1
|
Using the Zebrafish as a Genetic Model to Study Erythropoiesis. Int J Mol Sci 2021; 22:ijms221910475. [PMID: 34638816 PMCID: PMC8508994 DOI: 10.3390/ijms221910475] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 09/25/2021] [Indexed: 11/30/2022] Open
Abstract
Vertebrates generate mature red blood cells (RBCs) via a highly regulated, multistep process called erythropoiesis. Erythropoiesis involves synthesis of heme and hemoglobin, clearance of the nuclei and other organelles, and remodeling of the plasma membrane, and these processes are exquisitely coordinated by specific regulatory factors including transcriptional factors and signaling molecules. Defects in erythropoiesis can lead to blood disorders such as congenital dyserythropoietic anemias, Diamond–Blackfan anemias, sideroblastic anemias, myelodysplastic syndrome, and porphyria. The molecular mechanisms of erythropoiesis are highly conserved between fish and mammals, and the zebrafish (Danio rerio) has provided a powerful genetic model for studying erythropoiesis. Studies in zebrafish have yielded important insights into RBC development and established a number of models for human blood diseases. Here, we focus on latest discoveries of the molecular processes and mechanisms regulating zebrafish erythropoiesis and summarize newly established zebrafish models of human anemias.
Collapse
|
2
|
Desjarlais M, Ruknudin P, Wirth M, Lahaie I, Dabouz R, Rivera JC, Habelrih T, Omri S, Hardy P, Rivard A, Chemtob S. Tyrosine-Protein Phosphatase Non-receptor Type 9 (PTPN9) Negatively Regulates the Paracrine Vasoprotective Activity of Bone-Marrow Derived Pro-angiogenic Cells: Impact on Vascular Degeneration in Oxygen-Induced Retinopathy. Front Cell Dev Biol 2021; 9:679906. [PMID: 34124069 PMCID: PMC8194284 DOI: 10.3389/fcell.2021.679906] [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: 03/12/2021] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Aim Insufficient post-ischemic neovascularization is an initial key step in the pathogenesis of Oxygen-Induced Retinopathy (OIR). During neovascularization, pro-angiogenic cells (PACs) are mobilized from the bone marrow and integrate into ischemic tissues to promote angiogenesis. However, the modulation of PAC paracrine activity during OIR and the specific mechanisms involved remain to be explored. Because Tyrosine-protein phosphatase non-receptor type 9 (PTPN9) is reported to be a negative regulator of stem cell differentiation and angiogenesis signaling, we investigated its effect on PAC activity in the context of OIR. Methods and Results In a rat model of OIR, higher levels of PTPN9 in the retina and in bone marrow derived PACs are associated with retinal avascular areas, lower levels of the mobilization factor SDF-1 and decreased number of CD34+/CD117+/CD133+ PACs. PACs exposed ex vivo to hyperoxia display increased PTPN9 expression, which is associated with impaired ability of PAC secretome to promote angiogenesis ex vivo (choroidal vascular sprouting) and in vitro (endothelial cell tubule formation) compared to the secretome of PACs maintained in normoxia. Suppression of PTPN9 (using siRNA) increases VEGF and SDF-1 expression to normalize PAC secretome during hyperoxia, leading to restored angiogenic ability of PAC secretome. Moreover, endothelial cells exposed to the secretome of siPTPN9-treated PACs expressed increased levels of activated form of VEGF receptor 2 (VEGFR2). In the rat model of OIR, intravitreal injection of secretome from siPTPN9-treated PACs significantly reduced retinal vaso-obliteration; this was associated with higher retinal levels of VEGF/SDF-1, and increased recruitment of PACs (CD34+ cells) to the retinal and choroidal vessels. Conclusion Our results suggest that hyperoxia alters the paracrine proangiogenic activity of BM-PACs by inducing PTPN9, which can contribute to impair post-ischemic revascularization in the context of OIR. Targeting PTPN9 restores PAC angiogenic properties, and provide a new target for vessel integrity in ischemic retinopathies.
Collapse
Affiliation(s)
- Michel Desjarlais
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Pakiza Ruknudin
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Maëlle Wirth
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Isabelle Lahaie
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Rabah Dabouz
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - José Carlos Rivera
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Tiffany Habelrih
- Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Samy Omri
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Pierre Hardy
- Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alain Rivard
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, QC, Canada
| | - Sylvain Chemtob
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| |
Collapse
|
3
|
Cendrowski J, Kaczmarek M, Mazur M, Kuzmicz-Kowalska K, Jastrzebski K, Brewinska-Olchowik M, Kominek A, Piwocka K, Miaczynska M. Splicing variation of BMP2K balances abundance of COPII assemblies and autophagic degradation in erythroid cells. eLife 2020; 9:e58504. [PMID: 32795391 PMCID: PMC7473771 DOI: 10.7554/elife.58504] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/13/2020] [Indexed: 12/26/2022] Open
Abstract
Intracellular transport undergoes remodeling upon cell differentiation, which involves cell type-specific regulators. Bone morphogenetic protein 2-inducible kinase (BMP2K) has been potentially implicated in endocytosis and cell differentiation but its molecular functions remained unknown. We discovered that its longer (L) and shorter (S) splicing variants regulate erythroid differentiation in a manner unexplainable by their involvement in AP-2 adaptor phosphorylation and endocytosis. However, both variants interact with SEC16A and could localize to the juxtanuclear secretory compartment. Variant-specific depletion approach showed that BMP2K isoforms constitute a BMP2K-L/S regulatory system that controls the distribution of SEC16A and SEC24B as well as SEC31A abundance at COPII assemblies. Finally, we found L to promote and S to restrict autophagic degradation and erythroid differentiation. Hence, we propose that BMP2K-L and BMP2K-S differentially regulate abundance and distribution of COPII assemblies as well as autophagy, possibly thereby fine-tuning erythroid differentiation.
Collapse
Affiliation(s)
- Jaroslaw Cendrowski
- Laboratory of Cell Biology, International Institute of Molecular and Cell BiologyWarsawPoland
| | - Marta Kaczmarek
- Laboratory of Cell Biology, International Institute of Molecular and Cell BiologyWarsawPoland
| | - Michał Mazur
- Laboratory of Cell Biology, International Institute of Molecular and Cell BiologyWarsawPoland
| | | | - Kamil Jastrzebski
- Laboratory of Cell Biology, International Institute of Molecular and Cell BiologyWarsawPoland
| | | | - Agata Kominek
- Laboratory of Cytometry, Nencki Institute of Experimental BiologyWarsawPoland
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental BiologyWarsawPoland
| | - Marta Miaczynska
- Laboratory of Cell Biology, International Institute of Molecular and Cell BiologyWarsawPoland
| |
Collapse
|
4
|
Wang D, Cheng Z, Zhao M, Jiao C, Meng Q, Pan H, Xie Y, Li L, Zhu Y, Wang W, Qu C, Liang D. PTPN9 induces cell apoptosis by mitigating the activation of Stat3 and acts as a tumor suppressor in colorectal cancer. Cancer Manag Res 2019; 11:1309-1319. [PMID: 30804683 PMCID: PMC6371942 DOI: 10.2147/cmar.s187001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Accumulating evidence has shown that protein tyrosine phosphatases (PTPs) are involved in regulating the transduction of many signaling pathways and play important roles in modulating the progression of some cancers, but the functions of PTPs in cancers have not been well elucidated until now. Here, we aimed to identify the roles of protein tyrosine phosphatase nonreceptor type 9 (PTPN9), a cytoplasmic PTP, in the development of colorectal cancer and elucidate the regulatory mechanism involved. Materials and methods Cell viability assessment, colony formation assay, caspase-3 and caspase-9 activity assay, real-time PCR, and Western blot analysis were applied. Results Our results showed that PTPN9 expression was frequently downregulated in colorectal cancer tissues compared with adjacent normal tissues. Overexpression of PTPN9 mitigated cell growth and colony formation and induced cell apoptosis in colorectal cancer. Conversely, PTPN9 knockdown promoted cell growth and survival. Moreover, PTPN9 negatively regulated the activation of Stat3 and depressed its nuclear translocation in colorectal cancer. The effects of PTPN9 knockdown on cell apoptosis were attenuated by inhibition of the Stat3 pathway. Conclusion These results indicate that PTPN9 inhibits cell growth and survival by repressing the activation of Stat3 in colorectal cancer, which suggests an important underlying mechanism of regulating cell growth and provides a novel candidate therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Dawei Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China,
| | - Zhuoxin Cheng
- Department of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi 154002, People's Republic of China.,Heilongjiang Provincial Key Laboratory of Metabolic Disease, Jiamusi 154002, People's Republic of China
| | - Ming Zhao
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China,
| | - Chengbin Jiao
- Department of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi 154002, People's Republic of China
| | - Qinghui Meng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China,
| | - Huayang Pan
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China,
| | - Yu Xie
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China,
| | - Long Li
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China,
| | - Yexing Zhu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China,
| | - Wei Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China,
| | - Chunlei Qu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China,
| | - Deshen Liang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, People's Republic of China,
| |
Collapse
|
5
|
Huang Y, Zhang Y, Ge L, Lin Y, Kwok HF. The Roles of Protein Tyrosine Phosphatases in Hepatocellular Carcinoma. Cancers (Basel) 2018; 10:cancers10030082. [PMID: 29558404 PMCID: PMC5876657 DOI: 10.3390/cancers10030082] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/12/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023] Open
Abstract
The protein tyrosine phosphatase (PTP) family is involved in multiple cellular functions and plays an important role in various pathological and physiological processes. In many chronic diseases, for example cancer, PTP is a potential therapeutic target for cancer treatment. In the last two decades, dozens of PTP inhibitors which specifically target individual PTP molecules were developed as therapeutic agents. Hepatocellular carcinoma (HCC) is one of the most common malignant tumors and is the second most lethal cancer worldwide due to a lack of effective therapies. Recent studies have unveiled both oncogenic and tumor suppressive functions of PTP in HCC. Here, we review the current knowledge on the involvement of PTP in HCC and further discuss the possibility of targeting PTP in HCC.
Collapse
Affiliation(s)
- Yide Huang
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China.
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Yafei Zhang
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China.
| | - Lilin Ge
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yao Lin
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China.
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| |
Collapse
|
6
|
Lei L, Yan SY, Yang R, Chen JY, Li Y, Bu Y, Chang N, Zhou Q, Zhu X, Li CY, Xiong JW. Spliceosomal protein eftud2 mutation leads to p53-dependent apoptosis in zebrafish neural progenitors. Nucleic Acids Res 2017; 45:3422-3436. [PMID: 27899647 PMCID: PMC5389467 DOI: 10.1093/nar/gkw1043] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/24/2016] [Indexed: 12/26/2022] Open
Abstract
Haploinsufficiency of EFTUD2 (Elongation Factor Tu GTP Binding Domain Containing 2) is linked to human mandibulofacial dysostosis, Guion-Almeida type (MFDGA), but the underlying cellular and molecular mechanisms remain to be addressed. We report here the isolation, cloning and functional analysis of the mutated eftud2 (snu114) in a novel neuronal mutant fn10a in zebrafish. This mutant displayed abnormal brain development with evident neuronal apoptosis while the development of other organs appeared less affected. Positional cloning revealed a nonsense mutation such that the mutant eftud2 mRNA encoded a truncated Eftud2 protein and was subjected to nonsense-mediated decay. Disruption of eftud2 led to increased apoptosis and mitosis of neural progenitors while it had little effect on differentiated neurons. Further RNA-seq and functional analyses revealed a transcriptome-wide RNA splicing deficiency and a large amount of intron-retaining and exon-skipping transcripts, which resulted in inadequate nonsense-mediated RNA decay and activation of the p53 pathway in fn10a mutants. Therefore, our study has established that eftud2 functions in RNA splicing during neural development and provides a suitable zebrafish model for studying the molecular pathology of the neurological disease MFDGA.
Collapse
Affiliation(s)
- Lei Lei
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Shou-Yu Yan
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Ran Yang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Jia-Yu Chen
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Yumei Li
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Ye Bu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Nannan Chang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Qinchao Zhou
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Xiaojun Zhu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Chuan-Yun Li
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | - Jing-Wei Xiong
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| |
Collapse
|
7
|
Xiao C, Gao L, Hou Y, Xu C, Chang N, Wang F, Hu K, He A, Luo Y, Wang J, Peng J, Tang F, Zhu X, Xiong JW. Chromatin-remodelling factor Brg1 regulates myocardial proliferation and regeneration in zebrafish. Nat Commun 2016; 7:13787. [PMID: 27929112 PMCID: PMC5476829 DOI: 10.1038/ncomms13787] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 11/01/2016] [Indexed: 12/15/2022] Open
Abstract
The zebrafish possesses a remarkable capacity of adult heart regeneration, but the underlying mechanisms are not well understood. Here we report that chromatin remodelling factor Brg1 is essential for adult heart regeneration. Brg1 mRNA and protein are induced during heart regeneration. Transgenic over-expression of dominant-negative Xenopus Brg1 inhibits the formation of BrdU+/Mef2C+ and Tg(gata4:EGFP) cardiomyocytes, leading to severe cardiac fibrosis and compromised myocardial regeneration. RNA-seq and RNAscope analyses reveal that inhibition of Brg1 increases the expression of cyclin-dependent kinase inhibitors such as cdkn1a and cdkn1c in the myocardium after ventricular resection; and accordingly, myocardial-specific expression of dn-xBrg1 blunts myocardial proliferation and regeneration. Mechanistically, injury-induced Brg1, via its interaction with Dnmt3ab, suppresses the expression of cdkn1c by increasing the methylation level of CpG sites at the cdkn1c promoter. Taken together, our results suggest that Brg1 promotes heart regeneration by repressing cyclin-dependent kinase inhibitors partly through Dnmt3ab-dependent DNA methylation.
Collapse
Affiliation(s)
- Chenglu Xiao
- Institute of Molecular Medicine, Peking University, Beijing 100871, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China.,State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100871, China
| | - Lu Gao
- Institute of Molecular Medicine, Peking University, Beijing 100871, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China.,State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100871, China
| | - Yu Hou
- Biodynamic Optical Imaging Center, Peking University, Beijing 100871, China.,College of Life Sciences, Peking University, Beijing 100871, China
| | - Congfei Xu
- School of Life Sciences, University of Science and Technology of China, Hefei 230026, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Nannan Chang
- Institute of Molecular Medicine, Peking University, Beijing 100871, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China.,State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100871, China
| | - Fang Wang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Keping Hu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Beijing 100193, China.,Peking Union Medical College, Beijing 100730, China
| | - Aibin He
- Institute of Molecular Medicine, Peking University, Beijing 100871, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Jun Wang
- School of Life Sciences, University of Science and Technology of China, Hefei 230026, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Jinrong Peng
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fuchou Tang
- Biodynamic Optical Imaging Center, Peking University, Beijing 100871, China.,College of Life Sciences, Peking University, Beijing 100871, China
| | - Xiaojun Zhu
- Institute of Molecular Medicine, Peking University, Beijing 100871, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China.,State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100871, China
| | - Jing-Wei Xiong
- Institute of Molecular Medicine, Peking University, Beijing 100871, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China.,State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100871, China
| |
Collapse
|
8
|
Ouédraogo ZG, Biau J, Kemeny JL, Morel L, Verrelle P, Chautard E. Role of STAT3 in Genesis and Progression of Human Malignant Gliomas. Mol Neurobiol 2016; 54:5780-5797. [PMID: 27660268 DOI: 10.1007/s12035-016-0103-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 09/06/2016] [Indexed: 12/23/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is aberrantly activated in glioblastoma and has been identified as a relevant therapeutic target in this disease and many other human cancers. After two decades of intensive research, there is not yet any approved STAT3-based glioma therapy. In addition to the canonical activation by tyrosine 705 phosphorylation, concordant reports described a potential therapeutic relevance of other post-translational modifications including mainly serine 727 phosphorylation. Such reports reinforce the need to refine the strategy of targeting STAT3 in each concerned disease. This review focuses on the role of serine 727 and tyrosine 705 phosphorylation of STAT3 in glioma. It explores their contribution to glial cell transformation and to the mechanisms that make glioma escape to both immune control and standard treatment.
Collapse
Affiliation(s)
- Zangbéwendé Guy Ouédraogo
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, Clermont-Ferrand, France.,Département de Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, Centre Jean Perrin, EA7283 CREaT - Université d'Auvergne, 58 rue Montalembert, F-63000-63011, Clermont Ferrand, France.,Laboratoire de Pharmacologie, de Toxicologie et de Chimie Thérapeutique, Université de Ouagadougou, 03, Ouagadougou, BP 7021, Burkina Faso
| | - Julian Biau
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, Clermont-Ferrand, France.,Département de Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, Centre Jean Perrin, EA7283 CREaT - Université d'Auvergne, 58 rue Montalembert, F-63000-63011, Clermont Ferrand, France.,Département de Radiothérapie, Institut Curie, 91405, Orsay, France
| | - Jean-Louis Kemeny
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, Clermont-Ferrand, France.,CHU Clermont-Ferrand, Service d'Anatomopathologie, F-63003, Clermont-Ferrand, France
| | - Laurent Morel
- Clermont Université, Université Blaise-Pascal, GReD, UMR CNRS 6293, INSERM U1103, 24 Avenue des Landais BP80026, 63171, Aubière, France
| | - Pierre Verrelle
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, Clermont-Ferrand, France.,Département de Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, Centre Jean Perrin, EA7283 CREaT - Université d'Auvergne, 58 rue Montalembert, F-63000-63011, Clermont Ferrand, France.,Département de Radiothérapie, Institut Curie, 91405, Orsay, France
| | - Emmanuel Chautard
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, Clermont-Ferrand, France. .,Département de Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, Centre Jean Perrin, EA7283 CREaT - Université d'Auvergne, 58 rue Montalembert, F-63000-63011, Clermont Ferrand, France.
| |
Collapse
|
9
|
Hendriks WJAJ, Böhmer FD. Non-transmembrane PTPs in Cancer. PROTEIN TYROSINE PHOSPHATASES IN CANCER 2016:47-113. [DOI: 10.1007/978-1-4939-3649-6_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
10
|
Hatzihristidis T, Desai N, Hutchins AP, Meng TC, Tremblay ML, Miranda-Saavedra D. A Drosophila-centric view of protein tyrosine phosphatases. FEBS Lett 2015; 589:951-66. [PMID: 25771859 DOI: 10.1016/j.febslet.2015.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 03/02/2015] [Accepted: 03/02/2015] [Indexed: 12/30/2022]
Abstract
Most of our knowledge on protein tyrosine phosphatases (PTPs) is derived from human pathologies and mouse knockout models. These models largely correlate well with human disease phenotypes, but can be ambiguous due to compensatory mechanisms introduced by paralogous genes. Here we present the analysis of the PTP complement of the fruit fly and the complementary view that PTP studies in Drosophila will accelerate our understanding of PTPs in physiological and pathological conditions. With only 44 PTP genes, Drosophila represents a streamlined version of the human complement. Our integrated analysis places the Drosophila PTPs into evolutionary and functional contexts, thereby providing a platform for the exploitation of the fly for PTP research and the transfer of knowledge onto other model systems.
Collapse
Affiliation(s)
- Teri Hatzihristidis
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, Montreal, Québec H3A 1A3, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Nikita Desai
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, Montreal, Québec H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Andrew P Hutchins
- Key Laboratory of Regenerative Biology and Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
| | - Tzu-Ching Meng
- Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan; Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Michel L Tremblay
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, Montreal, Québec H3A 1A3, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada; Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
| | - Diego Miranda-Saavedra
- World Premier International (WPI) Immunology Frontier Research Center (IFReC), Osaka University, 3-1 Yamadaoka, Suita 565-0871, Osaka, Japan; Centro de Biología Molecular Severo Ochoa, CSIC/Universidad Autónoma de Madrid, 28049 Madrid, Spain; IE Business School, IE University, María de Molina 31 bis, 28006 Madrid, Spain.
| |
Collapse
|
11
|
Jiang H, Sui Y, Cui Y, Lin P, Li W, Xing S, Wang D, Hu M, Fu X. Expression, purification, and characterization of human osteoclastic protein-tyrosine phosphatase catalytic domain in Escherichia coli. Protein Expr Purif 2015; 107:7-12. [DOI: 10.1016/j.pep.2014.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/10/2014] [Accepted: 11/16/2014] [Indexed: 10/24/2022]
|
12
|
Yang R, Yan S, Zhu X, Li CY, Liu Z, Xiong JW. Antimalarial drug artemisinin depletes erythrocytes by activating apoptotic pathways in zebrafish. Exp Hematol 2015; 43:331-41.e8. [PMID: 25584867 DOI: 10.1016/j.exphem.2014.11.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/02/2014] [Accepted: 11/11/2014] [Indexed: 11/15/2022]
Abstract
Despite its extraordinary efficacy, administration of the major antimalarial drug artemisinin leads to anemia, and the underlying cellular and molecular mechanisms are not well understood. Here, we report the effects of artemisinin on erythroid development and apoptosis in zebrafish and human cells. By performing a small-molecule screen with zebrafish embryos, we found that artemisinin treatment depleted red blood cells and slightly decreased definitive hematopoietic stem cells, but had no effect on primitive hematopoietic progenitors. RNA-Seq revealed that artemisinin suppressed a cluster of genes in the heme biosynthesis and globin synthesis pathways. Furthermore, artemisinin induced apoptosis in erythrocytes in zebrafish embryos, as assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling assay, and preferentially acted on differentiated erythrocytes by elevating caspase 8 and caspase 9 activity in differentiated human K562 cells. Consistently, artemisinin suppressed the ectopic expression of erythroid genes in jak2aV581F-injected embryos, a zebrafish model for human polycythemia vera in which the bone marrow makes too many red blood cells. Taken together, our data suggested that artemisinin, in addition to killing parasites, has a direct action on differentiated erythrocytes other than definitive hematopoietic stem cells and causes erythroid apoptosis by interfering with the heme biosynthesis pathway in zebrafish and human cells.
Collapse
Affiliation(s)
- Ran Yang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Shouyu Yan
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xiaojun Zhu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Chuan-Yun Li
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Jing-Wei Xiong
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China.
| |
Collapse
|