCT and HSCT-based gene therapy.
Collapse
|
20
|
Morris R, Zhang Y, Ellyard JI, Vinuesa CG, Murphy JM, Laktyushin A, Kershaw NJ, Babon JJ. Structural and functional analysis of target recognition by the lymphocyte adaptor protein LNK. Nat Commun 2021; 12:6110. [PMID: 34671038 PMCID: PMC8528861 DOI: 10.1038/s41467-021-26394-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/30/2021] [Indexed: 01/17/2023] Open
Abstract
The SH2B family of adaptor proteins, SH2-B, APS, and LNK are key modulators of cellular signalling pathways. Whilst SH2-B and APS have been partially structurally and biochemically characterised, to date there has been no such characterisation of LNK. Here we present two crystal structures of the LNK substrate recognition domain, the SH2 domain, bound to phosphorylated motifs from JAK2 and EPOR, and biochemically define the basis for target recognition. The LNK SH2 domain adopts a canonical SH2 domain fold with an additional N-terminal helix. Targeted analysis of binding to phosphosites in signalling pathways indicated that specificity is conferred by amino acids one- and three-residues downstream of the phosphotyrosine. Several mutations in LNK showed impaired target binding in vitro and a reduced ability to inhibit signalling, allowing an understanding of the molecular basis of LNK dysfunction in variants identified in patients with myeloproliferative disease.
Collapse
Affiliation(s)
- Rhiannon Morris
- grid.1042.7Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Royal Parade, Parkville, VIC 3052 Australia
| | - Yaoyuan Zhang
- grid.1001.00000 0001 2180 7477Australia Department of Immunology and Infectious Diseases, Australian National University, Canberra, ACT Australia ,grid.1001.00000 0001 2180 7477Australia Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT Australia
| | - Julia I. Ellyard
- grid.1001.00000 0001 2180 7477Australia Department of Immunology and Infectious Diseases, Australian National University, Canberra, ACT Australia ,grid.1001.00000 0001 2180 7477Australia Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT Australia
| | - Carola G. Vinuesa
- grid.1001.00000 0001 2180 7477Australia Department of Immunology and Infectious Diseases, Australian National University, Canberra, ACT Australia ,grid.1001.00000 0001 2180 7477Australia Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT Australia
| | - James M. Murphy
- grid.1042.7Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Royal Parade, Parkville, VIC 3052 Australia
| | - Artem Laktyushin
- grid.1042.7Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Royal Parade, Parkville, VIC 3052 Australia
| | - Nadia J. Kershaw
- grid.1042.7Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Royal Parade, Parkville, VIC 3052 Australia
| | - Jeffrey J. Babon
- grid.1042.7Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Royal Parade, Parkville, VIC 3052 Australia
| |
Collapse
|
21
|
Guijarro-Hernández A, Vizmanos JL. A Broad Overview of Signaling in Ph-Negative Classic Myeloproliferative Neoplasms. Cancers (Basel) 2021; 13:cancers13050984. [PMID: 33652860 PMCID: PMC7956519 DOI: 10.3390/cancers13050984] [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: 02/08/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary There is growing evidence that Ph-negative myeloproliferative neoplasms are disorders in which multiple signaling pathways are significantly disturbed. The heterogeneous phenotypes observed among patients have highlighted the importance of having a comprehensive knowledge of the molecular mechanisms behind these diseases. This review aims to show a broad overview of the signaling involved in myeloproliferative neoplasms (MPNs) and other processes that can modify them, which could be helpful to better understand these diseases and develop more effective targeted treatments. Abstract Ph-negative myeloproliferative neoplasms (polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF)) are infrequent blood cancers characterized by signaling aberrations. Shortly after the discovery of the somatic mutations in JAK2, MPL, and CALR that cause these diseases, researchers extensively studied the aberrant functions of their mutant products. In all three cases, the main pathogenic mechanism appears to be the constitutive activation of JAK2/STAT signaling and JAK2-related pathways (MAPK/ERK, PI3K/AKT). However, some other non-canonical aberrant mechanisms derived from mutant JAK2 and CALR have also been described. Moreover, additional somatic mutations have been identified in other genes that affect epigenetic regulation, tumor suppression, transcription regulation, splicing and other signaling pathways, leading to the modification of some disease features and adding a layer of complexity to their molecular pathogenesis. All of these factors have highlighted the wide variety of cellular processes and pathways involved in the pathogenesis of MPNs. This review presents an overview of the complex signaling behind these diseases which could explain, at least in part, their phenotypic heterogeneity.
Collapse
Affiliation(s)
- Ana Guijarro-Hernández
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
| | - José Luis Vizmanos
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Correspondence:
| |
Collapse
|
22
|
Jacquelin S, Kramer F, Mullally A, Lane SW. Murine Models of Myelofibrosis. Cancers (Basel) 2020; 12:cancers12092381. [PMID: 32842500 PMCID: PMC7563264 DOI: 10.3390/cancers12092381] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 01/22/2023] Open
Abstract
Myelofibrosis (MF) is subtype of myeloproliferative neoplasm (MPN) characterized by a relatively poor prognosis in patients. Understanding the factors that drive MF pathogenesis is crucial to identifying novel therapeutic approaches with the potential to improve patient care. Driver mutations in three main genes (janus kinase 2 (JAK2), calreticulin (CALR), and myeloproliferative leukemia virus oncogene (MPL)) are recurrently mutated in MPN and are sufficient to engender MPN using animal models. Interestingly, animal studies have shown that the underlying molecular mutation and the acquisition of additional genetic lesions is associated with MF outcome and transition from early stage MPN such as essential thrombocythemia (ET) and polycythemia vera (PV) to secondary MF. In this issue, we review murine models that have contributed to a better characterization of MF pathobiology and identification of new therapeutic opportunities in MPN.
Collapse
Affiliation(s)
- Sebastien Jacquelin
- Cancer program QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- Correspondence: (S.J.); (S.W.L.)
| | - Frederike Kramer
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.K.); (A.M.)
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.K.); (A.M.)
| | - Steven W. Lane
- Cancer program QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- Cancer Care Services, The Royal Brisbane and Women’s Hospital, Brisbane 4029, Australia
- University of Queensland, St Lucia, QLD 4072, Australia
- Correspondence: (S.J.); (S.W.L.)
| |
Collapse
|
23
|
Palomero L, Galván-Femenía I, de Cid R, Espín R, Barnes DR, Cimba, Blommaert E, Gil-Gil M, Falo C, Stradella A, Ouchi D, Roso-Llorach A, Violan C, Peña-Chilet M, Dopazo J, Extremera AI, García-Valero M, Herranz C, Mateo F, Mereu E, Beesley J, Chenevix-Trench G, Roux C, Mak T, Brunet J, Hakem R, Gorrini C, Antoniou AC, Lázaro C, Pujana MA. Immune Cell Associations with Cancer Risk. iScience 2020; 23:101296. [PMID: 32622267 PMCID: PMC7334419 DOI: 10.1016/j.isci.2020.101296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/23/2020] [Accepted: 06/15/2020] [Indexed: 01/21/2023] Open
Abstract
Proper immune system function hinders cancer development, but little is known about whether genetic variants linked to cancer risk alter immune cells. Here, we report 57 cancer risk loci associated with differences in immune and/or stromal cell contents in the corresponding tissue. Predicted target genes show expression and regulatory associations with immune features. Polygenic risk scores also reveal associations with immune and/or stromal cell contents, and breast cancer scores show consistent results in normal and tumor tissue. SH2B3 links peripheral alterations of several immune cell types to the risk of this malignancy. Pleiotropic SH2B3 variants are associated with breast cancer risk in BRCA1/2 mutation carriers. A retrospective case-cohort study indicates a positive association between blood counts of basophils, leukocytes, and monocytes and age at breast cancer diagnosis. These findings broaden our knowledge of the role of the immune system in cancer and highlight promising prevention strategies for individuals at high risk.
Collapse
Affiliation(s)
- Luis Palomero
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain
| | - Ivan Galván-Femenía
- GCAT-Genomes for Life, Germans Trias i Pujol Health Sciences Research Institute (IGTP), Program for Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, Catalonia 08916, Spain
| | - Rafael de Cid
- GCAT-Genomes for Life, Germans Trias i Pujol Health Sciences Research Institute (IGTP), Program for Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, Catalonia 08916, Spain
| | - Roderic Espín
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain
| | - Daniel R Barnes
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Cimba
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Eline Blommaert
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain
| | - Miguel Gil-Gil
- Department of Medical Oncology, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain
| | - Catalina Falo
- Department of Medical Oncology, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain
| | - Agostina Stradella
- Department of Medical Oncology, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain
| | - Dan Ouchi
- Jordi Gol University Institute for Research Primary Healthcare (IDIAP Jordi Gol), Barcelona, Catalonia 08007, Spain; Autonomous University of Barcelona, Bellaterra, Catalonia 08913, Spain
| | - Albert Roso-Llorach
- Jordi Gol University Institute for Research Primary Healthcare (IDIAP Jordi Gol), Barcelona, Catalonia 08007, Spain; Autonomous University of Barcelona, Bellaterra, Catalonia 08913, Spain
| | - Concepció Violan
- Jordi Gol University Institute for Research Primary Healthcare (IDIAP Jordi Gol), Barcelona, Catalonia 08007, Spain; Autonomous University of Barcelona, Bellaterra, Catalonia 08913, Spain
| | - María Peña-Chilet
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), Bioinformatics in Rare Diseases (BiER), CIBERER, INB-ELIXIR-es, Hospital Virgen del Rocío, Seville 41013, Spain
| | - Joaquín Dopazo
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), Bioinformatics in Rare Diseases (BiER), CIBERER, INB-ELIXIR-es, Hospital Virgen del Rocío, Seville 41013, Spain
| | - Ana Isabel Extremera
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain
| | - Mar García-Valero
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain
| | - Carmen Herranz
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain
| | - Francesca Mateo
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain
| | - Elisabetta Mereu
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Catalonia 08003, Spain
| | - Jonathan Beesley
- Cancer Division, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | | | - Cecilia Roux
- Princess Margaret Cancer Centre, The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Tak Mak
- Princess Margaret Cancer Centre, The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Joan Brunet
- Hereditary Cancer Program, Catalan Institute of Oncology, Biomedical Research Institute of Girona (IDIBGI), Girona, Catalonia 17190, Spain
| | - Razq Hakem
- Princess Margaret Cancer Centre, Department of Medical Biophysics, University Health Network and University of Toronto, Toronto, ON M5G 2C1, Canada
| | - Chiara Gorrini
- Princess Margaret Cancer Centre, The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.
| | - Conxi Lázaro
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, and Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Miquel Angel Pujana
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia 08908, Spain.
| |
Collapse
|
24
|
Dondi E, Sibarita JB, Varin-Blank N, Velazquez L. The adaptor protein APS modulates BCR signalling in mature B cells. Cell Signal 2020; 73:109673. [PMID: 32470518 DOI: 10.1016/j.cellsig.2020.109673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 11/19/2022]
Abstract
Activation process of mature B cell is predominantly driven by specific BCR-mediated pathways, switched on and off all through late B cell differentiation stages. Mice deficient for APS, a member of the Lnk/SH2B family of adaptor proteins, showed that this adaptor plays a BCR-mediated regulatory role in mature B cells. However, the intermediates involved in this adaptor modulating functions in B cells are still unknown. In the present study, we investigated the role of APS in regulating BCR signalling notably through cytoskeleton remodeling in mature B cells. Herein, we showed that APS function is stage specific, as it exclusively intervenes in mature B cells. Upon activation, APS colocalizes with the BCR and associates with important regulators of BCR signalling, such as Syk and Cbl kinase. Importantly, APS interferes, as a scaffold protein, with the stability of Syk kinase by recruiting Cbl. This function is mainly mediated by APS SH2 domain, which regulates BCR-evoked cell dynamics. Our findings thus reveal that APS plays a regulatory role in BCR-induced responses by specifically modulating its interacting partners, which positions APS as a relevant modulator of BCR signalling in mature B cells.
Collapse
Affiliation(s)
- Elisabetta Dondi
- INSERM, U978, UFR SMBH, Bobigny, France; Comue USPC, Labex Inflamex, Université Paris 13, UFR SMBH, Bobigny, France
| | - Jean-Baptiste Sibarita
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, Bordeaux, France; CNRS UMR, 5297 Bordeaux, France
| | - Nadine Varin-Blank
- INSERM, U978, UFR SMBH, Bobigny, France; Comue USPC, Labex Inflamex, Université Paris 13, UFR SMBH, Bobigny, France.
| | - Laura Velazquez
- INSERM, U978, UFR SMBH, Bobigny, France; Comue USPC, Labex Inflamex, Université Paris 13, UFR SMBH, Bobigny, France.
| |
Collapse
|
25
|
Bellanné-Chantelot C, Rabadan Moraes G, Schmaltz-Panneau B, Marty C, Vainchenker W, Plo I. Germline genetic factors in the pathogenesis of myeloproliferative neoplasms. Blood Rev 2020; 42:100710. [PMID: 32532454 DOI: 10.1016/j.blre.2020.100710] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 04/08/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Myeloproliferative neoplasms (MPN) are clonal hematological malignancies that lead to overproduction of mature myeloid cells. They are due to acquired mutations in genes encoding for AK2, MPL and CALR that result in the activation of the cytokine receptor/JAK2 signaling pathway. In addition, it exists germline variants that can favor the initiation of the disease or may affect its phenotype. First, they can be common risk alleles, which correspond to frequent single nucleotide variants present in control population and that contribute to the development of either sporadic or familial MPN. Second, some variants predispose to the onset of MPN with a higher penetrance and lead to familial clustering of MPN. Finally, some extremely rare genetic variants can induce MPN-like hereditary disease. We will review these different subtypes of germline genetic variants and discuss how they impact the initiation and/or development of the MPN disease.
Collapse
Affiliation(s)
- Christine Bellanné-Chantelot
- Department of Genetics, Assistance Publique-Hôpitaux de Paris (APHP), Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Sorbonne Université, Paris, France; INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France
| | - Graciela Rabadan Moraes
- INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France; Université Paris Diderot (Paris 7), UMR1287, Gustave Roussy, Villejuif, France; Gustave Roussy, Villejuif, France
| | - Barbara Schmaltz-Panneau
- INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France; Gustave Roussy, Villejuif, France; Université Paris XI, UMR1287, Gustave Roussy, Villejuif, France
| | - Caroline Marty
- INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France; Gustave Roussy, Villejuif, France; Université Paris XI, UMR1287, Gustave Roussy, Villejuif, France
| | - William Vainchenker
- INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France; Gustave Roussy, Villejuif, France; Université Paris XI, UMR1287, Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France; Gustave Roussy, Villejuif, France; Université Paris XI, UMR1287, Gustave Roussy, Villejuif, France.
| |
Collapse
|
26
|
LNK promotes the growth and metastasis of triple negative breast cancer via activating JAK/STAT3 and ERK1/2 pathway. Cancer Cell Int 2020; 20:124. [PMID: 32322171 PMCID: PMC7160949 DOI: 10.1186/s12935-020-01197-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 03/31/2020] [Indexed: 11/17/2022] Open
Abstract
Background LNK adaptor protein is a crucial regulator of normal hematopoiesis, which down-regulates activated tyrosine kinases at the cell surface resulting in an antitumor effect. To date, little studies have examined activities of LNK in solid tumors except ovarian cancer. Methods Clinical tissue chips were obtained from 16 clinical patients after surgery. Western blotting assay and quantitative real time PCR was performed to measure the expression of LNK. We investigate the in vivo and vitro effect of LNK in Triple Negative Breast Cancer by using cell proliferation、migration assays and an in vivo murine xenograft model. Western blotting assay was performed to investigate the mechanism of LNK in triple negative breast cancer. Results We found that the levels of LNK expression were elevated in high grade triple-negative breast cancer through Clinical tissue chips. Remarkably, overexpression of LNK can promote breast cancer cell proliferation and migration in vivo and vitro, while silencing of LNK show the opposite phenomenon. We also found that LNK can promote breast cancer cell to proliferate and migrate via activating JAK/STAT3 and ERK1/2 pathway. Conclusions Our results suggest that the adaptor protein LNK acts as a positive signal transduction modulator in TNBC.
Collapse
|
27
|
Marneth AE, Mullally A. The Molecular Genetics of Myeloproliferative Neoplasms. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a034876. [PMID: 31548225 DOI: 10.1101/cshperspect.a034876] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Activated JAK-STAT signaling is central to the pathogenesis of BCR-ABL-negative myeloproliferative neoplasms (MPNs) and occurs as a result of MPN phenotypic driver mutations in JAK2, CALR, or MPL The spectrum of concomitant somatic mutations in other genes has now largely been defined in MPNs. With the integration of targeted next-generation sequencing (NGS) panels into clinical practice, the clinical significance of concomitant mutations in MPNs has become clearer. In this review, we describe the consequences of concomitant mutations in the most frequently mutated classes of genes in MPNs: (1) DNA methylation pathways, (2) chromatin modification, (3) RNA splicing, (4) signaling pathways, (5) transcription factors, and (6) DNA damage response/stress signaling. The increased use of molecular genetics for early risk stratification of patients brings the possibility of earlier intervention to prevent disease progression in MPNs. However, additional studies are required to decipher underlying molecular mechanisms and effectively target them.
Collapse
Affiliation(s)
- Anna E Marneth
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.,Broad Institute, Cambridge, Massachusetts 02142, USA.,Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
28
|
Zhong ZM, Chen X, Qi X, Wang XM, Li CY, Qin RJ, Wang SQ, Liang J, Zeng MS, Sun CZ. Adaptor protein LNK promotes anaplastic thyroid carcinoma cell growth via 14-3-3 ε/γ binding. Cancer Cell Int 2020; 20:11. [PMID: 31938019 PMCID: PMC6953139 DOI: 10.1186/s12935-019-1090-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/28/2019] [Indexed: 01/08/2023] Open
Abstract
Background Rapid progression contributes to treatment failure in anaplastic thyroid carcinoma (ATC) patients. In a preliminary study, we demonstrated that some hematopoietic factors may be involved in the progression of ATC. The adaptor protein LNK, which is a negative regulator of hematopoietic cytokine signalling, has been studied extensively in malignant hematopoietic cells. However, there are few studies on LNK in solid tumours. Methods Real-time PCR, immunohistochemistry (IHC) and western blot analysis of LNK were performed on ATC cells, differentiated thyroid cancer (DTC) cells and normal thyroid cells. In vitro assays (including pull-down, liquid chromatography-mass spectrometry (LC–MS), co-IP, MTT and colony formation) were performed to validate the effect of LNK on ATC progression and elucidate the molecular mechanisms. Results Compared with DTC cells and normal thyroid cells, ATC cells exhibit overexpression of LNK. In addition, LNK overexpression results in increased proliferation of ATC cells. Conversely, LNK knockdown significantly suppresses ATC cell proliferation. LC–MS identified the 14-3-3 ε/γ protein as a LNK binding partner. Finally, the results indicate that LNK overexpression significantly enhances the anti-apoptotic ability of ATC cells via the Akt-NFκB-Bcl-2/Bcl-xL pathway and that the oncogenic effect of LNK largely depends on 14-3-3 ε/γ binding. Conclusions The present study elucidated the important role of LNK in the growth of ATC opposite to its behaviour in the hematopoietic system and indicates that LNK is a potential target for the treatment of ATC.
Collapse
Affiliation(s)
- Zhao-Ming Zhong
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital, 519 Kunzhou Road, Kunming, China.,2Department of Medical Oncology, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, China
| | - Xue Chen
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital, 519 Kunzhou Road, Kunming, China
| | - Xiao Qi
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital, 519 Kunzhou Road, Kunming, China
| | - Xue-Min Wang
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital, 519 Kunzhou Road, Kunming, China
| | - Chun-Yan Li
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital, 519 Kunzhou Road, Kunming, China
| | - Ru-Jia Qin
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital, 519 Kunzhou Road, Kunming, China
| | - Shi-Qi Wang
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital, 519 Kunzhou Road, Kunming, China
| | - Jin Liang
- 2Department of Medical Oncology, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, China
| | - Mu-Sheng Zeng
- 3State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, China
| | - Chuan-Zheng Sun
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital, 519 Kunzhou Road, Kunming, China
| |
Collapse
|
29
|
Cheng Y, Duan C, Zhang C. New perspective on SH2B1: An accelerator of cancer progression. Biomed Pharmacother 2019; 121:109651. [PMID: 31739166 DOI: 10.1016/j.biopha.2019.109651] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/22/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
SH2B1 is well-known as an adaptor protein, and deletion of SH2B1 results in severe obesity and both leptin and insulin resistance. Some studies have revealed that SH2B1 is involved in the progression of lung cancer, esophageal cancer, gastric cancer, oropharyngeal cancer, and so on. Biological function experiments have proven that SH2B1 can regulate cellular morphology, motility and adhesion by modifying the actin cytoskeletal reorganization, and it can promote cell mitogenesis, transformation, survival and differentiation via different signal pathways by enhancing the kinase activity of several receptor tyrosine kinases. In addition, SH2B1 is an obesity-related gene, and epidemiological surveys suggest a complex relationship between obesity and cancer. Therefore, what is the relationship between SH2B1 and cancer? Herein, we attempt to provide a mini overview of the roles of SH2B1 in cancer.
Collapse
Affiliation(s)
- Yuanda Cheng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Xiangya Road 87th, Changsha, 410008, Hunan, PR China
| | - Chaojun Duan
- Institute of Medical Sciences, Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Xiangya Road 87th, Changsha, 410008, Hunan, PR China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Xiangya Road 87th, Changsha, 410008, Hunan, PR China.
| |
Collapse
|
30
|
Cielo D, Galatola M, Fernandez-Jimenez N, De Leo L, Garcia-Etxebarria K, Loganes C, Tommasini A, Not T, Auricchio R, Greco L, Bilbao JR. Combined Analysis of Methylation and Gene Expression Profiles in Separate Compartments of Small Bowel Mucosa Identified Celiac Disease Patients' Signatures. Sci Rep 2019; 9:10020. [PMID: 31292504 PMCID: PMC6620355 DOI: 10.1038/s41598-019-46468-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 06/14/2019] [Indexed: 02/07/2023] Open
Abstract
By GWAS studies on celiac disease, gene expression was studied at the level of the whole intestinal mucosa, composed by two different compartments: epithelium and lamina propria. Our aim is to analyse the gene-expression and DNA methylation of candidate genes in each of these compartments. Epithelium was separated from lamina propria in biopsies of CeD patients and CTRs using magnetic beads. Gene-expression was analysed by RT-PC; methylation analysis required bisulfite conversion and NGS. Reverse modulation of gene-expression and methylation in the same cellular compartment was observed for the IL21 and SH2B3 genes in CeD patients relative to CTRs. Bioinformatics analysis highlighted the regulatory elements in the genomic region of SH2B3 that altered methylation levels. The cREL and TNFAIP3 genes showed methylation patterns that were significantly different between CeD patients and CTRs. In CeD, the genes linked to inflammatory processes are up-regulated, whereas the genes involved in the cell adhesion/integrity of the intestinal barrier are down-regulated. These findings suggest a correlation between gene-expression and methylation profile for the IL21 and SH2B3 genes. We identified a “gene-expression phenotype” of CeD and showed that the abnormal response to dietary antigens in CeD might be related not to abnormalities of gene structure but to the regulation of molecular pathways.
Collapse
Affiliation(s)
- D Cielo
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy.,European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples "Federico II", Naples, Italy
| | - M Galatola
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy. .,European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples "Federico II", Naples, Italy.
| | - N Fernandez-Jimenez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV-EHU), BioCruces Health Research Institute, Leioa, Spain
| | - L De Leo
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - K Garcia-Etxebarria
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV-EHU), BioCruces Health Research Institute, Leioa, Spain
| | - C Loganes
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - A Tommasini
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - T Not
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV-EHU), BioCruces Health Research Institute, Leioa, Spain.,Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - R Auricchio
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy.,European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples "Federico II", Naples, Italy
| | - L Greco
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy.,European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples "Federico II", Naples, Italy
| | - J R Bilbao
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV-EHU), BioCruces Health Research Institute, Leioa, Spain
| |
Collapse
|
31
|
Sinclair PB, Ryan S, Bashton M, Hollern S, Hanna R, Case M, Schwalbe EC, Schwab CJ, Cranston RE, Young BD, Irving JAE, Vora AJ, Moorman AV, Harrison CJ. SH2B3 inactivation through CN-LOH 12q is uniquely associated with B-cell precursor ALL with iAMP21 or other chromosome 21 gain. Leukemia 2019; 33:1881-1894. [PMID: 30816328 PMCID: PMC6756024 DOI: 10.1038/s41375-019-0412-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/24/2019] [Indexed: 12/17/2022]
Abstract
In more than 30% of B-cell precursor acute lymphoblastic leukaemia (B-ALL), chromosome 21 sequence is overrepresented through aneuploidy or structural rearrangements, exemplified by intrachromosomal amplification of chromosome 21 (iAMP21). Although frequent, the mechanisms by which these abnormalities promote B-ALL remain obscure. Intriguingly, we found copy number neutral loss of heterozygosity (CN-LOH) of 12q was recurrent in iAMP21-ALL, but never observed in B-ALL without some form of chromosome 21 gain. As a consequence of CN-LOH 12q, mutations or deletions of the adaptor protein, SH2B3, were converted to homozygosity. In patients without CN-LOH 12q, bi-allelic abnormalities of SH2B3 occurred, but only in iAMP21-ALL, giving an overall incidence of 18% in this sub-type. Review of published data confirmed a tight association between overrepresentation of chromosome 21 and both CN-LOH 12q and SH2B3 abnormalities in B-ALL. Despite relatively small patient numbers, preliminary analysis linked 12q abnormalities to poor outcome in iAMP21-ALL (p = 0.03). Homology modelling of a leukaemia-associated SH2 domain mutation and in vitro analysis of patient-derived xenograft cells implicated the JAK/STAT pathway as one likely target for SH2B3 tumour suppressor activity in iAMP21-ALL.
Collapse
Affiliation(s)
- Paul B Sinclair
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK.
| | - Sarra Ryan
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Matthew Bashton
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Shaun Hollern
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Rebecca Hanna
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Marian Case
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Edward C Schwalbe
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Claire J Schwab
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Ruth E Cranston
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Brian D Young
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Julie A E Irving
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Ajay J Vora
- Great Ormond Street Hospital for Children NHS trust, London, UK
| | - Anthony V Moorman
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Christine J Harrison
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK.
| |
Collapse
|
32
|
Balcerek J, Jiang J, Li Y, Jiang Q, Holdreith N, Singh B, Chandra V, Lv K, Ren JG, Rozenova K, Li W, Greenberg RA, Tong W. Lnk/Sh2b3 deficiency restores hematopoietic stem cell function and genome integrity in Fancd2 deficient Fanconi anemia. Nat Commun 2018; 9:3915. [PMID: 30254368 PMCID: PMC6156422 DOI: 10.1038/s41467-018-06380-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/03/2018] [Indexed: 12/20/2022] Open
Abstract
Fanconi anemia (FA) is a bone marrow failure (BMF) syndrome that arises from mutations in a network of FA genes essential for DNA interstrand crosslink (ICL) repair and replication stress tolerance. While allogeneic stem cell transplantation can replace defective HSCs, interventions to mitigate HSC defects in FA do not exist. Remarkably, we reveal here that Lnk (Sh2b3) deficiency restores HSC function in Fancd2−/− mice. Lnk deficiency does not impact ICL repair, but instead stabilizes stalled replication forks in a manner, in part, dependent upon alleviating blocks to cytokine−mediated JAK2 signaling. Lnk deficiency restores proliferation and survival of Fancd2−/− HSCs, while reducing replication stress and genomic instability. Furthermore, deletion of LNK in human FA-like HSCs promotes clonogenic growth. These findings highlight a new role for cytokine/JAK signaling in promoting replication fork stability, illuminate replication stress as a major underlying origin of BMF in FA, and have strong therapeutic implications. Loss of Fancd2 leads to replication stress intolerance and Fanconi Anemia, where haematopoietic stem cell (HSC) function is compromised. Here, the authors show that Lnk/Sh2b3 loss restores HSC proliferation and survival in Fancd2 knockout mice and ameliorates replication stress in a cytokine/JAK2 signaling dependent manner.
Collapse
Affiliation(s)
- Joanna Balcerek
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jing Jiang
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.,Institute of Translational Medicine, School of Medicine, Yangzhou University, 225001, Yangzhou, Jiangsu, China
| | - Yang Li
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pathology & Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qinqin Jiang
- Department of Cancer Biology, Abramson Cancer Research Institute and Basser Center for BRCA, and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nicholas Holdreith
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Brijendra Singh
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vemika Chandra
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kaosheng Lv
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jian-Gang Ren
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Krasimira Rozenova
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Weihua Li
- Department of Cancer Biology, Abramson Cancer Research Institute and Basser Center for BRCA, and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Roger A Greenberg
- Department of Cancer Biology, Abramson Cancer Research Institute and Basser Center for BRCA, and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wei Tong
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA. .,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
33
|
Baughn LB, Meredith MM, Oseth L, Smolarek TA, Hirsch B. SH2B3 aberrations enriched in iAMP21 B lymphoblastic leukemia. Cancer Genet 2018; 226-227:30-35. [PMID: 30005852 DOI: 10.1016/j.cancergen.2018.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/07/2018] [Accepted: 05/19/2018] [Indexed: 11/18/2022]
Abstract
Acute lymphoblastic leukemia (ALL) represents the most common childhood malignancy. Although survival for pediatric B-ALL has approached 90%, variability in outcome among and within cytogenetically defined subgroups persists. While G-banding and fluorescence in situ hybridization (FISH) have been used to characterize leukemic clones, there is added value of chromosomal microarray and next generation sequencing in screening genome-wide for copy number aberrations, copy neutral loss of heterozygosity and nucleotide variations. Evaluation of novel genetic aberrations can provide information about the biologic mechanisms of cytogenetically defined subgroups associated with poor prognosis, explain heterogeneity in patient outcome and identify novel targets for therapeutic intervention. The high risk B-ALL intrachromosomal amplification of chromosome 21, (iAMP21), subtype is characterized by amplification of a region of chromosome 21 that typically encompasses the RUNX1 gene and is associated with poor prognosis. Analysis of chromosomal microarray and FISH data revealed that deletions of SH2B3, encoding a negative regulator of multiple tyrosine kinase and cytokine signaling pathways, are enriched among leukemias harboring iAMP21. Enrichment of SH2B3 aberrations in the iAMP21 subtype may indicate that loss of SH2B3 contributes to disease progression and raises the possibility that these leukemias may be sensitive to tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- L B Baughn
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN, United States
| | - M M Meredith
- Department of Laboratory Medicine and Pathology and Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, United States
| | - L Oseth
- Department of Laboratory Medicine and Pathology and Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, United States
| | - T A Smolarek
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, TCHRF 1042, Cincinnati, OH, United States
| | - B Hirsch
- Department of Laboratory Medicine and Pathology and Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, United States.
| |
Collapse
|
34
|
Luque Paz D, Boyer F, Beucher A, Bouvier A, Jouanneau-Courville R, Guardiola P, Lambert D, Delneste Y, Hunault M, Blanchet O, Ugo V. Concomitant CALR and LNK mutations leading to essential thrombocythemia with erythrocytosis. Blood Cells Mol Dis 2018; 71:75-76. [PMID: 29703677 DOI: 10.1016/j.bcmd.2018.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 10/17/2022]
Affiliation(s)
- Damien Luque Paz
- CHU Angers, Laboratoire d'Hématologie, Institut de Biologie en Santé, Angers, France; Université d'Angers, UFR Santé, Angers, France; CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France; Fédération Hospitalo-Universitaire (FHU) GOAL, 'Grand Ouest Against Leukemia', France
| | | | - Annaëlle Beucher
- CHU Angers, Laboratoire d'Hématologie, Institut de Biologie en Santé, Angers, France
| | - Anne Bouvier
- CHU Angers, Laboratoire d'Hématologie, Institut de Biologie en Santé, Angers, France; CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France; Fédération Hospitalo-Universitaire (FHU) GOAL, 'Grand Ouest Against Leukemia', France
| | - Rebecca Jouanneau-Courville
- CHU Angers, Laboratoire d'Hématologie, Institut de Biologie en Santé, Angers, France; Fédération Hospitalo-Universitaire (FHU) GOAL, 'Grand Ouest Against Leukemia', France
| | - Philippe Guardiola
- Université d'Angers, UFR Santé, Angers, France; CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France; Fédération Hospitalo-Universitaire (FHU) GOAL, 'Grand Ouest Against Leukemia', France; CHU Angers, Service de Génomique Onco-Hématologique, Angers, France
| | - Diane Lambert
- CHU Angers, Service de Génomique Onco-Hématologique, Angers, France
| | - Yves Delneste
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Mathilde Hunault
- Université d'Angers, UFR Santé, Angers, France; CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France; Fédération Hospitalo-Universitaire (FHU) GOAL, 'Grand Ouest Against Leukemia', France; CHU Angers, Service des Maladies du Sang, Angers, France
| | - Odile Blanchet
- CHU Angers, Laboratoire d'Hématologie, Institut de Biologie en Santé, Angers, France; Université d'Angers, UFR Santé, Angers, France; CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France; Fédération Hospitalo-Universitaire (FHU) GOAL, 'Grand Ouest Against Leukemia', France; CHU Angers, Centre de Ressources Biologiques, BB-0033-00038, Angers, France
| | - Valérie Ugo
- CHU Angers, Laboratoire d'Hématologie, Institut de Biologie en Santé, Angers, France; Université d'Angers, UFR Santé, Angers, France; CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France; Fédération Hospitalo-Universitaire (FHU) GOAL, 'Grand Ouest Against Leukemia', France.
| |
Collapse
|
35
|
Abstract
Stem cells self-renew and differentiate to generate all tissues and cells in the body. Stem cell health promotes adaptive responses to tissue damage or loss and is essential for tissue regeneration with age. In the past decade, the evolutionarily conserved transcription factors FOXO with known functions in promoting healthy aging have emerged as key regulators of stem cell homeostasis in various tissues, including blood, neural, and muscle stem cells. Aberrant FOXO functions have been implicated in a variety of disorders including neurodegenerative, blood, cancer, and diabetes some of which are fostered by abnormal stem cell function. As discussed in this chapter, at least in some stem cells FOXO regulatory mechanisms and applied functions follow a complex set of rules distinct from that operating in progenitor cell populations and in cultured cell lines. Elucidating the exact nature of FOXO properties in stem cells will be critical for identifying and targeting aberrant FOXO-mediated mechanisms that promote stem cell-derived disease specifically with age.
Collapse
Affiliation(s)
- Raymond Liang
- Icahn School of Medicine at Mount Sinai, New York, NY, United States; Developmental and Stem Cell Biology, Multidisciplinary Training Area, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Saghi Ghaffari
- Icahn School of Medicine at Mount Sinai, New York, NY, United States; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
36
|
Naudin C, Chevalier C, Roche S. The role of small adaptor proteins in the control of oncogenic signalingr driven by tyrosine kinases in human cancer. Oncotarget 2017; 7:11033-55. [PMID: 26788993 PMCID: PMC4905456 DOI: 10.18632/oncotarget.6929] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/01/2016] [Indexed: 12/15/2022] Open
Abstract
Protein phosphorylation on tyrosine (Tyr) residues has evolved as an important mechanism to coordinate cell communication in multicellular organisms. The importance of this process has been revealed by the discovery of the prominent oncogenic properties of tyrosine kinases (TK) upon deregulation of their physiological activities, often due to protein overexpression and/or somatic mutation. Recent reports suggest that TK oncogenic signaling is also under the control of small adaptor proteins. These cytosolic proteins lack intrinsic catalytic activity and signal by linking two functional members of a catalytic pathway. While most adaptors display positive regulatory functions, a small group of this family exerts negative regulatory functions by targeting several components of the TK signaling cascade. Here, we review how these less studied adaptor proteins negatively control TK activities and how their loss of function induces abnormal TK signaling, promoting tumor formation. We also discuss the therapeutic consequences of this novel regulatory mechanism in human oncology.
Collapse
Affiliation(s)
- Cécile Naudin
- CNRS UMR5237, University Montpellier, CRBM, Montpellier, France.,Present address: INSERM U1016, CNRS UMR8104, Institut Cochin, Paris, France
| | - Clément Chevalier
- CNRS UMR5237, University Montpellier, CRBM, Montpellier, France.,Present address: SFR Biosit (UMS CNRS 3480/US INSERM 018), MRic Photonics Platform, University Rennes, Rennes, France
| | - Serge Roche
- CNRS UMR5237, University Montpellier, CRBM, Montpellier, France.,Equipe Labellisée LIGUE 2014, Ligue Contre le Cancer, Paris, France
| |
Collapse
|
37
|
Wang W, Hang C, Zhang Y, Chen M, Meng X, Cao Q, Song N, Itkow J, Shen F, Yu D. Dietary miR-451 protects erythroid cells from oxidative stress via increasing the activity of Foxo3 pathway. Oncotarget 2017; 8:107109-107124. [PMID: 29291015 PMCID: PMC5739800 DOI: 10.18632/oncotarget.22346] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 08/26/2017] [Indexed: 11/25/2022] Open
Abstract
One fundamental issue in public health is the safety of food products derived from plants and animals. A recent study raised a concern that microRNAs, which widely exist in everyday foods, may alter consumers' functions. However, some studies have strongly questioned the likelihood of dietary uptake of functional microRNAs in mammals. Here we use a microRNA gene knockout animal model to show that miR-144/451 null mice can orally uptake miR-451 from a daily chow diet, and ingestion of wild type blood, that contains abundant miR-451, also enhances the level of miR-451 in the circulating blood of knockout mice. Moreover, reducing miR-451 level in miR-144/451 knockout blood by consuming food lacking miR-451 reduces the anti-oxidant capacity of miR-144/451 null red blood cells by targeting the 14-3-3ζ/Foxo3 pathway, while increasing miR-451 level via gavage-feeding of wild type blood increases the anti-oxidant capacity of miR-144/451 null red blood cells. We conclude that 1) some miRNAs in food can pass through the gastrointestinal tract into the blood to affect consumers' function and 2) microRNA knockout animals such as miR-144/451 null mice can acquire the deleted genetic information from daily foods, which might alter the results and conclusions from the studies using such animals.
Collapse
Affiliation(s)
- Wanchen Wang
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou 225001, China
| | - Chengwen Hang
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou 225001, China
| | - Yanqing Zhang
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou 225001, China
| | - Mingshi Chen
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou 225001, China
| | - Xinyu Meng
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou 225001, China
| | - Qing Cao
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou 225001, China
| | - Nana Song
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou 225001, China
| | - Jacobi Itkow
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou 225001, China
| | - Feiyang Shen
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou 225001, China
| | - Duonan Yu
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University School of Medicine, Yangzhou 225001, China.,Institute of Comparative Medicine, Yangzhou University, Yangzhou 225001, China.,Institute of Translational Medicine, Yangzhou University School of Medicine, Yangzhou 225001, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou 225001, China
| |
Collapse
|
38
|
Schischlik F, Kralovics R. Mutations in myeloproliferative neoplasms - their significance and clinical use. Expert Rev Hematol 2017; 10:961-973. [PMID: 28914569 DOI: 10.1080/17474086.2017.1380515] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Clonal hematologic diseases of the blood such as polycythemia vera, essential thrombocythemia and primary myelofibrosis belong to the BCR-ABL negative Myeloproliferative Neoplasms (MPN). These diseases are characterized by clonal expansion of hematopoietic precursor cells followed by increased production of differentiated cells of the myeloid lineage. Initiation of clonal hematopoiesis, formation of a clinical phenotype as well as disease progression form part of MPN disease evolution. The disease is driven by acquired somatic mutations in critical pathways such as cytokine signaling, epigenetic regulation, RNA splicing, and transcription factor signaling. Areas covered: The following review aims to provide an overview of the mutational landscape of MPN, the impact of these mutations in MPN pathogenesis as well as their prognostic value. Finally, a summary of how these mutations are being used or could potentially be used for the treatment of MPN patients is presented. Expert commentary: The genetic landscape of MPN patients has been successfully dissected within the past years with the advent of new sequencing technologies. Integrating the genetic information within a clinical setting is already benefitting patients in terms of disease monitoring and prognostic information of disease progression but will be further intensified within the next years.
Collapse
Affiliation(s)
- Fiorella Schischlik
- a CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences , Vienna , Austria
| | - Robert Kralovics
- a CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences , Vienna , Austria
| |
Collapse
|
39
|
Christiansen MK, Larsen SB, Nyegaard M, Neergaard-Petersen S, Würtz M, Grove EL, Hvas AM, Jensen HK, Kristensen SD. The SH2B3 and KCNK5 loci may be implicated in regulation of platelet count, volume, and maturity. Thromb Res 2017; 158:86-92. [PMID: 28865245 DOI: 10.1016/j.thromres.2017.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/21/2017] [Accepted: 08/17/2017] [Indexed: 01/11/2023]
Abstract
INTRODUCTION In recent genome-wide association studies, coronary artery disease (CAD) and myocardial infarction (MI) have been linked to a number of genetic variants, but their role in thrombopoiesis is largely unknown. AIM We investigated the association between CAD and MI-associated genetic variants and five thrombopoiesis-related indices: platelet count (PC), mean platelet volume (MPV), immature platelet count (IPC), immature platelet fraction (IPF), and serum thrombopoietin (TPO). METHODS We genotyped 45 genome-wide significant CAD/MI-markers in 879 stable CAD patients. A genetic risk score was calculated to assess the combined risk associated with all the genetic variants. Platelet indices were analysed using the Sysmex XE-2100 haematology analyser. TPO was measured by ELISA. RESULTS Two variants were nominally associated with several indices; for rs10947789 (KCNK5), the adjusted geometric mean was 2% higher for MPV (95% confidence interval: 1-2%, p=0.002), 6% for IPC (0-12%, p=0.033), and 9% for IPF (3-16%, p=0.004) per CAD risk allele. Moreover, an 11% lower TPO (3-19%, p=0.010) was observed. Rs3184504 (SH2B3) was associated with a higher adjusted geometric mean of 3% (1-6%, p=0.003) per CAD risk allele for PC, and an 11% (5-17%, p<0.001) lower TPO. Furthermore, the adjusted IPC was 5% (0-9%, p=0.037) lower per CAD risk allele for PC, whereas IPF levels did not vary across genotypes. CONCLUSION As a novel finding, our study suggests a role for KCNK5 in the regulation of platelet size and maturity. Furthermore, our findings confirm an association between the SH2B3-locus and platelet count.
Collapse
Affiliation(s)
- Morten K Christiansen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark; Faculty of Health, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sanne B Larsen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark.
| | - Mette Nyegaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Morten Würtz
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Erik L Grove
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark; Faculty of Health, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anne-Mette Hvas
- Faculty of Health, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Henrik K Jensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark; Faculty of Health, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Steen D Kristensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark; Faculty of Health, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
40
|
Umemura T, Joshita S, Hamano H, Yoshizawa K, Kawa S, Tanaka E, Ota M. Association of autoimmune hepatitis with Src homology 2 adaptor protein 3 gene polymorphisms in Japanese patients. J Hum Genet 2017; 62:963-967. [DOI: 10.1038/jhg.2017.74] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/14/2017] [Accepted: 06/14/2017] [Indexed: 12/24/2022]
|
41
|
Lv K, Jiang J, Donaghy R, Riling CR, Cheng Y, Chandra V, Rozenova K, An W, Mohapatra BC, Goetz BT, Pillai V, Han X, Todd EA, Jeschke GR, Langdon WY, Kumar S, Hexner EO, Band H, Tong W. CBL family E3 ubiquitin ligases control JAK2 ubiquitination and stability in hematopoietic stem cells and myeloid malignancies. Genes Dev 2017; 31:1007-1023. [PMID: 28611190 PMCID: PMC5495118 DOI: 10.1101/gad.297135.117] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 05/23/2017] [Indexed: 01/08/2023]
Abstract
Here, Lv et al. report that the CBL family E3 ubiquitin ligases down-regulate JAK2 stability and signaling via the adaptor protein LNK/SH2B3. Their results reveal a novel signaling axis that regulates JAK2 in normal and malignant HSPCs and suggest new therapeutic strategies for treating CBLmut myeloid malignancies. Janus kinase 2 (JAK2) is a central kinase in hematopoietic stem/progenitor cells (HSPCs), and its uncontrolled activation is a prominent oncogenic driver of hematopoietic neoplasms. However, molecular mechanisms underlying the regulation of JAK2 have remained elusive. Here we report that the Casitas B-cell lymphoma (CBL) family E3 ubiquitin ligases down-regulate JAK2 stability and signaling via the adaptor protein LNK/SH2B3. We demonstrated that depletion of CBL/CBL-B or LNK abrogated JAK2 ubiquitination, extended JAK2 half-life, and enhanced JAK2 signaling and cell growth in human cell lines as well as primary murine HSPCs. Built on these findings, we showed that JAK inhibitor (JAKi) significantly reduced aberrant HSPCs and mitigated leukemia development in a mouse model of aggressive myeloid leukemia driven by loss of Cbl and Cbl-b. Importantly, primary human CBL mutated (CBLmut) leukemias exhibited increased JAK2 protein levels and signaling and were hypersensitive to JAKi. Loss-of-function mutations in CBL E3 ubiquitin ligases are found in a wide range of myeloid malignancies, which are diseases without effective treatment options. Hence, our studies reveal a novel signaling axis that regulates JAK2 in normal and malignant HSPCs and suggest new therapeutic strategies for treating CBLmut myeloid malignancies.
Collapse
Affiliation(s)
- Kaosheng Lv
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Jing Jiang
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Ryan Donaghy
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | - Ying Cheng
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Vemika Chandra
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Krasimira Rozenova
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Wei An
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 6819, USA.,Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 6819, USA
| | - Bhopal C Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 6819, USA.,Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 6819, USA
| | - Benjamin T Goetz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 6819, USA.,Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 6819, USA
| | - Vinodh Pillai
- Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Xu Han
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Emily A Todd
- Progenra, Inc., Malvern, Pennsylvania 19355, USA
| | - Grace R Jeschke
- Division of Hematology and Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Wallace Y Langdon
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Suresh Kumar
- Progenra, Inc., Malvern, Pennsylvania 19355, USA
| | - Elizabeth O Hexner
- Division of Hematology and Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 6819, USA.,Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska 6819, USA
| | - Wei Tong
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
42
|
Maslah N, Cassinat B, Verger E, Kiladjian JJ, Velazquez L. The role of LNK/SH2B3 genetic alterations in myeloproliferative neoplasms and other hematological disorders. Leukemia 2017; 31:1661-1670. [PMID: 28484264 DOI: 10.1038/leu.2017.139] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/10/2017] [Accepted: 04/24/2017] [Indexed: 12/11/2022]
Abstract
Malignant hematological diseases are mainly because of the occurrence of molecular abnormalities leading to the deregulation of signaling pathways essential for precise cell behavior. High-resolution genome analysis using microarray and large-scale sequencing have helped identify several important acquired gene mutations that are responsible for such signaling deregulations across different hematological malignancies. In particular, the genetic landscape of classical myeloproliferative neoplasms (MPNs) has been in large part completed with the identification of driver mutations (targeting the cytokine receptor/Janus-activated kinase 2 (JAK2) pathway) that determine MPN phenotype, as well as additional mutations mainly affecting the regulation of gene expression (epigenetics or splicing regulators) and signaling. At present, most efforts concentrate in understanding how all these genetic alterations intertwine together to influence disease evolution and/or dictate clinical phenotype in order to use them to personalize diagnostic and clinical care. However, it is now evident that factors other than somatic mutations also play an important role in MPN disease initiation and progression, among which germline predisposition (single-nucleotide polymorphisms and haplotypes) may strongly influence the occurrence of MPNs. In this context, the LNK inhibitory adaptor protein encoded by the LNK/SH2B adaptor protein 3 (SH2B3) gene is the target of several genetic variations, acquired or inherited in MPNs, lymphoid leukemia and nonmalignant hematological diseases, underlying its importance in these pathological processes. As LNK adaptor is a key regulator of normal hematopoiesis, understanding the consequences of LNK variants on its protein functions and on driver or other mutations could be helpful to correlate genotype and phenotype of patients and to develop therapeutic strategies to target this molecule. In this review we summarize the current knowledge of LNK function in normal hematopoiesis, the different SH2B3 mutations reported to date and discuss how these genetic variations may influence the development of hematological malignancies.
Collapse
Affiliation(s)
- N Maslah
- APHP, Laboratoire de Biologie Cellulaire, Hôpital Saint-Louis, Paris, France.,Inserm UMRS 1131, IUH, Université Paris-Diderot, Paris, France
| | - B Cassinat
- APHP, Laboratoire de Biologie Cellulaire, Hôpital Saint-Louis, Paris, France.,Inserm UMRS 1131, IUH, Université Paris-Diderot, Paris, France
| | - E Verger
- APHP, Laboratoire de Biologie Cellulaire, Hôpital Saint-Louis, Paris, France.,Inserm UMRS 1131, IUH, Université Paris-Diderot, Paris, France
| | - J-J Kiladjian
- Inserm UMRS 1131, IUH, Université Paris-Diderot, Paris, France.,APHP, Centre d'investigations Cliniques, Hôpital Saint-Louis, Paris, France
| | - L Velazquez
- INSERM UMRS-MD1197, Institut André Lwoff/Université Paris XI, Hôpital Paul Brousse, Villejuif, France
| |
Collapse
|
43
|
Olivos DJ, Alvarez M, Cheng YH, Hooker RA, Ciovacco WA, Bethel M, McGough H, Yim C, Chitteti BR, Eleniste PP, Horowitz MC, Srour EF, Bruzzaniti A, Fuchs RK, Kacena MA. Lnk Deficiency Leads to TPO-Mediated Osteoclastogenesis and Increased Bone Mass Phenotype. J Cell Biochem 2017; 118:2231-2240. [PMID: 28067429 DOI: 10.1002/jcb.25874] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/06/2017] [Indexed: 11/11/2022]
Abstract
The Lnk adapter protein negatively regulates the signaling of thrombopoietin (TPO), the main megakaryocyte (MK) growth factor. Lnk-deficient (-/-) mice have increased TPO signaling and increased MK number. Interestingly, several mouse models exist in which increased MK number leads to a high bone mass phenotype. Here we report the bone phenotype of these mice. MicroCT and static histomorphometric analyses at 20 weeks showed the distal femur of Lnk-/- mice to have significantly higher bone volume fraction and trabecular number compared to wild-type (WT) mice. Notably, despite a significant increase in the number of osteoclasts (OC), and decreased bone formation rate in Lnk-/- mice compared to WT mice, Lnk-/- mice demonstrated a 2.5-fold greater BV/TV suggesting impaired OC function in vivo. Additionally, Lnk-/- mouse femurs exhibited non-significant increases in mid-shaft cross-sectional area, yet increased periosteal BFR compared to WT femurs was observed. Lnk-/- femurs also had non-significant increases in polar moment of inertia and decreased cortical bone area and thickness, resulting in reduced bone stiffness, modulus, and strength compared to WT femurs. Of note, Lnk is expressed by OC lineage cells and when Lnk-/- OC progenitors are cultured in the presence of TPO, significantly more OC are observed than in WT cultures. Lnk is also expressed in osteoblast (OB) cells and in vitro reduced alkaline phosphatase activity was observed in Lnk-/- cultures. These data suggest that both direct effects on OB and OC as well as indirect effects of MK in regulating OB contributes to the observed high bone mass. J. Cell. Biochem. 118: 2231-2240, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David J Olivos
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Marta Alvarez
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ying-Hua Cheng
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Richard Adam Hooker
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Wendy A Ciovacco
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, Connecticut
| | - Monique Bethel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Haley McGough
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Christopher Yim
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Pierre P Eleniste
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, Connecticut
| | - Edward F Srour
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Angela Bruzzaniti
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, Indiana
| | - Robyn K Fuchs
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, Indiana
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
44
|
Plo I, Bellanné-Chantelot C, Mosca M, Mazzi S, Marty C, Vainchenker W. Genetic Alterations of the Thrombopoietin/MPL/JAK2 Axis Impacting Megakaryopoiesis. Front Endocrinol (Lausanne) 2017; 8:234. [PMID: 28955303 PMCID: PMC5600916 DOI: 10.3389/fendo.2017.00234] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/28/2017] [Indexed: 12/31/2022] Open
Abstract
Megakaryopoiesis is an original and complex cell process which leads to the formation of platelets. The homeostatic production of platelets is mainly regulated and controlled by thrombopoietin (TPO) and the TPO receptor (MPL)/JAK2 axis. Therefore, any hereditary or acquired abnormality affecting this signaling axis can result in thrombocytosis or thrombocytopenia. Thrombocytosis can be due to genetic alterations that affect either the intrinsic MPL signaling through gain-of-function (GOF) activity (MPL, JAK2, CALR) and loss-of-function (LOF) activity of negative regulators (CBL, LNK) or the extrinsic MPL signaling by THPO GOF mutations leading to increased TPO synthesis. Alternatively, thrombocytosis may paradoxically result from mutations of MPL leading to an abnormal MPL trafficking, inducing increased TPO levels by alteration of its clearance. In contrast, thrombocytopenia can also result from LOF THPO or MPL mutations, which cause a complete defect in MPL trafficking to the cell membrane, impaired MPL signaling or stability, defects in the TPO/MPL interaction, or an absence of TPO production.
Collapse
Affiliation(s)
- Isabelle Plo
- INSERM UMR 1170, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1170, Villejuif, France
| | - Christine Bellanné-Chantelot
- INSERM UMR 1170, Gustave Roussy, Villejuif, France
- Department of Genetics, AP-HP Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, UPMC Univ Paris 06, Paris, France
| | - Matthieu Mosca
- INSERM UMR 1170, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1170, Villejuif, France
| | - Stefania Mazzi
- INSERM UMR 1170, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1170, Gustave Roussy, Villejuif, France
- Université Paris-Diderot, Paris, France
| | - Caroline Marty
- INSERM UMR 1170, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1170, Villejuif, France
| | - William Vainchenker
- INSERM UMR 1170, Gustave Roussy, Villejuif, France
- Université Paris-Saclay, UMR1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, UMR1170, Villejuif, France
- *Correspondence: William Vainchenker,
| |
Collapse
|
45
|
Grinfeld J, Nangalia J, Green AR. Molecular determinants of pathogenesis and clinical phenotype in myeloproliferative neoplasms. Haematologica 2017; 102:7-17. [PMID: 27909216 PMCID: PMC5210228 DOI: 10.3324/haematol.2014.113845] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/27/2016] [Indexed: 12/22/2022] Open
Abstract
The myeloproliferative neoplasms are a heterogeneous group of clonal disorders characterized by the overproduction of mature cells in the peripheral blood, together with an increased risk of thrombosis and progression to acute myeloid leukemia. The majority of patients with Philadelphia-chromosome negative myeloproliferative neoplasms harbor somatic mutations in Janus kinase 2, leading to constitutive activation. Acquired mutations in calreticulin or myeloproliferative leukemia virus oncogene are found in a significant number of patients with essential thrombocythemia or myelofibrosis, and mutations in numerous epigenetic regulators and spliceosome components are also seen. Although the cellular and molecular consequences of many of these mutations remain unclear, it seems likely that they interact with germline and microenvironmental factors to influence disease pathogenesis. This review will focus on the determinants of specific myeloproliferative neoplasm phenotypes as well as on how an improved understanding of molecular mechanisms can inform our understanding of the disease entities themselves.
Collapse
Affiliation(s)
- Jacob Grinfeld
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, Addenbrooke's Hospital, Cambridge, UK
| | - Jyoti Nangalia
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, Addenbrooke's Hospital, Cambridge, UK
| | - Anthony R Green
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
46
|
Hu X. CRISPR/Cas9 system and its applications in human hematopoietic cells. Blood Cells Mol Dis 2016; 62:6-12. [PMID: 27736664 DOI: 10.1016/j.bcmd.2016.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 09/19/2016] [Accepted: 09/23/2016] [Indexed: 12/26/2022]
Abstract
Since 2012, the CRISPR-Cas9 system has been quickly and successfully tested in a broad range of organisms and cells including hematopoietic cells. The application of CRISPR-Cas9 in human hematopoietic cells mainly involves the genes responsible for HIV infection, β-thalassemia and sickle cell disease (SCD). The successful disruption of CCR5 and CXCR4 genes in T cells by CRISPR-Cas9 promotes the prospect of the technology in the functional cure of HIV. More recently, eliminating CCR5 and CXCR4 in induced pluripotent stem cells (iPSCs) derived from patients and targeting the HIV genome have been successfully carried out in several laboratories. The outcome from these approaches bring us closer to the goal of eradicating HIV infection. For hemoglobinopathies the ability to produce iPSC-derived from patients with the correction of hemoglobin (HBB) mutations by CRISPR-Cas9 has been tested in a number of laboratories. These corrected iPSCs also show the potential to differentiate into mature erythrocytes expressing high-level and normal HBB. In light of the initial success of CRESPR-Cas9 in target mutated gene(s) in the iPSCs, a combination of genomic editing and autogenetic stem cell transplantation would be the best strategy for root treatment of the diseases, which could replace traditional allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Xiaotang Hu
- Department of Biology, College of Arts & Sciences, Barry University, 11300 Northeast Second Avenue, Miami Shores, FL 33161, United States.
| |
Collapse
|
47
|
Mosca M, Vertenoeil G, Toppaldoddi KR, Plo I, Vainchenker W. [Not Available]. Bull Cancer 2016; 103:S16-28. [PMID: 27494969 DOI: 10.1016/s0007-4551(16)30142-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BIOLOGICAL ASPECTS OF JAK/STAT SIGNALING IN BCR-ABL-NEGATIVE MYELOPROLIFERATIVE NEOPLASMS: Myeloproliferative disorders more recently named Myeloproliferative neoplasms (MPN) display several clinical entities: chronic myeloid leukemia (CML), the classical MPN including polycythemia vera (PV), essential thrombocythemia (ET), primary myelofibrosis (PMF) and atypical and unclassifiable NMP. The term MPN is mostly used for classical BCR-ABL-negative (myeloproliferative disorder) (ET, PV, PMF). These are clonal diseases resulting from the transformation of an hematopoietic stem cell and leading to an abnormal production of myeloid cells. The genetic defects responsible for the myeloproliferative abnormalities are called « driver » mutations and all result in deregulation of the cytokine receptor / JAK2 / STAT axis. Among them, JAK2, the thrombopoietin receptor (MPL) and calreticulin (CALR) mutations are found in around 90% of the cases. These driver MPN mutations can be associated with other driver mutations also found in other hematological malignancies, especially in PMFs. These are chronic diseases with major risks being thrombosis, hemorrhage and cytopenias for PMF and the long-term progression to myelofibrosis and the transformation to leukemia. Most recent therapeutic have focused on targeting the JAK2 signaling pathway directly by inhibitors of JAK2 or indirectly. Interferon a allows in some cases hematologic and molecular remission patients.
Collapse
Affiliation(s)
- Matthieu Mosca
- Inserm UMR 1170, Gustave Roussy, Villejuif, 94805, France; Université Paris-Saclay, UMR 1170, Gustave Roussy, Villejuif, 94805, France; Gustave Roussy, UMR 1170, Villejuif, 94805, France
| | - Gaëlle Vertenoeil
- Signal Transduction & Molecular Hematology Unit, Ludwig Institute for Cancer Research, Brussels B-1200, Belgium; De Duve Institute, Université catholique de Louvain, Brussels B-1200, Belgium
| | - Katte Rao Toppaldoddi
- Inserm UMR 1170, Gustave Roussy, Villejuif, 94805, France; Université Paris-Saclay, UMR 1170, Gustave Roussy, Villejuif, 94805, France; Gustave Roussy, UMR 1170, Villejuif, 94805, France
| | - Isabelle Plo
- Inserm UMR 1170, Gustave Roussy, Villejuif, 94805, France; Université Paris-Saclay, UMR 1170, Gustave Roussy, Villejuif, 94805, France; Gustave Roussy, UMR 1170, Villejuif, 94805, France
| | - William Vainchenker
- Inserm UMR 1170, Gustave Roussy, Villejuif, 94805, France; Université Paris-Saclay, UMR 1170, Gustave Roussy, Villejuif, 94805, France; Gustave Roussy, UMR 1170, Villejuif, 94805, France.
| |
Collapse
|
48
|
Wang W, Tang Y, Wang Y, Tascau L, Balcerek J, Tong W, Levine RL, Welch C, Tall AR, Wang N. LNK/SH2B3 Loss of Function Promotes Atherosclerosis and Thrombosis. Circ Res 2016; 119:e91-e103. [PMID: 27430239 DOI: 10.1161/circresaha.116.308955] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/15/2016] [Indexed: 01/01/2023]
Abstract
RATIONALE Human genome-wide association studies have revealed novel genetic loci that are associated with coronary heart disease. One such locus resides in LNK/SH2B3, which in mice is expressed in hematopoietic cells and suppresses thrombopoietin signaling via its receptor myeloproliferative leukemia virus oncogene. However, the mechanisms underlying the association of LNK single-nucleotide polymorphisms with coronary heart disease are poorly understood. OBJECTIVE To understand the functional effects of LNK single-nucleotide polymorphisms and explore the mechanisms whereby LNK loss of function impacts atherosclerosis and thrombosis. METHODS AND RESULTS Using human cord blood, we show that the common TT risk genotype (R262W) of LNK is associated with expansion of hematopoietic stem cells and enhanced megakaryopoiesis, demonstrating reduced LNK function and increased myeloproliferative leukemia virus oncogene signaling. In mice, hematopoietic Lnk deficiency leads to accelerated arterial thrombosis and atherosclerosis, but only in the setting of hypercholesterolemia. Hypercholesterolemia acts synergistically with LNK deficiency to increase interleukin 3/granulocyte-macrophage colony-stimulating factor receptor signaling in bone marrow myeloid progenitors, whereas in platelets cholesterol loading combines with Lnk deficiency to increase activation. Platelet LNK deficiency increases myeloproliferative leukemia virus oncogene signaling and AKT activation, whereas cholesterol loading decreases SHIP-1 phosphorylation, acting convergently to increase AKT and platelet activation. Together with increased myelopoiesis, platelet activation promotes prothrombotic and proatherogenic platelet/leukocyte aggregate formation. CONCLUSIONS LNK (R262W) is a loss-of-function variant that promotes thrombopoietin/myeloproliferative leukemia virus oncogene signaling and platelet and leukocyte production. In mice, LNK deficiency is associated with both increased platelet production and activation. Hypercholesterolemia acts in platelets and hematopoietic progenitors to exacerbate thrombosis and atherosclerosis associated with LNK deficiency.
Collapse
Affiliation(s)
- Wei Wang
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (W.W., Y.T., Y.W., L.T., C.W., A.R.T., N.W.); Division of Hematology, Children's Hospital of Philadelphia, PA (W.T.); Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia (J.B., W.T.); and Human Oncology and Pathogenesis Program (R.L.L.) and Leukemia Service, Department of Medicine (R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yang Tang
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (W.W., Y.T., Y.W., L.T., C.W., A.R.T., N.W.); Division of Hematology, Children's Hospital of Philadelphia, PA (W.T.); Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia (J.B., W.T.); and Human Oncology and Pathogenesis Program (R.L.L.) and Leukemia Service, Department of Medicine (R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ying Wang
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (W.W., Y.T., Y.W., L.T., C.W., A.R.T., N.W.); Division of Hematology, Children's Hospital of Philadelphia, PA (W.T.); Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia (J.B., W.T.); and Human Oncology and Pathogenesis Program (R.L.L.) and Leukemia Service, Department of Medicine (R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Liana Tascau
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (W.W., Y.T., Y.W., L.T., C.W., A.R.T., N.W.); Division of Hematology, Children's Hospital of Philadelphia, PA (W.T.); Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia (J.B., W.T.); and Human Oncology and Pathogenesis Program (R.L.L.) and Leukemia Service, Department of Medicine (R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Joanna Balcerek
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (W.W., Y.T., Y.W., L.T., C.W., A.R.T., N.W.); Division of Hematology, Children's Hospital of Philadelphia, PA (W.T.); Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia (J.B., W.T.); and Human Oncology and Pathogenesis Program (R.L.L.) and Leukemia Service, Department of Medicine (R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wei Tong
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (W.W., Y.T., Y.W., L.T., C.W., A.R.T., N.W.); Division of Hematology, Children's Hospital of Philadelphia, PA (W.T.); Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia (J.B., W.T.); and Human Oncology and Pathogenesis Program (R.L.L.) and Leukemia Service, Department of Medicine (R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ross L Levine
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (W.W., Y.T., Y.W., L.T., C.W., A.R.T., N.W.); Division of Hematology, Children's Hospital of Philadelphia, PA (W.T.); Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia (J.B., W.T.); and Human Oncology and Pathogenesis Program (R.L.L.) and Leukemia Service, Department of Medicine (R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carrie Welch
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (W.W., Y.T., Y.W., L.T., C.W., A.R.T., N.W.); Division of Hematology, Children's Hospital of Philadelphia, PA (W.T.); Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia (J.B., W.T.); and Human Oncology and Pathogenesis Program (R.L.L.) and Leukemia Service, Department of Medicine (R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alan R Tall
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (W.W., Y.T., Y.W., L.T., C.W., A.R.T., N.W.); Division of Hematology, Children's Hospital of Philadelphia, PA (W.T.); Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia (J.B., W.T.); and Human Oncology and Pathogenesis Program (R.L.L.) and Leukemia Service, Department of Medicine (R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nan Wang
- From the Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (W.W., Y.T., Y.W., L.T., C.W., A.R.T., N.W.); Division of Hematology, Children's Hospital of Philadelphia, PA (W.T.); Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia (J.B., W.T.); and Human Oncology and Pathogenesis Program (R.L.L.) and Leukemia Service, Department of Medicine (R.L.L.), Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
49
|
Germ line variants predispose to both JAK2 V617F clonal hematopoiesis and myeloproliferative neoplasms. Blood 2016; 128:1121-8. [PMID: 27365426 DOI: 10.1182/blood-2015-06-652941] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 06/05/2016] [Indexed: 12/11/2022] Open
Abstract
We conducted a genome-wide association study (GWAS) to identify novel predisposition alleles associated with Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs) and JAK2 V617F clonal hematopoiesis in the general population. We recruited a web-based cohort of 726 individuals with polycythemia vera, essential thrombocythemia, and myelofibrosis and 252 637 population controls unselected for hematologic phenotypes. Using a single-nucleotide polymorphism (SNP) array platform with custom probes for the JAK2 V617F mutation (V617F), we identified 497 individuals (0.2%) among the population controls who were V617F carriers. We performed a combined GWAS of the MPN cases plus V617F carriers in the control population (n = 1223) vs the remaining controls who were noncarriers for V617F (n = 252 140). For these MPN cases plus V617F carriers, we replicated the germ line JAK2 46/1 haplotype (rs59384377: odds ratio [OR] = 2.4, P = 6.6 × 10(-89)), previously associated with V617F-positive MPN. We also identified genome-wide significant associations in the TERT gene (rs7705526: OR = 1.8, P = 1.1 × 10(-32)), in SH2B3 (rs7310615: OR = 1.4, P = 3.1 × 10(-14)), and upstream of TET2 (rs1548483: OR = 2.0, P = 2.0 × 10(-9)). These associations were confirmed in a separate replication cohort of 446 V617F carriers vs 169 021 noncarriers. In a joint analysis of the combined GWAS and replication results, we identified additional genome-wide significant predisposition alleles associated with CHEK2, ATM, PINT, and GFI1B All SNP ORs were similar for MPN patients and controls who were V617F carriers. These data indicate that the same germ line variants endow individuals with a predisposition not only to MPN, but also to JAK2 V617F clonal hematopoiesis, a more common phenomenon that may foreshadow the development of an overt neoplasm.
Collapse
|
50
|
Chen Y, Fang F, Hu Y, Liu Q, Bu D, Tan M, Wu L, Zhu P. The Polymorphisms in LNK Gene Correlated to the Clinical Type of Myeloproliferative Neoplasms. PLoS One 2016; 11:e0154183. [PMID: 27111338 PMCID: PMC4844169 DOI: 10.1371/journal.pone.0154183] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/11/2016] [Indexed: 01/10/2023] Open
Abstract
Objective LNK is an adapter protein negatively regulating the JAK/STAT cell signaling pathway. In this study, we observed the correlation between variation in LNK gene and the clinical type of myeloproliferative neoplasms (MPN). Methods A total of 285 MPN cases were recruited, including essential thrombocythemia (ET) 154 cases, polycythemia vera (PV) 76 cases, primary myelofibrosis (PMF) 19 cases, and chronic myeloid leukemia (CML) 36 cases. Ninety-three healthy individuals were used as normal controls. V617F mutation in JAK2 was identified by allele-specific PCR method, RT-PCR was used for the detection of BCR/ABL1 fusion gene, and mutations and variations in coding exons and their flanking sequences of LNK gene were examined by PCR-sequencing. Results Missense mutations of A300V, V402M, and R415H in LNK were found in 8 patients including ET (4 cases, all combined with JAK2-V617F mutation), PV (2 cases, one combined with JAK2-V617F mutation), PMF (one case, combined with JAK2-V617F mutation) and CML (one case, combined with BCR/ABL1 fusion gene). The genotype and allele frequencies of the three SNPs (rs3184504, rs111340708 and rs78894077) in LNK were significantly different between MPN patients and controls. For rs3184504 (T/C, in exon2), the T allele (p.262W) and TT genotype were frequently seen in ET, PV and PMF (P<0.01), and C allele (p.262R) and CC genotype were frequently seen in CML (P<0.01). For rs78894077 (T/C, in exon1), the T allele (p.242S) was frequently found in ET (P<0.05). For rs111340708 (TGGGGx5/TGGGGx4, in intron 5), the TGGGG x4 allele was infrequently found in ET, PMF and CML(P<0.01). Conclusion Mutations in LNK could be found in some of MPN patients in the presence or absence of JAK2-V617F mutation. Several polymorphisms in LNK gene may affect the clinical type or the genetic predisposition of MPN.
Collapse
MESH Headings
- 3' Flanking Region
- 5' Flanking Region
- Adaptor Proteins, Signal Transducing
- Adult
- Aged
- Alleles
- Base Sequence
- Case-Control Studies
- Exons
- Female
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Gene Expression Regulation
- Gene Frequency
- Genetic Predisposition to Disease
- Genotype
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Intracellular Signaling Peptides and Proteins
- Janus Kinase 2/genetics
- Janus Kinase 2/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Male
- Middle Aged
- Mutation
- Open Reading Frames
- Phenotype
- Polycythemia Vera/diagnosis
- Polycythemia Vera/genetics
- Polycythemia Vera/metabolism
- Polycythemia Vera/pathology
- Polymorphism, Single Nucleotide
- Primary Myelofibrosis/diagnosis
- Primary Myelofibrosis/genetics
- Primary Myelofibrosis/metabolism
- Primary Myelofibrosis/pathology
- Proteins/genetics
- Proteins/metabolism
- Signal Transduction
- Thrombocythemia, Essential/diagnosis
- Thrombocythemia, Essential/genetics
- Thrombocythemia, Essential/metabolism
- Thrombocythemia, Essential/pathology
Collapse
Affiliation(s)
- Yan Chen
- Department of Hematology, Peking University First Hospital, Beijing, China
- Zunyi Medical College Affiliated Hospital, Zunyi, Guizhou, China
| | - Fang Fang
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Yang Hu
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Qian Liu
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Dingfang Bu
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Mei Tan
- Zunyi Medical College Affiliated Hospital, Zunyi, Guizhou, China
| | - Liusong Wu
- Zunyi Medical College Affiliated Hospital, Zunyi, Guizhou, China
| | - Ping Zhu
- Department of Hematology, Peking University First Hospital, Beijing, China
- * E-mail:
| |
Collapse
|