1
|
Vicario R, Fragkogianni S, Weber L, Lazarov T, Hu Y, Hayashi SY, Craddock B, Socci ND, Alberdi A, Baako A, Ay O, Ogishi M, Lopez-Rodrigo E, Kappagantula R, Viale A, Iacobuzio-Donahue CA, Zhou T, Ransohoff RM, Chesworth R, Abdel-Wahab O, Boisson B, Elemento O, Casanova JL, Miller WT, Geissmann F. A microglia clonal inflammatory disorder in Alzheimer's disease. eLife 2025; 13:RP96519. [PMID: 40085681 PMCID: PMC11908784 DOI: 10.7554/elife.96519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025] Open
Abstract
Somatic genetic heterogeneity resulting from post-zygotic DNA mutations is widespread in human tissues and can cause diseases, however, few studies have investigated its role in neurodegenerative processes such as Alzheimer's disease (AD). Here, we report the selective enrichment of microglia clones carrying pathogenic variants, that are not present in neuronal, glia/stromal cells, or blood, from patients with AD in comparison to age-matched controls. Notably, microglia-specific AD-associated variants preferentially target the MAPK pathway, including recurrent CBL ring-domain mutations. These variants activate ERK and drive a microglia transcriptional program characterized by a strong neuro-inflammatory response, both in vitro and in patients. Although the natural history of AD-associated microglial clones is difficult to establish in humans, microglial expression of a MAPK pathway activating variant was previously shown to cause neurodegeneration in mice, suggesting that AD-associated neuroinflammatory microglial clones may contribute to the neurodegenerative process in patients.
Collapse
Affiliation(s)
- Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Stamatina Fragkogianni
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Leslie Weber
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Yang Hu
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell New YorkNew YorkUnited States
| | - Samantha Y Hayashi
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony BrookNew YorkUnited States
| | - Barbara Craddock
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony BrookNew YorkUnited States
| | - Nicholas D Socci
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Araitz Alberdi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Ann Baako
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Oyku Ay
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New YorkNew YorkUnited States
| | - Estibaliz Lopez-Rodrigo
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Rajya Kappagantula
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Agnes Viale
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Christine A Iacobuzio-Donahue
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Ting Zhou
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | | | | | | | - Omar Abdel-Wahab
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New YorkNew YorkUnited States
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell New YorkNew YorkUnited States
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New YorkNew YorkUnited States
| | - W Todd Miller
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony BrookNew YorkUnited States
| | - Frédéric Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New YorkNew YorkUnited States
| |
Collapse
|
2
|
Bohlen J, Bagarić I, Vatovec T, Ogishi M, Ahmed SF, Cederholm A, Buetow L, Sobrino S, Le Floc’h C, Arango-Franco CA, Seabra L, Michelet M, Barzaghi F, Leardini D, Saettini F, Vendemini F, Baccelli F, Catala A, Gambineri E, Veltroni M, Aguilar de la Red Y, Rice GI, Consonni F, Berteloot L, Largeaud L, Conti F, Roullion C, Masson C, Bessot B, Seeleuthner Y, Le Voyer T, Rinchai D, Rosain J, Neehus AL, Erazo-Borrás L, Li H, Janda Z, Cho EJ, Muratore E, Soudée C, Lainé C, Delabesse E, Goulvestre C, Ma CS, Puel A, Tangye SG, André I, Bole-Feysot C, Abel L, Erlacher M, Zhang SY, Béziat V, Lagresle-Peyrou C, Six E, Pasquet M, Alsina L, Aiuti A, Zhang P, Crow YJ, Landegren N, Masetti R, Huang DT, Casanova JL, Bustamante J. Autoinflammation in patients with leukocytic CBL loss of heterozygosity is caused by constitutive ERK-mediated monocyte activation. J Clin Invest 2024; 134:e181604. [PMID: 39403923 PMCID: PMC11475086 DOI: 10.1172/jci181604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
Patients heterozygous for germline CBL loss-of-function (LOF) variants can develop myeloid malignancy, autoinflammation, or both, if some or all of their leukocytes become homozygous for these variants through somatic loss of heterozygosity (LOH) via uniparental isodisomy. We observed an upregulation of the inflammatory gene expression signature in whole blood from these patients, mimicking monogenic inborn errors underlying autoinflammation. Remarkably, these patients had constitutively activated monocytes that secreted 10 to 100 times more inflammatory cytokines than those of healthy individuals and CBL LOF heterozygotes without LOH. CBL-LOH hematopoietic stem and progenitor cells (HSPCs) outgrew the other cells, accounting for the persistence of peripheral monocytes homozygous for the CBL LOF variant. ERK pathway activation was required for the excessive production of cytokines by both resting and stimulated CBL-LOF monocytes, as shown in monocytic cell lines. Finally, we found that about 1 in 10,000 individuals in the UK Biobank were heterozygous for CBL LOF variants and that these carriers were at high risk of hematological and inflammatory conditions.
Collapse
Affiliation(s)
- Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Ivan Bagarić
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - Taja Vatovec
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Syed F. Ahmed
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Lori Buetow
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
| | - Steicy Sobrino
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Laboratory of Chromatin and Gene Regulation during Development, Paris Cité University, INSERM U1163, Imagine Institute, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM U1163, Imagine Institute, Paris, France
| | - Corentin Le Floc’h
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Carlos A. Arango-Franco
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Luis Seabra
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Marine Michelet
- Unit of Allergy and Pneumology, Children’s Hospital, Toulouse, France
| | - Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Davide Leardini
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
| | - Francesco Saettini
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | | | - Francesco Baccelli
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
| | - Albert Catala
- Pediatric Hematology and Oncology Department, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - Eleonora Gambineri
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Marinella Veltroni
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, Florence, Italy
| | | | - Gillian I. Rice
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Filippo Consonni
- Centre of Excellence, Division of Pediatric Oncology/Hematology, Meyer Children’s Hospital IRCCS, Florence, Italy
- “Mario Serio” Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Laureline Berteloot
- Department of Pediatric Imaging, Necker Hospital for Sick Children, Paris, France
- INSERM U1163, Paris, France
| | - Laetitia Largeaud
- Laboratory of Hematology, Hospital Center of the University of Toulouse, Toulouse, France
| | - Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Cécile Roullion
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Genomics Core Facility and
| | - Cécile Masson
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Bioinformatic Plateform, INSERM U1163 and INSERM US24/CNRS UAR3633, Paris Cité University, Paris, France
| | - Boris Bessot
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Clinical Immunology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Saint-Louis Hospital, Paris, France
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children–AP-HP, Paris, France
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Lucia Erazo-Borrás
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Primary Immunodeficiencies Group, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia, Medellín, Colombia
| | - Hailun Li
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Zarah Janda
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - En-Jui Cho
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Heidelberg University, Heidelberg, Germany
| | - Edoardo Muratore
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero–Universitaria di Bologna, Bologna, Italy
| | - Camille Soudée
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Candice Lainé
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Eric Delabesse
- Department of Hematology, CHU and Centre de Recherche de Cancérologie de Toulouse, Paul-Sabatier University, Toulouse, France
| | | | - Cindy S. Ma
- Garvan Institute of Medical Research, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, Australia
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, Australia
| | - Isabelle André
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
| | - Christine Bole-Feysot
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Genomics Core Facility and
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Chantal Lagresle-Peyrou
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Emmanuelle Six
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM U1163, Imagine Institute, Paris, France
| | - Marlène Pasquet
- Department of Pediatric Hematology and Oncology, Centre Hospitalo–Universitaire de Toulouse, Toulouse, France
| | - Laia Alsina
- Clinical Immunology and Primary Immunodeficiencies Unit, Pediatric Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Peng Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
| | - Yanick J. Crow
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Centre for Molecular Medicine, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Riccardo Masetti
- Unit of Allergy and Pneumology, Children’s Hospital, Toulouse, France
| | - Danny T. Huang
- Cancer Research UK Scotland Institute, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Department of Pediatrics, Necker Hospital for Sick Children–AP-HP, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, INSERM U1163, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children–AP-HP, Paris, France
| |
Collapse
|
3
|
Vicario R, Fragkogianni S, Weber L, Lazarov T, Hu Y, Hayashi SY, Craddock BP, Socci ND, Alberdi A, Baako A, Ay O, Ogishi M, Lopez-Rodrigo E, Kappagantula R, Viale A, Iacobuzio-Donahue CA, Zhou T, Ransohoff RM, Chesworth R, Abdel-Wahab O, Boisson B, Elemento O, Casanova JL, Miller WT, Geissmann F. A microglia clonal inflammatory disorder in Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577216. [PMID: 38328106 PMCID: PMC10849735 DOI: 10.1101/2024.01.25.577216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Somatic genetic heterogeneity resulting from post-zygotic DNA mutations is widespread in human tissues and can cause diseases, however few studies have investigated its role in neurodegenerative processes such as Alzheimer's Disease (AD). Here we report the selective enrichment of microglia clones carrying pathogenic variants, that are not present in neuronal, glia/stromal cells, or blood, from patients with AD in comparison to age-matched controls. Notably, microglia-specific AD-associated variants preferentially target the MAPK pathway, including recurrent CBL ring-domain mutations. These variants activate ERK and drive a microglia transcriptional program characterized by a strong neuro-inflammatory response, both in vitro and in patients. Although the natural history of AD-associated microglial clones is difficult to establish in human, microglial expression of a MAPK pathway activating variant was previously shown to cause neurodegeneration in mice, suggesting that AD-associated neuroinflammatory microglial clones may contribute to the neurodegenerative process in patients.
Collapse
Affiliation(s)
- Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Stamatina Fragkogianni
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Leslie Weber
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Yang Hu
- Department of Physiology and Biophysics, Institute for Computational Biomedicine,Weill Cornell New York, NY 10021, USA
| | - Samantha Y. Hayashi
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Barbara P. Craddock
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Nicholas D. Socci
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Araitz Alberdi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Ann Baako
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Oyku Ay
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
| | - Estibaliz Lopez-Rodrigo
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Rajya Kappagantula
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Agnes Viale
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Christine A. Iacobuzio-Donahue
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Ting Zhou
- SKI Stem Cell Research Core, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | | | | | | | - Omar Abdel-Wahab
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine,Weill Cornell New York, NY 10021, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, 10065 NY, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY, 11794-8661
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
4
|
Nguyen LT, Zimmermann K, Kowenz-Leutz E, Lim R, Hofstätter M, Mildner A, Leutz A. C/EBPβ-induced lymphoid-to-myeloid transdifferentiation emulates granulocyte-monocyte progenitor biology. Stem Cell Reports 2024; 19:112-125. [PMID: 38157851 PMCID: PMC10828814 DOI: 10.1016/j.stemcr.2023.11.011] [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: 12/22/2022] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
CCAAT/enhancer-binding protein beta (C/EBPβ) induces primary v-Abl immortalized mouse B cells to transdifferentiate (BT, B cell transdifferentiation) into granulocyte-macrophage progenitor-like cells (GMPBTs). GMPBTs maintain cytokine-independent self-renewal, lineage choice, and multilineage differentiation. Single-cell transcriptomics demonstrated that GMPBTs comprise a continuum of myelomonopoietic differentiation states that seamlessly fit into state-to-fate maps of normal granulocyte-macrophage progenitors (GMPs). Inactivating v-Abl kinase revealed the dependence on activated CSF2-JAK2-STAT5 signaling. Deleting IRF8 diminished monopoiesis and enhanced granulopoiesis while removing C/EBPβ-abrogated self-renewal and granulopoiesis but permitted macrophage differentiation. The GMPBT culture system is easily scalable to explore the basics of GMP biology and lineage commitment and largely reduces ethically and legislatively debatable, labor-intensive, and costly animal experiments.
Collapse
Affiliation(s)
- Linh Thuy Nguyen
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, Berlin, Germany; Berlin School of Integrative Oncology (BSIO), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Karin Zimmermann
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, Berlin, Germany
| | - Elisabeth Kowenz-Leutz
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, Berlin, Germany
| | - Ramonique Lim
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, Berlin, Germany
| | - Maria Hofstätter
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, Berlin, Germany
| | - Alexander Mildner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, Berlin, Germany; Institute of Biomedicine at University of Turku, Turku, Finland; InFLAMES Research Flagship, University of Turku, 20014 Turku, Finland
| | - Achim Leutz
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Roessle-Str. 10, Berlin, Germany.
| |
Collapse
|
5
|
Du L, Wang B, Wu M, Chen W, Wang W, Diao W, Ding M, Chen W, Cao W, Guo H, Zhang G. LINC00926 promotes progression of renal cell carcinoma via regulating miR-30a-5p/SOX4 axis and activating IFNγ-JAK2-STAT1 pathway. Cancer Lett 2023; 578:216463. [PMID: 37866544 DOI: 10.1016/j.canlet.2023.216463] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
The role of long non-coding RNA (lncRNA) in the progression of renal cell carcinoma (RCC) remains further study. Whether lncRNA may be used to predict the immunotherapy efficacy of RCC is less studied. In this study, LINC00926 was found to be mainly located in cytoplasm by FISH and RNA nuclear-cytoplasmic fractionation. Downregulation of LINC00926 in RCC cell lines inhibited the progression and metastasis of RCC cells. RNA pull-down assay and dual-luciferase reporter assay demonstrated that LINC00926 functioned as miR-30a-5p sponge to facilitate SOX4 expression. LINC00926 overexpression in BALB/c mice enhanced PD-L1 surface expression and resulted in immune escape. Mechanistic investigations showed that LINC00926 competitively bound to Lyn, leading to the inhibition of CBL-mediated ubiquitination and degradation, and stabilized Lyn, contributing to the activation of IFNγ-JAK2-STAT1 signaling pathway. Moreover, LINC00926, together with PD-L1 or PD-1 expression, may predict the overall survival in RCC patients. Therefore, LINC00926 has the potential to be a novel therapeutic target and a biomarker to predict ICB immunotherapy response in RCC.
Collapse
Affiliation(s)
- Lin Du
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Southeast University, Nanjing, 210008, Jiangsu, China; Department of Urology, The First People's Hospital of Yancheng, Yancheng, 224006, Jiangsu, China
| | - Baojun Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230039, Anhui, China; Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Mengtong Wu
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Weixu Chen
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Wendi Wang
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Southeast University, Nanjing, 210008, Jiangsu, China
| | - Wenli Diao
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Meng Ding
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Wei Chen
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Wenmin Cao
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Hongqian Guo
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Gutian Zhang
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Southeast University, Nanjing, 210008, Jiangsu, China; Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
6
|
Mendoza-Castrejon J, Magee JA. Layered immunity and layered leukemogenicity: Developmentally restricted mechanisms of pediatric leukemia initiation. Immunol Rev 2023; 315:197-215. [PMID: 36588481 PMCID: PMC10301262 DOI: 10.1111/imr.13180] [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] [Indexed: 01/03/2023]
Abstract
Hematopoietic stem cells (HSCs) and multipotent progenitor cells (MPPs) arise in successive waves during ontogeny, and their properties change significantly throughout life. Ontological changes in HSCs/MPPs underlie corresponding changes in mechanisms of pediatric leukemia initiation. As HSCs and MPPs progress from fetal to neonatal, juvenile and adult stages of life, they undergo transcriptional and epigenetic reprogramming that modifies immune output to meet age-specific pathogenic challenges. Some immune cells arise exclusively from fetal HSCs/MPPs. We propose that this layered immunity instructs cell fates that underlie a parallel layered leukemogenicity. Indeed, some pediatric leukemias, such as juvenile myelomonocytic leukemia, myeloid leukemia of Down syndrome, and infant pre-B-cell acute lymphoblastic leukemia, are age-restricted. They only present during infancy or early childhood. These leukemias likely arise from fetal progenitors that lose competence for transformation as they age. Other childhood leukemias, such as non-infant pre-B-cell acute lymphoblastic leukemia and acute myeloid leukemia, have mutation profiles that are common in childhood but rare in morphologically similar adult leukemias. These differences could reflect temporal changes in mechanisms of mutagenesis or changes in how progenitors respond to a given mutation at different ages. Interactions between leukemogenic mutations and normal developmental switches offer potential targets for therapy.
Collapse
Affiliation(s)
- Jonny Mendoza-Castrejon
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
| | - Jeffrey A. Magee
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
| |
Collapse
|
7
|
An Independent Assessment of a Commercial Clinical Interpretation Software Indicates That Software Can Mitigate Variation in Human Assessment. JOURNAL OF MOLECULAR PATHOLOGY 2022. [DOI: 10.3390/jmp3030012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Comprehensive next-generation sequencing (NGS) panels for cancer diagnostics create a bottleneck for interpretation. QIAGEN Clinical Insights Interpret One (QCI) is a clinical decision support software that supports molecular pathologists in the classification of oncology-related variants. This study compares variant assessments by QCI to assessments utilizing current laboratory methods. Eight laboratories were recruited by the external quality assessment organization GenQA. The laboratories submitted VCFs from sequencing studies performed on both hematological disorders and solid tumors for analysis by QCI and an independent laboratory. Results were compared and conflicts were resolved using a panel of experts. In total, 14/149 variants (9%) reported as Tier 1 or Tier 2 by either QCI or the submitting laboratory were found to be discordant after expert panel review. In contrast, 41/149 variants (28%) reflected discrepancy among human reviewers. The expert panel was unable to reach resolution on eight variants. QCI demonstrates high concordance in the classification of actionable mutations with independent laboratory methods and expert assessment. The rate of disagreement among laboratories and the expert panel was greater than the disagreement between QCI and expert assessment. Disagreement among experts highlights the subjectivity of classifying variants. The study demonstrates that QCI interpretation supports streamlining and standardization of NGS variant interpretation.
Collapse
|
8
|
Genomic and Epigenomic Landscape of Juvenile Myelomonocytic Leukemia. Cancers (Basel) 2022; 14:cancers14051335. [PMID: 35267643 PMCID: PMC8909150 DOI: 10.3390/cancers14051335] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Juvenile myelomonocytic leukemia (JMML) is a rare pediatric myelodysplastic/myeloproliferative neoplasm characterized by the constitutive activation of the RAS pathway. In spite of the recent progresses in the molecular characterization of JMML, this disease is still a clinical challenge due to its heterogeneity, difficult diagnosis, poor prognosis, and the lack of curative treatment options other than hematopoietic stem cell transplantation (HSCT). In this review, we will provide a detailed overview of the genetic and epigenetic alterations occurring in JMML, and discuss their clinical relevance in terms of disease prognosis and risk of relapse after HSCT. We will also present the most recent advances on novel preclinical and clinical therapeutic approaches directed against JMML molecular targets. Finally, we will outline future research perspectives to further explore the oncogenic mechanism driving JMML leukemogenesis and progression, with special attention to the application of single-cell next-generation sequencing technologies. Abstract Juvenile myelomonocytic leukemia (JMML) is a rare myelodysplastic/myeloproliferative neoplasm of early childhood. Most of JMML patients experience an aggressive clinical course of the disease and require hematopoietic stem cell transplantation, which is currently the only curative treatment. JMML is characterized by RAS signaling hyperactivation, which is mainly driven by mutations in one of five genes of the RAS pathway, including PTPN11, KRAS, NRAS, NF1, and CBL. These driving mutations define different disease subtypes with specific clinico-biological features. Secondary mutations affecting other genes inside and outside the RAS pathway contribute to JMML pathogenesis and are associated with a poorer prognosis. In addition to these genetic alterations, JMML commonly presents aberrant epigenetic profiles that strongly correlate with the clinical outcome of the patients. This observation led to the recent publication of an international JMML stratification consensus, which defines three JMML clinical groups based on DNA methylation status. Although the characterization of the genomic and epigenomic landscapes in JMML has significantly contributed to better understand the molecular mechanisms driving the disease, our knowledge on JMML origin, cell identity, and intratumor and interpatient heterogeneity is still scarce. The application of new single-cell sequencing technologies will be critical to address these questions in the future.
Collapse
|
9
|
Role of CBL Mutations in Cancer and Non-Malignant Phenotype. Cancers (Basel) 2022; 14:cancers14030839. [PMID: 35159106 PMCID: PMC8833995 DOI: 10.3390/cancers14030839] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary CBL mutations are progressively being described as involved in different clinical manifestations. Somatic CBL mutations can be found in different type of cancer. The clinical spectrum of germline mutations configures the so-called CBL syndrome, a cancer-predisposing condition that includes multisystemic involvement characterized by variable phenotypic expression and expressivity. In this review we provide an up-to-date review of the clinical manifestation of CBL mutations and of the molecular mechanisms in which CBL exerts its pathogenic role. Abstract CBL plays a key role in different cell pathways, mainly related to cancer onset and progression, hematopoietic development and T cell receptor regulation. Somatic CBL mutations have been reported in a variety of malignancies, ranging from acute myeloid leukemia to lung cancer. Growing evidence have defined the clinical spectrum of germline CBL mutations configuring the so-called CBL syndrome; a cancer-predisposing condition that also includes multisystemic involvement characterized by variable phenotypic expression and expressivity. This review provides a comprehensive overview of the molecular mechanisms in which CBL exerts its function and describes the clinical manifestation of CBL mutations in humans.
Collapse
|
10
|
Schreck KC, Morin A, Zhao G, Allen AN, Flannery P, Glantz M, Green AL, Jones C, Jones KL, Kilburn LB, Nazemi KJ, Samuel D, Sanford B, Solomon DA, Wang J, Pratilas CA, Nicolaides T, Mulcahy Levy JM. Deconvoluting Mechanisms of Acquired Resistance to RAF Inhibitors in BRAF V600E-Mutant Human Glioma. Clin Cancer Res 2021; 27:6197-6208. [PMID: 34433654 PMCID: PMC8595717 DOI: 10.1158/1078-0432.ccr-21-2660] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/12/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Selective RAF-targeted therapy is effective in some patients with BRAFV600E-mutated glioma, though emergent and adaptive resistance occurs through ill-defined mechanisms. EXPERIMENTAL DESIGN Paired pre-/post- RAF inhibitor (RAFi)-treated glioma samples (N = 15) were obtained and queried for treatment-emergent genomic alterations using DNA and RNA sequencing (RNA-seq). Functional validation of putative resistance mechanisms was performed using established and patient-derived BRAFV600E-mutant glioma cell lines. RESULTS Analysis of 15 tissue sample pairs identified 13 alterations conferring putative resistance were identified among nine paired samples (including mutations involving ERRFI1, BAP1, ANKHD1, and MAP2K1). We performed functional validation of mechanisms of resistance, including loss of NF1, PTEN, or CBL, in BRAFV600E-mutant glioma lines, and demonstrate they are capable of conferring resistance in vitro. Knockdown of CBL resulted in increased EGFR expression and phosphorylation, a possible mechanism for maintaining ERK signaling within the cell. Combination therapy with a MEKi or EGFR inhibitor was able to overcome resistance to BRAFi, in NF1 knockdown and CBL knockdown, respectively. Restoration of wild-type PTEN in B76 cells (PTEN-/-) restored sensitivity to BRAFi. We identified and validated CRAF upregulation as a mechanism of resistance in one resistant sample. RNA-seq analysis identified two emergent expression patterns in resistant samples, consistent with expression patterns of known glioma subtypes. CONCLUSIONS Resistance mechanisms to BRAFi in glioma are varied and may predict effective precision combinations of targeted therapy, highlighting the importance of a personalized approach.
Collapse
Affiliation(s)
- Karisa C Schreck
- Department of Neurology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Neurosurgery, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Andrew Morin
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, Colorado
| | - Guisheng Zhao
- Department of Pediatrics, NYU Langone Health, New York, New York
| | - Amy N Allen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Pediatrics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Patrick Flannery
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, Colorado
| | - Michael Glantz
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania
- Department of Oncology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Adam L Green
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, Colorado
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | | | - Lindsay B Kilburn
- Division of Oncology and the Brain Tumor Institute, Children's National Hospital, Washington, DC
| | - Kellie J Nazemi
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon
| | - David Samuel
- Department of Hematology-Oncology, Valley Children's Healthcare, Madera, California
| | - Bridget Sanford
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - David A Solomon
- Department of Pathology, University of California, San Francisco, California
| | - Jiawan Wang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Pediatrics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Christine A Pratilas
- Department of Neurology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
- Department of Pediatrics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | | | - Jean M Mulcahy Levy
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado.
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, Colorado
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, Colorado
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
11
|
Melas M, Mathew MT, Mori M, Jayaraman V, Wilson SA, Martin C, Jacobson-Kelly AE, Kelly BJ, Magrini V, Mardis ER, Cottrell CE, Lee K. Somatic Variation as an Incidental Finding in the Pediatric Next Generation Sequencing Era. Cold Spring Harb Mol Case Stud 2021; 7:mcs.a006135. [PMID: 34716204 PMCID: PMC8751410 DOI: 10.1101/mcs.a006135] [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: 07/29/2021] [Accepted: 10/27/2021] [Indexed: 11/28/2022] Open
Abstract
The methodologic approach used in next-generation sequencing (NGS) affords a high depth of coverage in genomic analysis. Inherent in the nature of genomic testing, there exists potential for identifying genomic findings that are incidental or secondary to the indication for clinical testing, with the frequency dependent on the breadth of analysis and the tissue sample under study. The interpretation and management of clinically meaningful incidental genomic findings is a pressing issue particularly in the pediatric population. Our study describes a 16-mo-old male who presented with profound global delays, brain abnormality, progressive microcephaly, and growth deficiency, as well as metopic craniosynostosis. Clinical exome sequencing (ES) trio analysis revealed the presence of two variants in the proband. The first was a de novo variant in the PPP2R1A gene (c.773G > A, p.Arg258His), which is associated with autosomal dominant (AD) intellectual disability, accounting for the proband's clinical phenotype. The second was a recurrent hotspot variant in the CBL gene (c.1111T > C, p.Tyr371His), which was present at a variant allele fraction of 11%, consistent with somatic variation in the peripheral blood sample. Germline pathogenic variants in CBL are associated with AD Noonan syndrome–like disorder with or without juvenile myelomonocytic leukemia. Molecular analyses using a different tissue source, buccal epithelial cells, suggest that the CBL alteration may represent a clonal population of cells restricted to leukocytes. This report highlights the laboratory methodologic and interpretative processes and clinical considerations in the setting of acquired variation detected during clinical ES in a pediatric patient.
Collapse
Affiliation(s)
- Marilena Melas
- The Steve and Cindy Rasmussen Inst for Genomic Medicine, Nationwide Children's Hospital
| | - Mariam T Mathew
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital; Dept of Pathology, The Ohio State Univ; Dept of Pediatrics, The Ohio State University
| | - Mari Mori
- Dept of Pediatrics, The Ohio State University; Genetic and Genomic Medicine, Nationwide Children's Hospital
| | - Vijayakumar Jayaraman
- The Steve and Cindy Rasmussen Inst for Genomic Medicine, Nationwide Children's Hospital
| | - Sarah A Wilson
- The Steve and Cindy Rasmussen Inst for Genomic Medicine, Nationwide Children's Hospital
| | | | - Amanda E Jacobson-Kelly
- Dept of Pediatrics, The Ohio State University; Division of Hematology/Oncology/BMT, Nationwide Children's Hospital
| | - Ben J Kelly
- The Steve and Cindy Rasmussen Inst for Genomic Medicine, Nationwide Children's Hospital
| | - Vincent Magrini
- The Steve and Cindy Rasmussen Inst for Genomic Medicine, Nationwide Children's Hospital; Dept of Pediatrics, The Ohio State University
| | - Elaine R Mardis
- The Steve and Cindy Rasmussen Inst for Genomic Medicine, Nationwide Children's Hospital; Dept of Pediatrics, The Ohio State University
| | - Catherine E Cottrell
- The Steve and Cindy Rasmussen Inst for Genomic Medicine, Nationwide Children's Hospital; Dept of Pathology, The Ohio State University; Dept of Pediatrics, The Ohio State University
| | - Kristy Lee
- The Steve and Cindy Rasmussen Inst for Genomic Medicine, Nationwide Children's Hospital; Dept of Pathology, The Ohio State University; Dept of Pediatrics, The Ohio State University
| |
Collapse
|
12
|
CBL mutations drive PI3K/AKT signaling via increased interaction with LYN and PIK3R1. Blood 2021; 137:2209-2220. [PMID: 33512474 DOI: 10.1182/blood.2020006528] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
Casitas B-lineage lymphoma (CBL) encodes an E3 ubiquitin ligase and signaling adaptor that regulates receptor and nonreceptor tyrosine kinases. Recurrent CBL mutations occur in myeloid neoplasms, including 10% to 20% of chronic myelomonocytic leukemia (CMML) cases, and selectively disrupt the protein's E3 ubiquitin ligase activity. CBL mutations have been associated with poor prognosis, but the oncogenic mechanisms and therapeutic implications of CBL mutations remain incompletely understood. We combined functional assays and global mass spectrometry to define the phosphoproteome, CBL interactome, and mechanism of signaling activation in a panel of cell lines expressing an allelic series of CBL mutations. Our analyses revealed that increased LYN activation and interaction with mutant CBL are key drivers of enhanced CBL phosphorylation, phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1) recruitment, and downstream phosphatidylinositol 3-kinase (PI3K)/AKT signaling in CBL-mutant cells. Signaling adaptor domains of CBL, including the tyrosine kinase-binding domain, proline-rich region, and C-terminal phosphotyrosine sites, were all required for the oncogenic function of CBL mutants. Genetic ablation or dasatinib-mediated inhibition of LYN reduced CBL phosphorylation, CBL-PIK3R1 interaction, and PI3K/AKT signaling. Furthermore, we demonstrated in vitro and in vivo antiproliferative efficacy of dasatinib in CBL-mutant cell lines and primary CMML. Overall, these mechanistic insights into the molecular function of CBL mutations provide rationale to explore the therapeutic potential of LYN inhibition in CBL-mutant myeloid malignancies.
Collapse
|
13
|
Wybenga-Groot LE, Tench AJ, Simpson CD, Germain JS, Raught B, Moran MF, McGlade CJ. SLAP2 Adaptor Binding Disrupts c-CBL Autoinhibition to Activate Ubiquitin Ligase Function. J Mol Biol 2021; 433:166880. [PMID: 33617900 DOI: 10.1016/j.jmb.2021.166880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/05/2021] [Accepted: 02/12/2021] [Indexed: 10/22/2022]
Abstract
CBL is a RING type E3 ubiquitin ligase that functions as a negative regulator of tyrosine kinase signaling and loss of CBL E3 function is implicated in several forms of leukemia. The Src-like adaptor proteins (SLAP/SLAP2) bind to CBL and are required for CBL-dependent downregulation of antigen receptor, cytokine receptor, and receptor tyrosine kinase signaling. Despite the established role of SLAP/SLAP2 in regulating CBL activity, the nature of the interaction and the mechanisms involved are not known. To understand the molecular basis of the interaction between SLAP/SLAP2 and CBL, we solved the crystal structure of CBL tyrosine kinase binding domain (TKBD) in complex with SLAP2. The carboxy-terminal region of SLAP2 adopts an α-helical structure which binds in a cleft between the 4H, EF-hand, and SH2 domains of the TKBD. This SLAP2 binding site is remote from the canonical TKBD phospho-tyrosine peptide binding site but overlaps with a region important for stabilizing CBL in its autoinhibited conformation. In addition, binding of SLAP2 to CBL in vitro activates the ubiquitin ligase function of autoinhibited CBL. Disruption of the CBL/SLAP2 interface through mutagenesis demonstrated a role for this protein-protein interaction in regulation of CBL E3 ligase activity in cells. Our results reveal that SLAP2 binding to a regulatory cleft of the TKBD provides an alternative mechanism for activation of CBL ubiquitin ligase function.
Collapse
Affiliation(s)
- Leanne E Wybenga-Groot
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Program in Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; SPARC BioCentre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada.
| | - Andrea J Tench
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Program in Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| | - Craig D Simpson
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Program in Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Jonathan St Germain
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Brian Raught
- Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, ON M5G 2M9, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Michael F Moran
- Program in Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; SPARC BioCentre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - C Jane McGlade
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Program in Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, ON M5G 2M9, Canada.
| |
Collapse
|
14
|
Hecht A, Meyer JA, Behnert A, Wong E, Chehab F, Olshen A, Hechmer A, Aftandilian C, Bhat R, Choi SW, Chonat S, Farrar JE, Fluchel M, Frangoul H, Han JH, Kolb EA, Kuo DJ, MacMillan ML, Maese L, Maloney KW, Narendran A, Oshrine B, Schultz KR, Sulis ML, Van Mater D, Tasian SK, Hofmann WK, Loh ML, Stieglitz E. Molecular and phenotypic diversity of CBL-mutated juvenile myelomonocytic leukemia. Haematologica 2020; 107:178-186. [PMID: 33375775 PMCID: PMC8719097 DOI: 10.3324/haematol.2020.270595] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Indexed: 11/22/2022] Open
Abstract
Mutations in the CBL gene were first identified in adults with various myeloid malignancies. Some patients with juvenile myelomonocytic leukemia (JMML) were also noted to harbor mutations in CBL, but were found to have generally less aggressive disease courses compared to patients with other forms of Ras pathway-mutant JMML. Importantly, and in contrast to most reports in adults, the majority of CBL mutations in JMML patients are germline with acquired uniparental disomy occurring in affected marrow cells. Here, we systematically studied a large cohort of 33 JMML patients with CBL mutations and found that this disease is highly diverse in presentation and overall outcome. Moreover, we discovered somatically acquired CBL mutations in 15% of pediatric patients who presented with more aggressive disease. Neither clinical features nor methylation profiling were able to distinguish patients with somatic CBL mutations from those with germline CBL mutations, highlighting the need for germline testing. Overall, we demonstrate that disease courses are quite heterogeneous even among patients with germline CBL mutations. Prospective clinical trials are warranted to find ideal treatment strategies for this diverse cohort of patients.
Collapse
Affiliation(s)
- Anna Hecht
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA; Department of Hematology/Oncology, University Hospital Mannheim, Heidelberg University
| | - Julia A Meyer
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, San Francisco
| | - Astrid Behnert
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, San Francisco
| | - Eric Wong
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, San Francisco
| | - Farid Chehab
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco
| | - Adam Olshen
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA; Department of Epidemiology and Biostatistics, University of California
| | - Aaron Hechmer
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco
| | | | - Rukhmi Bhat
- Northwestern University Feinberg School of Medicine, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Sung Won Choi
- Blood and Marrow Transplantation Program, University of Michigan, Ann Arbor, MI
| | - Satheesh Chonat
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Jason E Farrar
- Arkansas Children's Research Institute, Little Rock, AR; Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Mark Fluchel
- University of Utah, Department of Pediatrics, Division of Pediatric Hematology-Oncology, Salt Lake City, UT
| | - Haydar Frangoul
- The Children's Hospital at TriStar Centennial and Sarah Cannon Research Institute, Nashville, TN
| | - Jennifer H Han
- Division of Pediatric Hematology-Oncology, University of California, San Diego/ Rady Children's Hospital San Diego
| | - Edward A Kolb
- Nemours Center for Cancer and Blood Disorders/Alfred I. DuPont Hospital for Children, Wilmington, DE
| | - Dennis J Kuo
- Division of Pediatric Hematology-Oncology, University of California, San Diego/ Rady Children's Hospital San Diego
| | - Margaret L MacMillan
- Blood and Marrow Transplant Program, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Luke Maese
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | | | - Aru Narendran
- Pediatric Hematology and Oncology, Alberta Children's Hospital, Calgary, Alberta
| | | | - Kirk R Schultz
- British Columbia Children's Hospital and Research Institute, Vancouver, British Columbia
| | - Maria L Sulis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center. 1275 York Avenue. 10065 New York, NY
| | - David Van Mater
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia; Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Wolf-Karsten Hofmann
- Department of Hematology/Oncology, University Hospital Mannheim, Heidelberg University
| | - Mignon L Loh
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco
| | - Elliot Stieglitz
- Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco.
| |
Collapse
|
15
|
Ali AM, Cooper J, Walker A, Jones D, Saad A. Adult-onset acute myeloid leukaemia in a patient with germline mutation of CBL. Br J Haematol 2020; 192:665-667. [PMID: 33216958 DOI: 10.1111/bjh.17234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Alaa M Ali
- Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Julia Cooper
- Department of Internal Medicine, Division of Human Genetics, The Ohio State University, Columbus, OH, USA
| | - Alison Walker
- Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Daniel Jones
- Department of Pathology, Division of Molecular Pathology, The Ohio State University, Columbus, OH, USA
| | - Ayman Saad
- Department of Internal Medicine, Division of Hematology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
16
|
Chen S, Sun Q, Sun D, Willette-Brown J, Anderson MJ, Gu Q, Lewandoski M, Hu Y, Zhu F, Wei F, Zhang J. C-CBL is required for inhibition of angiogenesis through modulating JAK2/STAT3 activity in ROP development. Biomed Pharmacother 2020; 132:110856. [PMID: 33125970 PMCID: PMC8336301 DOI: 10.1016/j.biopha.2020.110856] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 11/29/2022] Open
Abstract
Purpose: The incidence of retinopathy of prematurity (ROP) has increased continuously in recent years. However, the therapeutic effects of current treatments still remain undesired. This study aims to investigate the role of C-CBL in retinal angiogenesis in ROP and its potential as a therapeutic target. Methods: Mouse retina microvascular endothelial cells (mRMECs) and induced experimental ROP/ oxygen-induced retinopathy (OIR) mice were employed to investigate the role of C-CBL in angiogenesis with combined molecular and cellular approaches, and histopathology methods. OIR mouse pups at postnatal day 12 (P12) were either injected intravitreally with adenovirus overexpressing c-Cbl or c-Cbl siRNA. Retinal neovascularization and avascular status were evaluated by retinal immunofluorescence (IF) staining, whole-mounts and hematoxylin and eosin (H&E) staining. Results: C-CBL inhibits neovascularization by negatively regulating JAK2/STAT3/VEGF signaling axis in a ubiquitination-dependent manner. Knockdown of c-Cbl by siRNA reduced ubiquitin-mediated JAK2 degradation and increased levels of p-JAK2, p-STAT3, VEGF, and neovascularization in mRMECs, which can be reversed by JAK2 inhibitor treatment. While knockdown of c-Cbl significantly increased neovascular (NV) zone in the retinas, c-Cbl overexpression inhibited neovascularization in the retinal tissues in OIR mice. Conclusion: We found that C-CBL is required for anti-neovascularization process in ROP development by inhibiting JAK2/STAT3-dependent angiogenesis. Thus, our finding strongly suggest that C-CBL may be a potential novel therapeutic target for treating ROP.
Collapse
Affiliation(s)
- Shimei Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200080, China; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai, 20080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 20080, China
| | - Qiao Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200080, China; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai, 20080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 20080, China
| | - Dandan Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200080, China; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai, 20080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 20080, China
| | - Jami Willette-Brown
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Matthew J Anderson
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Qing Gu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200080, China; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
| | - Mark Lewandoski
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Yinling Hu
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Feng Zhu
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA.
| | - Fang Wei
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200080, China; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai, 20080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 20080, China.
| | - Jian Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200080, China; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai, 20080, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 20080, China.
| |
Collapse
|
17
|
Garcia-Barcena C, Osinalde N, Ramirez J, Mayor U. How to Inactivate Human Ubiquitin E3 Ligases by Mutation. Front Cell Dev Biol 2020; 8:39. [PMID: 32117970 PMCID: PMC7010608 DOI: 10.3389/fcell.2020.00039] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/16/2020] [Indexed: 12/24/2022] Open
Abstract
E3 ubiquitin ligases are the ultimate enzymes involved in the transfer of ubiquitin to substrate proteins, a process that determines the fate of the modified protein. Numerous diseases are caused by defects in the ubiquitin-proteasome machinery, including when the activity of a given E3 ligase is hampered. Thus, inactivation of E3 ligases and the resulting effects at molecular or cellular level have been the focus of many studies during the last few years. For this purpose, site-specific mutation of key residues involved in either protein interaction, substrate recognition or ubiquitin transfer have been reported to successfully inactivate E3 ligases. Nevertheless, it is not always trivial to predict which mutation(s) will block the catalytic activity of a ligase. Here we review over 250 site-specific inactivating mutations that have been carried out in 120 human E3 ubiquitin ligases. We foresee that the information gathered here will be helpful for the design of future experimental strategies.
Collapse
Affiliation(s)
- Cristina Garcia-Barcena
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Nerea Osinalde
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Juanma Ramirez
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Ugo Mayor
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain.,Ikerbasque - Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
18
|
After 95 years, it's time to eRASe JMML. Blood Rev 2020; 43:100652. [PMID: 31980238 DOI: 10.1016/j.blre.2020.100652] [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: 06/21/2019] [Revised: 12/07/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022]
Abstract
Juvenile myelomonocytic leukaemia (JMML) is a rare clonal disorder of early childhood. Constitutive activation of the RAS pathway is the initial event in JMML. Around 90% of patients diagnosed with JMML carry a mutation in the PTPN11, NRAS, KRAS, NF1 or CBL genes. It has been demonstrated that after this first genetic event, an additional somatic mutation or epigenetic modification is involved in disease progression. The available genetic and clinical data have enabled researchers to establish relationships between JMML and several clinical conditions, including Noonan syndrome, Ras-associated lymphoproliferative disease, and Moyamoya disease. Despite scientific progress and the development of more effective treatments, JMML is still a deadly disease: the 5-year survival rate is ~50%. Here, we report on recent research having led to a better understanding of the genetic and molecular mechanisms involved in JMML.
Collapse
|
19
|
Identification of diverse activating mutations of the RAS-MAPK pathway in histiocytic sarcoma. Mod Pathol 2019; 32:830-843. [PMID: 30626916 DOI: 10.1038/s41379-018-0200-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/24/2018] [Accepted: 11/24/2018] [Indexed: 01/23/2023]
Abstract
Recent studies have demonstrated recurrent activating mutations involving the classical MAPK and PI3K signaling pathways in a large proportion of histiocytic neoplasms, such as Langerhans cell histiocytosis. However, very little is known about the molecular genetics of histiocytic sarcoma, a rare aggressive malignant neoplasm that shows pathologic characteristics of mature macrophages. Here we report the genomic characteristics of a large cohort of histiocytic sarcomas (n = 28) using a targeted next-generation sequencing approach to identify driver alterations. We identified recurrent mutations involving the RAS-MAPK signaling pathway (MAP2K1, KRAS, NRAS, BRAF, PTPN11, NF1, CBL) in a majority (57%) of histiocytic sarcoma cases and report a clinical response to a MEK inhibitor (Cobimetinib) in a patient with a NF1-mutated histiocytic sarcoma. A smaller subset of cases (21%) also showed mutations resulting in activation of the PI3K signaling pathway (PTEN, MTOR, PIK3R1, PIK3CA). In addition, the tumor-suppressor gene CDKN2A was the most frequently altered gene (46%). Further, a subset of histiocytic sarcoma cases shows striking molecular genetic similarities to B cell lymphomas, supporting a clonal relationship between B cell neoplasms and a subset of histiocytic sarcomas. These findings support a cooperative role for MAPK, PI3K, and cyclin-CDK4/6-INK4 signaling in the pathogenesis of histiocytic sarcoma and provide a rational basis for targeting these pathways.
Collapse
|
20
|
Tsuda Y, Hirata M, Katayama K, Motoi T, Matsubara D, Oda Y, Fujita M, Kobayashi H, Kawano H, Nishida Y, Sakai T, Okuma T, Goto T, Ogura K, Kawai A, Ae K, Anazawa U, Suehara Y, Iwata S, Miyano S, Imoto S, Shibata T, Nakagawa H, Yamaguchi R, Tanaka S, Matsuda K. Massively parallel sequencing of tenosynovial giant cell tumors reveals novel CSF1 fusion transcripts and novel somatic CBL mutations. Int J Cancer 2019; 145:3276-3284. [PMID: 31107544 DOI: 10.1002/ijc.32421] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/10/2019] [Accepted: 04/30/2019] [Indexed: 12/17/2022]
Abstract
Tenosynovial giant cell tumor (TSGCT) is a rare neoplasm. Although surgical resection is the widely accepted primary treatment for TSGCT, recurrences are frequent, and patients' joint function may be severely compromised. Previous studies reported that CSF1-COL6A3 fusion genes were identified in approximately 30% of TSGCTs. The aim of our study was to comprehensively clarify the genomic abnormalities in TSGCTs. We performed whole exome sequencing in combination with target sequence validation on 34 TSGCT samples. RNA sequencing was also performed on 18 samples. RNA sequencing revealed fusion transcripts involving CSF1, including novel CSF1-VCAM1, CSF1-FN1 and CSF1-CDH1 fusions, in 13/18 (72%) cases. These fusion genes were validated by chromogenic in situ hybridization. All CSF1 fusions resulted in the deletion of CSF1 exon 9, which was previously shown to be an important negative regulator of CSF1 expression. We also found that 12 (35%) of the 34 TSGCT samples harbored CBL missense mutations. All mutations were detected in exons 8 or 9, which encode the linker and RING finger domain. Among these mutations, C404Y, L380P and R420Q were recurrent. CBL-mutated cases showed higher JAK2 expression than wild-type CBL cases (p = 0.013). CSF1 fusion genes and CBL mutations were not mutually exclusive, and both alterations were detected in six of the 18 (33%) tumors. The frequent deletion of CSF1 exon 9 in the fusion transcripts suggested the importance of this event in the etiology of TSGCT. Our results may contribute to the development of new targeted therapies using JAK2 inhibitors for CBL-mutated TSGCT.
Collapse
Affiliation(s)
- Yusuke Tsuda
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Orthopedic Surgery, University of Tokyo, Tokyo, Japan
| | - Makoto Hirata
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kotoe Katayama
- Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Toru Motoi
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Daisuke Matsubara
- Division of Integrative Pathology, Jichi Medical University, Tochigi, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masashi Fujita
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | | | - Hirotaka Kawano
- Department of Orthopedic Surgery, University of Tokyo, Tokyo, Japan
| | - Yoshihiro Nishida
- Department of Orthopedic Surgery, University of Nagoya, Nagoya, Japan
| | - Tomohisa Sakai
- Department of Orthopedic Surgery, University of Nagoya, Nagoya, Japan
| | - Tomotake Okuma
- Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Takahiro Goto
- Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Koichi Ogura
- Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akira Kawai
- Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Keisuke Ae
- Department of Orthopedic Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ukei Anazawa
- Department of Orthopedic Surgery, Tokyo Dental College, Ichikawa General Hospital, Ichikawa, Japan
| | | | - Shintaro Iwata
- Division of Orthopedic Surgery, Chiba Cancer Center, Chiba, Japan
| | - Satoru Miyano
- Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Laboratory of DNA information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Seiya Imoto
- Health Intelligence Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Tatsuhiro Shibata
- Laboratory of Molecular Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Rui Yamaguchi
- Laboratory of Sequence Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Sakae Tanaka
- Department of Orthopedic Surgery, University of Tokyo, Tokyo, Japan
| | - Koichi Matsuda
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| |
Collapse
|
21
|
Liu CS, Yang-Yen HF, Suen CS, Hwang MJ, Yen JJY. Cbl-mediated K63-linked ubiquitination of JAK2 enhances JAK2 phosphorylation and signal transduction. Sci Rep 2017; 7:4613. [PMID: 28676638 PMCID: PMC5496907 DOI: 10.1038/s41598-017-04078-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/09/2017] [Indexed: 01/18/2023] Open
Abstract
JAK2 activation is crucial for cytokine receptor signal transduction and leukemogenesis. However, the underlying processes that lead to full activation of JAK2 are unclear. Here, we report a positive role for ubiquitination of JAK2 during GM-CSF-induced activation. Upon GM-CSF stimulation, JAK2 ubiquitination is significantly enhanced through K63-linked poly-ubiquitination. Studies employing both knockout and overexpression of Cbl, an E3 ubiquitin ligase, led to the conclusion that Cbl specifically promotes JAK2 ubiquitination, and this was further confirmed in vitro using a Cbl ubiquitination assay. Moreover, following GM-CSF stimulation, the levels of phospho-JAK2 and -STAT5 and a STAT5 luciferase reporter assay were all reduced in Cbl knockout cells and this effect could be rescued by Cbl expression. Mechanistically, Cbl can interact with, and ubiquitinate JAK2 FERM and kinase domains via the Cbl TKB domain. Using lysine-to-arginine site-directed mutagenesis, K970 in the kinase domain of JAK2 was identified as the ubiquitination site important for promoting full JAK2 activation by Cbl via K63-conjugated poly-ubiquitination. Our study suggests that GM-CSF-induced JAK2 activation is enhanced by Cbl-mediated ubiquitination of JAK2. Targeting ubiquitination of JAK2 might offer a novel therapeutic strategy against JAK2-mediated disorders.
Collapse
Affiliation(s)
- Chun-Shan Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | | | - Ching-Shu Suen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Ming-Jing Hwang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Jeffrey Jong-Young Yen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC. .,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC.
| |
Collapse
|
22
|
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
|
23
|
Nadeau SA, An W, Mohapatra BC, Mushtaq I, Bielecki TA, Luan H, Zutshi N, Ahmad G, Storck MD, Sanada M, Ogawa S, Band V, Band H. Structural Determinants of the Gain-of-Function Phenotype of Human Leukemia-associated Mutant CBL Oncogene. J Biol Chem 2017; 292:3666-3682. [PMID: 28082680 DOI: 10.1074/jbc.m116.772723] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Indexed: 01/19/2023] Open
Abstract
Mutations of the tyrosine kinase-directed ubiquitin ligase CBL cause myeloid leukemias, but the molecular determinants of the dominant leukemogenic activity of mutant CBL oncogenes are unclear. Here, we first define a gain-of-function attribute of the most common leukemia-associated CBL mutant, Y371H, by demonstrating its ability to increase proliferation of hematopoietic stem/progenitor cells (HSPCs) derived from CBL-null and CBL/CBL-B-null mice. Next, we express second-site point/deletion mutants of CBL-Y371H in CBL/CBL-B-null HSPCs or the cytokine-dependent human leukemic cell line TF-1 to show that individual or combined Tyr → Phe mutations of established phosphotyrosine residues (Tyr-700, Tyr-731, and Tyr-774) had little impact on the activity of the CBL-Y371H mutant in HSPCs, and the triple Tyr → Phe mutant was only modestly impaired in TF-1 cells. In contrast, intact tyrosine kinase-binding (TKB) domain and proline-rich region (PRR) were critical in both cell models. PRR deletion reduced the stem cell factor (SCF)-induced hyper-phosphorylation of the CBL-Y371H mutant and the c-KIT receptor and eliminated the sustained p-ERK1/2 and p-AKT induction by SCF. GST fusion protein pulldowns followed by phospho-specific antibody array analysis identified distinct CBL TKB domains or PRR-binding proteins that are phosphorylated in CBL-Y371H-expressing TF-1 cells. Our results support a model of mutant CBL gain-of-function in which mutant CBL proteins effectively compete with the remaining wild type CBL-B and juxtapose TKB domain-associated PTKs with PRR-associated signaling proteins to hyper-activate signaling downstream of hematopoietic growth factor receptors. Elucidation of mutant CBL domains required for leukemogenesis should facilitate targeted therapy approaches for patients with mutant CBL-driven leukemias.
Collapse
Affiliation(s)
- Scott A Nadeau
- From the Eppley Institute for Research in Cancer and Allied Diseases.,the Departments of Genetics, Cell Biology and Anatomy
| | - Wei An
- From the Eppley Institute for Research in Cancer and Allied Diseases.,the Departments of Genetics, Cell Biology and Anatomy
| | - Bhopal C Mohapatra
- From the Eppley Institute for Research in Cancer and Allied Diseases.,Biochemistry and Molecular Biology
| | - Insha Mushtaq
- From the Eppley Institute for Research in Cancer and Allied Diseases.,Pathology and Microbiology, College of Medicine, and
| | | | - Haitao Luan
- From the Eppley Institute for Research in Cancer and Allied Diseases.,the Departments of Genetics, Cell Biology and Anatomy
| | - Neha Zutshi
- From the Eppley Institute for Research in Cancer and Allied Diseases.,Pathology and Microbiology, College of Medicine, and
| | - Gulzar Ahmad
- From the Eppley Institute for Research in Cancer and Allied Diseases
| | - Matthew D Storck
- From the Eppley Institute for Research in Cancer and Allied Diseases
| | - Masashi Sanada
- the Department of Pathology and Tumor Biology, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Seishi Ogawa
- the Department of Pathology and Tumor Biology, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Vimla Band
- From the Eppley Institute for Research in Cancer and Allied Diseases.,the Departments of Genetics, Cell Biology and Anatomy.,the Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198 and
| | - Hamid Band
- From the Eppley Institute for Research in Cancer and Allied Diseases, .,the Departments of Genetics, Cell Biology and Anatomy.,Biochemistry and Molecular Biology.,Pathology and Microbiology, College of Medicine, and.,the Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198 and
| |
Collapse
|
24
|
Sperling AS, Gibson CJ, Ebert BL. The genetics of myelodysplastic syndrome: from clonal haematopoiesis to secondary leukaemia. Nat Rev Cancer 2017; 17:5-19. [PMID: 27834397 PMCID: PMC5470392 DOI: 10.1038/nrc.2016.112] [Citation(s) in RCA: 426] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Myelodysplastic syndrome (MDS) is a clonal disease that arises from the expansion of mutated haematopoietic stem cells. In a spectrum of myeloid disorders ranging from clonal haematopoiesis of indeterminate potential (CHIP) to secondary acute myeloid leukaemia (sAML), MDS is distinguished by the presence of peripheral blood cytopenias, dysplastic haematopoietic differentiation and the absence of features that define acute leukaemia. More than 50 recurrently mutated genes are involved in the pathogenesis of MDS, including genes that encode proteins involved in pre-mRNA splicing, epigenetic regulation and transcription. In this Review we discuss the molecular processes that lead to CHIP and further clonal evolution to MDS and sAML. We also highlight the ways in which these insights are shaping the clinical management of MDS, including classification schemata, prognostic scoring systems and therapeutic approaches.
Collapse
Affiliation(s)
- Adam S Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Christopher J Gibson
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| |
Collapse
|
25
|
Katzav S, Schmitz ML. Mutations of c-Cbl in myeloid malignancies. Oncotarget 2016; 6:10689-96. [PMID: 26028666 PMCID: PMC4484412 DOI: 10.18632/oncotarget.3986] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/15/2015] [Indexed: 12/18/2022] Open
Abstract
Next generation sequencing has shown the frequent occurrence of point mutations in the ubiquitin E3 ligase c-Cbl in myeloid malignancies. Mouse models revealed a causal contribution of c-Cbl for the onset of such neoplasms. The point mutations typically cluster in the linker region and RING finger domain and affect both alleles by acquired uniparental disomy. The fast progress in the detection of c-Cbl mutations is contrasted by our scarce knowledge on their functional consequences. The c-Cbl protein displays several enzymatic functions by promoting the attachment of differentially composed ubiquitin chains and of the ubiquitin-like protein NEDD8 to its target proteins. In addition, c-Cbl functions as an adapter protein and undergoes phosphorylation-dependent inducible conformation changes. Studies on the impact of c-Cbl mutations on its functions as a dynamic and versatile adapter protein, its interactomes and on its various enzymatic activities are now important to allow the identification of druggable targets within the c-Cbl signaling network.
Collapse
Affiliation(s)
- Shulamit Katzav
- Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - M Lienhard Schmitz
- Institute of Biochemistry, University of Giessen, Friedrichstrasse, Giessen, Germany
| |
Collapse
|
26
|
Choi J, Polcher A, Joas A. Systematic literature review on Parkinson's disease and Childhood Leukaemia and mode of actions for pesticides. ACTA ACUST UNITED AC 2016. [DOI: 10.2903/sp.efsa.2016.en-955] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
27
|
Smad7 Modulates Epidermal Growth Factor Receptor Turnover through Sequestration of c-Cbl. Mol Cell Biol 2015. [PMID: 26055326 DOI: 10.1128/mcb.00274‐15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Epidermal growth factor (EGF) regulates various cellular events, including proliferation, differentiation, migration, and tumorigenesis. For the maintenance of homeostasis, EGF signaling should be tightly regulated to prevent the aberrant activation. Smad7 has been known as inhibitory Smad that blocks the signal transduction of transforming growth factor β. In the process of cell proliferation or transformation, Smad7 has been shown the opposite activities as a promoter or suppressor depending on cell types or microenvironments. We found that the overexpression of Smad7 in human HaCaT keratinocyte cells and mouse skin tissues elevated EGF receptor (EGFR) activity by impairing ligand-induced ubiquitination and degradation of activated receptor, which is induced by the E3 ubiquitin ligase c-Cbl. The C-terminal MH2 region but not MH1 region of Smad7 is critical for interaction with c-Cbl to inhibit the ubiquitination of EGFR. Interestingly, wild-type Smad7, but not Smad6 or mutant Smad7, destabilized the EGF-induced complex formation of c-Cbl and EGFR. These data suggest a novel role for Smad7 as a promoter for prolonging the EGFR signal in keratinocyte and skin tissue by reducing its ligand-induced ubiquitination and degradation.
Collapse
|
28
|
Smad7 Modulates Epidermal Growth Factor Receptor Turnover through Sequestration of c-Cbl. Mol Cell Biol 2015; 35:2841-50. [PMID: 26055326 DOI: 10.1128/mcb.00274-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/02/2015] [Indexed: 01/05/2023] Open
Abstract
Epidermal growth factor (EGF) regulates various cellular events, including proliferation, differentiation, migration, and tumorigenesis. For the maintenance of homeostasis, EGF signaling should be tightly regulated to prevent the aberrant activation. Smad7 has been known as inhibitory Smad that blocks the signal transduction of transforming growth factor β. In the process of cell proliferation or transformation, Smad7 has been shown the opposite activities as a promoter or suppressor depending on cell types or microenvironments. We found that the overexpression of Smad7 in human HaCaT keratinocyte cells and mouse skin tissues elevated EGF receptor (EGFR) activity by impairing ligand-induced ubiquitination and degradation of activated receptor, which is induced by the E3 ubiquitin ligase c-Cbl. The C-terminal MH2 region but not MH1 region of Smad7 is critical for interaction with c-Cbl to inhibit the ubiquitination of EGFR. Interestingly, wild-type Smad7, but not Smad6 or mutant Smad7, destabilized the EGF-induced complex formation of c-Cbl and EGFR. These data suggest a novel role for Smad7 as a promoter for prolonging the EGFR signal in keratinocyte and skin tissue by reducing its ligand-induced ubiquitination and degradation.
Collapse
|
29
|
Ku M, Wall M, MacKinnon RN, Walkley CR, Purton LE, Tam C, Izon D, Campbell L, Cheng HC, Nandurkar H. Src family kinases and their role in hematological malignancies. Leuk Lymphoma 2015; 56:577-86. [PMID: 24898666 DOI: 10.3109/10428194.2014.907897] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The Src family protein tyrosine kinases (SFKs) are non-receptor intracellular kinases that have important roles in both hematopoiesis and leukemogenesis. The derangement of their expression or activation has been demonstrated to contribute to hematological malignancies. This review first examines the mechanisms of SFK overexpression and hyperactivation, emphasizing the dysregulation of the upstream modulators. Subsequently, the role of SFK up-regulation in the initiation, progression and therapy resistance of many hematological malignancies is also analyzed. The presented evidence endeavors to highlight the influence of SFK up-regulation on an extensive number of hematological malignancies and the need to consider them as candidates in targeted anticancer therapy.
Collapse
Affiliation(s)
- Matthew Ku
- Haematology Department and Victorian Cancer Cytogenetics Service, St Vincent's Hospital , Fitzroy , Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bedside to bench in juvenile myelomonocytic leukemia: insights into leukemogenesis from a rare pediatric leukemia. Blood 2014; 124:2487-97. [PMID: 25163700 DOI: 10.1182/blood-2014-03-300319] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a typically aggressive myeloid neoplasm of childhood that is clinically characterized by overproduction of monocytic cells that can infiltrate organs, including the spleen, liver, gastrointestinal tract, and lung. JMML is categorized as an overlap myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) by the World Health Organization and also shares some clinical and molecular features with chronic myelomonocytic leukemia, a similar disease in adults. Although the current standard of care for patients with JMML relies on allogeneic hematopoietic stem cell transplant, relapse is the most frequent cause of treatment failure. Tremendous progress has been made in defining the genomic landscape of JMML. Insights from cancer predisposition syndromes have led to the discovery of nearly 90% of driver mutations in JMML, all of which thus far converge on the Ras signaling pathway. This has improved our ability to accurately diagnose patients, develop molecular markers to measure disease burden, and choose therapeutic agents to test in clinical trials. This review emphasizes recent advances in the field, including mapping of the genomic and epigenome landscape, insights from new and existing disease models, targeted therapeutics, and future directions.
Collapse
|