1
|
Fonódi M, Nagy L, Boratkó A. Role of Protein Phosphatases in Tumor Angiogenesis: Assessing PP1, PP2A, PP2B and PTPs Activity. Int J Mol Sci 2024; 25:6868. [PMID: 38999976 PMCID: PMC11241275 DOI: 10.3390/ijms25136868] [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: 05/16/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Tumor angiogenesis, the formation of new blood vessels to support tumor growth and metastasis, is a complex process regulated by a multitude of signaling pathways. Dysregulation of signaling pathways involving protein kinases has been extensively studied, but the role of protein phosphatases in angiogenesis within the tumor microenvironment remains less explored. However, among angiogenic pathways, protein phosphatases play critical roles in modulating signaling cascades. This review provides a comprehensive overview of the involvement of protein phosphatases in tumor angiogenesis, highlighting their diverse functions and mechanisms of action. Protein phosphatases are key regulators of cellular signaling pathways by catalyzing the dephosphorylation of proteins, thereby modulating their activity and function. This review aims to assess the activity of the protein tyrosine phosphatases and serine/threonine phosphatases. These phosphatases exert their effects on angiogenic signaling pathways through various mechanisms, including direct dephosphorylation of angiogenic receptors and downstream signaling molecules. Moreover, protein phosphatases also crosstalk with other signaling pathways involved in angiogenesis, further emphasizing their significance in regulating tumor vascularization, including endothelial cell survival, sprouting, and vessel maturation. In conclusion, this review underscores the pivotal role of protein phosphatases in tumor angiogenesis and accentuate their potential as therapeutic targets for anti-angiogenic therapy in cancer.
Collapse
Affiliation(s)
| | | | - Anita Boratkó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (M.F.); (L.N.)
| |
Collapse
|
2
|
Anandakrishnan N, Yi Z, Sun Z, Liu T, Haydak J, Eddy S, Jayaraman P, DeFronzo S, Saha A, Sun Q, Yang D, Mendoza A, Mosoyan G, Wen HH, Schaub JA, Fu J, Kehrer T, Menon R, Otto EA, Godfrey B, Suarez-Farinas M, Leffters S, Twumasi A, Meliambro K, Charney AW, García-Sastre A, Campbell KN, Gusella GL, He JC, Miorin L, Nadkarni GN, Wisnivesky J, Li H, Kretzler M, Coca SG, Chan L, Zhang W, Azeloglu EU. Integrated multiomics implicates dysregulation of ECM and cell adhesion pathways as drivers of severe COVID-associated kidney injury. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.18.24304401. [PMID: 38562892 PMCID: PMC10984064 DOI: 10.1101/2024.03.18.24304401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
COVID-19 has been a significant public health concern for the last four years; however, little is known about the mechanisms that lead to severe COVID-associated kidney injury. In this multicenter study, we combined quantitative deep urinary proteomics and machine learning to predict severe acute outcomes in hospitalized COVID-19 patients. Using a 10-fold cross-validated random forest algorithm, we identified a set of urinary proteins that demonstrated predictive power for both discovery and validation set with 87% and 79% accuracy, respectively. These predictive urinary biomarkers were recapitulated in non-COVID acute kidney injury revealing overlapping injury mechanisms. We further combined orthogonal multiomics datasets to understand the mechanisms that drive severe COVID-associated kidney injury. Functional overlap and network analysis of urinary proteomics, plasma proteomics and urine sediment single-cell RNA sequencing showed that extracellular matrix and autophagy-associated pathways were uniquely impacted in severe COVID-19. Differentially abundant proteins associated with these pathways exhibited high expression in cells in the juxtamedullary nephron, endothelial cells, and podocytes, indicating that these kidney cell types could be potential targets. Further, single-cell transcriptomic analysis of kidney organoids infected with SARS-CoV-2 revealed dysregulation of extracellular matrix organization in multiple nephron segments, recapitulating the clinically observed fibrotic response across multiomics datasets. Ligand-receptor interaction analysis of the podocyte and tubule organoid clusters showed significant reduction and loss of interaction between integrins and basement membrane receptors in the infected kidney organoids. Collectively, these data suggest that extracellular matrix degradation and adhesion-associated mechanisms could be a main driver of COVID-associated kidney injury and severe outcomes.
Collapse
|
3
|
Li Y, Liu Z, Han X, Liang F, Zhang Q, Huang X, Shi X, Huo H, Han M, Liu X, Zhu H, He L, Shen L, Hu X, Wang J, Wang QD, Smart N, Zhou B, He B. Dynamics of Endothelial Cell Generation and Turnover in Arteries During Homeostasis and Diseases. Circulation 2024; 149:135-154. [PMID: 38084582 DOI: 10.1161/circulationaha.123.064301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 10/06/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Endothelial cell (EC) generation and turnover by self-proliferation contributes to vascular repair and regeneration. The ability to accurately measure the dynamics of EC generation would advance our understanding of cellular mechanisms of vascular homeostasis and diseases. However, it is currently challenging to evaluate the dynamics of EC generation in large vessels such as arteries because of their infrequent proliferation. METHODS By using dual recombination systems based on Cre-loxP and Dre-rox, we developed a genetic system for temporally seamless recording of EC proliferation in vivo. We combined genetic recording of EC proliferation with single-cell RNA sequencing and gene knockout to uncover cellular and molecular mechanisms underlying EC generation in arteries during homeostasis and disease. RESULTS Genetic proliferation tracing reveals that ≈3% of aortic ECs undergo proliferation per month in adult mice during homeostasis. The orientation of aortic EC division is generally parallel to blood flow in the aorta, which is regulated by the mechanosensing protein Piezo1. Single-cell RNA sequencing analysis reveals 4 heterogeneous aortic EC subpopulations with distinct proliferative activity. EC cluster 1 exhibits transit-amplifying cell features with preferential proliferative capacity and enriched expression of stem cell markers such as Sca1 and Sox18. EC proliferation increases in hypertension but decreases in type 2 diabetes, coinciding with changes in the extent of EC cluster 1 proliferation. Combined gene knockout and proliferation tracing reveals that Hippo/vascular endothelial growth factor receptor 2 signaling pathways regulate EC proliferation in large vessels. CONCLUSIONS Genetic proliferation tracing quantitatively delineates the dynamics of EC generation and turnover, as well as EC division orientation, in large vessels during homeostasis and disease. An EC subpopulation in the aorta exhibits more robust cell proliferation during homeostasis and type 2 diabetes, identifying it as a potential therapeutic target for vascular repair and regeneration.
Collapse
Affiliation(s)
- Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Zixin Liu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Qianyu Zhang
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
| | - Xiuzhen Huang
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Xin Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Huanhuan Huo
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Maoying Han
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
| | - Xiuxiu Liu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Huan Zhu
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China (L.H.)
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H., J.W.)
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (X.H., J.W.)
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Q.D.W.)
| | - Nicola Smart
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, UK (N.S.)
| | - Bin Zhou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, China (Y.L., Z.L., X. Han, X. Huang, M.H., X.L., H.Z., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (Q.Z., M.H., B.Z.)
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, China (B.Z.)
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (Y.L., X. Han, F.L., X.S., H.H., L.S., B.Z., B.H.)
| |
Collapse
|
4
|
Gabriele C, Aracri F, Prestagiacomo LE, Rota MA, Alba S, Tradigo G, Guzzi PH, Cuda G, Damiano R, Veltri P, Gaspari M. Development of a predictive model to distinguish prostate cancer from benign prostatic hyperplasia by integrating serum glycoproteomics and clinical variables. Clin Proteomics 2023; 20:52. [PMID: 37990292 PMCID: PMC10662699 DOI: 10.1186/s12014-023-09439-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/18/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Prostate Cancer (PCa) represents the second leading cause of cancer-related death in men. Prostate-specific antigen (PSA) serum testing, currently used for PCa screening, lacks the necessary sensitivity and specificity. New non-invasive diagnostic tools able to discriminate tumoral from benign conditions and aggressive (AG-PCa) from indolent forms of PCa (NAG-PCa) are required to avoid unnecessary biopsies. METHODS In this work, 32 formerly N-glycosylated peptides were quantified by PRM (parallel reaction monitoring) in 163 serum samples (79 from PCa patients and 84 from individuals affected by benign prostatic hyperplasia (BPH)) in two technical replicates. These potential biomarker candidates were prioritized through a multi-stage biomarker discovery pipeline articulated in: discovery, LC-PRM assay development and verification phases. Because of the well-established involvement of glycoproteins in cancer development and progression, the proteomic analysis was focused on glycoproteins enriched by TiO2 (titanium dioxide) strategy. RESULTS Machine learning algorithms have been applied to the combined matrix comprising proteomic and clinical variables, resulting in a predictive model based on six proteomic variables (RNASE1, LAMP2, LUM, MASP1, NCAM1, GPLD1) and five clinical variables (prostate dimension, proPSA, free-PSA, total-PSA, free/total-PSA) able to distinguish PCa from BPH with an area under the Receiver Operating Characteristic (ROC) curve of 0.93. This model outperformed PSA alone which, on the same sample set, was able to discriminate PCa from BPH with an AUC of 0.79. To improve the clinical managing of PCa patients, an explorative small-scale analysis (79 samples) aimed at distinguishing AG-PCa from NAG-PCa was conducted. A predictor of PCa aggressiveness based on the combination of 7 proteomic variables (FCN3, LGALS3BP, AZU1, C6, LAMB1, CHL1, POSTN) and proPSA was developed (AUC of 0.69). CONCLUSIONS To address the impelling need of more sensitive and specific serum diagnostic tests, a predictive model combining proteomic and clinical variables was developed. A preliminary evaluation to build a new tool able to discriminate aggressive presentations of PCa from tumors with benign behavior was exploited. This predictor displayed moderate performances, but no conclusions can be drawn due to the limited number of the sample cohort. Data are available via ProteomeXchange with identifier PXD035935.
Collapse
Affiliation(s)
- Caterina Gabriele
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy.
| | - Federica Aracri
- Department of Surgical and Medical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Licia Elvira Prestagiacomo
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | | | | | | | - Pietro Hiram Guzzi
- Department of Surgical and Medical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Giovanni Cuda
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Rocco Damiano
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Pierangelo Veltri
- Department of Surgical and Medical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
- Department of Computer Engineering, Modeling, Electronics and Systems, University of Calabria, 87036 Rende, Italy
| | - Marco Gaspari
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy.
| |
Collapse
|
5
|
Lv Z, Wang T, Cao X, Sun M, Qu Y. The role of receptor‐type protein tyrosine phosphatases in cancer. PRECISION MEDICAL SCIENCES 2023. [DOI: 10.1002/prm2.12090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Zhengyuan Lv
- Department of Medical Genetics, School of Basic Medical Science Nanjing Medical University Nanjing China
| | - Tianming Wang
- Department of Medical Genetics, School of Basic Medical Science Nanjing Medical University Nanjing China
- Central Laboratory, Translational Medicine Research Center The Affiliated Jiangning Hospital with Nanjing Medical University Nanjing China
| | - Xin Cao
- Department of Medical Genetics, School of Basic Medical Science Nanjing Medical University Nanjing China
| | - Mengting Sun
- Biobank of Jiangsu Institute of Cancer Research The Affiliated Cancer Hospital of Nanjing Medical University Nanjing China
| | - Yuan Qu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research The Affiliated Cancer Hospital of Nanjing Medical University Nanjing China
| |
Collapse
|
6
|
The Structure, Function and Regulation of Protein Tyrosine Phosphatase Receptor Type J and Its Role in Diseases. Cells 2022; 12:cells12010008. [PMID: 36611803 PMCID: PMC9818648 DOI: 10.3390/cells12010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Protein tyrosine phosphatase receptor type J (PTPRJ), also known as DEP-1, HPTPη, or CD148, belongs to the R3 subfamily of receptor protein tyrosine phosphatases (RPTPs). It was first identified as an antioncogene due to its protein level being significantly downregulated in most epithelial tumors and cancer cell lines (e.g., colon, lung, thyroid, breast, and pancreas). PTPRJ regulates mouse optic nerve projection by inhibiting the phosphorylation of the erythropoietin-producing hepatocellular carcinoma (Eph) receptor and abelson murine leukemia viral oncogene homolog 1 (c-Abl). PTPRJ is crucial for metabolism. Recent studies have demonstrated that PTPRJ dephosphorylates JAK2 at positions Y813 and Y868 to inhibit leptin signaling. Akt is more phosphorylated at the Ser473 and Thr308 sites in Ptprj-/- mice, suggesting that PTPRJ may be a novel negative regulator of insulin signaling. PTPRJ also plays an important role in balancing the pro- and anti-osteoclastogenic activity of the M-CSF receptor (M-CSFR), and in maintaining NFATc1 expression during the late stages of osteoclastogenesis to promote bone-resorbing osteoclast (OCL) maturation. Furthermore, multiple receptor tyrosine kinases (RTKs) as substrates of PTPRJ are probably a potential therapeutic target for many types of diseases, such as cancer, neurodegenerative diseases, and metabolic diseases, by inhibiting their phosphorylation activity. In light of the important roles that PTPRJ plays in many diseases, this review summarizes the structural features of the protein, its expression pattern, and the physiological and pathological functions of PTPRJ, to provide new ideas for treating PTPRJ as a potential therapeutic target for related metabolic diseases and cancer.
Collapse
|
7
|
Jimenez Y, Paulsen C, Turner E, Iturra S, Cuevas O, Lay-son G, Repetto GM, Rojas M, Calderon JF. Exome Sequencing Identifies Genetic Variants Associated with Extreme Manifestations of the Cardiovascular Phenotype in Marfan Syndrome. Genes (Basel) 2022; 13:genes13061027. [PMID: 35741789 PMCID: PMC9223058 DOI: 10.3390/genes13061027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/06/2022] [Accepted: 06/02/2022] [Indexed: 11/26/2022] Open
Abstract
Marfan Syndrome (MFS) is an autosomal dominant condition caused by variants in the fibrillin-1 (FBN1) gene. Cardinal features of MFS include ectopia lentis (EL), musculoskeletal features and aortic root aneurysm and dissection. Although dissection of the ascending aorta is the main cause of mortality in MFS, the clinical course differs considerably in age of onset and severity, even among individuals who share the same causative variant, suggesting the existence of additional genetic variants that modify the severity of the cardiovascular phenotype in MFS. We recruited MFS patients and classified them into severe (n = 8) or mild aortic phenotype (n = 14) according to age of presentation of the first aorta-related incident. We used Exome Sequencing to identify the genetic variants associated with the severity of aortic manifestations and we performed linkage analysis where suitable. We found five genes associated with severe aortic phenotype and three genes that could be protective for this phenotype in MFS. These genes regulate components of the extracellular matrix, TGFβ pathway and other signaling pathways that are involved in the maintenance of the ECM or angiogenesis. Further studies will be required to understand the functional effect of these variants and explore novel, personalized risk management and, potentially, therapies for these patients.
Collapse
Affiliation(s)
- Yanireth Jimenez
- Doctorado en Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 8320000, Chile; (Y.J.); (M.R.)
| | - Cesar Paulsen
- Servicio de Cirugía Cardiovascular, Instituto Nacional del Tórax, Santiago 7500808, Chile; (C.P.); (E.T.); (S.I.); (O.C.)
| | - Eduardo Turner
- Servicio de Cirugía Cardiovascular, Instituto Nacional del Tórax, Santiago 7500808, Chile; (C.P.); (E.T.); (S.I.); (O.C.)
| | - Sebastian Iturra
- Servicio de Cirugía Cardiovascular, Instituto Nacional del Tórax, Santiago 7500808, Chile; (C.P.); (E.T.); (S.I.); (O.C.)
| | - Oscar Cuevas
- Servicio de Cirugía Cardiovascular, Instituto Nacional del Tórax, Santiago 7500808, Chile; (C.P.); (E.T.); (S.I.); (O.C.)
- Departamento de Cirugía Cardiovascular, Clínica Alemana, Universidad del Desarrollo, Santiago 8320000, Chile
| | - Guillermo Lay-son
- Unidad de Genética, División de Pediatría, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
| | - Gabriela M. Repetto
- Programa de Enfermedades Poco Frecuentes, Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 8320000, Chile;
| | - Marcelo Rojas
- Doctorado en Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 8320000, Chile; (Y.J.); (M.R.)
| | - Juan F. Calderon
- Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago 8320000, Chile
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 8320000, Chile
- Correspondence: ; Tel.: +56-22-578-5778
| |
Collapse
|
8
|
Zhao Y, Gan L, Ren L, Lin Y, Ma C, Lin X. Factors influencing the blood-brain barrier permeability. Brain Res 2022; 1788:147937. [PMID: 35568085 DOI: 10.1016/j.brainres.2022.147937] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) is a dynamic structure that protects the brain from harmful blood-borne, endogenous and exogenous substances and maintains the homeostatic microenvironment. All constituent cell types play indispensable roles in the BBB's integrity, and other structural BBB components, such as tight junction proteins, adherens junctions, and junctional proteins, can control the barrier permeability. Regarding the need to exchange nutrients and toxic materials, solute carriers, ATP-binding case families, and ion transporter, as well as transcytosis regulate the influx and efflux transport, while the difference in localisation and expression can contribute to functional differences in transport properties. Numerous chemical mediators and other factors such as non-physicochemical factors have been identified to alter BBB permeability by mediating the structural components and barrier function, because of the close relationship with inflammation. In this review, we highlight recently gained mechanistic insights into the maintenance and disruption of the BBB. A better understanding of the factors influencing BBB permeability could contribute to supporting promising potential therapeutic targets for protecting the BBB and the delivery of central nervous system drugs via BBB permeability interventions under pathological conditions.
Collapse
Affiliation(s)
- Yibin Zhao
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Gan
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Ren
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yubo Lin
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Congcong Ma
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianming Lin
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
9
|
Hao L, Wu Y, Zhang Y, Ma T, Weng P, Zhang B, Wang J, Pei Q. SdrG gene activated joint surface membrane protein PTPRJ to induce periprosthetic joint infection after total hip arthroplasty. J Clin Pharm Ther 2022; 47:517-522. [PMID: 35014055 DOI: 10.1111/jcpt.13573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/25/2021] [Accepted: 11/03/2021] [Indexed: 11/28/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Up to now, no study focused on the role of SdrG in PJI after THA. To explore the mechanism of periprosthetic joint infection (PJI) after total hip arthroplasty (THA). METHODS Joint fluid and blood were collected from patients with PJI after THA, aseptic loosening of the joints or bacterial infection after traumatic fractures of the extremities alone. The expression of SdrG in the 3 groups was determined by agarose gel electrophoresis. The expression of protein tyrosine phosphatase receptor J (PTPRJ) was measured by immunohistochemistry method. The platelet adhesion rate was determined by biochemical analysis. The content of D-dimer, CRP and ESR in blood was determined by latex agglutination method. Multiple linear correlation analysis was used to analyse the correlation between PJI and the expression of PTPRJ protein, platelet adhesion rate, D-dimer content, CRP and ESR. RESULTS AND DISCUSSION The expression of SdrG and PTPRJ in PJI group was markedly increased compared to the other 2 groups. The platelet adhesion rate in PJI group was markedly larger compared to aseptic loosening and simple wound infection group, and the rate in simple wound infection group was larger than aseptic loosening group. The level of D-dimer, CRP and ESR in PJI group was higher than that of the other groups. The expression of PTPRJ protein, D-dimer content, CRP and ESR was all closely related to PJI, while platelet adhesion rate had no correlation with PJI. WHAT IS NEW AND CONCLUSION SDRG gene around joint prosthesis was over-expressed, which activated joint surface membrane protein PTPRJ and then induced platelet aggregation to reduce joint function.
Collapse
Affiliation(s)
- Linjie Hao
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yanjie Wu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yumin Zhang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Tao Ma
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Pengfei Weng
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Binfei Zhang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Junwei Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qi Pei
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
10
|
Redente EF. How Do We Know What We Are Missing? Loss of Signaling through CD148 Drives Fibroblast Activation in Pulmonary Fibrosis. Am J Respir Crit Care Med 2021; 204:249-251. [PMID: 33891825 PMCID: PMC8513589 DOI: 10.1164/rccm.202103-0737ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Elizabeth F Redente
- Department of Pediatrics National Jewish Health Denver, Colorado and.,Department of Medicine University of Colorado School of Medicine Aurora, Colorado
| |
Collapse
|
11
|
Tsoyi K, Liang X, De Rossi G, Ryter SW, Xiong K, Chu SG, Liu X, Ith B, Celada LJ, Romero F, Robertson MJ, Esposito AJ, Poli S, El-Chemaly S, Perrella MA, Shi Y, Whiteford J, Rosas IO. CD148 Deficiency in Fibroblasts Promotes the Development of Pulmonary Fibrosis. Am J Respir Crit Care Med 2021; 204:312-325. [PMID: 33784491 DOI: 10.1164/rccm.202008-3100oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: CD148/PTRJ (receptor-like protein tyrosine phosphatase η) exerts antifibrotic effects in experimental pulmonary fibrosis via interactions with its ligand syndecan-2; however, the role of CD148 in human pulmonary fibrosis remains incompletely characterized.Objectives: We investigated the role of CD148 in the profibrotic phenotype of fibroblasts in idiopathic pulmonary fibrosis (IPF).Methods: Conditional CD148 fibroblast-specific knockout mice were generated and exposed to bleomycin and then assessed for pulmonary fibrosis. Lung fibroblasts (mouse lung and human IPF lung), and precision-cut lung slices from human patients with IPF were isolated and subjected to experimental treatments. A CD148-activating 18-aa mimetic peptide (SDC2-pep) derived from syndecan-2 was evaluated for its therapeutic potential.Measurements and Main Results: CD148 expression was downregulated in IPF lungs and fibroblasts. In human IPF lung fibroblasts, silencing of CD148 increased extracellular matrix production and resistance to apoptosis, whereas overexpression of CD148 reversed the profibrotic phenotype. CD148 fibroblast-specific knockout mice displayed increased pulmonary fibrosis after bleomycin challenge compared with control mice. CD148-deficient fibroblasts exhibited hyperactivated PI3K/Akt/mTOR signaling, reduced autophagy, and increased p62 accumulation, which induced NF-κB activation and profibrotic gene expression. SDC2-pep reduced pulmonary fibrosis in vivo and inhibited IPF-derived fibroblast activation. In precision-cut lung slices from patients with IPF and control patients, SDC2-pep attenuated profibrotic gene expression in IPF and normal lungs stimulated with profibrotic stimuli.Conclusions: Lung fibroblast CD148 activation reduces p62 accumulation, which exerts antifibrotic effects by inhibiting NF-κB-mediated profibrotic gene expression. Targeting the CD148 phosphatase with activating ligands such as SDC2-pep may represent a potential therapeutic strategy in IPF.
Collapse
Affiliation(s)
- Konstantin Tsoyi
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Xiaoliang Liang
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Giulia De Rossi
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Stefan W Ryter
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Kevin Xiong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Sarah G Chu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Bonna Ith
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Lindsay J Celada
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Freddy Romero
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Matthew J Robertson
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Anthony J Esposito
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Sergio Poli
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - YuanYuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - James Whiteford
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Ivan O Rosas
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|
12
|
Magouliotis DE, Fergadi MP, Christodoulidis G, Svokos AA, Svokos KA, Bareka M, Athanasiou T. In-depth bioinformatic study of the cadherin 5 interactome in patients with thoracic aortic aneurysm unveils 8 novel biomarkers. Eur J Cardiothorac Surg 2021; 61:11-18. [PMID: 34293135 DOI: 10.1093/ejcts/ezab338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/25/2021] [Accepted: 06/12/2021] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVES Thoracic aortic aneurysm (TAA) is characterized by the dilation of the aorta and is associated with poor prognosis if not diagnosed and treated early. In this context, the identification of biomarkers regarding the TAA diagnosis, monitoring and prognosis is crucial. The purpose of the current study was to investigate the differential gene expression profile of the cadherin 5 (CDH5 or VE-Cadherin) gene network in patients with TAA, to propose novel biomarkers. METHODS In silico techniques were used to construct the interactome of the CDH5 network, identify the differentially expressed genes (DEGs) in TAA as compared to healthy controls, and uncover the related molecular functions and the regulating miRNAs. RESULTS Transcriptomic data of one microarray dataset were included, incorporating 43 TAA and 43 control samples. Eight DEGs were identified; 7 were up-regulated and 1 was down-regulated. A molecular signature of 8 genes (CDH5; Calcitonin Receptor-Like Receptor-CALCRL; Activin A Receptor-Like Type 1-ACVRL1, Tryptophanyl-TRNA Synthetase 1-WARS; Junction Plakoglobin-JUP, Protein Tyrosine Phosphatase Receptor Type J-PTPRJ, Purinergic Receptor P2X 4-P2RX4, Kinase Insert Domain Receptor-KDR) were identified as biomarkers associated with TAA. PTPRJ was associated with excellent discrimination and calibration in predicting TAA presentation. Positive correlations were reported regarding the expression of CDH5-CALCRL, CDH5-ACVRL1, CDH5-WARS and CDH5-PTPRJ. Finally, gene set enrichment analysis indicated the molecular functions and miRNA families (hsa-miR-296-5p, hsa-miR-6836-5p, hsa-miR-6132, hsa-miR-27a-5p and hsa-miR-6773-5p) relevant to the 8 biomarkers. CONCLUSIONS These outcomes propose an 8-gene molecular panel associated with TAA.
Collapse
Affiliation(s)
- Dimitrios E Magouliotis
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, UCL, London, UK
- Department of Cardiothoracic Surgery, University of Thessaly, Biopolis, Larissa, Greece
| | - Maria P Fergadi
- Department of Radiology, University of Thessaly, Biopolis, Larissa, Greece
| | | | - Alexis A Svokos
- Department of Obstetrics and Gynecology, Geisinger Medical Center, Danville, PA, USA
| | - Konstantina A Svokos
- Department of Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Metaxia Bareka
- Department of Anesthesiology, University of Thessaly, Biopolis, Larissa, Greece
| | - Thanos Athanasiou
- Department of Surgery and Cancer, Imperial College London, St Mary's Hospital, London, UK
| |
Collapse
|
13
|
Delprat V, Michiels C. A bi-directional dialog between vascular cells and monocytes/macrophages regulates tumor progression. Cancer Metastasis Rev 2021; 40:477-500. [PMID: 33783686 PMCID: PMC8213675 DOI: 10.1007/s10555-021-09958-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/23/2021] [Indexed: 02/06/2023]
Abstract
Cancer progression largely depends on tumor blood vessels as well on immune cell infiltration. In various tumors, vascular cells, namely endothelial cells (ECs) and pericytes, strongly regulate leukocyte infiltration into tumors and immune cell activation, hence the immune response to cancers. Recently, a lot of compelling studies unraveled the molecular mechanisms by which tumor vascular cells regulate monocyte and tumor-associated macrophage (TAM) recruitment and phenotype, and consequently tumor progression. Reciprocally, TAMs and monocytes strongly modulate tumor blood vessel and tumor lymphatic vessel formation by exerting pro-angiogenic and lymphangiogenic effects, respectively. Finally, the interaction between monocytes/TAMs and vascular cells is also impacting several steps of the spread of cancer cells throughout the body, a process called metastasis. In this review, the impact of the bi-directional dialog between blood vascular cells and monocytes/TAMs in the regulation of tumor progression is discussed. All together, these data led to the design of combinations of anti-angiogenic and immunotherapy targeting TAMs/monocyte whose effects are briefly discussed in the last part of this review.
Collapse
Affiliation(s)
- Victor Delprat
- Biochemistry and Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium
| | - Carine Michiels
- Biochemistry and Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium.
| |
Collapse
|
14
|
Shalev M, Arman E, Stein M, Cohen-Sharir Y, Brumfeld V, Kapishnikov S, Royal I, Tuckermann J, Elson A. PTPRJ promotes osteoclast maturation and activity by inhibiting Cbl-mediated ubiquitination of NFATc1 in late osteoclastogenesis. FEBS J 2021; 288:4702-4723. [PMID: 33605542 DOI: 10.1111/febs.15778] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 01/22/2021] [Accepted: 02/18/2021] [Indexed: 11/28/2022]
Abstract
Bone-resorbing osteoclasts (OCLs) are multinucleated phagocytes, whose central roles in regulating bone formation and homeostasis are critical for normal health and development. OCLs are produced from precursor monocytes in a multistage process that includes initial differentiation, cell-cell fusion, and subsequent functional and morphological maturation; the molecular regulation of osteoclastogenesis is not fully understood. Here, we identify the receptor-type protein tyrosine phosphatase PTPRJ as an essential regulator specifically of OCL maturation. Monocytes from PTPRJ-deficient (JKO) mice differentiate and fuse normally, but their maturation into functional OCLs and their ability to degrade bone are severely inhibited. In agreement, mice lacking PTPRJ throughout their bodies or only in OCLs exhibit increased bone mass due to reduced OCL-mediated bone resorption. We further show that PTPRJ promotes OCL maturation by dephosphorylating the M-CSF receptor (M-CSFR) and Cbl, thus reducing the ubiquitination and degradation of the key osteoclastogenic transcription factor NFATc1. Loss of PTPRJ increases ubiquitination of NFATc1 and reduces its amounts at later stages of osteoclastogenesis, thereby inhibiting OCL maturation. PTPRJ thus fulfills an essential and cell-autonomous role in promoting OCL maturation by balancing between the pro- and anti-osteoclastogenic activities of the M-CSFR and maintaining NFATc1 expression during late osteoclastogenesis.
Collapse
Affiliation(s)
- Moran Shalev
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Esther Arman
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Merle Stein
- Institute of Comparative Molecular Endocrinology, University of Ulm, Germany
| | - Yael Cohen-Sharir
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Vlad Brumfeld
- Department of Chemical Research Support, The Weizmann Institute of Science, Rehovot, Israel
| | - Sergey Kapishnikov
- Department of Chemical Research Support, The Weizmann Institute of Science, Rehovot, Israel
| | - Isabelle Royal
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, QC, Canada.,Institut du Cancer de Montréal, QC, Canada.,Department of Medicine, University of Montreal, QC, Canada
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Germany
| | - Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
15
|
Mercier C, Rousseau M, Geraldes P. Growth Factor Deregulation and Emerging Role of Phosphatases in Diabetic Peripheral Artery Disease. Front Cardiovasc Med 2021; 7:619612. [PMID: 33490120 PMCID: PMC7817696 DOI: 10.3389/fcvm.2020.619612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/10/2020] [Indexed: 01/25/2023] Open
Abstract
Peripheral artery disease is caused by atherosclerosis of lower extremity arteries leading to the loss of blood perfusion and subsequent critical ischemia. The presence of diabetes mellitus is an important risk factor that greatly increases the incidence, the progression and the severity of the disease. In addition to accelerated disease progression, diabetic patients are also more susceptible to develop serious impairment of their walking abilities through an increased risk of lower limb amputation. Hyperglycemia is known to alter the physiological development of collateral arteries in response to ischemia. Deregulation in the production of several critical pro-angiogenic factors has been reported in diabetes along with vascular cell unresponsiveness in initiating angiogenic processes. Among the multiple molecular mechanisms involved in the angiogenic response, protein tyrosine phosphatases are potent regulators by dephosphorylating pro-angiogenic tyrosine kinase receptors. However, evidence has indicated that diabetes-induced deregulation of phosphatases contributes to the progression of several micro and macrovascular complications. This review provides an overview of growth factor alterations in the context of diabetes and peripheral artery disease, as well as a description of the role of phosphatases in the regulation of angiogenic pathways followed by an analysis of the effects of hyperglycemia on the modulation of protein tyrosine phosphatase expression and activity. Knowledge of the role of phosphatases in diabetic peripheral artery disease will help the development of future therapeutics to locally regulate phosphatases and improve angiogenesis.
Collapse
Affiliation(s)
- Clément Mercier
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marina Rousseau
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pedro Geraldes
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
16
|
The translation attenuating arginine-rich sequence in the extended signal peptide of the protein-tyrosine phosphatase PTPRJ/DEP1 is conserved in mammals. PLoS One 2020; 15:e0240498. [PMID: 33296397 PMCID: PMC7725344 DOI: 10.1371/journal.pone.0240498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/23/2020] [Indexed: 11/19/2022] Open
Abstract
The signal peptides, present at the N-terminus of many proteins, guide the proteins into cell membranes. In some proteins, the signal peptide is with an extended N-terminal region. Previously, it was demonstrated that the N-terminally extended signal peptide of the human PTPRJ contains a cluster of arginine residues, which attenuates translation. The analysis of the mammalian orthologous sequences revealed that this sequence is highly conserved. The PTPRJ transcripts in placentals, marsupials, and monotremes encode a stretch of 10–14 arginine residues, positioned 11–12 codons downstream of the initiating AUG. The remarkable conservation of the repeated arginine residues in the PTPRJ signal peptides points to their key role. Further, the presence of an arginine cluster in the extended signal peptides of other proteins (E3 ubiquitin-protein ligase, NOTCH3) is noted and indicates a more general importance of this cis-acting mechanism of translational suppression.
Collapse
|
17
|
High expression of PTPRM predicts poor prognosis and promotes tumor growth and lymph node metastasis in cervical cancer. Cell Death Dis 2020; 11:687. [PMID: 32826853 PMCID: PMC7443137 DOI: 10.1038/s41419-020-02826-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 01/01/2023]
Abstract
The prognosis for cervical cancer (CCa) patients with lymph node metastasis (LNM) is dismal. Elucidation of the molecular mechanisms underlying LNM may provide clinical therapeutic strategies for CCa patients with LNM. However, the precise mechanism of LNM in CCa remains unclear. Herein, we demonstrated that protein tyrosine phosphatase receptor type M (PTPRM), identified from TCGA dataset, was markedly upregulated in CCa with LNM and correlated with LNM. Moreover, PTPRM was an independent prognostic factor of CCa patients in multivariate Cox′s proportional hazards model analysis and associated with poor prognosis. Furthermore, through gain-of-function and loss-of-function approaches, we found that PTPRM promoted CCa cells proliferation, migration, invasion, lymphangiogenesis, and LNM. Mechanistically, PTPRM promoted epithelial–mesenchymal transition (EMT) via Src-AKT signaling pathway and induced lymphangiogenesis in a VEGF-C dependent manner, resulting in LNM of CCa. Importantly, knockdown of PTPRM dramatically reduced LNM in vivo, suggesting that PTPRM plays an important role in the LNM of CCa. Taken together, our findings uncover a novel molecular mechanism in the LNM of CCa and identify PTPRM as a novel prognostic factor and potential therapeutic target for LNM in CCa.
Collapse
|
18
|
Luu VZ, Luu AZ, Chowdhury B, Elbardisy O, Pan Y, Al-Omran M, Quan A, Teoh H, Hess DA, Verma S. Disruption of endothelial cell intraflagellar transport protein 88 exacerbates doxorubicin-induced cardiotoxicity. Life Sci 2020; 260:118216. [PMID: 32768582 DOI: 10.1016/j.lfs.2020.118216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/03/2020] [Indexed: 12/21/2022]
Abstract
AIMS Doxorubicin (DOX) is a potent anticancer drug with severe dose-dependent cardiotoxicity. To address this issue, previous research primarily focused on DOX-induced toxicity on cardiomyocytes. However, more recent research has looked into the endothelium as a therapeutic target due to the emerging role of endothelial cells in the support of cardiomyocyte survival and function. MAIN METHODS We investigated a novel role of endothelial cell (EC) primary cilia in the prevention of DOX-mediated cardiotoxicity. Mice lacking EC primary cilia, via the deletion of EC-specific intraflagellar protein 88 (IFT88) expression, were administered DOX (20 mg/kg i.p.), and assessed for survival, cardiac function, cardiac structure changes, and indices of cardiomyocyte injury. KEY FINDINGS DOX-treatment resulted in reduced survival and cardiac function (ejection fraction and fractional shortening) in EC-IFT88-/- mice vs. their similarly treated wild-type littermates. Cardiomyocyte vacuolization, cardiac fibrosis, and serum CK-MB levels were also increased in DOX-treated mice compared to saline-treated controls. However, these parameters were not significantly different when comparing WT and EC-IFT88-/- mice after DOX treatment. SIGNIFICANCE The loss of EC primary cilia accelerated DOX-mediated mortality and reduced cardiac function, suggesting pathways downstream of ciliary-mediated signal transduction as potential targets to promote EC support of cardiomyocyte function during DOX treatment.
Collapse
Affiliation(s)
- Vincent Z Luu
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Albert Z Luu
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Biswajit Chowdhury
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Omar Elbardisy
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Department of Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Yi Pan
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Mohammed Al-Omran
- Division of Vascular Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Quan
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Hwee Teoh
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada; Division of Endocrinology and Metabolism, Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - David A Hess
- Division of Vascular Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Subodh Verma
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
19
|
Sun Y, Li S, Yu W, Chen C, Liu T, Li L, Zhang D, Zhao Z, Gao J, Wang X, Shi D, Liu L. CD148 Serves as a Prognostic Marker of Gastric Cancer and Hinders Tumor Progression by Dephosphorylating EGFR. J Cancer 2020; 11:2667-2678. [PMID: 32201537 PMCID: PMC7065996 DOI: 10.7150/jca.40955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 01/24/2020] [Indexed: 01/06/2023] Open
Abstract
CD148 is a member of the receptor-type protein tyrosine phosphatase family encoded by the PTPRJ gene and has controversial impacts on cancers. In this study, we investigated the clinical significance of CD148 in gastric cancer and the possible mechanisms. Suppressed CD148 expression indicated adverse pathological features and poor outcomes in gastric cancer patients. CD148 overexpression impeded tumor proliferation, motility, and invasiveness, while CD148 knock-down or knockout promoted the ability of gastric cancer cells to grow and metastasize in vitro and in vivo. Mechanistically, CD148 negatively regulated EGFR phosphorylation of multiple tyrosine residues, including Y1173, Y1068, and Y1092, and remarkably inhibited downstream PI3K/AKT and MEK/ERK pathways. In silico analysis revealed that gene deletions or missense/truncated mutations of PTPRJ gene rarely occurred in gastric cancers. Instead, a 3' UTR-specific methylation might regulate CD148 expression, and the potential regulators were TET2 and TET3. Collectively, our results suggest that CD148 is a convincing prognostic marker as well as a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Yiting Sun
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.,Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Song Li
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Wenbin Yu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Cheng Chen
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Teng Liu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Lanbo Li
- Animal Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Di Zhang
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Zeyi Zhao
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Jing Gao
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiao Wang
- Department of Pathology, School of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Duanbo Shi
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Lian Liu
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
20
|
The protein tyrosine phosphatase PTPRJ/DEP-1 contributes to the regulation of the Notch-signaling pathway and sprouting angiogenesis. Angiogenesis 2019; 23:145-157. [DOI: 10.1007/s10456-019-09683-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022]
|
21
|
Lu H, Deng S, Zheng M, Hu K. iTRAQ plasma proteomics analysis for candidate biomarkers of type 2 incipient diabetic nephropathy. Clin Proteomics 2019; 16:33. [PMID: 31384238 PMCID: PMC6668123 DOI: 10.1186/s12014-019-9253-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022] Open
Abstract
Background Diabetic nephropathy is the most frequent cause of end-stage renal disease worldwide. Identification of biomarkers for diabetic nephropathy for early diagnosis may be the key to avoiding damage from this condition. Methods Proteomic iTRAQ technology was first used to identify differentially expressed plasma proteins in type 2 incipient diabetic nephropathy (IDN) using a Q-Exactive mass spectrometer. Results Compared with controls, 57 proteins (32 upregulated and 25 downregulated proteins) were identified. Furthermore, the gelsolin, collectin-11, PTPRJ, and AKAP-7 proteins were confirmed by Western blots as candidate biomarkers for type 2 IDN through ROC analysis. Conclusions These findings offer a theoretical basis for the early treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Hongmei Lu
- 1The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808 China
| | - Shaodong Deng
- 1The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808 China
| | - Minghui Zheng
- 2Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120 China
| | - Kunhua Hu
- 3Proteomics Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| |
Collapse
|
22
|
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: 2.8] [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
|
23
|
Eirin A, Zhu XY, Woollard JR, Tang H, Dasari S, Lerman A, Lerman LO. Metabolic Syndrome Interferes with Packaging of Proteins within Porcine Mesenchymal Stem Cell-Derived Extracellular Vesicles. Stem Cells Transl Med 2019; 8:430-440. [PMID: 30707002 PMCID: PMC6477000 DOI: 10.1002/sctm.18-0171] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 12/29/2018] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) release extracellular vesicles (EVs), which shuttle proteins to recipient cells, promoting cellular repair. We hypothesized that cardiovascular risk factors may alter the pattern of proteins packed within MSC‐derived EVs. To test this, we compared the protein cargo of EVs to their parent MSCs in pigs with metabolic syndrome (MetS) and Lean controls. Porcine MSCs were harvested from abdominal fat after 16 weeks of Lean‐ or MetS‐diet (n = 5 each), and their EVs isolated. Following liquid chromatography mass spectrometry proteomic analysis, proteins were classified based on cellular component, molecular function, and protein class. Five candidate proteins were validated by Western blot. Clustering analysis was performed to identify primary functional categories of proteins enriched in or excluded from EVs. Proteomics analysis identified 6,690 and 6,790 distinct proteins in Lean‐ and MetS‐EVs, respectively. Differential expression analysis revealed that 146 proteins were upregulated and 273 downregulated in Lean‐EVs versus Lean‐MSCs, whereas 787 proteins were upregulated and 185 downregulated in MetS‐EVs versus MetS‐MSCs. Proteins enriched in both Lean‐ and MetS‐EVs participate in vesicle‐mediated transport and cell‐to‐cell communication. Proteins enriched exclusively in Lean‐EVs modulate pathways related to the MSC reparative capacity, including cell proliferation, differentiation, and activation, as well as transforming growth factor‐β signaling. Contrarily, proteins enriched only in MetS‐EVs are linked to proinflammatory pathways, including acute inflammatory response, leukocyte transendothelial migration, and cytokine production. Coculture with MetS‐EVs increased renal tubular cell inflammation. MetS alters the protein cargo of porcine MSC‐derived EVs, selectively packaging specific proinflammatory signatures that may impair their ability to repair damaged tissues. stem cells translational medicine2019;8:430–440
Collapse
Affiliation(s)
- Alfonso Eirin
- Divisions of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiang-Yang Zhu
- Divisions of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - John R Woollard
- Divisions of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Hui Tang
- Divisions of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Surendra Dasari
- Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Amir Lerman
- Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Lilach O Lerman
- Divisions of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
24
|
Meta-analysis of association between Arg326Gln (rs1503185) and Gln276Pro (rs1566734) polymorphisms of PTPRJ gene and cancer risk. J Appl Genet 2019; 60:57-62. [PMID: 30661225 PMCID: PMC6373398 DOI: 10.1007/s13353-019-00481-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/21/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022]
Abstract
Protein tyrosine phosphatase receptor type J (PTPRJ, DEP1) is a tumour suppressor gene that negatively regulates such processes as angiogenesis, cell proliferation and migration and is one of the genes important for tumour development. Similar to other phosphatase genes, PTPRJ is also described as an oncogene. Among various genetic changes characteristic for this gene, single nucleotide polymorphisms (SNPs) constituting benign genetic variants that can modulate its function have been described. We focused on Gln276Pro and Arg326Gln missense polymorphisms and performed a meta-analysis using data from 2930 and 852 patients for Gln276Pro and Arg326Gln respectively in different cancers. A meta-analysis was performed based on five articles accessed via the PubMed and Research Gate databases. Our meta-analysis revealed that for Arg326Gln, the presence of the Arg (C) allele was associated with lower risk of some cancers, the strongest association was observed for colorectal cancer patients, and there was no association between Gln276Pro (G>T) polymorphism and cancer risk. The polymorphisms Arg326Gln and Gln276Pro of the PTPRJ gene are not associated with an increased risk of cancer except for the Arg326Gln polymorphism in colorectal cancer. Large-scale studies should be performed to verify the impact of this SNP on individual susceptibility to colorectal cancer for given individuals.
Collapse
|
25
|
Bilotta A, Dattilo V, D'Agostino S, Belviso S, Scalise S, Bilotta M, Gaudio E, Paduano F, Perrotti N, Florio T, Fusco A, Iuliano R, Trapasso F. A novel splice variant of the protein tyrosine phosphatase PTPRJ that encodes for a soluble protein involved in angiogenesis. Oncotarget 2018; 8:10091-10102. [PMID: 28052032 PMCID: PMC5354644 DOI: 10.18632/oncotarget.14350] [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] [Received: 09/20/2016] [Accepted: 12/13/2016] [Indexed: 02/01/2023] Open
Abstract
PTPRJ is a receptor protein tyrosine phosphatase with tumor suppressor activity. Very little is known about the role of PTPRJ ectodomain, although recently both physiological and synthetic PTPRJ ligands have been identified. A putative shorter spliced variant, coding for a 539 aa protein corresponding to the extracellular N-terminus of PTPRJ, is reported in several databases but, currently, no further information is available. Here, we confirmed that the PTPRJ short isoform (named sPTPRJ) is a soluble protein secreted into the supernatant of both endothelial and tumor cells. Like PTPRJ, also sPTPRJ undergoes post-translational modifications such as glycosylation, as assessed by sPTPRJ immunoprecipitation. To characterize its functional activity, we performed an endothelial cell tube formation assay and a wound healing assay on HUVEC cells overexpressing sPTPRJ and we found that sPTPRJ has a proangiogenic activity. We also showed that sPTPRJ expression down-regulates endothelial adhesion molecules, that is a hallmark of proangiogenic activity. Moreover, sPTPRJ mRNA levels in human high-grade glioma, one of the most angiogenic tumors, are higher in tumor samples compared to controls. Further studies will be helpful not only to clarify the way sPTPRJ works but also to supply clues to circumvent its activity in cancer therapy.
Collapse
Affiliation(s)
- Anna Bilotta
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Vincenzo Dattilo
- Department of Scienze della Salute, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Sabrina D'Agostino
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Stefania Belviso
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Stefania Scalise
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Mariaconcetta Bilotta
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Eugenio Gaudio
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy.,Lymphoma and Genomics Research Program, Institute of Oncology Research (IOR), Bellinzona, Switzerland
| | - Francesco Paduano
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy.,Tecnologica Research Institute, Biomedical Section, Crotone, Italy
| | - Nicola Perrotti
- Department of Scienze della Salute, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Tullio Florio
- Laboratory of Pharmacology, Dept. of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy
| | - Alfredo Fusco
- Istituto di Endocrinologia e Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University Federico II of Napoli, Napoli, Italy
| | - Rodolfo Iuliano
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Francesco Trapasso
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
26
|
Wu P, Li T, Li R, Jia L, Zhu P, Liu Y, Chen Q, Tang D, Yu Y, Li C. 3D genome of multiple myeloma reveals spatial genome disorganization associated with copy number variations. Nat Commun 2017; 8:1937. [PMID: 29203764 PMCID: PMC5715138 DOI: 10.1038/s41467-017-01793-w] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 10/17/2017] [Indexed: 01/06/2023] Open
Abstract
The Hi-C method is widely used to study the functional roles of the three-dimensional (3D) architecture of genomes. Here, we integrate Hi-C, whole-genome sequencing (WGS) and RNA-seq to study the 3D genome architecture of multiple myeloma (MM) and how it associates with genomic variation and gene expression. Our results show that Hi-C interaction matrices are biased by copy number variations (CNVs) and can be used to detect CNVs. Also, combining Hi-C and WGS data can improve the detection of translocations. We find that CNV breakpoints significantly overlap with topologically associating domain (TAD) boundaries. Compared to normal B cells, the numbers of TADs increases by 25% in MM, the average size of TADs is smaller, and about 20% of genomic regions switch their chromatin A/B compartment types. In summary, we report a 3D genome interaction map of aneuploid MM cells and reveal the relationship among CNVs, translocations, 3D genome reorganization, and gene expression regulation. Chromosome conformation capture techniques enable the study of genome organization in cancer cells. Here, the authors use Hi-C, WGS, and RNA-seq to study the 3D genome of multiple myeloma and find that genome disorganization is associated with copy number variations and changes in gene expression.
Collapse
Affiliation(s)
- Pengze Wu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Bioinformatics, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Tingting Li
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Bioinformatics, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ruifeng Li
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Bioinformatics, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Lumeng Jia
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Bioinformatics, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ping Zhu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Bioinformatics, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Yifang Liu
- PKU-Tsinghua-NIBS Graduate Program, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qing Chen
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Bioinformatics, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Daiwei Tang
- PKU-Tsinghua-NIBS Graduate Program, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuezhou Yu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Bioinformatics, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Cheng Li
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Bioinformatics, School of Life Sciences, Peking University, Beijing, 100871, China. .,Center for Statistical Science, Peking University, Beijing, 100871, China.
| |
Collapse
|
27
|
Recent advances in understanding the role of protein-tyrosine phosphatases in development and disease. Dev Biol 2017; 428:283-292. [PMID: 28728679 DOI: 10.1016/j.ydbio.2017.03.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 01/15/2023]
Abstract
Protein-tyrosine phosphatases (PTPs) remove phosphate groups from tyrosine residues, and thereby propagate or inhibit signal transduction, and hence influence cellular processes such as cell proliferation and differentiation. The importance of tightly controlled PTP activity is reflected by the numerous mechanisms employed by the cell to control PTP activity, including a variety of post-translational modifications, and restricted subcellular localization. This review highlights the strides made in the last decade and discusses the important role of PTPs in key aspects of embryonic development: the regulation of stem cell self-renewal and differentiation, gastrulation and somitogenesis during early embryonic development, osteogenesis, and angiogenesis. The tentative importance of PTPs in these processes is highlighted by the diseases that present upon aberrant activity.
Collapse
|
28
|
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: 31] [Impact Index Per Article: 4.4] [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
|
29
|
Zhang XF, Tu R, Li K, Ye P, Cui X. Tumor Suppressor PTPRJ Is a Target of miR-155 in Colorectal Cancer. J Cell Biochem 2017; 118:3391-3400. [PMID: 28316102 DOI: 10.1002/jcb.25995] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/16/2017] [Indexed: 01/02/2023]
Abstract
PTPRJ is known for its antiproliferative role. Loss of heterozygosity (LOH) of PTPRJ has frequently been observed in various human cancers including colorectal cancer (CRC), lung cancer, and breast cancer. However, the function and mechanism of PTPRJ in CRC are not well understood. At the present study, we show that ectopic expression of PTPRJ inhibits cell growth, migration, and invasiveness in CRC cell line HCT116. Moreover, PTPRJ inhibits the tumorigenecity of HCT116 in a xenograft tumor model. MiR-155, the well-known oncomiR in CRC, is identified as an upstream factor of PTPRJ. MiR-155 directly binds to the 3' untranslated region of PTPRJ mRNA and suppresses the mRNA and protein levels of PTPRJ. Furthermore, the growth-promoting and AKT signaling activation effect of miR-155 was abrogated by PTPRJ overexpression, and vice versa. Our study reveals the crucial role of miR-155/PTPRJ/AKT axis in proliferation and migration of CRC cells and suggests a therapeutic potential of PTPRJ. J. Cell. Biochem. 118: 3391-3400, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiao-Fei Zhang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430072, P. R. China
| | - Rongfu Tu
- College of Life Sciences, Wuhan University, Wuhan 430070, P. R. China
| | - Keke Li
- College of Life Sciences, Wuhan University, Wuhan 430070, P. R. China
| | - Pengxiang Ye
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430072, P. R. China
| | - Xiaofeng Cui
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430072, P. R. China
| |
Collapse
|
30
|
Laczmanska I, Skiba P, Karpinski P, Bebenek M, Sasiadek MM. Customized Array Comparative Genomic Hybridization Analysis of 25 Phosphatase-encoding Genes in Colorectal Cancer Tissues. Cancer Genomics Proteomics 2017; 14:69-74. [PMID: 28031238 DOI: 10.21873/cgp.20019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/05/2016] [Accepted: 12/06/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND/AIM Molecular mechanisms of alterations in protein tyrosine phosphatases (PTPs) genes in cancer have been previously described and include chromosomal aberrations, gene mutations, and epigenetic silencing. However, little is known about small intragenic gains and losses that may lead to either changes in expression or enzyme activity and even loss of protein function. MATERIALS AND METHODS The aim of this study was to investigate 25 phosphatase genes using customized array comparative genomic hybridization in 16 sporadic colorectal cancer tissues. RESULTS The analysis revealed two unique small alterations: of 2 kb in PTPN14 intron 1 and of 1 kb in PTPRJ intron 1. We also found gains and losses of whole PTPs gene sequences covered by large chromosome aberrations. CONCLUSION In our preliminary studies using high-resolution custom microarray we confirmed that PTPs are frequently subjected to whole-gene rearrangements in colorectal cancer, and we revealed that non-polymorphic intragenic changes are rare.
Collapse
Affiliation(s)
| | - Pawel Skiba
- Genetics Department, Wroclaw Medical University, Wroclaw, Poland
| | - Pawel Karpinski
- Genetics Department, Wroclaw Medical University, Wroclaw, Poland
| | - Marek Bebenek
- 1st Department of Surgical Oncology, Lower Silesian Oncology Center, Wroclaw, Poland
| | - Maria M Sasiadek
- Genetics Department, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
31
|
Corti F, Simons M. Modulation of VEGF receptor 2 signaling by protein phosphatases. Pharmacol Res 2017; 115:107-123. [PMID: 27888154 PMCID: PMC5205541 DOI: 10.1016/j.phrs.2016.11.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022]
Abstract
Phosphorylation of serines, threonines, and tyrosines is a central event in signal transduction cascades in eukaryotic cells. The phosphorylation state of any particular protein reflects a balance of activity between kinases and phosphatases. Kinase biology has been exhaustively studied and is reasonably well understood, however, much less is known about phosphatases. A large body of evidence now shows that protein phosphatases do not behave as indiscriminate signal terminators, but can function both as negative or positive regulators of specific signaling pathways. Genetic models have also shown that different protein phosphatases play precise biological roles in health and disease. Finally, genome sequencing has unveiled the existence of many protein phosphatases and associated regulatory subunits comparable in number to kinases. A wide variety of roles for protein phosphatase roles have been recently described in the context of cancer, diabetes, hereditary disorders and other diseases. In particular, there have been several recent advances in our understanding of phosphatases involved in regulation of vascular endothelial growth factor receptor 2 (VEGFR2) signaling. The receptor is the principal signaling molecule mediating a wide spectrum of VEGF signal and, thus, is of paramount significance in a wide variety of diseases ranging from cancer to cardiovascular to ophthalmic. This review focuses on the current knowledge about protein phosphatases' regulation of VEGFR2 signaling and how these enzymes can modulate its biological effects.
Collapse
Affiliation(s)
- Federico Corti
- Yale Cardiovascular Research Center, Department of Internal Medicine and Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine and Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|