1
|
Maciel SVSA, Oliveira IPP, Senes BB, Silva JAIDV, Feitosa FLB, Alves JS, Costa RB, de Camargo GMF. Genomic regions associated with coat color in Gir cattle. Genome 2024; 67:233-242. [PMID: 38579337 DOI: 10.1139/gen-2023-0115] [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] [Indexed: 04/07/2024]
Abstract
Indicine cattle breeds are adapted to the tropical climate, and their coat plays an important role in this process. Coat color influences thermoregulation and the adhesion of ectoparasites and may be associated with productive and reproductive traits. Furthermore, coat color is used for breed qualification, with breeders preferring certain colors. The Gir cattle is characterized by a wide variety of coat colors. Therefore, we performed genome-wide association studies to identify candidate genes for coat color in Gir cattle. Different phenotype scenarios were considered in the analyses and regions were identified on eight chromosomes. Some regions and many candidate genes are influencing coat color in the Gir cattle, which was found to be a polygenic trait. The candidate genes identified have been associated with white spotting patterns and base coat color in cattle and other species. In addition, a possible epistatic effect on coat color determination in the Gir cattle was suggested. This is the first published study that identified genomic regions and listed candidate genes associated with coat color in Gir cattle. The findings provided a better understanding of the genetic architecture of the trait in the breed and will allow to guide future fine-mapping studies for the development of genetic markers for selection.
Collapse
|
2
|
Abdellateif MS, Bayoumi AK, Mohammed MA. c-Kit Receptors as a Therapeutic Target in Cancer: Current Insights. Onco Targets Ther 2023; 16:785-799. [PMID: 37790582 PMCID: PMC10544070 DOI: 10.2147/ott.s404648] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/19/2023] [Indexed: 10/05/2023] Open
Abstract
c-Kit is a type III receptor tyrosine kinase (RTK) that has an essential role in various biological functions including gametogenesis, melanogenesis, hematopoiesis, cell survival, and apoptosis. c-KIT aberrations, either overexpression or loss-of-function mutations, have been implicated in the pathogenesis and development of many cancers, including gastrointestinal stromal tumors, mastocytosis, acute myeloid leukemia, breast, thyroid, and colorectal cancer, making c-KIT an attractive molecular target for the treatment of cancers. Therefore, a lot of effort has been put into investigating the utility of tyrosine kinase inhibitors for the management of c-KIT mutated tumors. This review of the literature illustrates the role of c-KIT mutations in many cancers, aiming to provide insights into the role of TKIs as a therapeutic option for cancer patients with c-KIT aberrations. In conclusion, c-KIT is implicated in different types of cancer, and it could be a successful molecular target; however, proper detection of the underlying mutation type is required before starting the appropriate personalized therapy.
Collapse
Affiliation(s)
- Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| | - Ahmed K Bayoumi
- Paediatric Oncology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
- Children’s Cancer Hospital 57357, Cairo, 11617, Egypt
| | - Mohammed Aly Mohammed
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| |
Collapse
|
3
|
Xiong Y, Taleb M, Misawa K, Hou Z, Banerjee S, Amador-Molina A, Jones DR, Chintala NK, Adusumilli PS. c-Kit signaling potentiates CAR T cell efficacy in solid tumors by CD28- and IL-2-independent co-stimulation. NATURE CANCER 2023; 4:1001-1015. [PMID: 37336986 PMCID: PMC10765546 DOI: 10.1038/s43018-023-00573-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 05/08/2023] [Indexed: 06/21/2023]
Abstract
The limited efficacy of chimeric antigen receptor (CAR) T cell therapy for solid tumors necessitates engineering strategies that promote functional persistence in an immunosuppressive environment. Herein, we use c-Kit signaling, a physiological pathway associated with stemness in hematopoietic progenitor cells (T cells lose expression of c-Kit during differentiation). CAR T cells with intracellular expression, but no cell-surface receptor expression, of the c-Kit D816V mutation (KITv) have upregulated STAT phosphorylation, antigen activation-dependent proliferation and CD28- and interleukin-2-independent and interferon-γ-mediated co-stimulation, augmenting the cytotoxicity of first-generation CAR T cells. This translates to enhanced survival, including in transforming growth factor-β-rich and low-antigen-expressing solid tumor models. KITv CAR T cells have equivalent or better in vivo efficacy than second-generation CAR T cells and are susceptible to tyrosine kinase inhibitors (safety switch). When combined with CD28 co-stimulation, KITv co-stimulation functions as a third signal, enhancing efficacy and providing a potent approach to treat solid tumors.
Collapse
Affiliation(s)
- Yuquan Xiong
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meriem Taleb
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kyohei Misawa
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhaohua Hou
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Srijita Banerjee
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alfredo Amador-Molina
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Navin K Chintala
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
4
|
Sheikh E, Tran T, Vranic S, Levy A, Bonfil RD. Role and significance of c-KIT receptor tyrosine kinase in cancer: A review. Bosn J Basic Med Sci 2022; 22:683-698. [PMID: 35490363 PMCID: PMC9519160 DOI: 10.17305/bjbms.2021.7399] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
c-kit is a classical proto-oncogene that encodes a receptor tyrosine kinase (RTK) that responds to stem cell factor (SCF). C-KIT signaling is a critical regulator of cell proliferation, survival, and migration and is implicated in several physiological processes, including pigmentation, hematopoiesis and gut movement. Accumulating evidence suggests that dysregulated c-KIT function, caused by either overexpression or mutations in c-kit, promotes tumor development and progression in various human cancers. In this review, we discuss the most important structural and biological features of c-KIT, as well as insights into the activation of intracellular signaling pathways following SCF binding to this RTK. We then illustrate how different c-kit alterations are associated with specific human cancers and describe recent studies that highlight the contribution of c-KIT to cancer stemness, epithelial-mesenchymal transition and progression to metastatic disease in different experimental models. The impact of tyrosine kinase inhibitors in treating c-KIT-positive tumors and limitations due to their propensity to develop drug resistance are summarized. Finally, we appraise the potential of novel therapeutic approaches targeting c-KIT more selectively while minimizing toxicity to normal tissue.
Collapse
Affiliation(s)
- Emana Sheikh
- OMS-III, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, United States
| | - Tony Tran
- OMS-III, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, United States
| | - Semir Vranic
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Arkene Levy
- Department of Medical Education, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, United States
| | - R. Daniel Bonfil
- Department of Medical Education, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, United States
| |
Collapse
|
5
|
Fé LXSGM, Cipolatti EP, Pinto MCC, Branco S, Nogueira FCS, Ortiz GMD, Pinheiro ADS, Manoel EA. Enzymes in the time of COVID-19: An overview about the effects in the human body, enzyme market, and perspectives for new drugs. Med Res Rev 2022; 42:2126-2167. [PMID: 35762498 PMCID: PMC9350392 DOI: 10.1002/med.21919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 01/27/2022] [Accepted: 06/08/2022] [Indexed: 12/11/2022]
Abstract
The rising pandemic caused by a coronavirus, resulted in a scientific quest to discover some effective treatments against its etiologic agent, the severe acute respiratory syndrome‐coronavirus 2 (SARS‐CoV‐2). This research represented a significant scientific landmark and resulted in many medical advances. However, efforts to understand the viral mechanism of action and how the human body machinery is subverted during the infection are still ongoing. Herein, we contributed to this field with this compilation of the roles of both viral and human enzymes in the context of SARS‐CoV‐2 infection. In this sense, this overview reports that proteases are vital for the infection to take place: from SARS‐CoV‐2 perspective, the main protease (Mpro) and papain‐like protease (PLpro) are highlighted; from the human body, angiotensin‐converting enzyme‐2, transmembrane serine protease‐2, and cathepsins (CatB/L) are pointed out. In addition, the influence of the virus on other enzymes is reported as the JAK/STAT pathway and the levels of lipase, enzymes from the cholesterol metabolism pathway, amylase, aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, and glyceraldehyde 3‐phosphate dehydrogenase are also be disturbed in SARS‐CoV‐2 infection. Finally, this paper discusses the importance of detailed enzymatic studies for future treatments against SARS‐CoV‐2, and how some issues related to the syndrome treatment can create opportunities in the biotechnological market of enzymes and the development of new drugs.
Collapse
Affiliation(s)
- Luana Xavier Soares Gomes Moura Fé
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eliane Pereira Cipolatti
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Engenharia Química, Instituto de Tecnologia, Universidade Federal Rural do Rio de Janeiro (UFRRJ), Seropédica, Rio de Janeiro, Brazil
| | - Martina Costa Cerqueira Pinto
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil.,Chemical Engineering Program, Instituto Alberto Luiz Coimbra de Pós-graduação e Pesquisa de Engenharia (COPPE), Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suema Branco
- Biofísica Ambiental, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio César Sousa Nogueira
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gisela Maria Dellamora Ortiz
- Departamento de Fármacos e Medicamentos, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anderson de Sá Pinheiro
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Evelin Andrade Manoel
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Klaus A, Clapes T, Yvernogeau L, Basu S, Weijts B, Maas J, Smal I, Galjart N, Robin C. CLASP2 safeguards hematopoietic stem cell properties during mouse and fish development. Cell Rep 2022; 39:110957. [PMID: 35705037 DOI: 10.1016/j.celrep.2022.110957] [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: 08/12/2021] [Revised: 01/28/2022] [Accepted: 05/23/2022] [Indexed: 11/27/2022] Open
Abstract
Hematopoietic stem cells (HSCs) express a large variety of cell surface receptors that are associated with acquisition of self-renewal and multipotent properties. Correct expression of these receptors depends on a delicate balance between cell surface trafficking, recycling, and degradation and is controlled by the microtubule network and Golgi apparatus, whose roles have hardly been explored during embryonic/fetal hematopoiesis. Here we show that, in the absence of CLASP2, a microtubule-associated protein, the overall production of HSCs is reduced, and the produced HSCs fail to self-renew and maintain their stemness throughout mouse and zebrafish development. This phenotype can be attributed to decreased cell surface expression of the hematopoietic receptor c-Kit, which originates from increased lysosomal degradation in combination with a reduction in trafficking to the plasma membrane. A dysfunctional Golgi apparatus in CLASP2-deficient HSCs seems to be the underlying cause of the c-Kit expression and signaling imbalance.
Collapse
Affiliation(s)
- Anna Klaus
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Thomas Clapes
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Laurent Yvernogeau
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Sreya Basu
- Department of Cell Biology, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, the Netherlands
| | - Bart Weijts
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Joris Maas
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Ihor Smal
- Theme Biomedical Sciences and Departments of Cell Biology and Molecular Genetics, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, the Netherlands
| | - Niels Galjart
- Department of Cell Biology, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, the Netherlands
| | - Catherine Robin
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands; Regenerative Medicine Center, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands.
| |
Collapse
|
7
|
Rigo R, Chelbi R, Agopian J, Letard S, Griffon A, Ghamlouch H, Vernerey J, Ladopoulos V, Voisset E, De Sepulveda P, Guittard G, Nunès JA, Bidaut G, Göttgens B, Weber M, Bernard OA, Dubreuil P, Soucie E. TET2 regulates immune tolerance in chronically activated mast cells. JCI Insight 2022; 7:154191. [PMID: 35393954 PMCID: PMC9057605 DOI: 10.1172/jci.insight.154191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Abstract
Mutation of the TET2 DNA-hydroxymethylase has been associated with a number of immune pathologies. The disparity in phenotype and clinical presentation among these pathologies leads to questions regarding the role of TET2 mutation in promoting disease evolution in different immune cell types. Here we show that, in primary mast cells, Tet2 expression is induced in response to chronic and acute activation signals. In TET2-deficient mast cells, chronic activation via the oncogenic KITD816V allele associated with mastocytosis, selects for a specific epigenetic signature characterized by hypermethylated DNA regions (HMR) at immune response genes. H3K27ac and transcription factor binding is consistent with priming or more open chromatin at both HMR and non-HMR in proximity to immune genes in these cells, and this signature coincides with increased pathological inflammation signals. HMR are also associated with a subset of immune genes that are direct targets of TET2 and repressed in TET2-deficient cells. Repression of these genes results in immune tolerance to acute stimulation that can be rescued with vitamin C treatment or reiterated with a Tet inhibitor. Overall, our data support a model where TET2 plays a direct role in preventing immune tolerance in chronically activated mast cells, supporting TET2 as a viable target to reprogram the innate immune response for innovative therapies.
Collapse
Affiliation(s)
- Riccardo Rigo
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Rabie Chelbi
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France.,Inovarion, Paris, France
| | - Julie Agopian
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Sebastien Letard
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Aurélien Griffon
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Hussein Ghamlouch
- INSERM, Mixed Research Unit (UMR) 1170, Institut Gustave Roussy, Facility of Medicine, Paris-Sud University, Paris-Saclay University, Equipe Labélisée Ligue Nationale Contre le Cancer, Villejuif, France
| | - Julien Vernerey
- CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Vasileios Ladopoulos
- Department of Haematology, Cambridge Institute for Medical Research, and.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Edwige Voisset
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Paulo De Sepulveda
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Geoffrey Guittard
- CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Jacques A Nunès
- CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Ghislain Bidaut
- CRCM, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille University, Marseille, France
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, and.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Michael Weber
- CNRS, University of Strasbourg, UMR7242 Biotechnology and Cell Signaling, Illkirch, France
| | - Olivier A Bernard
- INSERM, Mixed Research Unit (UMR) 1170, Institut Gustave Roussy, Facility of Medicine, Paris-Sud University, Paris-Saclay University, Equipe Labélisée Ligue Nationale Contre le Cancer, Villejuif, France
| | - Patrice Dubreuil
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| | - Erinn Soucie
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Institut Paoli-Calmettes, Equipe Labélisée Ligue Nationale Contre le Cancer, Marseille, France
| |
Collapse
|
8
|
Luo W, Wang J, Zhou Y, Pang M, Yu X, Tong J. Dynamic mRNA and miRNA expression of the head during early development in bighead carp (Hypophthalmichthys nobilis). BMC Genomics 2022; 23:168. [PMID: 35232381 PMCID: PMC8887032 DOI: 10.1186/s12864-022-08387-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 02/09/2022] [Indexed: 11/24/2022] Open
Abstract
Background Head of fish species, an exquisitely complex anatomical system, is important not only for studying fish evolution and development, but also for economic values. Currently, although some studies have been made on fish growth and body shapes, very limited information is available on the molecular mechanism of head development. Results In this study, RNA sequencing (RNA–Seq) and small RNA sequencing (sRNA–Seq) technologies were used to conduct integrated analysis for the head of bighead carp at different development stages, including 1, 3, 5, 15 and 30 Dph (days post hatch). By RNA-Seq data, 26 pathways related to growth and bone formation were identified as the main physiological processes during early development. Coupling this to sRNA–Seq data, we picked out six key pathways that may be responsible for head development, namely ECM receptor interaction, TNF signaling pathway, osteoclast differentiation, PI3K–Akt signaling pathway, Neuroactive ligand–receptor interaction and Jak–STAT signaling pathway. Totally, 114 important candidate genes from the six pathways were obtained. Then we found the top 20 key genes according to the degree value by cytohubba, which regulated cell growth, skeletal formation and blood homeostasis, such as pik3ca, pik3r1, egfr, vegfa, igf1 and itga2b. Finally, we also acquired 19 key miRNAs playing multiple roles in the perfection of various tissues in the head (such as brain, eye and mouth) and mineralization of head bone system, such as let–7e, miR–142a–5p, miR–144–3p, miR–23a–3p and miR–223. Conclusions Results of this study will be informative for genetic mechanisms of head development and also provide potential candidate targets for the interaction regulation during early growth in bighead carp. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08387-x.
Collapse
Affiliation(s)
- Weiwei Luo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, Institute of Hydrobiology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Junru Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, Institute of Hydrobiology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Ying Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, Institute of Hydrobiology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Meixia Pang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, Institute of Hydrobiology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China.,Postdoctoral Innovation Practice Base, Shenzhen Polytechnic, Shenzhen, 518055, China
| | - Xiaomu Yu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, Institute of Hydrobiology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Jingou Tong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, Institute of Hydrobiology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
9
|
Chi SG, Minami Y. Emerging Targeted Therapy for Specific Genomic Abnormalities in Acute Myeloid Leukemia. Int J Mol Sci 2022; 23:2362. [PMID: 35216478 PMCID: PMC8879537 DOI: 10.3390/ijms23042362] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/12/2022] [Accepted: 02/17/2022] [Indexed: 11/17/2022] Open
Abstract
We describe recent updates of existing molecular-targeting agents and emerging novel gene-specific strategies. FLT3 and IDH inhibitors are being tested in combination with conventional chemotherapy for both medically fit patients and patients who are ineligible for intensive therapy. FLT3 inhibitors combined with non-cytotoxic agents, such as BCL-2 inhibitors, have potential therapeutic applicability. The menin-MLL complex pathway is an emerging therapeutic target. The pathway accounts for the leukemogenesis in AML with MLL-rearrangement, NPM1 mutation, and NUP98 fusion genes. Potent menin-MLL inhibitors have demonstrated promising anti-leukemic effects in preclinical studies. The downstream signaling molecule SYK represents an additional target. However, the TP53 mutation continues to remain a challenge. While the p53 stabilizer APR-246 in combination with azacitidine failed to show superiority compared to azacitidine monotherapy in a phase 3 trial, next-generation p53 stabilizers are now under development. Among a number of non-canonical approaches to TP53-mutated AML, the anti-CD47 antibody magrolimab in combination with azacitidine showed promising results in a phase 1b trial. Further, the efficacy was somewhat better in patients with the TP53 mutation. Although clinical evidence has not been accumulated sufficiently, targeting activating KIT mutations and RAS pathway-related molecules can be a future therapeutic strategy.
Collapse
Affiliation(s)
| | - Yosuke Minami
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 2778577, Japan;
| |
Collapse
|
10
|
Marega LF, Sabino JS, Pedroni MV, Teocchi M, Lanaro C, de Albuquerque DM, Dos Santos IP, Costa FF, Dos Santos Vilela MM. Phenotypes of STAT3 gain-of-function variant related to disruptive regulation of CXCL8/STAT3, KIT/STAT3, and IL-2/CD25/Treg axes. Immunol Res 2021; 69:445-456. [PMID: 34390446 DOI: 10.1007/s12026-021-09225-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/27/2021] [Indexed: 12/01/2022]
Abstract
STAT3 is a cytokine-signaling transcription factor critical for gene regulation. Gain-of-function (GOF) mutations in STAT3 are associated with lymphoproliferation, autoimmune cytopenias, increased susceptibility to infection, early-onset solid-organ autoimmunity, short stature, and eczema. We studied the JAK/STAT signaling pathway gene expression and the cytokine profile in two families carrying STAT3-GOF variants to shed light on the STAT3-GOF-associated variable expressivity, including the identification of disease markers. Considering 92 target genes, KIT and IL2RA were downregulated only in patients with high clinical penetrance, while CXCL8 was markedly downregulated for all of them. Unlike previous studies, SOCS3-a STAT3 inhibitor-was not upregulated in patients. In addition, low levels of IL-2 and a reduced numbers of Tregs cells were strikingly prevalent in patients. This study shows a disruptive role of STAT3-GOF variants in the regulatory axis activities CXCL8/STAT3, KIT/STAT3, IL2/CD25/Treg, which, by slightly different mechanisms, underlie the broad clinical spectrum seen in the studied patients. In addition, we suggest the investigation of CXCL8 as a biomarker for identifying STAT3-GOF mutation.
Collapse
Affiliation(s)
- Lia Furlaneto Marega
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Janine Schincariol Sabino
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Marcus Vinicius Pedroni
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Marcelo Teocchi
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Carolina Lanaro
- Hematology and Hemotherapy Center - Hemocentro Campinas, University of Campinas (UNICAMP), SP, Campinas, Brazil
| | | | - Irene Pereira Dos Santos
- Flow Cytometry Laboratory, Hematology and Hemotherapy Center, University of Campinas (UNICAMP), SP, Campinas, Brazil
| | - Fernando Ferreira Costa
- Hematology and Hemotherapy Center - Hemocentro Campinas, University of Campinas (UNICAMP), SP, Campinas, Brazil
| | - Maria Marluce Dos Santos Vilela
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil.
| |
Collapse
|
11
|
Nintedanib targets KIT D816V neoplastic cells derived from induced pluripotent stem cells of systemic mastocytosis. Blood 2021; 137:2070-2084. [PMID: 33512435 DOI: 10.1182/blood.2019004509] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 12/08/2020] [Indexed: 01/10/2023] Open
Abstract
The KIT D816V mutation is found in >80% of patients with systemic mastocytosis (SM) and is key to neoplastic mast cell (MC) expansion and accumulation in affected organs. Therefore, KIT D816V represents a prime therapeutic target for SM. Here, we generated a panel of patient-specific KIT D816V induced pluripotent stem cells (iPSCs) from patients with aggressive SM and mast cell leukemia to develop a patient-specific SM disease model for mechanistic and drug-discovery studies. KIT D816V iPSCs differentiated into neoplastic hematopoietic progenitor cells and MCs with patient-specific phenotypic features, thereby reflecting the heterogeneity of the disease. CRISPR/Cas9n-engineered KIT D816V human embryonic stem cells (ESCs), when differentiated into hematopoietic cells, recapitulated the phenotype observed for KIT D816V iPSC hematopoiesis. KIT D816V causes constitutive activation of the KIT tyrosine kinase receptor, and we exploited our iPSCs and ESCs to investigate new tyrosine kinase inhibitors targeting KIT D816V. Our study identified nintedanib, a US Food and Drug Administration-approved angiokinase inhibitor that targets vascular endothelial growth factor receptor, platelet-derived growth factor receptor, and fibroblast growth factor receptor, as a novel KIT D816V inhibitor. Nintedanib selectively reduced the viability of iPSC-derived KIT D816V hematopoietic progenitor cells and MCs in the nanomolar range. Nintedanib was also active on primary samples of KIT D816V SM patients. Molecular docking studies show that nintedanib binds to the adenosine triphosphate binding pocket of inactive KIT D816V. Our results suggest nintedanib as a new drug candidate for KIT D816V-targeted therapy of advanced SM.
Collapse
|
12
|
Liu M, Etherington MS, Hanna A, Medina BD, Vitiello GA, Bowler TG, Param NJ, Levin L, Rossi F, DeMatteo RP. Oncogenic KIT Modulates Type I IFN-Mediated Antitumor Immunity in GIST. Cancer Immunol Res 2021; 9:542-553. [PMID: 33648985 PMCID: PMC8102332 DOI: 10.1158/2326-6066.cir-20-0692] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/17/2020] [Accepted: 02/25/2021] [Indexed: 11/16/2022]
Abstract
Type I IFNs are implicated in tumor immunogenicity and response to systemic therapy, but their interaction with oncogene signaling is not well understood. Here, we studied oncogenic KIT, which drives gastrointestinal stromal tumor (GIST), the most common sarcoma. Using mouse models of GIST, we found that KIT inhibition reduced type I IFN production and signaling, which downregulated tumor MHC class I expression. Absence of type I IFN signaling increased tumor size, in part due to CD8+ T-cell impairment. Oncogenic KIT was required for GIST type I IFN signal transduction via STAT1. In human GIST cell lines and surgical specimens, type I IFN signaling contributed to human lymphocyte antigen class I expression and correlated with tumor immunogenicity. Augmenting the type I IFN response partially compensated for the immunosuppressive effects of KIT inhibition. Thus, KIT signaling contributes to type I IFN signaling, whereas KIT inhibition attenuates tumor immunogenicity and is partly rescued by innate immune stimulation.See related Spotlight on p. 489.
Collapse
Affiliation(s)
- Mengyuan Liu
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mark S Etherington
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew Hanna
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Benjamin D Medina
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gerardo A Vitiello
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Timothy G Bowler
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nesteene J Param
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lillian Levin
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ferdinand Rossi
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ronald P DeMatteo
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
13
|
Li J, Guo M, Tian X, Wang X, Yang X, Wu P, Liu C, Xiao Z, Qu Y, Yin Y, Wang C, Zhang Y, Zhu Z, Liu Z, Peng C, Zhu T, Liang Q. Virus-Host Interactome and Proteomic Survey Reveal Potential Virulence Factors Influencing SARS-CoV-2 Pathogenesis. MED 2021. [PMID: 32838362 DOI: 10.1101/2020.03.31.019216] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BACKGROUND The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is a global public health concern due to relatively easy person-to-person transmission and the current lack of effective antiviral therapy. However, the exact molecular mechanisms of SARS-CoV-2 pathogenesis remain largely unknown. METHODS Genome-wide screening was used to establish intraviral and viral-host interactomes. Quantitative proteomics was used to investigate the peripheral blood mononuclear cell (PBMC) proteome signature in COVID-19. FINDINGS We elucidated 286 host proteins targeted by SARS-CoV-2 and >350 host proteins that are significantly perturbed in COVID-19-derived PBMCs. This signature in severe COVID-19 PBMCs reveals a significant upregulation of cellular proteins related to neutrophil activation and blood coagulation, as well as a downregulation of proteins mediating T cell receptor signaling. From the interactome, we further identified that non-structural protein 10 interacts with NF-κB-repressing factor (NKRF) to facilitate interleukin-8 (IL-8) induction, which potentially contributes to IL-8-mediated chemotaxis of neutrophils and the overexuberant host inflammatory response observed in COVID-19 patients. CONCLUSIONS Our study not only presents a systematic examination of SARS-CoV-2-induced perturbation of host targets and cellular networks but it also reveals insights into the mechanisms by which SARS-CoV-2 triggers cytokine storms, representing a powerful resource in the pursuit of therapeutic interventions. FUNDING National Key Research and Development Project of China, National Natural Science Foundation of China, National Science and Technology Major Project, Program for Professor of Special Appointment (Eastern Scholar) at Shanghai Institutions of Higher Learning, Shanghai Science and Technology Commission, Shanghai Municipal Health Commission, Shanghai Municipal Key Clinical Specialty, Innovative Research Team of High-level Local Universities in Shanghai, Interdisciplinary Program of Shanghai Jiao Tong University, SII Challenge Fund for COVID-19 Research, Chinese Academy of Sciences (CAS) Large Research Infrastructure of Maintenance and Remolding Project, and Chinese Academy of Sciences Key Technology Talent Program.
Collapse
Affiliation(s)
- Jingjiao Li
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Mingquan Guo
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xiaoxu Tian
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Xin Wang
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xing Yang
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ping Wu
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Chengrong Liu
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zixuan Xiao
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Yafei Qu
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yue Yin
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Chunxia Wang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Yucai Zhang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Zhaoqin Zhu
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Zhenshan Liu
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Tongyu Zhu
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qiming Liang
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
14
|
The V654A second-site KIT mutation increases tumor oncogenesis and STAT activation in a mouse model of gastrointestinal stromal tumor. Oncogene 2020; 39:7153-7165. [PMID: 33024275 PMCID: PMC7718339 DOI: 10.1038/s41388-020-01489-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/19/2020] [Accepted: 09/24/2020] [Indexed: 01/02/2023]
Abstract
Gastrointestinal stromal tumor (GIST) is the most common human sarcoma and arises in the gastrointestinal tract. Most GISTs are caused by activating mutations in the KIT receptor tyrosine kinase, such as the exon 11 KIT V559Δ mutation. The small molecule imatinib inhibits KIT and has been a mainstay of therapy in GIST. Unfortunately, imatinib-treated patients typically relapse, most often due to clonal emergence of the resistance-associated KIT V654A mutation. To determine the biologic impact of this second-site mutation in vivo, we created a mouse model with the corresponding V558Δ;V653A Kit double mutation restricted (a) spatially to ETV1+ cells, which include the interstitial cells of Cajal (ICCs) from which GISTs presumably originate, and (b) temporally through tamoxifen treatment after birth. This resulted in the first in vivo model of the most common second-site mutation associated with imatinib resistance in GIST and the first in vivo demonstration that cell-autonomous expression of mutant KIT in the ICC lineage leads to GIST. GISTs driven by the V558Δ;V653A Kit double mutation were resistant to imatinib, while cabozantinib was more effective in overcoming resistance than sunitinib. Compared to control mice with a single V558Δ Kit mutation, mice with a double V558Δ; V653A Kit mutation had increased tumor oncogenesis and associated KIT-dependent STAT activation. Our findings demonstrate that the biologic consequences of a second-site mutation in an oncogenic driver may include not only a mechanism for drug resistance, but changes in tumor oncogenic potential and differential activation of signaling pathways.
Collapse
|
15
|
Voisset E, Brenet F, Lopez S, de Sepulveda P. SRC-Family Kinases in Acute Myeloid Leukaemia and Mastocytosis. Cancers (Basel) 2020; 12:cancers12071996. [PMID: 32708273 PMCID: PMC7409304 DOI: 10.3390/cancers12071996] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 11/16/2022] Open
Abstract
Protein tyrosine kinases have been recognized as important actors of cell transformation and cancer progression, since their discovery as products of viral oncogenes. SRC-family kinases (SFKs) play crucial roles in normal hematopoiesis. Not surprisingly, they are hyperactivated and are essential for membrane receptor downstream signaling in hematological malignancies such as acute myeloid leukemia (AML) and mastocytosis. The precise roles of SFKs are difficult to delineate due to the number of substrates, the functional redundancy among members, and the use of tools that are not selective. Yet, a large num ber of studies have accumulated evidence to support that SFKs are rational therapeutic targets in AML and mastocytosis. These two pathologies are regulated by two related receptor tyrosine kinases, which are well known in the field of hematology: FLT3 and KIT. FLT3 is one of the most frequently mutated genes in AML, while KIT oncogenic mutations occur in 80-90% of mastocytosis. Studies on oncogenic FLT3 and KIT signaling have shed light on specific roles for members of the SFK family. This review highlights the central roles of SFKs in AML and mastocytosis, and their interconnection with FLT3 and KIT oncoproteins.
Collapse
|
16
|
Li J, Guo M, Tian X, Wang X, Yang X, Wu P, Liu C, Xiao Z, Qu Y, Yin Y, Wang C, Zhang Y, Zhu Z, Liu Z, Peng C, Zhu T, Liang Q. Virus-Host Interactome and Proteomic Survey Reveal Potential Virulence Factors Influencing SARS-CoV-2 Pathogenesis. MED 2020; 2:99-112.e7. [PMID: 32838362 PMCID: PMC7373048 DOI: 10.1016/j.medj.2020.07.002] [Citation(s) in RCA: 223] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/27/2020] [Accepted: 07/15/2020] [Indexed: 02/05/2023]
Abstract
Background The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is a global public health concern due to relatively easy person-to-person transmission and the current lack of effective antiviral therapy. However, the exact molecular mechanisms of SARS-CoV-2 pathogenesis remain largely unknown. Methods Genome-wide screening was used to establish intraviral and viral-host interactomes. Quantitative proteomics was used to investigate the peripheral blood mononuclear cell (PBMC) proteome signature in COVID-19. Findings We elucidated 286 host proteins targeted by SARS-CoV-2 and >350 host proteins that are significantly perturbed in COVID-19-derived PBMCs. This signature in severe COVID-19 PBMCs reveals a significant upregulation of cellular proteins related to neutrophil activation and blood coagulation, as well as a downregulation of proteins mediating T cell receptor signaling. From the interactome, we further identified that non-structural protein 10 interacts with NF-κB-repressing factor (NKRF) to facilitate interleukin-8 (IL-8) induction, which potentially contributes to IL-8-mediated chemotaxis of neutrophils and the overexuberant host inflammatory response observed in COVID-19 patients. Conclusions Our study not only presents a systematic examination of SARS-CoV-2-induced perturbation of host targets and cellular networks but it also reveals insights into the mechanisms by which SARS-CoV-2 triggers cytokine storms, representing a powerful resource in the pursuit of therapeutic interventions. Funding National Key Research and Development Project of China, National Natural Science Foundation of China, National Science and Technology Major Project, Program for Professor of Special Appointment (Eastern Scholar) at Shanghai Institutions of Higher Learning, Shanghai Science and Technology Commission, Shanghai Municipal Health Commission, Shanghai Municipal Key Clinical Specialty, Innovative Research Team of High-level Local Universities in Shanghai, Interdisciplinary Program of Shanghai Jiao Tong University, SII Challenge Fund for COVID-19 Research, Chinese Academy of Sciences (CAS) Large Research Infrastructure of Maintenance and Remolding Project, and Chinese Academy of Sciences Key Technology Talent Program. Genome-wide screens identify 58 binary interactions between 29 SARS-CoV-2 proteins Virus-host interactome identifies 286 host targets for SARS-CoV-2 proteins Quantitative analysis depicts the overall proteome signature in COVID-19 PBMCs Nsp10 targets NKRF to facilitate IL-8 induction
The COVID-19 pandemic is caused by SARS-CoV-2, but little is known about the functions of its viral proteins. The authors characterized the SARS-CoV-2 intraviral and virus-host interaction networks in human cells and identified 286 potential host targets. Quantitative proteomic analysis revealed elevated levels of IL-6 and IL-8 in PBMCs collected from severe COVID-19 patients compared with mild ones, and the functional annotation of differentially expressed proteins implicate pathways involved in neutrophil activation, T cell receptor signaling, and the coagulation cascade. Combining virus-host interactome with COVID-19 proteomic analysis, the authors found that nsp10 interacts with NKRF to mediate IL-8 expression, providing a potential molecular mechanism for SARS-CoV-2-induced cytokine storm and marking it as a possible emerging therapeutic target.
Collapse
Affiliation(s)
- Jingjiao Li
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Mingquan Guo
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xiaoxu Tian
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Xin Wang
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xing Yang
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ping Wu
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Chengrong Liu
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zixuan Xiao
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Yafei Qu
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yue Yin
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Chunxia Wang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Yucai Zhang
- Department of Critical Care Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Zhaoqin Zhu
- Department of Laboratory Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Zhenshan Liu
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Tongyu Zhu
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qiming Liang
- Research Center of Translational Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
17
|
Brachet-Botineau M, Polomski M, Neubauer HA, Juen L, Hédou D, Viaud-Massuard MC, Prié G, Gouilleux F. Pharmacological Inhibition of Oncogenic STAT3 and STAT5 Signaling in Hematopoietic Cancers. Cancers (Basel) 2020; 12:E240. [PMID: 31963765 PMCID: PMC7016966 DOI: 10.3390/cancers12010240] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
Signal Transducer and Activator of Transcription (STAT) 3 and 5 are important effectors of cellular transformation, and aberrant STAT3 and STAT5 signaling have been demonstrated in hematopoietic cancers. STAT3 and STAT5 are common targets for different tyrosine kinase oncogenes (TKOs). In addition, STAT3 and STAT5 proteins were shown to contain activating mutations in some rare but aggressive leukemias/lymphomas. Both proteins also contribute to drug resistance in hematopoietic malignancies and are now well recognized as major targets in cancer treatment. The development of inhibitors targeting STAT3 and STAT5 has been the subject of intense investigations during the last decade. This review summarizes the current knowledge of oncogenic STAT3 and STAT5 functions in hematopoietic cancers as well as advances in preclinical and clinical development of pharmacological inhibitors.
Collapse
Affiliation(s)
- Marie Brachet-Botineau
- Leukemic Niche and Oxidative metabolism (LNOx), CNRS ERL 7001, University of Tours, 37000 Tours, France;
| | - Marion Polomski
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, A-1210 Vienna, Austria;
| | - Ludovic Juen
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Damien Hédou
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Marie-Claude Viaud-Massuard
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Gildas Prié
- Innovation Moléculaire et Thérapeutique (IMT), EA 7501, University of Tours, 37000 Tours, France; (M.P.); (L.J.); (D.H.); (M.-C.V.-M.); (G.P.)
| | - Fabrice Gouilleux
- Leukemic Niche and Oxidative metabolism (LNOx), CNRS ERL 7001, University of Tours, 37000 Tours, France;
| |
Collapse
|
18
|
Kieslinger M, Swoboda A, Kramer N, Pratscher B, Wolfesberger B, Burgener IA. Companion Animals as Models for Inhibition of STAT3 and STAT5. Cancers (Basel) 2019; 11:cancers11122035. [PMID: 31861073 PMCID: PMC6966487 DOI: 10.3390/cancers11122035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/28/2022] Open
Abstract
The use of transgenic mouse models has revolutionized the study of many human diseases. However, murine models are limited in their representation of spontaneously arising tumors and often lack key clinical signs and pathological changes. Thus, a closer representation of complex human diseases is of high therapeutic relevance. Given the high failure rate of drugs at the clinical trial phase (i.e., around 90%), there is a critical need for additional clinically relevant animal models. Companion animals like cats and dogs display chronic inflammatory or neoplastic diseases that closely resemble the human counterpart. Cat and dog patients can also be treated with clinically approved inhibitors or, if ethics and drug safety studies allow, pilot studies can be conducted using, e.g., inhibitors of the evolutionary conserved JAK-STAT pathway. The incidence by which different types of cancers occur in companion animals as well as mechanisms of disease are unique between humans and companion animals, where one can learn from each other. Taking advantage of this situation, existing inhibitors of known oncogenic STAT3/5 or JAK kinase signaling pathways can be studied in the context of rare human diseases, benefitting both, the development of drugs for human use and their application in veterinary medicine.
Collapse
|
19
|
Hazzan T, Eberle J, Worm M, Babina M. Thymic Stromal Lymphopoietin Interferes with the Apoptosis of Human Skin Mast Cells by a Dual Strategy Involving STAT5/Mcl-1 and JNK/Bcl-x L. Cells 2019; 8:E829. [PMID: 31387206 PMCID: PMC6721763 DOI: 10.3390/cells8080829] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/29/2019] [Accepted: 08/01/2019] [Indexed: 12/21/2022] Open
Abstract
Mast cells (MCs) play critical roles in allergic and inflammatory reactions and contribute to multiple pathologies in the skin, in which they show increased numbers, which frequently correlates with severity. It remains ill-defined how MC accumulation is established by the cutaneous microenvironment, in part because research on human MCs rarely employs MCs matured in the tissue, and extrapolations from other MC subsets have limitations, considering the high level of MC heterogeneity. Thymic stromal lymphopoietin (TSLP)-released by epithelial cells, like keratinocytes, following disturbed homeostasis and inflammation-has attracted much attention, but its impact on skin MCs remains undefined, despite the vast expression of the TSLP receptor by these cells. Using several methods, each detecting a distinct component of the apoptotic process (membrane alterations, DNA degradation, and caspase-3 activity), our study pinpoints TSLP as a novel survival factor of dermal MCs. TSLP confers apoptosis resistance via concomitant activation of the TSLP/ signal transducer and activator of transcription (STAT)-5 / myeloid cell leukemia (Mcl)-1 route and a newly uncovered TSLP/ c-Jun-N-terminal kinase (JNK)/ B-cell lymphoma (Bcl)-xL axis, as evidenced by RNA interference and pharmacological inhibition. Our findings highlight the potential contribution of TSLP to the MC supportive niche of the skin and, vice versa, highlight MCs as crucial responders to TSLP in the context of TSLP-driven disorders.
Collapse
Affiliation(s)
- Tarek Hazzan
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Eberle
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Margitta Worm
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | - Magda Babina
- Department of Dermatology, Venerology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
20
|
Oncogenic KIT mutations induce STAT3-dependent autophagy to support cell proliferation in acute myeloid leukemia. Oncogenesis 2019; 8:39. [PMID: 31311917 PMCID: PMC6635375 DOI: 10.1038/s41389-019-0148-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 04/10/2019] [Accepted: 05/31/2019] [Indexed: 12/14/2022] Open
Abstract
Autophagy is associated with both survival and cell death in myeloid malignancies. Therefore, deciphering its role in different genetically defined subtypes of acute myeloid leukemia (AML) is critical. Activating mutations of the KIT receptor tyrosine kinase are frequently detected in core-binding factor AML and are associated with a greater risk of relapse. Herein, we report that basal autophagy was significantly increased by the KITD816V mutation in AML cells and contributed to support their cell proliferation and survival. Invalidation of the key autophagy protein Atg12 strongly reduced tumor burden and improved survival of immunocompromised NSG mice engrafted with KITD816V TF-1 cells. Downstream of KITD816V, STAT3, but not AKT or ERK pathways, was identified as a major regulator of autophagy. Accordingly, STAT3 pharmacological inhibition or downregulation inhibited autophagy and reduced tumor growth both in vitro and in vivo. Taken together, our results support the notion that targeting autophagy or STAT3 opens up an exploratory pathway for finding new therapeutic opportunities for patients with CBF-AML or others malignancies with KITD816V mutations.
Collapse
|
21
|
The upregulation of Pim kinases is essential in coordinating the survival, proliferation, and migration of KIT D816V-mutated neoplastic mast cells. Leuk Res 2019; 83:106166. [PMID: 31203104 DOI: 10.1016/j.leukres.2019.106166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/19/2019] [Accepted: 06/06/2019] [Indexed: 11/23/2022]
Abstract
About ˜80% of mast cell neoplasm patients harbor the c-Kit activating mutation D816 V, which is associated with c-Kit inhibitor resistance and poor prognosis. However, the molecular basis for these effects is not fully known. To address this issue, in this study we screened molecules whose expression is altered by KIT D816 V mutation and found that Pim kinases were overexpressed in D816V-mutant neoplastic mast cells. This was accompanied by upregulation of signal transducer and activator of transcription (STAT) and mammalian target of rapamycin (mTOR) and downregulation of Akt and extracellular signal-regulated kinase (ERK1/2). Activated Pim kinases promoted the survival of D816 V cells by maintaining mTOR and p70S6K activation even under nutrient starvation. Conversely, cell proliferation was suppressed by inhibiting Pim kinases. The mRNA level of C-X-C chemokine receptor type 4 (CXCR4) was about 2-fold higher in D816 V cells; this was associated with a 2-fold increase in migratory capacity, which was modulated by Pim kinases. We also confirmed that upregulation of Pim kinases is a feature specific to cells with the D816 V mutation and is not observed in cells with the c-Kit activating N822 K mutation. These data suggest Pim kinases as a promising therapeutic target for the treatment of mast cell neoplasms with KIT D816 V mutation.
Collapse
|
22
|
Pearson S, Blance R, Somervaille TC, Whetton AD, Pierce A. AXL Inhibition Extinguishes Primitive JAK2 Mutated Myeloproliferative Neoplasm Progenitor Cells. Hemasphere 2019; 3:e233. [PMID: 31723838 PMCID: PMC6746025 DOI: 10.1097/hs9.0000000000000233] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/25/2019] [Accepted: 03/25/2019] [Indexed: 12/13/2022] Open
Abstract
Myeloproliferative neoplasms (MPN) are clonal stem cell associated disorders inclusive of chronic myeloid leukemia (CML), Polycythaemia vera (PV), myelofibrosis (MF), and essential thrombocythemia (ET). They are characterized by increased production of myeloid cells with minimal effects on terminal differentiation but can undergo transformation to acute leukemias. PV is the most common chronic myeloproliferative neoplasm and in the majority of cases is characterized by a V617F point mutation in JAK2. This JAK2 activating mutation is also found in about half the patients with MF and ET. Such aberrant proteins offer great potential for the treatment of these diseases however inhibitors to JAK2 have had limited success in the clinic in terms of curing the disease. We have previously used advanced proteomic techniques to identify drug targets and thus develop novel treatment strategies to distinguish the leukemic clone in both CML and PV. Here, we build on our proteomic data sets to characterize a new target, the receptor tyrosine kinase AXL. AXL is overexpressed in acute myeloid leukemia and importantly small molecule inhibitors have been developed which are currently in clinical trial hence offer the opportunity to repurpose this drug for the treatment of MPNs. We demonstrate that AXL is upregulated and activated in JAK2 associated MPNs. Further we show that inhibition of AXL preferentially kills early hemopoietic stem cells from PV patients and as such represents a promising therapeutic approach for JAK2 driven MPNs.
Collapse
Affiliation(s)
- Stella Pearson
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, The University of Manchester, UK
| | - Rognvald Blance
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, The University of Manchester, UK
| | | | - Anthony D. Whetton
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, The University of Manchester, UK
- Stoller Biomarker Discovery Centre, The University of Manchester, UK
| | - Andrew Pierce
- Stem Cell and Leukaemia Proteomics Laboratory, Manchester Academic Health Science Centre, The University of Manchester, UK
| |
Collapse
|
23
|
Youn A, Kim KI, Rabadan R, Tycko B, Shen Y, Wang S. A pan-cancer analysis of driver gene mutations, DNA methylation and gene expressions reveals that chromatin remodeling is a major mechanism inducing global changes in cancer epigenomes. BMC Med Genomics 2018; 11:98. [PMID: 30400878 PMCID: PMC6218985 DOI: 10.1186/s12920-018-0425-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 10/19/2018] [Indexed: 01/08/2023] Open
Abstract
Background Recent large-scale cancer sequencing studies have discovered many novel cancer driver genes (CDGs) in human cancers. Some studies also suggest that CDG mutations contribute to cancer-associated epigenomic and transcriptomic alterations across many cancer types. Here we aim to improve our understanding of the connections between CDG mutations and altered cancer cell epigenomes and transcriptomes on pan-cancer level and how these connections contribute to the known association between epigenome and transcriptome. Method Using multi-omics data including somatic mutation, DNA methylation, and gene expression data of 20 cancer types from The Cancer Genome Atlas (TCGA) project, we conducted a pan-cancer analysis to identify CDGs, when mutated, have strong associations with genome-wide methylation or expression changes across cancer types, which we refer as methylation driver genes (MDGs) or expression driver genes (EDGs), respectively. Results We identified 32 MDGs, among which, eight are known chromatin modification or remodeling genes. Many of the remaining 24 MDGs are connected to chromatin regulators through either regulating their transcription or physically interacting with them as potential co-factors. We identified 29 EDGs, 26 of which are also MDGs. Further investigation on target genes’ promoters methylation and expression alteration patterns of these 26 overlapping driver genes shows that hyper-methylation of target genes’ promoters are significantly associated with down-regulation of the same target genes and hypo-methylation of target genes’ promoters are significantly associated with up-regulation of the same target genes. Conclusion This finding suggests a pivotal role for genetically driven changes in chromatin remodeling in shaping DNA methylation and gene expression patterns during tumor development. Electronic supplementary material The online version of this article (10.1186/s12920-018-0425-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ahrim Youn
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York, USA.,The Jackson Laboratory For Genomic Medicine, Farmington, Connecticut, USA
| | - Kyung In Kim
- The Jackson Laboratory For Genomic Medicine, Farmington, Connecticut, USA
| | - Raul Rabadan
- Department of System Biology, Columbia University, New York, New York, USA.,Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | - Benjamin Tycko
- Division of Genetics & Epigenetics, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Yufeng Shen
- Department of System Biology, Columbia University, New York, New York, USA.,Department of Biomedical Informatics, Columbia University, New York, New York, USA.,Columbia Genome Center, Columbia University, New York, New York, USA
| | - Shuang Wang
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York, USA.
| |
Collapse
|
24
|
Arock M, Wedeh G, Hoermann G, Bibi S, Akin C, Peter B, Gleixner KV, Hartmann K, Butterfield JH, Metcalfe DD, Valent P. Preclinical human models and emerging therapeutics for advanced systemic mastocytosis. Haematologica 2018; 103:1760-1771. [PMID: 29976735 PMCID: PMC6278969 DOI: 10.3324/haematol.2018.195867] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/27/2018] [Indexed: 12/19/2022] Open
Abstract
Mastocytosis is a term used to denote a group of rare diseases characterized by an abnormal accumulation of neoplastic mast cells in various tissues and organs. In most patients with systemic mastocytosis, the neoplastic cells carry activating mutations in KIT Progress in mastocytosis research has long been hindered by the lack of suitable in vitro models, such as permanent human mast cell lines. In fact, only a few human mast cell lines are available to date: HMC-1, LAD1/2, LUVA, ROSA and MCPV-1. The HMC-1 and LAD1/2 cell lines were derived from patients with mast cell leukemia. By contrast, the more recently established LUVA, ROSA and MCPV-1 cell lines were derived from CD34+ cells of non-mastocytosis donors. While some of these cell lines (LAD1/2, LUVA, ROSAKIT WT and MCPV-1) do not harbor KIT mutations, HMC-1 and ROSAKIT D816V cells exhibit activating KIT mutations found in mastocytosis and have thus been used to study disease pathogenesis. In addition, these cell lines are increasingly employed to validate new therapeutic targets and to screen for effects of new targeted drugs. Recently, the ROSAKIT D816V subclone has been successfully used to generate a unique in vivo model of advanced mastocytosis by injection into immunocompromised mice. Such a model may allow in vivo validation of data obtained in vitro with targeted drugs directed against mastocytosis. In this review, we discuss the major characteristics of all available human mast cell lines, with particular emphasis on the use of HMC-1 and ROSAKIT D816V cells in preclinical therapeutic research in mastocytosis.
Collapse
Affiliation(s)
- Michel Arock
- LBPA CNRS UMR8113, Ecole Normale Supérieure Paris-Saclay, Cachan, France .,Laboratory of Hematology, Pitié-Salpêtrière Hospital, Paris, France
| | - Ghaith Wedeh
- LBPA CNRS UMR8113, Ecole Normale Supérieure Paris-Saclay, Cachan, France
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria
| | - Siham Bibi
- LBPA CNRS UMR8113, Ecole Normale Supérieure Paris-Saclay, Cachan, France
| | - Cem Akin
- Michigan Medicine Allergy Clinic, University of Michigan, Ann Arbor, MI, USA
| | - Barbara Peter
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Karoline V Gleixner
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Karin Hartmann
- Department of Dermatology, University of Luebeck, Germany
| | | | - Dean D Metcalfe
- Laboratory of Allergic Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Peter Valent
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| |
Collapse
|
25
|
Liu L, Zuo X, Zhu Z, Wen L, Yang C, Zhu C, Tang L, Cheng Y, Chen M, Zhou F, Zheng X, Wang W, Yin X, Tang H, Sun L, Yang S, Sheng Y, Cui Y, Zhang X. Genome-wide association study identifies three novel susceptibility loci for systemic lupus erythematosus in Han Chinese. Br J Dermatol 2018; 179:506-508. [PMID: 29494758 DOI: 10.1111/bjd.16500] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- L Liu
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.,Institute of Dermatology and Department of Dermatology, Huashan Hospital of Fudan University, Shanghai, 200040, China
| | - X Zuo
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Z Zhu
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - L Wen
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - C Yang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - C Zhu
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - L Tang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Y Cheng
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - M Chen
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - F Zhou
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - X Zheng
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - W Wang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - X Yin
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.,Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, 48109, U.S.A
| | - H Tang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - L Sun
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - S Yang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Y Sheng
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Y Cui
- Department of Dermatology, China-Japan Friendship Hospital, East Street Cherry Park, Chaoyang District, Beijing, 100029, China
| | - X Zhang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, 81 Meishan Road, Hefei, Anhui, 230032, China.,Key Laboratory of Dermatology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.,Institute of Dermatology and Department of Dermatology, Huashan Hospital of Fudan University, Shanghai, 200040, China
| |
Collapse
|
26
|
Yim S, Yu H, Jang D, Lee D. Annotating activation/inhibition relationships to protein-protein interactions using gene ontology relations. BMC SYSTEMS BIOLOGY 2018; 12:9. [PMID: 29671402 PMCID: PMC5907154 DOI: 10.1186/s12918-018-0535-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Signaling pathways can be reconstructed by identifying ‘effect types’ (i.e. activation/inhibition) of protein-protein interactions (PPIs). Effect types are composed of ‘directions’ (i.e. upstream/downstream) and ‘signs’ (i.e. positive/negative), thereby requiring directions as well as signs of PPIs to predict signaling events from PPI networks. Here, we propose a computational method for systemically annotating effect types to PPIs using relations between functional information of proteins. Results We used regulates, positively regulates, and negatively regulates relations in Gene Ontology (GO) to predict directions and signs of PPIs. These relations indicate both directions and signs between GO terms so that we can project directions and signs between relevant GO terms to PPIs. Independent test results showed that our method is effective for predicting both directions and signs of PPIs. Moreover, our method outperformed a previous GO-based method that did not consider the relations between GO terms. We annotated effect types to human PPIs and validated several highly confident effect types against literature. The annotated human PPIs are available in Additional file 2 to aid signaling pathway reconstruction and network biology research. Conclusions We annotated effect types to PPIs by using regulates, positively regulates, and negatively regulates relations in GO. We demonstrated that those relations are effective for predicting not only signs, but also directions of PPIs. The usefulness of those relations suggests their potential applications to other types of interactions such as protein-DNA interactions. Electronic supplementary material The online version of this article (10.1186/s12918-018-0535-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Soorin Yim
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.,Bio-Synergy Research Center, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hasun Yu
- Bio-Synergy Research Center, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Dongjin Jang
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea. .,Bio-Synergy Research Center, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
27
|
Lopez S, Voisset E, Tisserand JC, Mosca C, Prebet T, Santamaria D, Dubreuil P, De Sepulveda P. An essential pathway links FLT3-ITD, HCK and CDK6 in acute myeloid leukemia. Oncotarget 2018; 7:51163-51173. [PMID: 27323399 PMCID: PMC5239466 DOI: 10.18632/oncotarget.9965] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 05/25/2016] [Indexed: 11/25/2022] Open
Abstract
CDK4/CDK6 and RB proteins drive the progression through the G1 phase of the cell cycle. In acute myeloid leukemia (AML), the activity of the CDK/Cyclin D complex is increased. The mechanism involved is unknown, as are the respective roles played by CDK4 or CDK6 in this process. Here, we report that AML cells carrying FLT3-ITD mutations are dependent on CDK6 for cell proliferation while CDK4 is not essential. We showed that FLT3-ITD signaling is responsible for CDK6 overexpression, through a pathway involving the SRC-family kinase HCK. Accordingly, FLT3-ITD failed to transform primary hematopoietic progenitor cells from Cdk6-/- mice. Our results demonstrate that CDK6 is the primary target of CDK4/CDK6 inhibitors in FLT3-ITD positive AML. Furthermore, we delineate an essential protein kinase pathway -FLT3/HCK/CDK6- in the context of AML with FLT3-ITD mutations.
Collapse
Affiliation(s)
- Sophie Lopez
- Inserm, Cancer Research Center of Marseille (CRCM), U1068, Marseille, France.,Institut Paoli-Calmettes (IPC), Marseille, France.,Aix-Marseille University, UM 105, Marseille, France.,CNRS, UMR7258, Marseille, France
| | - Edwige Voisset
- Inserm, Cancer Research Center of Marseille (CRCM), U1068, Marseille, France.,Institut Paoli-Calmettes (IPC), Marseille, France.,Aix-Marseille University, UM 105, Marseille, France.,CNRS, UMR7258, Marseille, France.,Present address: Department of Medical and Molecular Genetics, King's College London, Faculty of Life Sciences and Medicine, London, United Kingdom
| | - Julie C Tisserand
- Inserm, Cancer Research Center of Marseille (CRCM), U1068, Marseille, France.,Institut Paoli-Calmettes (IPC), Marseille, France.,Aix-Marseille University, UM 105, Marseille, France.,CNRS, UMR7258, Marseille, France
| | - Cyndie Mosca
- Inserm, Cancer Research Center of Marseille (CRCM), U1068, Marseille, France.,Institut Paoli-Calmettes (IPC), Marseille, France.,Aix-Marseille University, UM 105, Marseille, France.,CNRS, UMR7258, Marseille, France
| | | | | | - Patrice Dubreuil
- Inserm, Cancer Research Center of Marseille (CRCM), U1068, Marseille, France.,Institut Paoli-Calmettes (IPC), Marseille, France.,Aix-Marseille University, UM 105, Marseille, France.,CNRS, UMR7258, Marseille, France
| | - Paulo De Sepulveda
- Inserm, Cancer Research Center of Marseille (CRCM), U1068, Marseille, France.,Institut Paoli-Calmettes (IPC), Marseille, France.,Aix-Marseille University, UM 105, Marseille, France.,CNRS, UMR7258, Marseille, France
| |
Collapse
|
28
|
The role of regulatory T cells and genes involved in their differentiation in pathogenesis of selected inflammatory and neoplastic skin diseases. Part III: Polymorphisms of genes involved in Tregs' activation and function. Postepy Dermatol Alergol 2017; 34:517-525. [PMID: 29422815 PMCID: PMC5799752 DOI: 10.5114/pdia.2017.67053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/10/2017] [Indexed: 12/29/2022] Open
Abstract
Regulatory T cells (Tregs) represent a cell type that promotes immune tolerance to autologous components and maintains immune system homeostasis. The abnormal function of Tregs is relevant to the pathogenesis of several skin diseases like psoriasis, atopic dermatitis, systemic lupus erythematosus, cutaneous T-cell lymphomas, and skin cancer and is also important in rheumatoid arthritis, diabetes and other autoimmune diseases. In this review, we will summarize the role of mutations and/or polymorphisms of genes involved in Tregs development, and functions in the pathogenesis of selected skin diseases.
Collapse
|
29
|
Peter B, Bibi S, Eisenwort G, Wingelhofer B, Berger D, Stefanzl G, Blatt K, Herrmann H, Hadzijusufovic E, Hoermann G, Hoffmann T, Schwaab J, Jawhar M, Willmann M, Sperr WR, Zuber J, Sotlar K, Horny HP, Moriggl R, Reiter A, Arock M, Valent P. Drug-induced inhibition of phosphorylation of STAT5 overrides drug resistance in neoplastic mast cells. Leukemia 2017; 32:1016-1022. [PMID: 29249817 PMCID: PMC6037300 DOI: 10.1038/leu.2017.338] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/31/2017] [Accepted: 11/13/2017] [Indexed: 12/20/2022]
Abstract
Systemic mastocytosis (SM) is a mast cell (MC) neoplasm with complex pathology and a variable clinical course. In aggressive SM (ASM) and MC leukemia (MCL) responses to conventional drugs are poor and the prognosis is dismal. R763 is a multi-kinase inhibitor that blocks the activity of Aurora-kinase-A/B, ABL1, AKT and FLT3. We examined the effects of R763 on proliferation and survival of neoplastic MC. R763 produced dose-dependent inhibition of proliferation in the human MC lines HMC-1.1 (IC50 5-50 nM), HMC-1.2 (IC50 1-10 nM), ROSAKIT WT (IC50 1-10 nM), ROSAKIT D816V (IC50 50-500 nM) and MCPV-1.1 (IC50 100-1000 nM). Moreover, R763 induced growth inhibition in primary neoplastic MC in patients with ASM and MCL. Growth-inhibitory effects of R763 were accompanied by signs of apoptosis and a G2/M cell cycle arrest. R763 also inhibited phosphorylation of KIT, BTK, AKT and STAT5 in neoplastic MC. The most sensitive target appeared to be STAT5. In fact, tyrosine phosphorylation of STAT5 was inhibited by R763 at 10 nM. At this low concentration, R763 produced synergistic growth-inhibitory effects on neoplastic MC when combined with midostaurin or dasatinib. Together, R763 is a novel promising multi-kinase inhibitor that blocks STAT5 activation and thereby overrides drug-resistance in neoplastic MC.
Collapse
Affiliation(s)
- B Peter
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - S Bibi
- Laboratoire de Biologie et Pharmacologie Appliquée, CNRS UMR 8113, Ecole Normale Superieure de Cachan, Cachan, France
| | - G Eisenwort
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - B Wingelhofer
- Ludwig Boltzmann Institute for Cancer Research, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Medical University of Vienna, Vienna, Austria
| | - D Berger
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - G Stefanzl
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - K Blatt
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - H Herrmann
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | - E Hadzijusufovic
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria.,Department for Companion Animals and Horses, Clinical Unit of Internal Medicine, University of Veterinary Medicine Vienna, Vienna, Austria
| | - G Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna,Austria
| | - T Hoffmann
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - J Schwaab
- Department of Hematology and Oncology, University Medical Center Mannheim and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - M Jawhar
- Department of Hematology and Oncology, University Medical Center Mannheim and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - M Willmann
- Department for Companion Animals and Horses, Clinical Unit of Internal Medicine, University of Veterinary Medicine Vienna, Vienna, Austria
| | - W R Sperr
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - J Zuber
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - K Sotlar
- University Institute of Pathology, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - H-P Horny
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - R Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Medical University of Vienna, Vienna, Austria
| | - A Reiter
- Department of Hematology and Oncology, University Medical Center Mannheim and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - M Arock
- Laboratoire de Biologie et Pharmacologie Appliquée, CNRS UMR 8113, Ecole Normale Superieure de Cachan, Cachan, France.,Laboratory of Hematology, Pitié-Salpêtrière Hospital, Paris, France
| | - P Valent
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
30
|
Obata Y, Horikawa K, Shiina I, Takahashi T, Murata T, Tasaki Y, Suzuki K, Yonekura K, Esumi H, Nishida T, Abe R. Oncogenic Kit signalling on the Golgi is suppressed by blocking secretory trafficking with M-COPA in gastrointestinal stromal tumours. Cancer Lett 2017; 415:1-10. [PMID: 29196126 DOI: 10.1016/j.canlet.2017.11.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/14/2017] [Accepted: 11/23/2017] [Indexed: 02/08/2023]
Abstract
Most gastrointestinal stromal tumours (GISTs) are caused by constitutively active mutations in Kit tyrosine kinase. The drug imatinib, a specific Kit inhibitor, improves the prognosis of metastatic GIST patients, but these patients become resistant to the drug by acquiring secondary mutations in the Kit kinase domain. We recently reported that a Kit mutant causes oncogenic signals only on the Golgi apparatus in GISTs. In this study, we show that in GIST, 2-methylcoprophilinamide (M-COPA, also known as "AMF-26"), an inhibitor of biosynthetic protein trafficking from the endoplasmic reticulum (ER) to the Golgi, suppresses Kit autophosphorylation at Y703/Y721/Y730/Y936, resulting in blockade of oncogenic signalling. Results of our M-COPA treatment assay show that Kit Y703/Y730/Y936 in the ER are dephosphorylated by protein tyrosine phosphatases (PTPs), thus the ER-retained Kit is unable to activate downstream molecules. ER-localized Kit Y721 is not phosphorylated, but not due to PTPs. Importantly, M-COPA can inhibit the activation of the Kit kinase domain mutant, resulting in suppression of imatinib-resistant GIST proliferation. Our study demonstrates that Kit autophosphorylation is spatio-temporally regulated and may offer a new strategy for treating imatinib-resistant GISTs.
Collapse
Affiliation(s)
- Yuuki Obata
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda 278-0022, Chiba, Japan
| | - Keita Horikawa
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda 278-0022, Chiba, Japan
| | - Isamu Shiina
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Shinjuku-ku 162-8601, Tokyo, Japan
| | - Tsuyoshi Takahashi
- Department of Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Osaka, Japan
| | - Takatsugu Murata
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Shinjuku-ku 162-8601, Tokyo, Japan
| | - Yasutaka Tasaki
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Shinjuku-ku 162-8601, Tokyo, Japan
| | - Kyohei Suzuki
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Shinjuku-ku 162-8601, Tokyo, Japan
| | - Keita Yonekura
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Shinjuku-ku 162-8601, Tokyo, Japan
| | - Hiroyasu Esumi
- Division of Clinical Research, Research Institute for Biomedical Sciences, Tokyo, University of Science, Japan
| | - Toshirou Nishida
- National Cancer Center Hospital, Chuo-ku, 104-0045, Tokyo, Japan
| | - Ryo Abe
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda 278-0022, Chiba, Japan.
| |
Collapse
|
31
|
Direct engagement of the PI3K pathway by mutant KIT dominates oncogenic signaling in gastrointestinal stromal tumor. Proc Natl Acad Sci U S A 2017; 114:E8448-E8457. [PMID: 28923937 DOI: 10.1073/pnas.1711449114] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) predominantly harbor activating mutations in the receptor tyrosine kinase KIT. To genetically dissect in vivo the requirement of different signal transduction pathways emanating from KIT for tumorigenesis, the oncogenic KitV558Δ mutation was combined with point mutations abrogating specific phosphorylation sites on KIT. Compared with single-mutant KitV558Δ/+ mice, double-mutant KitV558Δ;Y567F/Y567F knock-in mice lacking the SRC family kinase-binding site on KIT (pY567) exhibited attenuated MAPK signaling and tumor growth. Surprisingly, abrogation of the PI3K-binding site (pY719) in KitV558Δ;Y719F/Y719F mice prevented GIST development, although the interstitial cells of Cajal (ICC), the cells of origin of GIST, were normal. Pharmacologic inhibition of the PI3K pathway in tumor-bearing KitV558Δ/+ mice with the dual PI3K/mTOR inhibitor voxtalisib, the pan-PI3K inhibitor pilaralisib, and the PI3K-alpha-restricted inhibitor alpelisib each diminished tumor proliferation. The addition of the MEK inhibitor PD-325901 or binimetinib further decreased downstream KIT signaling. Moreover, combining PI3K and MEK inhibition was effective against imatinib-resistant KitV558Δ;T669I/+ tumors.
Collapse
|
32
|
Delyon J, Chevret S, Jouary T, Dalac S, Dalle S, Guillot B, Arnault JP, Avril MF, Bedane C, Bens G, Pham-Ledard A, Mansard S, Grange F, Machet L, Meyer N, Legoupil D, Saiag P, Idir Z, Renault V, Deleuze JF, Hindie E, Battistella M, Dumaz N, Mourah S, Lebbe C. STAT3 Mediates Nilotinib Response in KIT-Altered Melanoma: A Phase II Multicenter Trial of the French Skin Cancer Network. J Invest Dermatol 2017; 138:58-67. [PMID: 28843487 DOI: 10.1016/j.jid.2017.07.839] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/18/2017] [Accepted: 07/14/2017] [Indexed: 01/30/2023]
Abstract
Mutated oncogenic KIT is a therapeutic target in melanoma. We conducted a multicenter phase II trial on the KIT inhibitor nilotinib in patients with unresectable melanoma harboring KIT alteration. The primary endpoint was the response rate (complete response or partial response following Response Evaluation Criteria in Solid Tumors criteria) at 6 months. Pharmacodynamic studies using KIT sequencing, qPCR array, and immunostaining of downstream KIT effectors were performed during treatment. Twenty-five patients were included and received 400 mg oral nilotinib twice daily. At 6 months, nilotinib induced tumor response in four patients. The best overall response rate was 20% and the disease control rate was 56%, limited to patients harboring exon 11 or 13 mutations. Four patients exhibited durable response, including three persisting (3.6 and 2.8 years for two patients with stage IIIC and 2.5 years for one with IVM1b melanoma). A reduction in signal transducer and activator of transcription (STAT) 3 phosphorylation and its effectors (BCL-2, MCL-1) in tumors during follow-up was significantly associated with clinical response. In the KIT-mutated melanoma cell line M230, nilotinib reduced STAT3 signaling and STAT inhibitors were as efficient as KIT inhibitors in reducing cell proliferation. Our study evidences a significant association between STAT3 inhibition and response to nilotinib, and provides a rationale for future research assessing STAT inhibitors in KIT-mutated melanoma.
Collapse
Affiliation(s)
- Julie Delyon
- Service de Dermatologie, and CIC (Centre d'Investigations Cliniques), AP-HP, Hôpital Saint-Louis, Paris, France; INSERM, UMR-976, AP-HP, Hôpital Saint-Louis, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France.
| | - Sylvie Chevret
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Service de Biostatistique et Information Médicale, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Thomas Jouary
- Unité Onco-dermatologie, Hôpital François Mitterrand, Pau, France
| | - Sophie Dalac
- Service de Dermatologie, CHU Dijon Bourgogne, Dijon, France
| | - Stephane Dalle
- Cancer Research Center of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard Lyon 1 University, Institut de Cancérologie des Hospices Civils de Lyon, Lyon, France
| | | | | | - Marie-Françoise Avril
- Service de Dermatologie, AP-HP, Hôpital Cochin, Paris, France; Université Paris Descartes, Paris, France
| | - Christophe Bedane
- Unité d'oncologie thoracique et cutanée, Hopital Dupuytren, Limoges, France
| | - Guido Bens
- Service de Dermatologie, Centre hospitalier régional d'Orléans, Orléans, France
| | | | - Sandrine Mansard
- Dermatology Department, CHU de Clermont Ferrand, Clermont Ferrand, France
| | - Florent Grange
- Dermatology Department, Reims University Hospital, Reims, France
| | - Laurent Machet
- Department of Dermatology, Centre Hospitalier Regional et Universitaire (CHRU) de Tours, Tours, France; Inserm U930, University Francois Rabelais de Tours, Tours, France
| | - Nicolas Meyer
- Dermatologie, Institut Universitaire du Cancer et CHU de Toulouse, Toulouse, France; Inserm UMR 1037, CRCT, Toulouse, France
| | | | - Philippe Saiag
- Université de Versailles St-Quentin, EA 4340, Boulogne-Billancourt, France; Service de Dermatologie Générale et Oncologique, AP-HP, Hôpital Ambroise Paré, Boulogne-Billancourt, France
| | - Zakia Idir
- AP-HP, Département de la Recherche Clinique et du Développement, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Victor Renault
- Laboratory for Bioinformatics, CEPH-Fondation Jean Dausset, Paris, France
| | - Jean-François Deleuze
- Centre National de Génotypage, CEA, Evry, France; CEPH-Fondation Jean Dausset, Paris, France
| | - Elif Hindie
- Service de Médecine Nucléaire, CHU de Bordeaux, LabEx TRAIL, Université de Bordeaux, Bordeaux, France
| | - Maxime Battistella
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France; INSERM, UMR_S1165, Paris, France; Pathology department, Hopital Saint-Louis, AP-HP, Paris, France
| | - Nicolas Dumaz
- INSERM, UMR-976, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Samia Mourah
- INSERM, UMR-976, AP-HP, Hôpital Saint-Louis, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Laboratoire de Pharmacologie Biologique, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Celeste Lebbe
- Service de Dermatologie, and CIC (Centre d'Investigations Cliniques), AP-HP, Hôpital Saint-Louis, Paris, France; INSERM, UMR-976, AP-HP, Hôpital Saint-Louis, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | |
Collapse
|
33
|
Zakiryanova GK, Kustova E, Urazalieva NT, Amirbekov A, Baimuchametov ET, Nakisbekov NN, Shurin MR. Alterations of oncogenes expression in NK cells in patients with cancer. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:493-502. [PMID: 28695716 PMCID: PMC5691306 DOI: 10.1002/iid3.179] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/08/2017] [Accepted: 05/23/2017] [Indexed: 12/20/2022]
Abstract
INTRODUCTION C-kit/SCF signaling plays a key role in regulating NK cell homeostasis, maturation, proliferation, and cytotoxicity. C-kit-deficiency in NK cells results in significant reduction of their number, suggesting an imperative role for c-kit signaling in NK cell biology. We have recently showed that human NK cells express not only c-kit-receptor, but also both membrane-bound and soluble forms of c-kit ligand-Stem cell factor. The goal of this study was to characterize the c-kit/SCF autocrine loop in peripheral blood NK cells obtained from patients with cancer. METHODS Using Smart Flare and qRT-PCR, we have characterized expression of c-kit and two forms of SCF in patients' NK cells and correlated these results with the expression of c-myc and STAT3. RESULTS Our results demonstrated that the expression of proto-oncogenes c-myc and c-kit was significantly decreased in NK cells from all cancer patients. Expression of membrane-bound SCF in NK cells correlated with the presence of remote metastases. CONCLUSIONS We suggest that the abnormal signaling and expression of c-kit/SCF, c-myc, and STAT3 in NK cells is responsible for the defect in their cytolytic activity in cancer and these defects at the gene expression level may be the cause rather than the result of tumor progression.
Collapse
Affiliation(s)
- Gulnur K Zakiryanova
- Scientific and Technological Park Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Elena Kustova
- Laboratory of Immunology, Scientific Center of Pediatric and Children Surgery, Almaty, Kazakhstan
| | - Nataliya T Urazalieva
- Laboratory of Immunology, Scientific Center of Pediatric and Children Surgery, Almaty, Kazakhstan
| | - Aday Amirbekov
- Joint Use Center, Atchabarov Scientific-research institute of fundamental and applied medicine, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | | | - Narymzhan N Nakisbekov
- Joint Use Center, Atchabarov Scientific-research institute of fundamental and applied medicine, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Michael R Shurin
- Clinical Immunopathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
34
|
Phung B, Kazi JU, Lundby A, Bergsteinsdottir K, Sun J, Goding CR, Jönsson G, Olsen JV, Steingrímsson E, Rönnstrand L. KIT D816V Induces SRC-Mediated Tyrosine Phosphorylation of MITF and Altered Transcription Program in Melanoma. Mol Cancer Res 2017; 15:1265-1274. [PMID: 28584020 DOI: 10.1158/1541-7786.mcr-17-0149] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/25/2017] [Accepted: 05/23/2017] [Indexed: 11/16/2022]
Abstract
The oncogenic D816V mutation of the KIT receptor is well characterized in systemic mastocytosis and acute myeloid leukemia. Although KITD816V has been found in melanoma, its function and involvement in this malignancy is not understood. Here we show that KITD816V induces tyrosine phosphorylation of MITF through a triple protein complex formation between KIT, MITF, and SRC family kinases. In turn, phosphorylated MITF activates target genes that are involved in melanoma proliferation, cell-cycle progression, suppression of senescence, survival, and invasion. By blocking the triple protein complex formation, thus preventing MITF phosphorylation, the cells became hypersensitive to SRC inhibitors. We have therefore delineated a mechanism behind the oncogenic effects of KITD816V in melanoma and provided a rationale for the heightened SRC inhibitor sensitivity in KITD816V transformed cells.Implications: This study demonstrates that an oncogenic tyrosine kinase mutant, KITD816V, can alter the transcriptional program of the transcription factor MITF in melanoma Mol Cancer Res; 15(9); 1265-74. ©2017 AACR.
Collapse
Affiliation(s)
- Bengt Phung
- Division of Translational Cancer Research, Lund Stem Cell Center, Lund University, Medicon Village and Department of Oncology, Skåne University Hospital, Lund, Sweden.,Department of Biochemistry and Molecular Biology, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland.,Melanoma Genomics, Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Julhash U Kazi
- Division of Translational Cancer Research, Lund Stem Cell Center, Lund University, Medicon Village and Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Alicia Lundby
- Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Kristin Bergsteinsdottir
- Department of Biochemistry and Molecular Biology, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Jianmin Sun
- Division of Translational Cancer Research, Lund Stem Cell Center, Lund University, Medicon Village and Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Colin R Goding
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| | - Göran Jönsson
- Melanoma Genomics, Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jesper V Olsen
- Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Eiríkur Steingrímsson
- Department of Biochemistry and Molecular Biology, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland.
| | - Lars Rönnstrand
- Division of Translational Cancer Research, Lund Stem Cell Center, Lund University, Medicon Village and Department of Oncology, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
35
|
D816V mutation in the KIT gene activation loop has greater cell-proliferative and anti-apoptotic ability than N822K mutation in core-binding factor acute myeloid leukemia. Exp Hematol 2017; 52:56-64.e4. [PMID: 28506695 DOI: 10.1016/j.exphem.2017.05.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 04/03/2017] [Accepted: 05/02/2017] [Indexed: 01/09/2023]
Abstract
In core-binding factor acute myeloid leukemia (CBF-AML), there have been conflicting reports regarding the status as an unfavorable prognostic factor of mutation in the KIT gene, the significance of which remains unclear. We previously reported that prognoses differ between the KIT D816V and N822K mutations. In the present study, we compared in vitro the cell-proliferative and anti-apoptotic ability of D816V and N822K. We transduced these KIT mutations into the interleukin-3-dependent cell line TF-1 (TF-1 KITD816V, TF-1 KITN822K). When these KIT mutations were transduced into TF-1 cells, the cells acquired a proliferative ability independent of growth factor, which was significantly higher in TF-1 KITD816V than in TF-1 KITN822K (p = 0.022). When Ara-C was added in the absence of growth factor, Annexin V assay revealed that TF-1 KITD816V was associated with a significantly lower proportion of apoptotic cells than TF-1 KITN822K (p < 0.001). Regarding signal transduction pathways, both KIT D816V and KIT N822K underwent autophosphorylation in the absence of growth factor. This was followed in KIT D816V by downstream activation of the SRC family kinase pathway in addition to the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway, and in KIT N822K by downstream activation of the mitogen-activated protein kinase (MAPK) pathway in addition to the JAK/STAT pathway. These findings establish that D816V and N822K mutations are situated closely on the KIT receptor activation loop, but D816V has greater cell-proliferative and anti-apoptotic ability than N822K.
Collapse
|
36
|
Hara Y, Obata Y, Horikawa K, Tasaki Y, Suzuki K, Murata T, Shiina I, Abe R. M-COPA suppresses endolysosomal Kit-Akt oncogenic signalling through inhibiting the secretory pathway in neoplastic mast cells. PLoS One 2017; 12:e0175514. [PMID: 28403213 PMCID: PMC5389679 DOI: 10.1371/journal.pone.0175514] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/27/2017] [Indexed: 01/28/2023] Open
Abstract
Gain-of-function mutations in Kit receptor tyrosine kinase result in the development of a variety of cancers, such as mast cell tumours, gastrointestinal stromal tumours (GISTs), acute myeloid leukemia, and melanomas. The drug imatinib, a selective inhibitor of Kit, is used for treatment of mutant Kit-positive cancers. However, mutations in the Kit kinase domain, which are frequently found in neoplastic mast cells, confer an imatinib resistance, and cancers expressing the mutants can proliferate in the presence of imatinib. Recently, we showed that in neoplastic mast cells that endogenously express an imatinib-resistant Kit mutant, Kit causes oncogenic activation of the phosphatidylinositol 3-kinase-Akt (PI3K-Akt) pathway and the signal transducer and activator of transcription 5 (STAT5) but only on endolysosomes and on the endoplasmic reticulum (ER), respectively. Here, we show a strategy for inhibition of the Kit-PI3K-Akt pathway in neoplastic mast cells by M-COPA (2-methylcoprophilinamide), an inhibitor of this secretory pathway. In M-COPA-treated cells, Kit localization in the ER is significantly increased, whereas endolysosomal Kit disappears, indicating that M-COPA blocks the biosynthetic transport of Kit from the ER. The drug greatly inhibits oncogenic Akt activation without affecting the association of Kit with PI3K, indicating that ER-localized Kit-PI3K complex is unable to activate Akt. Importantly, M-COPA but not imatinib suppresses neoplastic mast cell proliferation through inhibiting anti-apoptotic Akt activation. Results of our M-COPA treatment assay show that Kit can activate Erk not only on the ER but also on other compartments. Furthermore, Tyr568/570, Tyr703, Tyr721, and Tyr936 in Kit are phosphorylated on the ER, indicating that these five tyrosine residues are all phosphorylated before mutant Kit reaches the plasma membrane (PM). Our study provides evidence that Kit is tyrosine-phosphorylated soon after synthesis on the ER but is unable to activate Akt and also demonstrates that M-COPA is efficacious for growth suppression of neoplastic mast cells.
Collapse
Affiliation(s)
- Yasushi Hara
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Yuuki Obata
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
- * E-mail:
| | - Keita Horikawa
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Yasutaka Tasaki
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Shinjuku-ku, Tokyo, Japan
| | - Kyohei Suzuki
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Shinjuku-ku, Tokyo, Japan
| | - Takatsugu Murata
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Shinjuku-ku, Tokyo, Japan
| | - Isamu Shiina
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Shinjuku-ku, Tokyo, Japan
| | - Ryo Abe
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| |
Collapse
|
37
|
Drube S, Weber F, Göpfert C, Loschinski R, Rothe M, Boelke F, Diamanti MA, Löhn T, Ruth J, Schütz D, Häfner N, Greten FR, Stumm R, Hartmann K, Krämer OH, Dudeck A, Kamradt T. TAK1 and IKK2, novel mediators of SCF-induced signaling and potential targets for c-Kit-driven diseases. Oncotarget 2016; 6:28833-50. [PMID: 26353931 PMCID: PMC4745695 DOI: 10.18632/oncotarget.5008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/20/2015] [Indexed: 12/25/2022] Open
Abstract
NF-κB activation depends on the IKK complex consisting of the catalytically active IKK1 and 2 subunits and the scaffold protein NEMO. Hitherto, IKK2 activation has always been associated with IκBα degradation, NF-κB activation, and cytokine production. In contrast, we found that in SCF-stimulated primary bone marrow-derived mast cells (BMMCs), IKK2 is alternatively activated. Mechanistically, activated TAK1 mediates the association between c-Kit and IKK2 and therefore facilitates the Lyn-dependent IKK2 activation which suffices to mediate mitogenic signaling but, surprisingly, does not result in NF-κB activation. Moreover, the c-Kit-mediated and Lyn-dependent IKK2 activation is targeted by MyD88-dependent pathways leading to enhanced IKK2 activation and therefore to potentiated effector functions. In neoplastic cells, expressing constitutively active c-Kit mutants, activated TAK1 and IKKs do also not induce NF-κB activation but mediate uncontrolled proliferation, resistance to apoptosis and enables IL-33 to mediate c-Kit-dependent signaling. Together, we identified the formation of the c-Kit-Lyn-TAK1 signalosome which mediates IKK2 activation. Unexpectedly, this IKK activation is uncoupled from the NF-κB-machinery but is critical to modulate functional cell responses in primary-, and mediates uncontrolled proliferation and survival of tumor-mast cells. Therefore, targeting TAK1 and IKKs might be a novel approach to treat c-Kit-driven diseases.
Collapse
Affiliation(s)
- Sebastian Drube
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Franziska Weber
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | | | - Romy Loschinski
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Mandy Rothe
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Franziska Boelke
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Michaela A Diamanti
- Georg-Speyer-Haus, Institute for Tumorbiology and Experimental Therapy, Frankfurt, Germany
| | - Tobias Löhn
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Julia Ruth
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Dagmar Schütz
- Institut für Pharmakologie, Universitätsklinikum Jena, Jena, Germany
| | - Norman Häfner
- Gynäkologische Molekularbiologie, Klinik für Frauenheilkunde und Geburtshilfe, Jena, Germany
| | - Florian R Greten
- Georg-Speyer-Haus, Institute for Tumorbiology and Experimental Therapy, Frankfurt, Germany
| | - Ralf Stumm
- Institut für Pharmakologie, Universitätsklinikum Jena, Jena, Germany
| | - Karin Hartmann
- Klinik und Poliklinik für Dermatologie und Venerologie, Universität zu Köln, Köln, Germany
| | - Oliver H Krämer
- Institut für Toxikologie, Universitätsmedizin Mainz, Mainz, Germany
| | - Anne Dudeck
- Institute for Immunology, Technische Universität Dresden, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Thomas Kamradt
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| |
Collapse
|
38
|
Chatterjee A, Ghosh J, Kapur R. Mastocytosis: a mutated KIT receptor induced myeloproliferative disorder. Oncotarget 2016; 6:18250-64. [PMID: 26158763 PMCID: PMC4621888 DOI: 10.18632/oncotarget.4213] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 05/23/2015] [Indexed: 12/20/2022] Open
Abstract
Although more than 90% systemic mastocytosis (SM) patients express gain of function mutations in the KIT receptor, recent next generation sequencing has revealed the presence of several additional genetic and epigenetic mutations in a subset of these patients, which confer poor prognosis and inferior overall survival. A clear understanding of how genetic and epigenetic mutations cooperate in regulating the tremendous heterogeneity observed in these patients will be essential for designing effective treatment strategies for this complex disease. In this review, we describe the clinical heterogeneity observed in patients with mastocytosis, the nature of relatively novel mutations identified in these patients, therapeutic strategies to target molecules downstream from activating KIT receptor and finally we speculate on potential novel strategies to interfere with the function of not only the oncogenic KIT receptor but also epigenetic mutations seen in these patients.
Collapse
Affiliation(s)
- Anindya Chatterjee
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Joydeep Ghosh
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Reuben Kapur
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Molecular Biology and Biochemistry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
39
|
Bibi S, Arslanhan MD, Langenfeld F, Jeanningros S, Cerny-Reiterer S, Hadzijusufovic E, Tchertanov L, Moriggl R, Valent P, Arock M. Co-operating STAT5 and AKT signaling pathways in chronic myeloid leukemia and mastocytosis: possible new targets of therapy. Haematologica 2015; 99:417-29. [PMID: 24598853 DOI: 10.3324/haematol.2013.098442] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chronic myeloid leukemia and systemic mastocytosis are myeloid neoplasms sharing a number of pathogenetic and clinical features. In both conditions, an aberrantly activated oncoprotein with tyrosine kinase activity, namely BCR-ABL1 in chronic myeloid leukemia, and mutant KIT, mostly KIT D816V, in systemic mastocytosis, is key to disease evolution. The appreciation of the role of such tyrosine kinases in these diseases has led to the development of improved therapies with tyrosine kinase-targeted inhibitors. However, most drugs, including new KIT D816V-blocking agents, have failed to achieve long-lasting remissions in advanced systemic mastocytosis, and there is a similar problem in chronic myeloid leukemia, where imatinib-resistant patients sometimes fail to achieve remission, even with second- or third-line BCR-ABL1 specific tyrosine kinase inhibitors. During disease progression, additional signaling pathways become activated in neoplastic cells, but most converge into major downstream networks. Among these, the AKT and STAT5 pathways appear most critical and may result in drug-resistant chronic myeloid leukemia and systemic mastocytosis. Inhibition of phosphorylation of these targets has proven their crucial role in disease-evolution in both malignancies. Together, these observations suggest that STAT5 and AKT are key drivers of oncogenesis in drug-resistant forms of the diseases, and that targeting STAT5 and AKT might be an interesting approach in these malignancies. The present article provides an overview of our current knowledge about the critical role of AKT and STAT5 in the pathophysiology of chronic myeloid leukemia and systemic mastocytosis and on their potential value as therapeutic targets in these neoplasms.
Collapse
|
40
|
Oncogenic Kit signals on endolysosomes and endoplasmic reticulum are essential for neoplastic mast cell proliferation. Nat Commun 2014; 5:5715. [PMID: 25493654 PMCID: PMC4284665 DOI: 10.1038/ncomms6715] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/30/2014] [Indexed: 11/08/2022] Open
Abstract
Kit is a receptor-type tyrosine kinase found on the plasma membrane. It can transform mast cells through activating mutations. Here, we show that a mutant Kit from neoplastic mast cells from mice, Kit(D814Y), is permanently active and allows cells to proliferate autonomously. It does so by activating two signalling pathways from different intracellular compartments. Mutant Kit from the cell surface accumulates on endolysosomes through clathrin-mediated endocytosis, which requires Kit's kinase activity. Kit(D814Y) is constitutively associated with phosphatidylinositol 3-kinase, but the complex activates Akt only on the cytoplasmic surface of endolysosomes. It resists destruction because it is under-ubiquitinated. Kit(D814Y) also appears in the endoplasmic reticulum soon after biosynthesis, and there, can activate STAT5 aberrantly. These mechanisms of oncogenic signalling are also seen in rat and human mast cell leukemia cells. Thus, oncogenic Kit signalling occurs from different intracellular compartments, and the mutation acts by altering Kit trafficking as well as activation.
Collapse
|
41
|
Agarwal S, Kazi JU, Mohlin S, Påhlman S, Rönnstrand L. The activation loop tyrosine 823 is essential for the transforming capacity of the c-Kit oncogenic mutant D816V. Oncogene 2014; 34:4581-90. [PMID: 25435369 DOI: 10.1038/onc.2014.383] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/25/2014] [Accepted: 09/30/2014] [Indexed: 12/31/2022]
Abstract
Oncogenic c-Kit mutations have been shown to display ligand-independent receptor activation and cell proliferation. A substitution of aspartate to valine at amino acid 816 (D816V) is one of the most commonly found oncogenic c-Kit mutations and is found in >90% of cases of mastocytosis and less commonly in germ-cell tumors, core-binding factor acute myeloid leukemia and mucosal melanomas. The mechanisms by which this mutation leads to constitutive activation and transformation are not fully understood. Previous studies have shown that the D816V mutation causes a structural change in the activation loop (A-loop), resulting in weaker binding of the A-loop to the juxtamembrane domain. In this paper, we have investigated the role of Y823, the only tyrosine residue in the A-loop, and its role in oncogenic transformation by c-Kit/D816V by introducing the Y823F mutation. Although dispensable for the kinase activity of c-Kit/D816V, the presence of Y823 was crucial for cell proliferation and survival. Furthermore, mutation of Y823 selectively downregulates the Ras/Erk and Akt pathways as well as the phosphorylation of STAT5 and reduces the transforming capacity of the D816V/c-Kit in vitro. We further show that mice injected with cells expressing c-Kit/D816V/Y823F display significantly reduced tumor size as well as tumor weight compared with controls. Finally, microarray analysis, comparing Y823F/D816V cells with cells expressing c-Kit/D816V, demonstrate that mutation of Y823 causes upregulation of proapoptotic genes, whereas genes of survival pathways are downregulated. Thus, phosphorylation of Y823 is not necessary for kinase activation, but essential for the transforming ability of the c-Kit/D816V mutant.
Collapse
Affiliation(s)
- S Agarwal
- Translational Cancer Research, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - J U Kazi
- Translational Cancer Research, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - S Mohlin
- Translational Cancer Research, Lund University, Lund, Sweden.,CREATE Health, Lund University, Lund, Sweden
| | - S Påhlman
- Translational Cancer Research, Lund University, Lund, Sweden.,CREATE Health, Lund University, Lund, Sweden
| | - L Rönnstrand
- Translational Cancer Research, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
42
|
Ranieri G, Marech I, Pantaleo M, Piccinno M, Roncetti M, Mutinati M, Rizzo A, Gadaleta CD, Introna M, Patruno R, Sciorsci RL. In vivo model for mastocytosis: A comparative review. Crit Rev Oncol Hematol 2014; 93:159-69. [PMID: 25465741 DOI: 10.1016/j.critrevonc.2014.10.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 10/01/2014] [Accepted: 10/22/2014] [Indexed: 12/27/2022] Open
Abstract
Human mastocytosis are heterogeneous group of neoplastic diseases characterized by a different degree of uncontrolled mast cell (MC) proliferation and activation. Interestingly, human mastocytosis share several biological and clinical features with canine mast cell disorders, so called canine mast cell tumors (CMCTs). These CMCTs are the most common spontaneous cutaneous tumors found in dogs representing a valid model to study neoplastic mast cell disorders. It has been discovered that the pathological activation of c-Kit receptor (c-KitR), expressed by MCs, has been involved in the pathogenesis of neoplastic MC disorders. In this review we have focused on human mastocytosis in terms of: (i) epidemiology and classification; (ii) pathogenesis at molecular levels; (iii) clinical presentation. In addition, we have summarized animal models useful to study neoplastic MC disorders including CMCTs and murine transgenic models. Finally, we have revised therapeutic approaches mostly common in human and canine MCTs and novel tyrosine kinase inhibitors approved for CMCTs and recently translated in human clinical trials.
Collapse
Affiliation(s)
- Girolamo Ranieri
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre Istituto Tumori "Giovanni Paolo II", Bari, Italy.
| | - Ilaria Marech
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Marianna Pantaleo
- Department of Emergency and Organ Transplantation (D.E.T.O.), Veterinary Medical School, Università "Aldo Moro", Bari, Italy
| | - Mariagrazia Piccinno
- Department of Emergency and Organ Transplantation (D.E.T.O.), Veterinary Medical School, Università "Aldo Moro", Bari, Italy
| | - Maria Roncetti
- Department of Emergency and Organ Transplantation (D.E.T.O.), Veterinary Medical School, Università "Aldo Moro", Bari, Italy
| | - Maddalena Mutinati
- Department of Emergency and Organ Transplantation (D.E.T.O.), Veterinary Medical School, Università "Aldo Moro", Bari, Italy
| | - Annalisa Rizzo
- Department of Emergency and Organ Transplantation (D.E.T.O.), Veterinary Medical School, Università "Aldo Moro", Bari, Italy
| | - Cosmo Damiano Gadaleta
- Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Marcello Introna
- Department of Pathology, Veterinary Medical School, Università "Aldo Moro", Bari, Italy
| | - Rosa Patruno
- Department of Prevention and Animal Health, ASL BAT, Barletta, Italy
| | - Raffaele Luigi Sciorsci
- Department of Emergency and Organ Transplantation (D.E.T.O.), Veterinary Medical School, Università "Aldo Moro", Bari, Italy
| |
Collapse
|
43
|
Allain A, Chauvot de Beauchêne I, Langenfeld F, Guarracino Y, Laine E, Tchertanov L. Allosteric pathway identification through network analysis: from molecular dynamics simulations to interactive 2D and 3D graphs. Faraday Discuss 2014; 169:303-21. [PMID: 25340971 DOI: 10.1039/c4fd00024b] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Allostery is a universal phenomenon that couples the information induced by a local perturbation (effector) in a protein to spatially distant regulated sites. Such an event can be described in terms of a large scale transmission of information (communication) through a dynamic coupling between structurally rigid (minimally frustrated) and plastic (locally frustrated) clusters of residues. To elaborate a rational description of allosteric coupling, we propose an original approach - MOdular NETwork Analysis (MONETA) - based on the analysis of inter-residue dynamical correlations to localize the propagation of both structural and dynamical effects of a perturbation throughout a protein structure. MONETA uses inter-residue cross-correlations and commute times computed from molecular dynamics simulations and a topological description of a protein to build a modular network representation composed of clusters of residues (dynamic segments) linked together by chains of residues (communication pathways). MONETA provides a brand new direct and simple visualization of protein allosteric communication. A GEPHI module implemented in the MONETA package allows the generation of 2D graphs of the communication network. An interactive PyMOL plugin permits drawing of the communication pathways between chosen protein fragments or residues on a 3D representation. MONETA is a powerful tool for on-the-fly display of communication networks in proteins. We applied MONETA for the analysis of communication pathways (i) between the main regulatory fragments of receptors tyrosine kinases (RTKs), KIT and CSF-1R, in the native and mutated states and (ii) in proteins STAT5 (STAT5a and STAT5b) in the phosphorylated and the unphosphorylated forms. The description of the physical support for allosteric coupling by MONETA allowed a comparison of the mechanisms of (a) constitutive activation induced by equivalent mutations in two RTKs and (b) allosteric regulation in the activated and non-activated STAT5 proteins. Our theoretical prediction based on results obtained with MONETA was validated for KIT by in vitro experiments. MONETA is a versatile analytical and visualization tool entirely devoted to the understanding of the functioning/malfunctioning of allosteric regulation in proteins - a crucial basis to guide the discovery of next-generation allosteric drugs.
Collapse
Affiliation(s)
- Ariane Allain
- Bioinformatics, Molecular Dynamics & Modeling (BiMoDyM), Laboratoire de Biologie et Pharmacologie Appliquée (LBPA UMR8113 CNRS), École Normale Supérieure de Cachan, 61 avenue du Président Wilson, 94235 Cachan, France.
| | | | | | | | | | | |
Collapse
|
44
|
A new human mast cell line expressing a functional IgE receptor converts to tumorigenic growth by KIT D816V transfection. Blood 2014; 124:111-20. [PMID: 24677542 DOI: 10.1182/blood-2013-10-534685] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In systemic mastocytosis (SM), clinical problems arise from factor-independent proliferation of mast cells (MCs) and the increased release of mediators by MCs, but no human cell line model for studying MC activation in the context of SM is available. We have created a stable stem cell factor (SCF) -dependent human MC line, ROSA(KIT WT), expressing a fully functional immunoglobulin E (IgE) receptor. Transfection with KIT D816V converted ROSA(KIT WT) cells into an SCF-independent clone, ROSA(KIT D816V), which produced a mastocytosis-like disease in NSG mice. Although several signaling pathways were activated, ROSA(KIT D816V) did not exhibit an increased, but did exhibit a decreased responsiveness to IgE-dependent stimuli. Moreover, NSG mice bearing ROSA(KIT D816V)-derived tumors did not show mediator-related symptoms, and KIT D816V-positive MCs obtained from patients with SM did not show increased IgE-dependent histamine release or CD63 upregulation. Our data show that KIT D816V is a disease-propagating oncoprotein, but it does not activate MCs to release proinflammatory mediators, which may explain why mediator-related symptoms in SM occur preferentially in the context of a coexisting allergy. ROSA(KIT D816V) may provide a valuable tool for studying the pathogenesis of mastocytosis and should facilitate the development of novel drugs for treating SM patients.
Collapse
|
45
|
Chan IJ, Kasprowicz S, Tharp MD. Distinct signalling pathways for mutated KIT(V560G) and KIT(D816V) in mastocytosis. Clin Exp Dermatol 2013; 38:538-44. [PMID: 23777495 DOI: 10.1111/ced.12000] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The activating mutations KIT(V560G) and KIT(D816V) are associated with mastocytosis. Thus, identifying and inhibiting the signalling pathways associated with mutated KIT gene offers a potentially important strategy for the treatment of mastocytosis. AIM To correlate KIT mutations with specific signalling pathways in human mast-cell lines using pathway inhibitors. METHODS Human mast-cell (HMC) lines expressing KIT(V560G) (the cell line HMC-1) and KIT(V560G and D816V) (HMC-1.2) were treated with specific signalling pathway inhibitors for 1-5 days, and the inhibitory effects on growth were determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cell-proliferation assay, western blotting and flow cytometry. RESULTS Growth inhibitory assays and western blot analyses showed that the Janus kinase 3/signal transducer and activator of transcription (JAK3/STAT) pathway is the preferential signalling pathway for KIT(V560G), whereas the mechanistic target of rapamycin complex 1/4E-binding protein 1 (mTORC1/4E-BP1) pathway is preferentially linked to KIT(D816V). Inhibition of these critical signalling pathways results in programmed cell death. CONCLUSIONS KIT(V560G) and KIT(D816V) use different signalling pathways that promote mast-cell growth. Inhibitors of these specific pathways might be effective in treating mastocytosis.
Collapse
Affiliation(s)
- I J Chan
- Department of Dermatology, Rush University Medical Center, Chicago, IL, USA.
| | | | | |
Collapse
|
46
|
Lu Y, Piao D, Zhang H, Li X, Chao GH, Park SJ, Chang YC, Kim CH, Murakami M, Jung SH, Choi JH, Son JK, Chang HW. Saucerneol F inhibits tumor necrosis factor-α and IL-6 production by suppressing Fyn-mediated pathways in FcεRI-mediated mast cells. Food Chem Toxicol 2013; 59:696-702. [PMID: 23851146 DOI: 10.1016/j.fct.2013.06.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/17/2013] [Accepted: 06/28/2013] [Indexed: 10/26/2022]
|
47
|
Kosan C, Ginter T, Heinzel T, Krämer OH. STAT5 acetylation: Mechanisms and consequences for immunological control and leukemogenesis. JAKSTAT 2013; 2:e26102. [PMID: 24416653 PMCID: PMC3876427 DOI: 10.4161/jkst.26102] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 08/08/2013] [Accepted: 08/09/2013] [Indexed: 12/30/2022] Open
Abstract
The cytokine-inducible transcription factors signal transducer and activator of transcription 5A and 5B (STAT5A and STAT5B) are important for the proper development of multicellular eukaryotes. Disturbed signaling cascades evoking uncontrolled expression of STAT5 target genes are associated with cancer and immunological failure. Here, we summarize how STAT5 acetylation is integrated into posttranslational modification networks within cells. Moreover, we focus on how inhibitors of deacetylases and tyrosine kinases can correct leukemogenic signaling nodes involving STAT5. Such small molecules can be exploited in the fight against neoplastic diseases and immunological disorders.
Collapse
Affiliation(s)
- Christian Kosan
- Center for Molecular Biomedicine (CMB); Institute of Biochemistry and Biophysics; University of Jena; Jena, Germany
| | - Torsten Ginter
- Center for Molecular Biomedicine (CMB); Institute of Biochemistry and Biophysics; University of Jena; Jena, Germany
| | - Thorsten Heinzel
- Center for Molecular Biomedicine (CMB); Institute of Biochemistry and Biophysics; University of Jena; Jena, Germany
| | - Oliver H Krämer
- Center for Molecular Biomedicine (CMB); Institute of Biochemistry and Biophysics; University of Jena; Jena, Germany ; Institute of Toxicology; Medical Center of the University Mainz; Mainz, Germany
| |
Collapse
|
48
|
Chaix A, Arcangeli ML, Lopez S, Voisset E, Yang Y, Vita M, Letard S, Audebert S, Finetti P, Birnbaum D, Bertucci F, Aurrand-Lions M, Dubreuil P, De Sepulveda P. KIT-D816V oncogenic activity is controlled by the juxtamembrane docking site Y568-Y570. Oncogene 2013; 33:872-81. [PMID: 23416972 DOI: 10.1038/onc.2013.12] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 11/20/2012] [Accepted: 12/14/2012] [Indexed: 02/06/2023]
Abstract
Mutation of KIT receptor tyrosine kinase at residue D816 results in ligand-independent constitutive kinase activity. This mutation occurs in most patients with mastocytosis, a myeloproliferative neoplasm, and is detected at lower frequencies in acute myeloid leukemia and in germ cell tumors. Other KIT mutations occur in gastrointestinal stromal tumors (GIST) and mucosal melanoma. KIT is considered as a bona fide therapeutic target as c-kit mutations are driving oncogenes in these pathologies. However, several evidences suggest that KIT-D816V mutant is not as aggressive as other KIT mutants. Here, we show that an intracellular docking site in the juxtamembrane region of KIT maintains a negative regulation on KIT-D816V transforming potential. Sixteen signaling proteins were shown to interact with this motif. We further demonstrate that mutation of this site results in signaling modifications, altered gene expression profile and increased transforming activity of KIT-D816V mutant. This result was unexpected as mutations of the homologous sites on wild-type (WT) KIT, or on the related oncogenic FLT3-ITD receptor, impair their function. Our results support the hypothesis that, KIT-D816V mutation is a mild oncogenic event that is sufficient to confer partial transforming properties, but requires additional mutations to acquire its full transforming potential.
Collapse
Affiliation(s)
- A Chaix
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - M-L Arcangeli
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - S Lopez
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - E Voisset
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - Y Yang
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - M Vita
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - S Letard
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - S Audebert
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - P Finetti
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - D Birnbaum
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - F Bertucci
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - M Aurrand-Lions
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - P Dubreuil
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| | - P De Sepulveda
- 1] INSERM, U1068, Centre de Recherche en Cancérologie de Marseille, Marseille, France [2] Institut Paoli-Calmettes, Marseille, France [3] Aix-Marseille University, Marseille, France
| |
Collapse
|
49
|
Cotargeting signaling pathways driving survival and cell cycle circumvents resistance to Kit inhibitors in leukemia. Blood 2012; 119:4228-41. [PMID: 22438255 DOI: 10.1182/blood-2011-07-368316] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Oncogenic mutations leading to persistent kinase activities are associated with malignancies. Therefore, deciphering the signaling networks downstream of these oncogenic stimuli remains a challenge to gather insights into targeted therapy. To elucidate the biochemical networks connecting the Kit mutant to leukemogenesis, in the present study, we performed a global profiling of tyrosine-phosphorylated proteins from mutant Kit-driven murine leukemia proerythroblasts and identified Shp2 and Stat5 as proximal effectors of Kit. Shp2 or Stat5 gene depletion by sh-RNA, combined with pharmacologic inhibition of PI3kinase or Mek/Erk activities, revealed 2 distinct and independent signaling pathways contributing to malignancy. We demonstrate that cell survival is driven by the Kit/Shp2/Ras/Mek/Erk1/2 pathway, whereas the G(1)/S transition during the cell cycle is accelerated by both the Kit/Stat5 and Kit/PI3K/Akt pathways. The combined use of the clinically relevant drugs NVP-BEZ235, which targets the cell cycle, and Obatoclax, which targets survival, demonstrated synergistic effects to inhibit leukemia cell growth. This synergy was confirmed with a human mast leukemia cell line (HMC-1.2) that expresses mutant Kit. The results of the present study using liquid chromatography/tandem mass spectrometry analysis have elucidated signaling networks downstream of an oncogenic kinase, providing a molecular rationale for pathway-targeted therapy to treat cancer cells refractory to tyrosine kinase inhibitors.
Collapse
|
50
|
Teng SP, Hsu WL, Chiu CY, Wong ML, Chang SC. Overexpression of P-glycoprotein, STAT3, phospho-STAT3 and KIT in spontaneous canine cutaneous mast cell tumours before and after prednisolone treatment. Vet J 2012; 193:551-6. [PMID: 22398132 DOI: 10.1016/j.tvjl.2012.01.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Revised: 01/29/2012] [Accepted: 01/30/2012] [Indexed: 10/28/2022]
Abstract
Prednisolone is a glucocorticoid (GC) commonly used in the treatment of canine mast cell tumours (MCTs); however, resistance to GCs develops in many MCTs following repeated treatment. P-glycoprotein (P-gp), signal transducer and activator of transcription 3 (STAT3) and KIT (CD117) are involved in GC resistance. The aim of this study was to evaluate the response to prednisolone treatment in canine cutaneous MCTs and to investigate the levels of P-gp, STAT3, phospho-STAT3 (pSTAT3) and KIT proteins in MCTs with or without prednisolone treatment. Immunohistochemistry was performed on tumour samples from 41 dogs with cutaneous MCTs. The overall objective response rate (including complete and partial responses) was 51.8% for dogs treated with prednisolone; poorly differentiated or higher stage MCTs had a lower response rate. The median time-span of tumours to reach maximal tumour regression was 14 d (range 3-77 d); 22 (81.5%) reached maximal regression at 21 d. The majority of MCTs overexpressed both P-gp and STAT3 before and after prednisolone treatment. Reduced expression of pSTAT3 and alterations in the KIT expression pattern were observed in MCTs post-treatment. Prednisolone treatment that caused a marked reduction in tumour volume was correlated with reduced pSTAT3 expression. A cytoplasmic KIT staining pattern was correlated with a lower tumour response rate to prednisolone treatment.
Collapse
Affiliation(s)
- Shr-Ping Teng
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | | | | | | | | |
Collapse
|