1
|
Dai N, Groenendyk J, Michalak M. Interplay between myotubularins and Ca 2+ homeostasis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119739. [PMID: 38710289 DOI: 10.1016/j.bbamcr.2024.119739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024]
Abstract
The myotubularin family, encompassing myotubularin 1 (MTM1) and 14 myotubularin-related proteins (MTMRs), represents a conserved group of phosphatases featuring a protein tyrosine phosphatase domain. Nine members are characterized by an active phosphatase domain C(X)5R, dephosphorylating the D3 position of PtdIns(3)P and PtdIns(3,5)P2. Mutations in myotubularin genes result in human myopathies, and several neuropathies including X-linked myotubular myopathy and Charcot-Marie-Tooth type 4B. MTM1, MTMR6 and MTMR14 also contribute to Ca2+ signaling and Ca2+ homeostasis that play a key role in many MTM-dependent myopathies and neuropathies. Here we explore the evolving roles of MTM1/MTMRs, unveiling their influence on critical aspects of Ca2+ signaling pathways.
Collapse
Affiliation(s)
- Ning Dai
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| |
Collapse
|
2
|
Mohren L, Doege A, Miroschnikov N, Dräger O, Busch MA, Dünker N. Role of Protein Tyrosine Phosphatase Receptor Type E (PTPRE) in Chemoresistant Retinoblastoma. Int J Mol Sci 2024; 25:4572. [PMID: 38674157 PMCID: PMC11049872 DOI: 10.3390/ijms25084572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Protein tyrosine phosphatase receptor type E (PTPRE) is a member of the "classical" protein tyrosine phosphatase subfamily and regulates a variety of cellular processes in a tissue-specific manner by antagonizing the function of protein tyrosine kinases. PTPRE plays a tumorigenic role in different human cancer cells, but its role in retinoblastoma (RB), the most common malignant eye cancer in children, remains to be elucidated. Etoposide-resistant RB cell lines and RB patients display significant higher PTPRE expression levels compared to chemosensitive counterparts and the healthy human retina, respectively. PTPRE promotor methylation analyses revealed that PTPRE expression in RB is not regulated via this mechanism. Lentiviral PTPRE knockdown (KD) induced a significant decrease in growth kinetics, cell viability, and anchorage-independent growth of etoposide-resistant Y79 and WERI RB cells. Caspase-dependent apoptosis rates were significantly increased and a re-sensitization for etoposide could be observed after PTPRE depletion. In vivo chicken chorioallantoic membrane (CAM) assays revealed decreased tumor formation capacity as well as reduced tumor size and weight following PTPRE KD. Expression levels of miR631 were significantly downregulated in etoposide-resistant RB cells and patients. Transient miR631 overexpression resulted in significantly decreased PTPRE levels and concomitantly decreased proliferation and increased apoptosis levels in etoposide-resistant RB cells. These impacts mirror PTPRE KD effects, indicating a regulation of PTPRE via this miR. Additionally, PTPRE KD led to altered phosphorylation of protein kinase SGK3 and-dependent on the cell line-AKT and ERK1/2, suggesting potential PTPRE downstream signaling pathways. In summary, these results indicate an oncogenic role of PTPRE in chemoresistant retinoblastoma.
Collapse
Affiliation(s)
- Lars Mohren
- Institute for Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Medical Faculty, University of Duisburg-Essen, 45122 Essen, Germany; (L.M.); (A.D.); (N.D.)
| | - Annika Doege
- Institute for Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Medical Faculty, University of Duisburg-Essen, 45122 Essen, Germany; (L.M.); (A.D.); (N.D.)
| | - Natalia Miroschnikov
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, Medical Faculty, University of Duisburg-Essen, 45122 Essen, Germany
| | - Oliver Dräger
- Medical School OWL, Cellular Neurophysiology, Bielefeld University, 33615 Bielefeld, Germany;
| | - Maike Anna Busch
- Institute for Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Medical Faculty, University of Duisburg-Essen, 45122 Essen, Germany; (L.M.); (A.D.); (N.D.)
| | - Nicole Dünker
- Institute for Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Medical Faculty, University of Duisburg-Essen, 45122 Essen, Germany; (L.M.); (A.D.); (N.D.)
| |
Collapse
|
3
|
Abstract
Phosphatases and kinases maintain an equilibrium of dephosphorylated and phosphorylated proteins, respectively, that are required for critical cellular functions. Imbalance in this equilibrium or irregularity in their function causes unfavorable cellular effects that have been implicated in the development of numerous diseases. Protein tyrosine phosphatases (PTPs) catalyze the dephosphorylation of protein substrates on tyrosine residues, and their involvement in cell signaling and diseases such as cancer and inflammatory and metabolic diseases has made them attractive therapeutic targets. However, PTPs have proved challenging in therapeutics development, garnering them the unfavorable reputation of being undruggable. Nonetheless, great strides have been made toward the inhibition of PTPs over the past decade. Here, we discuss the advancement in small-molecule inhibition for the PTP subfamily known as the mitogen-activated protein kinase (MAPK) phosphatases (MKPs). We review strategies and inhibitor discovery tools that have proven successful for small-molecule inhibition of the MKPs and discuss what the future of MKP inhibition potentially might yield.
Collapse
Affiliation(s)
- Shanelle R Shillingford
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
| | - Anton M Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA;
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
4
|
Liu Y, Zhang J, Du Z, Huang J, Cheng Y, Yi W, Li T, Yang J, Chen C. Comprehensive analysis of PTPN family expression and prognosis in acute myeloid leukemia. Front Genet 2023; 13:1087938. [PMID: 36699453 PMCID: PMC9868563 DOI: 10.3389/fgene.2022.1087938] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023] Open
Abstract
Background: Tyrosyl phosphorylation is carried out by a group of enzymes known as non-receptor protein tyrosine phosphatases (PTPNs). In the current investigation, it is hoped to shed light on the relationships between the expression patterns of PTPN family members and the prognosis of acute myeloid leukemia (AML). Methods: PTPN expression was examined using GEPIA and GEO databases. To investigate the connection between PTPN expression and survival in AML patients, we downloaded data from the Broad TCGA Firehose and Clinical Proteomic Tumor Analysis (CPTAC) of the Cancer Genome Atlas (TCGA). We used quantitative real-time PCR (qRT-PCR) to confirm that essential genes were performed in clinical samples and cell lines. We then used western blot to verify that the genes expressed in the above databases were positive in normal tissues, AML patient samples, and AML cell lines. Next, we investigated associations between genome-wide expression profiles and PTPN6 expression using the GEO datasets. We investigated the interactive exploration of multidimensional cancer genomics using the cBioPortal datasets. Using the DAVID database, a study of gene ontology enrichment was performed. The protein-protein interaction (PPI) network was created using the STRING portal, and the gene-gene interaction network was performed using GeneMANIA. Results: Data from GEO and GEPIA revealed that most PTPN family members were linked to AML. Patients with leukemia have elevated levels of several PTPN members. All of the AML patients' poor overall survival (OS, p < .05) was significantly linked with higher expression of PTPN1, PTPN6, and PTPN7. Additionally, clinical samples showed that the expression of PTPN 6, PTPN 7, PTPN 13, and PTPN 14 was higher than normal in AML patients (p = .0116, p = .0034, p = .0092, and p = .0057, respectively) and AML cell lines (p = .0004, p = .0035, p = .0357, and p = .0177, respectively). Western blotting results showed that the expression of PTPN6 in AML samples and AML cell lines was significantly higher than that in normal control samples. Conclusion: Differentially expressed PTPN family members were found in AML. The prognosis of patients and PTPN gene expression were shown to be correlated. PTPN6 is one of these members and may be used as an AML diagnostic and prognostic marker.
Collapse
Affiliation(s)
- Yong Liu
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Jing Zhang
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Zefan Du
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Junbin Huang
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Yucai Cheng
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Wenfang Yi
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Tianwen Li
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Jing Yang
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Chun Chen
- Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
5
|
Liu D, Zhang Y, Fang H, Yuan J, Ji L. The progress of research into pseudophosphatases. Front Public Health 2022; 10:965631. [PMID: 36106167 PMCID: PMC9464862 DOI: 10.3389/fpubh.2022.965631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/09/2022] [Indexed: 01/24/2023] Open
Abstract
Pseudophosphatases are a class of phosphatases that mutate at the catalytically active site. They play important parts in many life processes and disorders, e.g., cell apoptosis, stress reaction, tumorigenesis, axon differentiation, Charcot-Marie-Tooth, and metabolic dysfunction. The present review considers the structures and action types of pseudophosphatases in four families, protein tyrosine phosphatases (PTPs), myotube protein phosphatases (MTMs), phosphatases and tensin homologues (PTENs) and dual specificity phosphatases (DUSPs), as well as their mechanisms in signaling and disease. We aimed to provide reference material for the research and treatment of related diseases.
Collapse
Affiliation(s)
- Deqiang Liu
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yiming Zhang
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Hui Fang
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jinxiang Yuan
- College of Life Sciences, Shandong Normal University, Jinan, China,The Collaborative Innovation Center, Jining Medical University, Jining, China,*Correspondence: Jinxiang Yuan
| | - Lizhen Ji
- College of Life Sciences, Shandong Normal University, Jinan, China,Lizhen Ji
| |
Collapse
|
6
|
Mattei AM, Smailys JD, Hepworth EMW, Hinton SD. The Roles of Pseudophosphatases in Disease. Int J Mol Sci 2021; 22:ijms22136924. [PMID: 34203203 PMCID: PMC8269279 DOI: 10.3390/ijms22136924] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/10/2021] [Accepted: 06/24/2021] [Indexed: 01/07/2023] Open
Abstract
The pseudophosphatases, atypical members of the protein tyrosine phosphatase family, have emerged as bona fide signaling regulators within the past two decades. Their roles as regulators have led to a renaissance of the pseudophosphatase and pseudoenyme fields, catapulting interest from a mere curiosity to intriguing and relevant proteins to investigate. Pseudophosphatases make up approximately fourteen percent of the phosphatase family, and are conserved throughout evolution. Pseudophosphatases, along with pseudokinases, are important players in physiology and pathophysiology. These atypical members of the protein tyrosine phosphatase and protein tyrosine kinase superfamily, respectively, are rendered catalytically inactive through mutations within their catalytic active signature motif and/or other important domains required for catalysis. This new interest in the pursuit of the relevant functions of these proteins has resulted in an elucidation of their roles in signaling cascades and diseases. There is a rapid accumulation of knowledge of diseases linked to their dysregulation, such as neuropathies and various cancers. This review analyzes the involvement of pseudophosphatases in diseases, highlighting the function of various role(s) of pseudophosphatases involvement in pathologies, and thus providing a platform to strongly consider them as key therapeutic drug targets.
Collapse
|
7
|
Transcriptional drug repositioning and cheminformatics approach for differentiation therapy of leukaemia cells. Sci Rep 2021; 11:12537. [PMID: 34131166 PMCID: PMC8206077 DOI: 10.1038/s41598-021-91629-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/21/2021] [Indexed: 02/05/2023] Open
Abstract
Differentiation therapy is attracting increasing interest in cancer as it can be more specific than conventional chemotherapy approaches, and it has offered new treatment options for some cancer types, such as treating acute promyelocytic leukaemia (APL) by retinoic acid. However, there is a pressing need to identify additional molecules which act in this way, both in leukaemia and other cancer types. In this work, we hence developed a novel transcriptional drug repositioning approach, based on both bioinformatics and cheminformatics components, that enables selecting such compounds in a more informed manner. We have validated the approach for leukaemia cells, and retrospectively retinoic acid was successfully identified using our method. Prospectively, the anti-parasitic compound fenbendazole was tested in leukaemia cells, and we were able to show that it can induce the differentiation of leukaemia cells to granulocytes in low concentrations of 0.1 μM and within as short a time period as 3 days. This work hence provides a systematic and validated approach for identifying small molecules for differentiation therapy in cancer.
Collapse
|
8
|
Measurable residual disease in elderly acute myeloid leukemia: results from the PETHEMA-FLUGAZA phase 3 clinical trial. Blood Adv 2021; 5:760-770. [PMID: 33560390 DOI: 10.1182/bloodadvances.2020003195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
The value of measurable residual disease (MRD) in elderly patients with acute myeloid leukemia (AML) is inconsistent between those treated with intensive vs hypomethylating drugs, and unknown after semi-intensive therapy. We investigated the role of MRD in refining complete remission (CR) and treatment duration in the phase 3 FLUGAZA clinical trial, which randomized 283 elderly AML patients to induction and consolidation with fludarabine plus cytarabine (FLUGA) vs 5-azacitidine. After consolidation, patients continued treatment if MRD was ≥0.01% or stopped if MRD was <0.01%, as assessed by multidimensional flow cytometry (MFC). On multivariate analysis including genetic risk and treatment arm, MRD status in patients achieving CR (N = 72) was the only independent prognostic factor for relapse-free survival (RFS) (HR, 3.45; P = .002). Achieving undetectable MRD significantly improved RFS of patients with adverse genetics (HR, 0.32; P = .013). Longer overall survival was observed in patients with undetectable MRD after induction though not after consolidation. Although leukemic cells from most patients displayed phenotypic aberrancies vs their normal counterpart (N = 259 of 265), CD34 progenitors from cases with undetectable MRD by MFC carried extensive genetic abnormalities identified by whole-exome sequencing. Interestingly, the number of genetic alterations significantly increased from diagnosis to MRD stages in patients treated with FLUGA vs 5-azacitidine (2.2-fold vs 1.1-fold; P = .001). This study supports MRD assessment to refine CR after semi-intensive therapy or hypomethylating agents, but unveils that improved sensitivity is warranted to individualize treatment and prolong survival of elderly AML patients achieving undetectable MRD.
Collapse
|
9
|
Samimi H, Mehta I, Docking TR, Zainulabadeen A, Karsan A, Zare H. DNA methylation analysis improves the prognostication of acute myeloid leukemia. EJHAEM 2021; 2:211-218. [PMID: 34308417 PMCID: PMC8294109 DOI: 10.1002/jha2.187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
Integration of orthogonal data could provide new opportunities to pinpoint the underlying molecular mechanisms of hematologic disorders. Using a novel gene network approach, we integrated DNA methylation data from The Cancer Genome Atlas (n = 194 cases) with the corresponding gene expression profile. Our integrated gene network analysis classified AML patients into low-, intermediate-, and high-risk groups. The identified high-risk group had significantly shorter overall survival compared to the low-risk group (p-value ≤10-11). Specifically, our approach identified a particular subgroup of nine high-risk AML cases that died within 2 years after diagnosis. These high-risk cases otherwise would be incorrectly classified as intermediate-risk solely based on cytogenetics, mutation profiles, and common molecular characteristics of AML. We confirmed the prognostic value of our integrative gene network approach using two independent datasets, as well as through comparison with European LeukemiaNet and LSC17 criteria. Our approach could be useful in the prognostication of a subset of borderline AML cases. These cases would not be classified into appropriate risk groups by other approaches that use gene expression, but not DNA methylation data. Our findings highlight the significance of epigenomic data, and they indicate integrating DNA methylation data with gene coexpression networks can have a synergistic effect.
Collapse
Affiliation(s)
- Hanie Samimi
- Department of Computer ScienceTexas State UniversitySan MarcosTexasUSA
| | - Isha Mehta
- Department of Cell Systems & AnatomyThe University of Texas Health Science CenterSan AntonioTexasUSA
| | - Thomas Roderick Docking
- Canada's Michael Smith Genome Sciences CentreBritish Columbia Cancer Research CentreVancouverBritish ColumbiaCanada
| | | | - Aly Karsan
- Canada's Michael Smith Genome Sciences CentreBritish Columbia Cancer Research CentreVancouverBritish ColumbiaCanada
| | - Habil Zare
- Department of Cell Systems & AnatomyThe University of Texas Health Science CenterSan AntonioTexasUSA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUniversity of Texas Health Sciences CenterSan AntonioTexasUSA
| |
Collapse
|
10
|
Khan AO, Stapley RJ, Pike JA, Wijesinghe SN, Reyat JS, Almazni I, Machlus KR, Morgan NV. Novel gene variants in patients with platelet-based bleeding using combined exome sequencing and RNAseq murine expression data. J Thromb Haemost 2021; 19:262-268. [PMID: 33021027 DOI: 10.1111/jth.15119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 01/12/2023]
Abstract
Essentials Identifying genetic variants in platelet disorders is challenging due to its heterogenous nature. We combine WES, RNAseq, and python-based bioinformatics to identify novel gene variants. We find novel candidates in patient data by cross-referencing against a murine RNAseq model of thrombopoiesis. This innovative combined bioinformatic approach provides novel data for future research in the field. ABSTRACT: Background The UK Genotyping and Phenotyping of Platelets study has recruited and analyzed 129 patients with suspected heritable bleeding. Previously, 55 individuals had a definitive genetic diagnosis based on whole exome sequencing (WES) and platelet morphological and functional testing. A significant challenge in this field is defining filtering criteria to identify the most likely candidate mutations for diagnosis and further study. Objective Identify candidate gene mutations for the remaining 74 patients with platelet-based bleeding with unknown genetic cause, forming the basis of future re-recruitment and further functional testing and assessment. Methods Using python-based data frame indexing, we first identify and filter all novel and rare variants using a panel of 116 genes known to cause bleeding across the full cohort of WES data. This identified new variants not previously reported in this cohort. We then index the remaining patients, with rare or novel variants in known bleeding genes against a murine RNA sequencing dataset that models proplatelet-forming megakaryocytes. Results Filtering against known genes identified candidate variants in 59 individuals, including novel variants in several known genes. In the remaining cohort of "unknown" patients, indexing against differentially expressed genes revealed candidate gene variants in several novel unreported genes, focusing on 14 patients with a severe clinical presentation. Conclusions We identified candidate mutations in a cohort of patients with no previous genetic diagnosis. This work involves innovative coupling of RNA sequencing and WES to identify candidate variants forming the basis of future study in a significant number of undiagnosed patients.
Collapse
Affiliation(s)
- Abdullah O Khan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rachel J Stapley
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jeremy A Pike
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Susanne N Wijesinghe
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jasmeet S Reyat
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Ibrahim Almazni
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Kellie R Machlus
- Hematology Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Neil V Morgan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
11
|
Quigley CA, Li YG, Brown MR, Pillai SG, Banerjee P, Scott RS, Blum WF, Parks JS. Genetic Polymorphisms Associated with Idiopathic Short Stature and First-Year Response to Growth Hormone Treatment. Horm Res Paediatr 2019; 91:164-174. [PMID: 30970347 DOI: 10.1159/000496989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/14/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS The term idiopathic short stature (ISS) describes short stature of unknown, but likely polygenic, etiology. This study aimed to identify genetic polymorphisms associated with the ISS phenotype, and with growth response to supplemental GH. METHODS Using a case-control analysis we compared the prevalence of "tall" versus "short" alleles at 52 polymorphic loci (17 in growth-related candidate genes, 35 identified in prior genome-wide association studies of adult height) in 94 children with ISS followed in the Genetics and Neuroendocrinology of Short Stature International Study, versus 143 controls from the Fels Longitudinal Study. RESULTS Four variants were nominally associated with ISS using a genotypic model, confirmed by a simultaneous confident inference approach: compared with controls children with ISS had lower odds of "tall" alleles (odds ratio, 95% CI) for GHR (0.52, 0.29-0.96); rs2234693/ESR1 (0.50, 0.25-0.98); rs967417/BMP2 (0.39, 0.17-0.93), and rs4743034/ZNF462 (0.40, 0.18-0.89). Children with ISS also had lower odds of the "tall" allele (A) at the IGFBP3 -202 promoter polymorphism (rs2855744; 0.40, 0.20-0.80) in the simultaneous confident inference analysis. A significant association with 1st-year height SD score increase during GH treatment was observed with rs11205277, located near 4 known genes: MTMR11, SV2A, HIST2H2AA3, and SF3B4; the latter, in which heterozygous mutations occur in Nager acrofacial dysostosis, appears the most relevant gene. CONCLUSIONS In children with ISS we identified associations with "short" alleles at a number of height-related loci. In addition, a polymorphic variant located near SF3B4 was associated with the GH treatment response in our cohort. The findings in our small study warrant further investigation.
Collapse
Affiliation(s)
- Charmian A Quigley
- Endocrinology, Sydney Children's Hospital, Sydney, New South Wales, Australia,
| | - Ying Grace Li
- Lilly Research Laboratories, Indianapolis, Indiana, USA
| | - Milton R Brown
- Pediatric Endocrinology, Emory University, Atlanta, Georgia, USA
| | | | | | | | | | - John S Parks
- Pediatric Endocrinology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
12
|
Liu Y, Yang N, Peng X, Liu G, Zhong H, Liu L. One-lincRNA and five-mRNA based signature for prognosis of multiple myeloma patients undergoing proteasome inhibitors therapy. Biomed Pharmacother 2019; 118:109254. [PMID: 31357080 DOI: 10.1016/j.biopha.2019.109254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/11/2019] [Accepted: 07/19/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma is the second largest malignant tumor of the blood system. Proteasome inhibitors (PIs) currently are effective drugs for some myeloma patients, but their prognosis varies. We extracted the transcriptome expression data and clinical information of myeloma patients from MMRF CoMMpass database, and used the Random Survival Forest Variable Hunting (RSF-VH) algorithm to select 6 highly prognosis-related genes and to develop a 6-genes scoring model, by which the risk score predicted were significantly associated with the progress-free survival (PFS, P<0.001). The median PFS of the high-risk group is 21 months, while it is 29 months in the low-risk group. The scoring model was further validated in the testing cohort. Furthermore, Analysis revealed that the risk score performed better in predicting the multiple myeloma patients' prognosis than the existed staging system, including R-ISS. The risk score is independent with the most existed clinical risk indicators, and the prognostic effectiveness of 6-genes scoring model is homogenous in patients with different clinical observations. Further bioinformatic analysis revealed that the risk score is not only significantly associated with multiple myeloma-related pathways, including immune response, but also with the infiltration of many kinds of immune cells that associated with clinical malignancy. Collectively, the model we developed using one lincRNA and five mRNAs is a robust and effective indicator for myeloma patients' prognosis undergoing proteasome inhibitors therapy.
Collapse
Affiliation(s)
- Yunhe Liu
- Institute of Biomedical Sciences, Fudan University, PR China
| | - Ning Yang
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, PR China
| | - Xueqing Peng
- Institute of Biomedical Sciences, Fudan University, PR China
| | - Gang Liu
- Institute of Biomedical Sciences, Fudan University, PR China.
| | - Hua Zhong
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, PR China.
| | - Lei Liu
- Institute of Biomedical Sciences, Fudan University, PR China.
| |
Collapse
|
13
|
Ruckert MT, de Andrade PV, Santos VS, Silveira VS. Protein tyrosine phosphatases: promising targets in pancreatic ductal adenocarcinoma. Cell Mol Life Sci 2019; 76:2571-2592. [PMID: 30982078 PMCID: PMC11105579 DOI: 10.1007/s00018-019-03095-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 03/25/2019] [Accepted: 04/08/2019] [Indexed: 12/21/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer. It is the fourth leading cause of cancer-related death and is associated with a very poor prognosis. KRAS driver mutations occur in approximately 95% of PDAC cases and cause the activation of several signaling pathways such as mitogen-activated protein kinase (MAPK) pathways. Regulation of these signaling pathways is orchestrated by feedback loops mediated by the balance between protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs), leading to activation or inhibition of its downstream targets. The human PTPome comprises 125 members, and these proteins are classified into three distinct families according to their structure. Since PTP activity description, it has become clear that they have both inhibitory and stimulatory effects on cancer-associated signaling processes and that deregulation of PTP function is closely associated with tumorigenesis. Several PTPs have displayed either tumor suppressor or oncogenic characteristics during the development and progression of PDAC. In this sense, PTPs have been presented as promising candidates for the treatment of human pancreatic cancer, and many PTP inhibitors have been developed since these proteins were first associated with cancer. Nevertheless, some challenges persist regarding the development of effective and safe methods to target these molecules and deliver these drugs. In this review, we discuss the role of PTPs in tumorigenesis as tumor suppressor and oncogenic proteins. We have focused on the differential expression of these proteins in PDAC, as well as their clinical implications and possible targeting for pharmacological inhibition in cancer therapy.
Collapse
Affiliation(s)
- Mariana Tannús Ruckert
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil
| | - Pamela Viani de Andrade
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil
| | - Verena Silva Santos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil
| | - Vanessa Silva Silveira
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
14
|
The Dual-Specificity Phosphatase 10 (DUSP10): Its Role in Cancer, Inflammation, and Immunity. Int J Mol Sci 2019; 20:ijms20071626. [PMID: 30939861 PMCID: PMC6480380 DOI: 10.3390/ijms20071626] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/28/2019] [Accepted: 03/30/2019] [Indexed: 12/22/2022] Open
Abstract
Cancer is one of the most diagnosed diseases in developed countries. Inflammation is a common response to different stress situations including cancer and infection. In those processes, the family of mitogen-activated protein kinases (MAPKs) has an important role regulating cytokine secretion, proliferation, survival, and apoptosis, among others. MAPKs regulate a large number of extracellular signals upon a variety of physiological as well as pathological conditions. MAPKs activation is tightly regulated by phosphorylation/dephosphorylation events. In this regard, the dual-specificity phosphatase 10 (DUSP10) has been described as a MAPK phosphatase that negatively regulates p38 MAPK and c-Jun N-terminal kinase (JNK) in several cellular types and tissues. Several studies have proposed that extracellular signal-regulated kinase (ERK) can be also modulated by DUSP10. This suggests a complex role of DUSP10 on MAPKs regulation and, in consequence, its impact in a wide variety of responses involved in both cancer and inflammation. Here, we review DUSP10 function in cancerous and immune cells and studies in both mouse models and patients that establish a clear role of DUSP10 in different processes such as inflammation, immunity, and cancer.
Collapse
|
15
|
Cullot G, Boutin J, Toutain J, Prat F, Pennamen P, Rooryck C, Teichmann M, Rousseau E, Lamrissi-Garcia I, Guyonnet-Duperat V, Bibeyran A, Lalanne M, Prouzet-Mauléon V, Turcq B, Ged C, Blouin JM, Richard E, Dabernat S, Moreau-Gaudry F, Bedel A. CRISPR-Cas9 genome editing induces megabase-scale chromosomal truncations. Nat Commun 2019; 10:1136. [PMID: 30850590 PMCID: PMC6408493 DOI: 10.1038/s41467-019-09006-2] [Citation(s) in RCA: 299] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/06/2019] [Indexed: 12/19/2022] Open
Abstract
CRISPR-Cas9 is a promising technology for genome editing. Here we use Cas9 nuclease-induced double-strand break DNA (DSB) at the UROS locus to model and correct congenital erythropoietic porphyria. We demonstrate that homology-directed repair is rare compared with NHEJ pathway leading to on-target indels and causing unwanted dysfunctional protein. Moreover, we describe unexpected chromosomal truncations resulting from only one Cas9 nuclease-induced DSB in cell lines and primary cells by a p53-dependent mechanism. Altogether, these side effects may limit the promising perspectives of the CRISPR-Cas9 nuclease system for disease modeling and gene therapy. We show that the single nickase approach could be safer since it prevents on- and off-target indels and chromosomal truncations. These results demonstrate that the single nickase and not the nuclease approach is preferable, not only for modeling disease but also and more importantly for the safe management of future CRISPR-Cas9-mediated gene therapies.
Collapse
MESH Headings
- CRISPR-Associated Protein 9/genetics
- CRISPR-Associated Protein 9/metabolism
- CRISPR-Cas Systems
- Chromosome Deletion
- Chromosomes, Human, Pair 10
- Clustered Regularly Interspaced Short Palindromic Repeats
- DNA/genetics
- DNA/metabolism
- DNA Breaks, Double-Stranded
- Deoxyribonuclease I/genetics
- Deoxyribonuclease I/metabolism
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Gene Editing/methods
- Genetic Therapy/methods
- Genome, Human
- HEK293 Cells
- High-Throughput Nucleotide Sequencing
- Humans
- K562 Cells
- Models, Biological
- Porphyria, Erythropoietic/genetics
- Porphyria, Erythropoietic/metabolism
- Porphyria, Erythropoietic/pathology
- Porphyria, Erythropoietic/therapy
- Primary Cell Culture
- RNA, Guide, CRISPR-Cas Systems/genetics
- RNA, Guide, CRISPR-Cas Systems/metabolism
- Recombinational DNA Repair
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Uroporphyrinogen III Synthetase/genetics
- Uroporphyrinogen III Synthetase/metabolism
Collapse
Affiliation(s)
- Grégoire Cullot
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
| | - Julian Boutin
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
- Biochemistry Laboratory, CHU Bordeaux, 33000, Bordeaux, France
| | - Jérôme Toutain
- Medical genetic laboratory, CHU Bordeaux, 33000, Bordeaux, France
| | - Florence Prat
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
| | - Perrine Pennamen
- Medical genetic laboratory, CHU Bordeaux, 33000, Bordeaux, France
| | - Caroline Rooryck
- Medical genetic laboratory, CHU Bordeaux, 33000, Bordeaux, France
| | - Martin Teichmann
- Univ. Bordeaux, 33000, Bordeaux, France
- UMR 5320, INSERM U1212, ARNA Laboratory, 33000, Bordeaux, France
| | - Emilie Rousseau
- Univ. Bordeaux, 33000, Bordeaux, France
- UMR 5320, INSERM U1212, ARNA Laboratory, 33000, Bordeaux, France
| | - Isabelle Lamrissi-Garcia
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
| | - Véronique Guyonnet-Duperat
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
- Vectorology Platform, 33000, Bordeaux, France
| | - Alice Bibeyran
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
- Vectorology Platform, 33000, Bordeaux, France
| | - Magalie Lalanne
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
| | | | - Béatrice Turcq
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1218, ACTION, 33000, Bordeaux, France
| | - Cécile Ged
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
- Biochemistry Laboratory, CHU Bordeaux, 33000, Bordeaux, France
- Laboratory of excellence, GR-Ex, Imagine institute, 75015, Paris, France
| | - Jean-Marc Blouin
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
- Biochemistry Laboratory, CHU Bordeaux, 33000, Bordeaux, France
- Laboratory of excellence, GR-Ex, Imagine institute, 75015, Paris, France
| | - Emmanuel Richard
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
- Biochemistry Laboratory, CHU Bordeaux, 33000, Bordeaux, France
- Laboratory of excellence, GR-Ex, Imagine institute, 75015, Paris, France
| | - Sandrine Dabernat
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
- Biochemistry Laboratory, CHU Bordeaux, 33000, Bordeaux, France
| | - François Moreau-Gaudry
- Univ. Bordeaux, 33000, Bordeaux, France.
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France.
- Biochemistry Laboratory, CHU Bordeaux, 33000, Bordeaux, France.
- Vectorology Platform, 33000, Bordeaux, France.
- Laboratory of excellence, GR-Ex, Imagine institute, 75015, Paris, France.
| | - Aurélie Bedel
- Univ. Bordeaux, 33000, Bordeaux, France
- INSERM U1035, Biotherapy of genetic diseases, inflammatory disorders and cancers, 33000, Bordeaux, France
- Biochemistry Laboratory, CHU Bordeaux, 33000, Bordeaux, France
- Laboratory of excellence, GR-Ex, Imagine institute, 75015, Paris, France
| |
Collapse
|
16
|
Elson A. Stepping out of the shadows: Oncogenic and tumor-promoting protein tyrosine phosphatases. Int J Biochem Cell Biol 2017; 96:135-147. [PMID: 28941747 DOI: 10.1016/j.biocel.2017.09.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 12/18/2022]
Abstract
Protein tyrosine phosphorylation is critical for proper function of cells and organisms. Phosphorylation is regulated by the concerted but generically opposing activities of tyrosine kinases (PTKs) and tyrosine phosphatases (PTPs), which ensure its proper regulation, reversibility, and ability to respond to changing physiological situations. Historically, PTKs have been associated mainly with oncogenic and pro-tumorigenic activities, leading to the generalization that protein dephosphorylation is anti-oncogenic and hence that PTPs are tumor-suppressors. In many cases PTPs do suppress tumorigenesis. However, a growing body of evidence indicates that PTPs act as dominant oncogenes and drive cell transformation in a number of contexts, while in others PTPs support transformation that is driven by other oncogenes. This review summarizes the known transforming and tumor-promoting activities of the classical, tyrosine specific PTPs and highlights their potential as drug targets for cancer therapy.
Collapse
Affiliation(s)
- Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
17
|
Laudier B, Epiais T, Pâris A, Menuet A, Briault S, Ozsancak C, Perche O. Molecular and clinical analyses with neuropsychological assessment of a case of del(10)(q26.2qter) without intellectual disability: Genomic and transcriptomic combined approach and review of the literature. Am J Med Genet A 2016; 170:1806-12. [PMID: 27113058 DOI: 10.1002/ajmg.a.37677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/03/2016] [Indexed: 11/10/2022]
Abstract
Terminal deletion of the long arm of the chromosome 10 is a rare but well known abnormality, with a large phenotypic variability. Very few data are available about subtelomeric deletion 10q26 patients without intellectual disability. Herein, we report the case of a young adult with a classical 10q26.2qter deletion. She exhibited mainly short stature at birth and in childhood/adulthood without intellectual disability or behavioral problems. After clinical and neuropsychological assessments, we performed genomic array and transcriptomic analysis and compared our results to the data available in the literature. The patient presents a 6.525 Mb heterozygous 10q26.2qter deletion, encompassed 48 genes. Among those genes, DOCK1, C10orf90, and CALY previously described as potential candidate genes for intellectual disability, were partially or completed deleted. Interestingly, they were not deregulated as demonstrated by transcriptomic analysis. This allowed us to suggest that the mechanism involved in the deletion 10qter phenotype is much more complex that only the haploinsufficiency of DOCK1 or other genes encompassed in the deletion. Genomic and transcriptomic combined approach has to be considered to understand this pathogenesis. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Béatrice Laudier
- UMR7355, CNRS, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France.,Department of Genetics, Regional Hospital, Orleans, France
| | | | - Arnaud Pâris
- UMR7355, CNRS, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Arnaud Menuet
- UMR7355, CNRS, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Sylvain Briault
- UMR7355, CNRS, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France.,Department of Genetics, Regional Hospital, Orleans, France
| | - Canan Ozsancak
- Department of Neurology, Regional Hospital, Orleans, France
| | - Olivier Perche
- UMR7355, CNRS, Orleans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France.,Department of Genetics, Regional Hospital, Orleans, France
| |
Collapse
|
18
|
Song L, Jiang W, Liu W, Ji J, Shi T, Zhang J, Xia C. Protein tyrosine phosphatases receptor type D is a potential tumour suppressor gene inactivated by deoxyribonucleic acid methylation in paediatric acute myeloid leukaemia. Acta Paediatr 2016; 105:e132-41. [PMID: 26607758 DOI: 10.1111/apa.13284] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/27/2015] [Accepted: 11/20/2015] [Indexed: 01/09/2023]
Abstract
AIM Protein tyrosine phosphatases receptor type D (PTPRD) is a tumour suppressor gene, and its epigenetic silencing is frequently found in glioblastoma. As aberrant deoxyribonucleic acid (DNA) methylation patterning has been shown to play a role in leukaemogenesis, we studied the promoter methylation, expression profiles and molecular functions of PTPRD in paediatric patients with acute myeloid leukaemia (AML). METHODS Bone marrow specimens were obtained from 32 Chinese patients with a mean age of 7.2 years (range 1.1-16.5). PTPRD and methylation status were evaluated by real-time polymerase chain reaction (PCR) and methylation-specific PCR. Western blot and flow cytometry techniques were also used. RESULTS PTPRD expression was decreased by promoter region methylation in six AML cells and methylated in 21 (65.6%) of the 32 samples. In addition, PTPRD expression could be induced by the DNA demethylating agent 5-aza-2'-deoxycytidine. Furthermore, functional studies showed that overexpression of PTPRD in AML cells inhibited cell proliferation and clonogenicity as well as inducing apoptosis. However, PTPRD knockdown increased cell proliferation. These effects were associated with downregulation of cyclin D1, c-myc and upregulation of Bax. CONCLUSION The results of this study demonstrated that PTPRD was a potential tumour suppressor gene inactivated by DNA methylation in paediatric AML.
Collapse
Affiliation(s)
- Lei Song
- Department of Paediatrics The Second Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Wen Jiang
- Department of Gastroenterology Nantong Third People's Hospital Nantong Jiangsu China
| | - Wei Liu
- Department of Surgery The Second Affiliated Hospital of Nantong University Nantong Jiangsu China
| | - Ju‐Hua Ji
- Department of Gastroenterology Nantong Third People's Hospital Nantong Jiangsu China
| | - Tai‐Feng Shi
- Department of Gastroenterology Nantong Third People's Hospital Nantong Jiangsu China
| | - Jie Zhang
- School of Medicine Nantong University Nantong Jiangsu China
| | - Chun‐Qiu Xia
- Department of Surgery The Second Affiliated Hospital of Nantong University Nantong Jiangsu China
| |
Collapse
|
19
|
St-Denis N, Gupta GD, Lin ZY, Gonzalez-Badillo B, Pelletier L, Gingras AC. Myotubularin-related proteins 3 and 4 interact with polo-like kinase 1 and centrosomal protein of 55 kDa to ensure proper abscission. Mol Cell Proteomics 2015; 14:946-60. [PMID: 25659891 PMCID: PMC4390272 DOI: 10.1074/mcp.m114.046086] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/22/2015] [Indexed: 11/06/2022] Open
Abstract
The myotubularins are a family of phosphatases that dephosphorylate the phosphatidylinositols phosphatidylinositol-3-phosphate and phosphatidylinositol-3,5-phosphate. Several family members are mutated in disease, yet the biological functions of the majority of myotubularins remain unknown. To gain insight into the roles of the individual enzymes, we have used affinity purification coupled to mass spectrometry to identify protein-protein interactions for the myotubularins. The myotubularin interactome comprises 66 high confidence (false discovery rate ≤1%) interactions, including 18 pairwise interactions between individual myotubularins. The results reveal a number of potential signaling contexts for this family of enzymes, including an intriguing, novel role for myotubularin-related protein 3 and myotubularin-related protein 4 in the regulation of abscission, the final step of mitosis in which the membrane bridge remaining between two daughter cells is cleaved. Both depletion and overexpression of either myotubularin-related protein 3 or myotubularin-related protein 4 result in abnormal midbody morphology and cytokinesis failure. Interestingly, myotubularin-related protein 3 and myotubularin-related protein 4 do not exert their effects through lipid regulation at the midbody, but regulate abscission during early mitosis, by interacting with the mitotic kinase polo-like kinase 1, and with centrosomal protein of 55 kDa (CEP55), an important regulator of abscission. Structure-function analysis reveals that, consistent with known intramyotubularin interactions, myotubularin-related protein 3 and myotubularin-related protein 4 interact through their respective coiled coil domains. The interaction between myotubularin-related protein 3 and polo-like kinase 1 relies on the divergent, nonlipid binding Fab1, YOTB, Vac1, and EEA1 domain of myotubularin-related protein 3, and myotubularin-related protein 4 interacts with CEP55 through a short GPPXXXY motif, analogous to endosomal sorting complex required for transport-I components. Disruption of any of these interactions results in abscission failure, by disrupting the proper recruitment of CEP55, and subsequently, of endosomal sorting complex required for transport-I, to the midbody. Our data suggest that myotubularin-related protein 3 and myotubularin-related protein 4 may act as a bridge between CEP55 and polo-like kinase 1, ensuring proper CEP55 phosphorylation and regulating CEP55 recruitment to the midbody. This work provides a novel role for myotubularin-related protein 3/4 heterodimers, and highlights the temporal and spatial complexity of the regulation of cytokinesis.
Collapse
Affiliation(s)
- Nicole St-Denis
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Gagan D Gupta
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Zhen Yuan Lin
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Beatriz Gonzalez-Badillo
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Laurence Pelletier
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; §Department of Molecular Genetics, University of Toronto, Toronto ON M5S 1A8, Canada
| | - Anne-Claude Gingras
- From the ‡Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada; §Department of Molecular Genetics, University of Toronto, Toronto ON M5S 1A8, Canada
| |
Collapse
|
20
|
Keratin 19 expression correlates with poor prognosis in breast cancer. Mol Biol Rep 2014; 41:7729-35. [DOI: 10.1007/s11033-014-3684-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 08/19/2014] [Indexed: 10/24/2022]
|
21
|
Reiterer V, Eyers PA, Farhan H. Day of the dead: pseudokinases and pseudophosphatases in physiology and disease. Trends Cell Biol 2014; 24:489-505. [PMID: 24818526 DOI: 10.1016/j.tcb.2014.03.008] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 03/25/2014] [Accepted: 03/27/2014] [Indexed: 12/19/2022]
Abstract
Pseudophosphatases and pseudokinases are increasingly viewed as integral elements of signaling pathways, and there is mounting evidence that they have frequently retained the ability to interact with cellular 'substrates', and can exert important roles in different diseases. However, these pseudoenzymes have traditionally received scant attention compared to classical kinases and phosphatases. In this review we explore new findings in the emerging pseudokinase and pseudophosphatase fields, and discuss their different modes of action which include exciting new roles as scaffolds, anchors, spatial modulators, traps, and ligand-driven regulators of canonical kinases and phosphatases. Thus, it is now apparent that pseudokinases and pseudophosphatases both support and drive a panoply of signaling networks. Finally, we highlight recent evidence on their involvement in human pathologies, marking them as potential novel drug targets.
Collapse
Affiliation(s)
- Veronika Reiterer
- Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland
| | - Patrick A Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK.
| | - Hesso Farhan
- Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland; Department of Biology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
22
|
Kazi JU, Kabir NN, Rönnstrand L. Protein kinase C (PKC) as a drug target in chronic lymphocytic leukemia. Med Oncol 2013; 30:757. [PMID: 24174318 DOI: 10.1007/s12032-013-0757-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 10/24/2013] [Indexed: 02/07/2023]
Abstract
Protein kinase C (PKC) belongs to a family of ten serine/threonine protein kinases encoded by nine genes. This family of proteins plays critical roles in signal transduction which results in cell proliferation, survival, differentiation and apoptosis. Due to differential subcellular localization and tissue distribution, each member displays distinct signaling characteristics. In this review, we have summarized the roles of PKC family members in chronic lymphocytic leukemia (CLL). CLL is a heterogeneous hematological disorder with survival ranging from months to decades. PKC isoforms are differentially expressed in CLL and play critical roles in CLL pathogenesis. Thus, isoform-specific PKC inhibitors may be an attractive option for CLL treatment.
Collapse
Affiliation(s)
- Julhash U Kazi
- Translational Cancer Research, Lund University, Medicon Village, Building 404:C3, 223 63, Lund, Sweden,
| | | | | |
Collapse
|
23
|
Kabir NN, Rönnstrand L, Kazi JU. The basic helix-loop-helix (bHLH) proteins in breast cancer progression. Med Oncol 2013; 30:666. [DOI: 10.1007/s12032-013-0666-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 07/12/2013] [Indexed: 11/28/2022]
|