1
|
Guarnaccia L, Navone SE, Begani L, Barilla E, Garzia E, Campanella R, Miozzo M, Fontana L, Alotta G, Cordiglieri C, Gaudino C, Schisano L, Ampollini A, Riboni L, Locatelli M, Marfia G. Testing calpain inhibition in tumor endothelial cells: novel targetable biomarkers against glioblastoma malignancy. Front Oncol 2024; 14:1355202. [PMID: 39156707 PMCID: PMC11327812 DOI: 10.3389/fonc.2024.1355202] [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: 12/13/2023] [Accepted: 07/01/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Glioblastoma IDH-wildtype (GBM) is the most malignant brain tumor in adults, with a poor prognosis of approximately 15 months after diagnosis. Most patients suffer from a recurrence in <1 year, and this renders GBM a life-threatening challenge. Among molecular mechanisms driving GBM aggressiveness, angiogenesis mediated by GBM endothelial cells (GECs) deserves consideration as a therapeutic turning point. In this scenario, calpains, a family of ubiquitously expressed calcium-dependent cysteine proteases, emerged as promising targets to be investigated as a novel therapeutic strategy and prognostic tissue biomarkers. Methods To explore this hypothesis, GECs were isolated from n=10 GBM biopsies and characterized phenotypically by immunofluorescence. The expression levels of calpains were evaluated by qRT-PCR and Western blot, and their association with patients' prognosis was estimated by Pearson correlation and Kaplan-Meier survival analysis. Calpain targeting efficacy was assessed by a time- and dose-dependent proliferation curve, MTT assay for viability, caspase-3/7 activity, migration and angiogenesis in vitro, and gene and protein expression level modification. Results Immunofluorescence confirmed the endothelial phenotype of our primary GECs. A significant overexpression was observed for calpain-1/2/3 (CAPN) and calpain-small-subunits-1/2 (CAPNS1), whereas calpastatin gene, the calpain natural inhibitor, was reported to be downregulated. A significant negative correlation was observed between CAPN1/CAPNS1 and patient overall survival. GEC challenging revealed that the inhibition of calpain-1 exerts the strongest proapoptotic efficacy, so GEC mortality reached the 80%, confirmed by the increased activity of caspase-3/7. Functional assays revealed a strong affection of in vitro migration and angiogenesis. Gene and protein expression proved a downregulation of MAPK, VEGF/VEGFRs, and Bcl-2, and an upregulation of caspases and Bax-family mediators. Conclusion Overall, the differential expression of calpains and their correlation with patient survival suggest a novel promising target pathway, whose blockade showed encouraging results toward precision medicine strategies.
Collapse
Affiliation(s)
- Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Begani
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Emanuele Garzia
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, Milan, Italy
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, Milan, Italy
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Monica Miozzo
- Medical Genetics, Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
- Medical Genetics Unit, ASST Santi Paolo e Carlo, Milan, Italy
| | - Laura Fontana
- Medical Genetics, Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
- Medical Genetics Unit, ASST Santi Paolo e Carlo, Milan, Italy
| | | | - Chiara Cordiglieri
- Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
| | - Chiara Gaudino
- Department of Neuroradiology, Azienda Ospedaliero-Universitaria Policlinico Umberto I, Rome, Italy
| | - Luigi Schisano
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonella Ampollini
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Marco Locatelli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Medical-Surgical Physiopathology and Transplantation, University of Milan, Milan, Italy
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, Milan, Italy
| |
Collapse
|
2
|
Song L, Shi X, Kovacs L, Han W, John J, Barman SA, Dong Z, Lucas R, Fulton DJR, Verin AD, Su Y. Calpain Promotes LPS-induced Lung Endothelial Barrier Dysfunction via Cleavage of Talin. Am J Respir Cell Mol Biol 2023; 69:678-688. [PMID: 37639326 PMCID: PMC10704117 DOI: 10.1165/rcmb.2023-0009oc] [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: 01/06/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023] Open
Abstract
Acute lung injury (ALI) is characterized by lung vascular endothelial cell (EC) barrier compromise resulting in increased endothelial permeability and pulmonary edema. The infection of gram-negative bacteria that produce toxins like LPS is one of the major causes of ALI. LPS activates Toll-like receptor 4, leading to cytoskeleton reorganization, resulting in lung endothelial barrier disruption and pulmonary edema in ALI. However, the signaling pathways that lead to the cytoskeleton reorganization and lung microvascular EC barrier disruption remain largely unexplored. Here we show that LPS induces calpain activation and talin cleavage into head and rod domains and that inhibition of calpain attenuates talin cleavage, RhoA activation, and pulmonary EC barrier disruption in LPS-treated human lung microvascular ECs in vitro and lung EC barrier disruption and pulmonary edema induced by LPS in ALI in vivo. Moreover, overexpression of calpain causes talin cleavage and RhoA activation, myosin light chain (MLC) phosphorylation, and increases in actin stress fiber formation. Furthermore, knockdown of talin attenuates LPS-induced RhoA activation and MLC phosphorylation and increased stress fiber formation and mitigates LPS-induced lung microvascular endothelial barrier disruption. Additionally, overexpression of talin head and rod domains increases RhoA activation, MLC phosphorylation, and stress fiber formation and enhances lung endothelial barrier disruption. Finally, overexpression of cleavage-resistant talin mutant reduces LPS-induced increases in MLC phosphorylation in human lung microvascular ECs and attenuates LPS-induced lung microvascular endothelial barrier disruption. These results provide the first evidence that calpain mediates LPS-induced lung microvascular endothelial barrier disruption in ALI via cleavage of talin.
Collapse
Affiliation(s)
| | | | - Laszlo Kovacs
- Department of Pharmacology & Toxicology
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | | | - Joseph John
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | | | - Zheng Dong
- Department of Cellular Biology and Anatomy, and
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Rudolf Lucas
- Department of Pharmacology & Toxicology
- Department of Medicine
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | - David J. R. Fulton
- Department of Pharmacology & Toxicology
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | - Alexander D. Verin
- Department of Medicine
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
| | - Yunchao Su
- Department of Pharmacology & Toxicology
- Department of Medicine
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; and
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| |
Collapse
|
3
|
PI3K Isoforms in Vascular Biology, A Focus on the Vascular System-Immune Response Connection. Curr Top Microbiol Immunol 2022; 436:289-309. [DOI: 10.1007/978-3-031-06566-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
4
|
Calpain suppresses cell growth and invasion of glioblastoma multiforme by producing the cleavage of filamin A. Int J Clin Oncol 2020; 25:1055-1066. [PMID: 32103382 DOI: 10.1007/s10147-020-01636-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/12/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Filamin A is the most widely expressed isoform of filamin in mammalian tissues. It can be hydrolyzed by Calpain, producing a 90-kDa carboxyl-terminal fragment (ABP90). Calpeptin is a chemical inhibitor of Calpain, which can inhibit this effect. It has been shown that ABP90 acts as a transcription factor which is involved in mediating cell signaling. However, the significance of ABP90 and its clinical signature with underlying mechanisms have not been well studied in glioblastoma multiforme (GBM). METHODS ABP90 protein was measured in 36 glioma patients by Western blot. Human GBM cell lines U87 and A172 were used to clarify the precise role of ABP90. CCK-8 assay was used to analyze the cell viability. Transwell invasion assay and wound healing assay were used to analyze the migration and invasion. Expression of matrix metalloproteinase 2/tissue inhibitors of metalloproteinase 2 (MMP2/TIMP2) protein was analyzed by Western blot. RESULTS ABP90 protein expression was lower in GBM tissues. The patients with low ABP90 protein expression had a shorter OS time (p = 0.046). After being treated with Calpain, the expression of ABP90 was upregulated, which led to a decline of cell viability, enhanced the efficacy of temozolomide and restrained the cell invasion. Calpeptin could inhibit the effect. The mechanism might be involved in the balance of MMP2/TIMP2. CONCLUSIONS Our present data suggest that ABP90 expression is a significant prognostic factor and may play an important role in cell viability, chemotherapeutic sensitivity and invasion of GBM.
Collapse
|
5
|
Dókus LE, Yousef M, Bánóczi Z. Modulators of calpain activity: inhibitors and activators as potential drugs. Expert Opin Drug Discov 2020; 15:471-486. [DOI: 10.1080/17460441.2020.1722638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Levente Endre Dókus
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Mo’ath Yousef
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Zoltán Bánóczi
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
6
|
Non-proteolytic calpain-6 interacts with VEGFA and promotes angiogenesis by increasing VEGF secretion. Sci Rep 2019; 9:15771. [PMID: 31673071 PMCID: PMC6823460 DOI: 10.1038/s41598-019-52364-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/16/2019] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis is involved in both normal physiological and pathological conditions. Vascular endothelial growth factor (VEGF) is a major factor for promoting angiogenesis. The current anti-VEGF therapies have limited efficacy and significant adverse effects. To find novel targets of VEGFA for angiogenesis inhibition, we performed yeast two-hybrid screening and identified calpain-6 as a novel VEGFA-interaction partner and confirmed the endogenous VEGFA–calpain-6 interaction in mammalian placenta. A domain mapping study revealed that the Gly321–Asp500 domain in calpain-6 is required for the interaction with the C-terminus of the VEGFA protein. The functional significance of the VEGFA–calpain-6 interaction was explored by assessing its effect on angiogenesis in vitro. Whereas forced overexpression of calpain-6 increased the secretion of the VEGF protein and tube formation, knockdown of calpain-6 expression abrogated the calpain-6-mediated VEGF secretion and tube formation in HUVECs. Consistent with the domain mapping result, overexpressing calpain-6 without the VEGFA-interacting domain III (Gly321–Asp500) failed to increase the secretion of VEGF protein. Our results identify calpain-6, an unconventional non-proteolytic calpain, as a novel VEGFA-interacting protein and demonstrate that their interaction is necessary to enhance VEGF secretion. Thus, calpain-6 might be a potential molecular target for angiogenesis inhibition in many diseases.
Collapse
|
7
|
Siuda D, Randriamboavonjy V, Fleming I. Regulation of calpain 2 expression by miR-223 and miR-145. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1862:194438. [PMID: 31634637 DOI: 10.1016/j.bbagrm.2019.194438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/26/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022]
Abstract
Calpain 2 (CAPN2) is a Ca2+-dependent cysteine-protease that is involved in different cellular processes. Despite its important role, little is known about how CAPN2 expression is regulated. This study addressed the potential regulation of CAPN2 by microRNAs (miRNAs) in human endothelial cells. Two miRNAs were found to regulate CAPN2 expression by two distinct mechanisms, one direct and the other indirect. MiR-223 directly targeted CAPN2 by binding to the CAPN2 3'-untranslated region. Mir-223 overexpression decreased CAPN2 protein levels in cultured cells and in mice miR-223 antagonism led to an increase in CAPN2 protein in lung tissue. MiR-145 overexpression also decreased CAPN2 expression but did not affect a CAPN2 luciferase construct, indicating that the effect was indirect. MiR-145 targets histone deacetylase (HDAC) 2, and HDAC inhibition transcriptionally regulated CAPN2 expression by hyperacetylation of the promoter of CAPN2 gene and a subsequent decrease in polymerase 2 binding. Indeed, down regulation of HDAC2 by miR-145 not only decreased CAPN2 protein expression and calpain activity, but also protected paxillin against calpain-dependent degradation. Thus, protein levels of CAPN2 are regulated by miR-223, acting directly on the 3'-untranslated region as well as by miR-145, which acts via an increase in HDAC2. ENZYMES: Calpain 2 (EC 3.4.22.53), histone deacetylase 2 (EC 3.5.1.98).
Collapse
Affiliation(s)
- Daniel Siuda
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany
| | - Voahanginirina Randriamboavonjy
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany.
| |
Collapse
|
8
|
Butler CT, Kennedy SA, Buckley A, Doyle R, Conroy E, Gallagher WM, O'Sullivan J, Kennedy BN. 1,4-dihydroxy quininib attenuates growth of colorectal cancer cells and xenografts and regulates the TIE-2 signaling pathway in patient tumours. Oncotarget 2019; 10:3725-3744. [PMID: 31217905 PMCID: PMC6557215 DOI: 10.18632/oncotarget.26966] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 04/21/2019] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer associated deaths in developed countries. Cancer progression and metastatic spread is reliant on new blood vasculature, or angiogenesis. Tumour-related angiogenesis is regulated by pro- and anti-angiogenic factors secreted from malignant tissue in a stepwise process. Previously we structurally modified the small anti-angiogenic molecule quininib and discovered a more potent anti-angiogenic compound 1, 4 dihydroxy quininib (Q8), an antagonist of cysteinyl leukotriene receptor-1 with VEGF-independent bioactivity. Here, Q8, quininib (Q1) and five structural analogues were assayed for anti-tumorigenic effects in pre-clinical cancer models. Q8 reduced clone formation of the human colorectal cancer cell line HT29-Luc2. Gene silencing of CysLT1 in HT29-Luc2 cells significantly reduced expression of calpain-2. In human ex vivo colorectal cancer tumour explants, Q8 significantly decreased the secretion of both TIE-2 and VCAM-1 expression. In vivo Q8 was well tolerated up to 50 mg/kg by Balb/C mice and significantly more effective at reducing tumour volume in colorectal tumour xenografts compared to the parent drug quininib. In tumour xenografts, Q8 significantly reduced expression of the angiogenic marker calpain-2. In summary, we propose Q8 may act on the TIE-2-Angiopoietin signalling pathway to significantly inhibit the process of tumour angiogenesis in colorectal cancer.
Collapse
Affiliation(s)
- Clare T Butler
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, Dublin, Ireland
| | - Susan A Kennedy
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Amy Buckley
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Ronan Doyle
- Department of Histopathology, Trinity College Dublin Central Pathology Laboratory, St James's Hospital, Dublin, Ireland
| | - Emer Conroy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, Dublin, Ireland
| | - William M Gallagher
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, Dublin, Ireland
| | - Jacintha O'Sullivan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin, Ireland.,These authors contributed equally to this work
| | - Breandán N Kennedy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, Dublin, Ireland.,These authors contributed equally to this work
| |
Collapse
|
9
|
Teng X, Ji C, Zhong H, Zheng D, Ni R, Hill DJ, Xiong S, Fan GC, Greer PA, Shen Z, Peng T. Selective deletion of endothelial cell calpain in mice reduces diabetic cardiomyopathy by improving angiogenesis. Diabetologia 2019; 62:860-872. [PMID: 30778623 PMCID: PMC6702672 DOI: 10.1007/s00125-019-4828-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 01/14/2019] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS The role of non-cardiomyocytes in diabetic cardiomyopathy has not been fully addressed. This study investigated whether endothelial cell calpain plays a role in myocardial endothelial injury and microvascular rarefaction in diabetes, thereby contributing to diabetic cardiomyopathy. METHODS Endothelial cell-specific Capns1-knockout (KO) mice were generated. Conditions mimicking prediabetes and type 1 and type 2 diabetes were induced in these KO mice and their wild-type littermates. Myocardial function and coronary flow reserve were assessed by echocardiography. Histological analyses were performed to determine capillary density, cardiomyocyte size and fibrosis in the heart. Isolated aortas were assayed for neovascularisation. Cultured cardiac microvascular endothelial cells were stimulated with high palmitate. Angiogenesis and apoptosis were analysed. RESULTS Endothelial cell-specific deletion of Capns1 disrupted calpain 1 and calpain 2 in endothelial cells, reduced cardiac fibrosis and hypertrophy, and alleviated myocardial dysfunction in mouse models of diabetes without significantly affecting systemic metabolic variables. These protective effects of calpain disruption in endothelial cells were associated with an increase in myocardial capillary density (wild-type vs Capns1-KO 3646.14 ± 423.51 vs 4708.7 ± 417.93 capillary number/high-power field in prediabetes, 2999.36 ± 854.77 vs 4579.22 ± 672.56 capillary number/high-power field in type 2 diabetes and 2364.87 ± 249.57 vs 3014.63 ± 215.46 capillary number/high-power field in type 1 diabetes) and coronary flow reserve. Ex vivo analysis of neovascularisation revealed more endothelial cell sprouts from aortic rings of prediabetic and diabetic Capns1-KO mice compared with their wild-type littermates. In cultured cardiac microvascular endothelial cells, inhibition of calpain improved angiogenesis and prevented apoptosis under metabolic stress. Mechanistically, deletion of Capns1 elevated the protein levels of β-catenin in endothelial cells of Capns1-KO mice and constitutive activity of calpain 2 suppressed β-catenin protein expression in cultured endothelial cells. Upregulation of β-catenin promoted angiogenesis and inhibited apoptosis whereas knockdown of β-catenin offset the protective effects of calpain inhibition in endothelial cells under metabolic stress. CONCLUSIONS/INTERPRETATION These results delineate a primary role of calpain in inducing cardiac endothelial cell injury and impairing neovascularisation via suppression of β-catenin, thereby promoting diabetic cardiomyopathy, and indicate that calpain is a promising therapeutic target to prevent diabetic cardiac complications.
Collapse
Affiliation(s)
- Xiaomei Teng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
- Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou, China
- Institute for Cardiovascular Science, Soochow University, Suzhou, China
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4G5, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Chen Ji
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Huiting Zhong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Dong Zheng
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4G5, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Rui Ni
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4G5, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - David J Hill
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4G5, Canada
- Department of Medicine, Western University, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Sidong Xiong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Peter A Greer
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Queen's University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital, Soochow University, Suzhou, China
- Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Tianqing Peng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China.
- Critical Illness Research, Lawson Health Research Institute, VRL 6th Floor, A6-140, 800 Commissioners Road, London, ON, N6A 4G5, Canada.
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada.
- Department of Medicine, Western University, London, ON, Canada.
| |
Collapse
|
10
|
Chen Z, Boor PJ, Finnerty CC, Herndon DN, Albrecht T. Calpain-mediated cleavage of p53 in human cytomegalovirus-infected lung fibroblasts. FASEB Bioadv 2019; 1:151-166. [PMID: 32123827 PMCID: PMC6996331 DOI: 10.1096/fba.1028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/23/2018] [Accepted: 10/26/2018] [Indexed: 12/14/2022] Open
Abstract
Endogenous fragments of p53 protein were identified in human cytomegalovirus (HCMV)-infected human lung fibroblasts, particularly a 44-kDa N-terminal fragment [hereafter referred to as p53(ΔCp44)], generated via calpain cleavage. The fragment abundance increased in a biphasic manner, peaking at 6-9 hours and 48 hours post infection. Treatment of LU cells with calpain inhibitors eliminated most detectable p53 fragments. In cell-free experiments, exogenous m-calpain cleavage generated p53(ΔCp44). Attempts to preserve p53 proteins by treating cells with the calpain inhibitor E64d for 6 hours before harvesting increased the sensitivity of p53 to calpain cleavage. p53 in mock-infected cell lysates was much more sensitive to cleavage and degradation by exogenous calpain than that in HCMV-infected cells. The proteasome inhibitor MG132 stabilized p53(ΔCp44), particularly in mock-infected cells. p53(ΔCp44) appeared to be tightly associated with a chromatin-rich fraction. The abundance of p53β was unchanged over a 96-h time course and very similar in mock- and HCMV-infected cells, making it unlikely that p53(ΔCp44) was p53β. The biological activities of this and other fragments lacking C-terminal sequences are unknown, but deserve further investigation, given the association of p53(ΔCp44) with the chromatin-rich (or buffer C insoluble) fraction in HCMV-infected cells.
Collapse
Affiliation(s)
- Zhenping Chen
- Department of Microbiology and ImmunologyUniversity of Texas Medical BranchGalvestonTexas
- Department of PathologyUniversity of Texas Medical BranchGalvestonTexas
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTexas
| | - Paul J. Boor
- Department of PathologyUniversity of Texas Medical BranchGalvestonTexas
- Shriners Hospitals for Children—GalvestonGalvestonTexas
| | - Celeste C. Finnerty
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTexas
- Shriners Hospitals for Children—GalvestonGalvestonTexas
| | - David N. Herndon
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTexas
- Shriners Hospitals for Children—GalvestonGalvestonTexas
| | - Thomas Albrecht
- Department of Microbiology and ImmunologyUniversity of Texas Medical BranchGalvestonTexas
- Infectious Disease and Toxicology Optical Imaging CoreUniversity of Texas Medical BranchGalvestonTexas
| |
Collapse
|
11
|
Calcium influx-mediated translocation of m-calpain induces Ku80 cleavage and enhances the Ku80-related DNA repair pathway. Oncotarget 2017; 7:30831-44. [PMID: 27121057 PMCID: PMC5058721 DOI: 10.18632/oncotarget.8791] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 04/01/2016] [Indexed: 01/14/2023] Open
Abstract
Proteomic analysis of ionomycin-treated and untreated mammary epithelial MCF10A cells elucidated differences in Ku80 cleavage. Ku80, a subunit of the Ku protein complex, is an initiator of the non-homologous, end-joining (NHEJ), double-strand breaks (DSBs) repair pathway. The nuclear Ku80 was cleaved in a calcium concentration-dependent manner by m-calpain but not by m-calpain. The cleavage of nuclear Ku80 at its α/β domain was validated by Western blotting analysis using flag-tagged expression vectors of truncated versions of Ku80 and a flag antibody and was confirmed in m-calpain knock-down cells and in vitro cell-free evaluation with recombinant proteins of calpains, Ku70, and Ku80. In addition, the cleaved Ku80 still formed a Ku heterodimer and promoted DNA DSB repair activity. Taken together, these findings indicate that translocated m-calpain enhances the NHEJ pathway through the cleavage of Ku80. Based on the present study, m-calpain in DNA repair pathways might be a novel anticancer drug target, or its mechanism might be a possible route for resistance acquisition of DNA damage-inducing chemotherapeutics.
Collapse
|
12
|
Abstract
Receptor signaling relays on intracellular events amplified by secondary and tertiary messenger molecules. In cardiomyocytes and smooth muscle cells, cyclic AMP (cAMP) and subsequent calcium (Ca2+) fluxes are the best characterized receptor-regulated signaling events. However, most of receptors able to modify contractility and other intracellular responses signal through a variety of other messengers, and whether these signaling events are interconnected has long remained unclear. For example, the PI3K (phosphoinositide 3-kinase) pathway connected to the production of the lipid second messenger PIP3/PtdIns(3,4,5)P3 (phosphatidylinositol (3,4,5)-trisphosphate) is potentially involved in metabolic regulation, activation of hypertrophy, and survival pathways. Recent studies, highlighted in this review, started to interconnect PI3K pathway activation to Ca2+ signaling. This interdependency, by balancing contractility with metabolic control, is crucial for cells of the cardiovascular system and is emerging to play key roles in disease development. Better understanding of the interplay between Ca2+ and PI3K signaling is, thus, expected to provide new ground for therapeutic intervention. This review explores the emerging molecular mechanisms linking Ca2+ and PI3K signaling in health and disease.
Collapse
Affiliation(s)
- Alessandra Ghigo
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue)
| | - Muriel Laffargue
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue)
| | - Mingchuan Li
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue)
| | - Emilio Hirsch
- From the Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Italy (A.G., M. Li, E.H.); and INSERM U1048, I2MC and Université Toulouse III, France (M. Laffargue).
| |
Collapse
|
13
|
Zhang Y, Liu NM, Wang Y, Youn JY, Cai H. Endothelial cell calpain as a critical modulator of angiogenesis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1326-1335. [PMID: 28366876 DOI: 10.1016/j.bbadis.2017.03.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 03/04/2017] [Accepted: 03/28/2017] [Indexed: 12/13/2022]
Abstract
Calpains are a family of calcium-dependent non-lysosomal cysteine proteases. In particular, calpains residing in the endothelial cells play important roles in angiogenesis. It has been shown that calpain activity can be increased in endothelial cells by growth factors, primarily vascular endothelial growth factor (VEGF). VEGF/VEGFR2 induces calpain 2 dependent activation of PI3K/AMPK/Akt/eNOS pathway, and consequent nitric oxide production and physiological angiogenesis. Under pathological conditions such as tumor angiogenesis, endothelial calpains can be activated by hypoxia. This review focuses on the molecular regulatory mechanisms of calpain activation, and the newly identified mechanistic roles and downstream signaling events of calpains in physiological angiogenesis, and in the conditions of pathological tumor angiogenesis and diabetic wound healing, as well as retinopathy and atherosclerosis that are also associated with an increase in calpain activity. Further discussed include the differential strategies of modulating angiogenesis through manipulating calpain expression/activity in different pathological settings. Targeted limitation of angiogenesis in cancer and targeted promotion of angiogenesis in diabetic wound healing via modulations of calpains and calpain-dependent signaling mechanisms are of significant translational potential. Emerging strategies of tissue-specific targeting, environment-dependent targeting, and genome-targeted editing may turn out to be effective regimens for targeted manipulation of angiogenesis through calpain pathways, for differential treatments including both attenuation of tumor angiogenesis and potentiation of diabetic angiogenesis.
Collapse
Affiliation(s)
- Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Norika Mengchia Liu
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Yongchen Wang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA.
| |
Collapse
|
14
|
Kovacs L, Su Y. Redox-Dependent Calpain Signaling in Airway and Pulmonary Vascular Remodeling in COPD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:139-160. [PMID: 29047085 PMCID: PMC7036267 DOI: 10.1007/978-3-319-63245-2_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The calcium-dependent cytosolic, neutral, thiol endopeptidases, calpains, perform limited cleavage of their substrates thereby irreversibly changing their functions. Calpains have been shown to be involved in several physiological processes such as cell motility, proliferation, cell cycle, signal transduction, and apoptosis. Overactivation of calpain or mutations in the calpain genes contribute to a number of pathological conditions including neurodegenerative disorders, rheumatoid arthritis, cancer, and lung diseases. High concentrations of reactive oxygen and nitrogen species (RONS) originated from cigarette smoke or released by numerous cell types such as activated inflammatory cells and other respiratory cells cause oxidative and nitrosative stress contributing to the pathogenesis of COPD. RONS and calpain play important roles in the development of airway and pulmonary vascular remodeling in COPD. Published data show that increased RONS production is associated with increased calpain activation and/or elevated calpain protein level, leading to epithelial or endothelial barrier dysfunction, neovascularization, lung inflammation, increased smooth muscle cell proliferation, and deposition of extracellular matrix protein. Further investigation of the redox-dependent calpain signaling may provide future targets for the prevention and treatment of COPD.
Collapse
Affiliation(s)
- Laszlo Kovacs
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, 30912, USA.
| |
Collapse
|
15
|
Butler CT, Reynolds AL, Tosetto M, Dillon ET, Guiry PJ, Cagney G, O'Sullivan J, Kennedy BN. A Quininib Analogue and Cysteinyl Leukotriene Receptor Antagonist Inhibits Vascular Endothelial Growth Factor (VEGF)-independent Angiogenesis and Exerts an Additive Antiangiogenic Response with Bevacizumab. J Biol Chem 2016; 292:3552-3567. [PMID: 28035003 DOI: 10.1074/jbc.m116.747766] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 12/19/2016] [Indexed: 12/31/2022] Open
Abstract
Excess blood vessel growth contributes to the pathology of metastatic cancers and age-related retinopathies. Despite development of improved treatments, these conditions are associated with high economic costs and drug resistance. Bevacizumab (Avastin®), a monoclonal antibody against vascular endothelial growth factor (VEGF), is used clinically to treat certain types of metastatic cancers. Unfortunately, many patients do not respond or inevitably become resistant to bevacizumab, highlighting the need for more effective antiangiogenic drugs with novel mechanisms of action. Previous studies discovered quininib, an antiangiogenic small molecule antagonist of cysteinyl leukotriene receptors 1 and 2 (CysLT1 and CysLT2). Here, we screened a series of quininib analogues and identified a more potent antiangiogenic novel chemical entity (IUPAC name (E)-2-(2-quinolin-2-yl-vinyl)-benzene-1,4-diol HCl) hereafter designated Q8. Q8 inhibits developmental angiogenesis in Tg(fli1:EGFP) zebrafish and inhibits human microvascular endothelial cell (HMEC-1) proliferation, tubule formation, and migration. Q8 elicits antiangiogenic effects in a VEGF-independent in vitro model of angiogenesis and exerts an additive antiangiogenic response with the anti-VEGF biologic bevacizumab. Cell-based receptor binding assays confirm that Q8 is a CysLT1 antagonist and is sufficient to reduce cellular levels of NF-κB and calpain-2 and secreted levels of the proangiogenic proteins intercellular adhesion molecule-1, vascular cell adhesion protein-1, and VEGF. Distinct reductions of VEGF by bevacizumab explain the additive antiangiogenic effects observed in combination with Q8. In summary, Q8 is a more effective antiangiogenic drug compared with quininib. The VEGF-independent activity coupled with the additive antiangiogenic response observed in combination with bevacizumab demonstrates that Q8 offers an alternative therapeutic strategy to combat resistance associated with conventional anti-VEGF therapies.
Collapse
Affiliation(s)
- Clare T Butler
- From the UCD School of Biomolecular and Biomedical Science, UCD Conway Institute and
| | - Alison L Reynolds
- From the UCD School of Biomolecular and Biomedical Science, UCD Conway Institute and
| | - Miriam Tosetto
- Centre for Colorectal Disease, St. Vincent's University Hospital, Dublin 4, Ireland, and
| | - Eugene T Dillon
- From the UCD School of Biomolecular and Biomedical Science, UCD Conway Institute and
| | - Patrick J Guiry
- UCD School of Chemistry, UCD Centre for Synthesis and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - Gerard Cagney
- From the UCD School of Biomolecular and Biomedical Science, UCD Conway Institute and
| | - Jacintha O'Sullivan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Breandán N Kennedy
- From the UCD School of Biomolecular and Biomedical Science, UCD Conway Institute and
| |
Collapse
|
16
|
Zhang Y, Li Q, Youn JY, Cai H. Protein Phosphotyrosine Phosphatase 1B (PTP1B) in Calpain-dependent Feedback Regulation of Vascular Endothelial Growth Factor Receptor (VEGFR2) in Endothelial Cells: IMPLICATIONS IN VEGF-DEPENDENT ANGIOGENESIS AND DIABETIC WOUND HEALING. J Biol Chem 2016; 292:407-416. [PMID: 27872190 DOI: 10.1074/jbc.m116.766832] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Indexed: 01/13/2023] Open
Abstract
The VEGF/VEGFR2/Akt/eNOS/NO pathway is essential to VEGF-induced angiogenesis. We have previously discovered a novel role of calpain in mediating VEGF-induced PI3K/AMPK/Akt/eNOS activation through Ezrin. Here, we sought to identify possible feedback regulation of VEGFR2 by calpain via its substrate protein phosphotyrosine phosphatase 1B (PTP1B), and the relevance of this pathway to VEGF-induced angiogenesis, especially in diabetic wound healing. Overexpression of PTP1B inhibited VEGF-induced VEGFR2 and Akt phosphorylation in bovine aortic endothelial cells, while PTP1B siRNA increased both, implicating negative regulation of VEGFR2 by PTP1B. Calpain inhibitor ALLN induced VEGFR2 activation, which can be completely blocked by PTP1B overexpression. Calpain activation induced by overexpression or Ca/A23187 resulted in PTP1B cleavage, which can be blocked by ALLN. Moreover, calpain activation inhibited VEGF-induced VEGFR2 phosphorylation, which can be restored by PTP1B siRNA. These data implicate calpain/PTP1B negative feedback regulation of VEGFR2, in addition to the primary signaling pathway of VEGF/VEGFR2/calpain/PI3K/AMPK/Akt/eNOS. We next examined a potential role of PTP1B in VEGF-induced angiogenesis. Endothelial cells transfected with PTP1B siRNA showed faster wound closure in response to VEGF. Aortic discs isolated from PTP1B siRNA-transfected mice also had augmented endothelial outgrowth. Importantly, PTP1B inhibition and/or calpain overexpression significantly accelerated wound healing in STZ-induced diabetic mice. In conclusion, our data for the first time demonstrate a calpain/PTP1B/VEGFR2 negative feedback loop in the regulation of VEGF-induced angiogenesis. Modulation of local PTP1B and/or calpain activities may prove beneficial in the treatment of impaired wound healing in diabetes.
Collapse
Affiliation(s)
- Yixuan Zhang
- From the Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), California 90095
| | - Qiang Li
- From the Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), California 90095
| | - Ji Youn Youn
- From the Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), California 90095
| | - Hua Cai
- From the Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), California 90095
| |
Collapse
|
17
|
Coly PM, Perzo N, Le Joncour V, Lecointre C, Schouft MT, Desrues L, Tonon MC, Wurtz O, Gandolfo P, Castel H, Morin F. Chemotactic G protein-coupled receptors control cell migration by repressing autophagosome biogenesis. Autophagy 2016; 12:2344-2362. [PMID: 27715446 DOI: 10.1080/15548627.2016.1235125] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chemotactic migration is a fundamental behavior of cells and its regulation is particularly relevant in physiological processes such as organogenesis and angiogenesis, as well as in pathological processes such as tumor metastasis. The majority of chemotactic stimuli activate cell surface receptors that belong to the G protein-coupled receptor (GPCR) superfamily. Although the autophagy machinery has been shown to play a role in cell migration, its mode of regulation by chemotactic GPCRs remains largely unexplored. We found that ligand-induced activation of 2 chemotactic GPCRs, the chemokine receptor CXCR4 and the urotensin 2 receptor UTS2R, triggers a marked reduction in the biogenesis of autophagosomes, in both HEK-293 and U87 glioblastoma cells. Chemotactic GPCRs exert their anti-autophagic effects through the activation of CAPNs, which prevent the formation of pre-autophagosomal vesicles from the plasma membrane. We further demonstrated that CXCR4- or UTS2R-induced inhibition of autophagy favors the formation of adhesion complexes to the extracellular matrix and is required for chemotactic migration. Altogether, our data reveal a new link between GPCR signaling and the autophagy machinery, and may help to envisage therapeutic strategies in pathological processes such as cancer cell invasion.
Collapse
Affiliation(s)
- Pierre-Michaël Coly
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| | - Nicolas Perzo
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| | - Vadim Le Joncour
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| | - Céline Lecointre
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| | - Marie-Thérèse Schouft
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| | - Laurence Desrues
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| | - Marie-Christine Tonon
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| | - Olivier Wurtz
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| | - Pierrick Gandolfo
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| | - Hélène Castel
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| | - Fabrice Morin
- a Normandie Univ, UNIROUEN, INSERM, DC2N , Rouen , France.,b Institute for Research and Innovation in Biomedicine (IRIB) , Rouen , France
| |
Collapse
|
18
|
The inhibition of calpains ameliorates vascular restenosis through MMP2/TGF-β1 pathway. Sci Rep 2016; 6:29975. [PMID: 27453531 PMCID: PMC4958998 DOI: 10.1038/srep29975] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/28/2016] [Indexed: 01/16/2023] Open
Abstract
Restenosis limits the efficacy of vascular percutaneous intervention, in which vascular smooth muscle cell (VSMC) proliferation and activation of inflammation are two primary causal factors. Calpains influence VSMC proliferation and collagen synthesis. However, the roles of calpastatin and calpains in vascular restenosis remain unclear. Here, restenosis was induced by ligating the left carotid artery, and VSMCs were pretreated with platelet-derived growth factor (PDGF)-BB. Adenovirus vector carrying MMP2 sequence and specific small interfering RNA against calpain-1/2 were introduced. Finally, restenosis enhanced the expression of calpain-1/2, but reduced calpastatin content. In calpastatin transgenic mice, lumen narrowing was attenuated gradually and peaked on days 14-21. Cell proliferation and migration as well as collagen synthesis were inhibited in transgenic mice, and expression of calpain-1/2 and MMP2/transforming growth factor-β1 (TGF-β1). Consistently, in VSMCs pretreated with PDGF-BB, calpastatin induction and calpains inhibition suppressed the proliferation and migration of VSMCs and collagen synthesis, and reduced expression of calpain-1/2 and MMP2/TGF-β1. Moreover, simvastatin improved restenosis indicators by suppressing the HIF-1α/calpains/MMP2/TGF-β1 pathway. However, MMP2 supplementation eliminated the vascular protection of calpastatin induction and simvastatin. Collectively, calpains inhibition plays crucial roles in vascular restenosis by preventing neointimal hyperplasia at the early stage via suppression of the MMP2/TGF-β1 pathway.
Collapse
|
19
|
Kovacs L, Han W, Rafikov R, Bagi Z, Offermanns S, Saido TC, Black SM, Su Y. Activation of Calpain-2 by Mediators in Pulmonary Vascular Remodeling of Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2016; 54:384-93. [PMID: 26248159 PMCID: PMC4821035 DOI: 10.1165/rcmb.2015-0151oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/30/2015] [Indexed: 12/27/2022] Open
Abstract
Calpain mediates collagen synthesis and cell proliferation and plays an important role in pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). In the present study, we investigated whether and how calpain is activated by PAH mediators in pulmonary artery smooth muscle cells (PASMCs). These data show that smooth muscle-specific knockout of calpain attenuated and knockout of calpastatin potentiated pulmonary vascular remodeling and pulmonary hypertension. Treatment of PASMCs with the PAH mediators platelet-derived growth factor (PDGF), serotonin, H2O2, endothelin-1, and IL-6 caused significant increases in calpain activity, cell proliferation, and collagen-I protein level without changes in protein levels of calpain-1 and -2. The calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis (acetoxymethyl ester) (BAPTA/AM) did not affect calpain activation, but the extracellular signal-regulated kinase (ERK) 1/2 inhibitor PD98059 and knocking down of calpain-2 prevented calpain activation in PAH mediator-treated PASMCs. Mass spectrometry data showed that the phosphorylation of calpain-2 at serine (Ser) 50 was increased and the phosphorylation of calpain-2 at Ser369 was decreased in PDGF-treated PASMCs. The PDGF-induced increase in Ser50 phosphorylation of calpain-2 was prevented by PD98059, whereas dephosphorylation of calpain-2 at Ser369 was blocked by the protein phosphatase 2A inhibitor fostriecin. Furthermore, smooth muscle of pulmonary arteries in PAH animal models and patients with PAH showed higher levels of phospho-Ser50-calpain-2 (P-Ser50) and lower levels of phospho-Ser369-calpain-2 (P-Ser369). These data support that calpain modulates pulmonary vascular remodeling in PAH. PAH mediator-induced activation of calpain is caused by ERK1/2-dependent phosphorylation of calpain-2 at Ser50 and protein phosphatase 2A-dependent dephosphorylation of calpain-2 at Ser369 in pulmonary vascular remodeling of PAH.
Collapse
MESH Headings
- Animals
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Calpain/genetics
- Calpain/metabolism
- Disease Models, Animal
- Enzyme Activation
- Enzyme Activators/pharmacology
- Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/metabolism
- HEK293 Cells
- Humans
- Hypertension, Pulmonary/enzymology
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypoxia/complications
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- Protein Phosphatase 2/antagonists & inhibitors
- Protein Phosphatase 2/metabolism
- Pulmonary Artery/drug effects
- Pulmonary Artery/enzymology
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- RNA Interference
- Signal Transduction
- Transfection
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
| | | | - Ruslan Rafikov
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Zsolt Bagi
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Goethe University, Frankfurt, Germany; and
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama, Japan
| | - Stephen M. Black
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Yunchao Su
- Departments of Pharmacology and Toxicology
- Medicine, and
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| |
Collapse
|
20
|
Yıldırım C, Favre J, Weijers EM, Fontijn RD, van Wijhe MH, van Vliet SJ, Boon RA, Koolwijk P, van der Pouw Kraan TCTM, Horrevoets AJG. IFN-β affects the angiogenic potential of circulating angiogenic cells by activating calpain 1. Am J Physiol Heart Circ Physiol 2015; 309:H1667-78. [DOI: 10.1152/ajpheart.00810.2014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 09/01/2015] [Indexed: 01/15/2023]
Abstract
Circulating angiogenic cells (CACs) are monocyte-derived cells with endothelial characteristics, which contribute to both angiogenesis and arteriogenesis in a paracrine way. Interferon-β (IFN-β) is known to inhibit these divergent processes in animals and patients. We hypothesized that IFN-β might act by affecting the differentiation and function of CACs. CACs were cultured from peripheral blood mononuclear cells and phenotypically characterized by surface expression of monocytic and endothelial markers. IFN-β significantly reduced the number of CACs by 18–64%. Apoptosis was not induced by IFN-β, neither in mononuclear cells during differentiation, nor after maturation to CACs. Rather, IFN-β impaired adhesion to, and spreading on, fibronectin, which was dependent on α5β1 (VLA-5)-integrin. IFN-β affected the function of VLA-5 in mature CACs, leading to rounding and detachment of cells, by induction of calpain 1 activity. Cell rounding and detachment was completely reversed by inhibition of calpain 1 activity in mature CACs. During in vitro capillary formation, CAC addition and calpain 1 inhibition enhanced sprouting of endothelial cells to a comparable extent, but were not sufficient to rescue tube formation in the presence of IFN-β. We show that the IFN-β-induced reduction of the numbers of in vitro differentiated CACs is based on activation of calpain 1, resulting in an attenuated adhesion to extracellular matrix proteins via VLA-5. In vivo, this could lead to inhibition of vessel formation due to reduction of the locally recruited CAC numbers and their paracrine angiogenic factors.
Collapse
Affiliation(s)
- Cansu Yıldırım
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Julie Favre
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ester M. Weijers
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; and
| | - Ruud D. Fontijn
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Michiel H. van Wijhe
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; and
| | - Sandra J. van Vliet
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Reinier A. Boon
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Pieter Koolwijk
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; and
| | | | - Anton J. G. Horrevoets
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Hutchinson TE, Patel JM. Peptide-stimulated angiogenesis: Role of lung endothelial caveolar signaling and nitric oxide. Nitric Oxide 2015; 51:43-51. [PMID: 26537637 DOI: 10.1016/j.niox.2015.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/29/2015] [Accepted: 10/26/2015] [Indexed: 10/22/2022]
Abstract
Endothelial nitric oxide (NO) synthase (eNOS)-derived NO plays a critical role in the modulation of angiogenesis in the pulmonary vasculature. We recently reported that an eleven amino acid (SSWRRKRKESS) cell penetrating synthetic peptide (P1) activates caveolar signaling, caveloae/eNOS dissociation, and enhance NO production in lung endothelial cells (EC). This study examines whether P1 promote angiogenesis via modulation of caveolar signaling and the level of NO generation in EC and pulmonary artery (PA) segments. P1-enhanced tube formation and cell sprouting were abolished by caveolae disruptor Filipin (FIL) in EC and PA, respectively. P1 enhanced eNOS activity and angiogenesis were attenuated by inhibition of eNOS as well as PLCγ-1, PKC-α but not PI3K-mediated caveolar signaling in intact EC and/or PA. P1 failed to enhance the catalytic activity of eNOS and angiogenesis in caveolae disrupted EC by FIL. Lower (0.01 mM) concentration of NOC-18 enhanced angiogenesis without inhibition of eNOS activity whereas higher concentration of NOC-18 (1.0 mM) inhibited eNOS activity and angiogenesis in EC. Inhibition of eNOS by l-NAME in the presence of P1 resulted in near total loss of tube formation in EC. Although P1 enhanced angiogenesis mimicked only by lower concentrations of NO generated by NOC-18, this response is independent of caveolar signaling/integrity. These results suggest that P1-enhanced angiogenesis is regulated by dynamic process involving caveolar signaling-mediated increased eNOS/NO activity or by the direct exposure to NOC-18 generating only physiologic range of NO independent of caveolae in lung EC and PA segments.
Collapse
Affiliation(s)
- Tarun E Hutchinson
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32608-1197, USA
| | - Jawaharlal M Patel
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32608-1197, USA; Research Service, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608-1197, USA.
| |
Collapse
|
22
|
Proteomic Study to Survey the CIGB-552 Antitumor Effect. BIOMED RESEARCH INTERNATIONAL 2015; 2015:124082. [PMID: 26576414 PMCID: PMC4630370 DOI: 10.1155/2015/124082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 08/26/2015] [Indexed: 11/17/2022]
Abstract
CIGB-552 is a cell-penetrating peptide that exerts in vitro and in vivo antitumor effect on cancer cells. In the present work, the mechanism involved in such anticancer activity was studied using chemical proteomics and expression-based proteomics in culture cancer cell lines. CIGB-552 interacts with at least 55 proteins, as determined by chemical proteomics. A temporal differential proteomics based on iTRAQ quantification method was performed to identify CIGB-552 modulated proteins. The proteomic profile includes 72 differentially expressed proteins in response to CIGB-552 treatment. Proteins related to cell proliferation and apoptosis were identified by both approaches. In line with previous findings, proteomic data revealed that CIGB-552 triggers the inhibition of NF-κB signaling pathway. Furthermore, proteins related to cell invasion were differentially modulated by CIGB-552 treatment suggesting new potentialities of CIGB-552 as anticancer agent. Overall, the current study contributes to a better understanding of the antitumor action mechanism of CIGB-552.
Collapse
|
23
|
LeBlanc ME, Wang W, Caberoy NB, Chen X, Guo F, Alvarado G, Shen C, Wang F, Wang H, Chen R, Liu ZJ, Webster K, Li W. Hepatoma-derived growth factor-related protein-3 is a novel angiogenic factor. PLoS One 2015; 10:e0127904. [PMID: 25996149 PMCID: PMC4440747 DOI: 10.1371/journal.pone.0127904] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/21/2015] [Indexed: 12/23/2022] Open
Abstract
Hepatoma-derived growth factor-related protein-3 (Hdgfrp3 or HRP-3) was recently reported as a neurotrophic factor and is upregulated in hepatocellular carcinoma to promote cancer cell survival. Here we identified HRP-3 as a new endothelial ligand and characterized its in vitro and in vivo functional roles and molecular signaling. We combined open reading frame phage display with multi-round in vivo binding selection to enrich retinal endothelial ligands, which were systematically identified by next generation DNA sequencing. One of the identified endothelial ligands was HRP-3. HRP-3 expression in the retina and brain was characterized by Western blot and immunohistochemistry. Cell proliferation assay showed that HRP-3 stimulated the growth of human umbilical vein endothelial cells (HUVECs). HRP-3 induced tube formation of HUVECs in culture. Wound healing assay indicated that HRP-3 promoted endothelial cell migration. HRP-3 was further confirmed for its in vitro angiogenic activity by spheroid sprouting assay. HRP-3 extrinsically activated the extracellular-signal-regulated kinase ½ (ERK1/2) pathway in endothelial cells. The angiogenic activity of HRP-3 was independently verified by mouse cornea pocket assay. Furthermore, in vivo Matrigel plug assay corroborated HRP-3 activity to promote new blood vessel formation. These results demonstrated that HRP-3 is a novel angiogenic factor.
Collapse
Affiliation(s)
- Michelle E. LeBlanc
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Weiwen Wang
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Nora B. Caberoy
- School of Life Sciences, University of Nevada Las Vegas, Las Vegas, Nevada, United States of America
| | - Xiuping Chen
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, Florida, United States of America
- Department of Ophthalmology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Feiye Guo
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Gabriela Alvarado
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Chen Shen
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Feng Wang
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hui Wang
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rui Chen
- Dept. of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Zhao-Jun Liu
- Vascular Biology Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Keith Webster
- Vascular Biology Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Wei Li
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, Florida, United States of America
- Vascular Biology Institute, University of Miami School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
24
|
Dave JM, Bayless KJ. Vimentin as an integral regulator of cell adhesion and endothelial sprouting. Microcirculation 2015; 21:333-44. [PMID: 24387004 DOI: 10.1111/micc.12111] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 12/30/2013] [Indexed: 12/11/2022]
Abstract
Angiogenesis is a multistep process that requires intricate changes in cell shape to generate new blood vessels. IF are a large family of proteins that play an important structural and functional role in forming and regulating the cytoskeleton. Vimentin, a major type III intermediate filament protein is expressed in endothelial and other mesenchymal cells. The structure of vimentin is conserved in mammals and shows dynamic expression profiles in various cell types and different developmental stages. Although initial studies with vimentin-deficient mice demonstrated a virtually normal phenotype, subsequent studies have revealed several defects in cell attachment, migration, signaling, neurite extension, and vascularization. Regulation of vimentin is highly complex and is driven by posttranslational modifications such as phosphorylation and cleavage by intracellular proteases. This review discusses various novel functions which are now known to be mediated by vimentin, summarizing structure, regulation and roles of vimentin in cell adhesion, migration, angiogenesis, neurite extension, and cancer. We specifically highlight a pathway involving growth factor-mediated calpain activation, vimentin cleavage, and MT1-MMP membrane translocation that is required for endothelial cell invasion in 3D environments. This pathway may also regulate the analogous processes of neurite extension and tumor cell invasion.
Collapse
Affiliation(s)
- Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | | |
Collapse
|
25
|
Protein phosphatase magnesium dependent 1A governs the wound healing-inflammation-angiogenesis cross talk on injury. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2936-50. [PMID: 25196308 DOI: 10.1016/j.ajpath.2014.07.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 06/25/2014] [Accepted: 07/18/2014] [Indexed: 01/09/2023]
Abstract
Protein phosphatase magnesium dependent 1A (PPM1A) has been implicated in fibrosis and skin wounding. We generated PPM1A knockout mice to study the role of PPM1A in the wound healing-inflammation-angiogenesis cross talk. The role of PPM1A in these processes was studied using the ocular alkali burn model system. In the injured cornea the absence of PPM1A led to enhanced inflammatory response, stromal keratocyte transactivation, fibrosis, increased p38 mitogen-activated protein kinase phosphorylation, elevated expression of transforming growth factor-β-related genes (including Acta2, TGF-β, Col1, MMP9, and VEGF) and subsequently to neovascularization. Augmented angiogenesis in the absence of PPM1A is a general process occurring in vivo in PPM1A knockout mice upon subcutaneous Matrigel injection and ex vivo in aortic ring Matrigel cultures. Using primary keratocyte cultures and various experimental approaches, we found that phospho-p38 is a favored PPM1A substrate and that by its dephosphorylation PPM1A participates in the regulation of the transforming growth factor-β signaling cascade, the hallmark of inflammation and the angiogenic process. On the whole, the studies presented here position PPM1A as a new player in the wound healing-inflammation-angiogenesis axis in mouse, reveal its crucial role in homeostasis on injury, and highlight its potential as a therapeutic mediator in pathologic conditions, such as inflammation and angiogenesis disorders, including cancer.
Collapse
|
26
|
Hempel C, Hoyer N, Kildemoes A, Jendresen CB, Kurtzhals JAL. Systemic and Cerebral Vascular Endothelial Growth Factor Levels Increase in Murine Cerebral Malaria along with Increased Calpain and Caspase Activity and Can be Reduced by Erythropoietin Treatment. Front Immunol 2014; 5:291. [PMID: 24995009 PMCID: PMC4062992 DOI: 10.3389/fimmu.2014.00291] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 06/03/2014] [Indexed: 12/20/2022] Open
Abstract
The pathogenesis of cerebral malaria (CM) includes compromised microvascular perfusion, increased inflammation, cytoadhesion, and endothelial activation. These events cause blood-brain barrier disruption and neuropathology and associations with the vascular endothelial growth factor (VEGF) signaling pathway have been shown. We studied this pathway in mice infected with Plasmodium berghei ANKA causing murine CM with or without the use of erythropoietin (EPO) as adjunct therapy. ELISA and western blotting was used for quantification of VEGF and relevant proteins in brain and plasma. CM increased levels of VEGF in brain and plasma and decreased plasma levels of soluble VEGF receptor 2. EPO treatment normalized VEGF receptor 2 levels and reduced brain VEGF levels. Hypoxia-inducible factor (HIF)-1α was significantly upregulated whereas cerebral HIF-2α and EPO levels remained unchanged. Furthermore, we noticed increased caspase-3 and calpain activity in terminally ill mice, as measured by protease-specific cleavage of α-spectrin and p35. In conclusion, we detected increased cerebral and systemic VEGF as well as HIF-1α, which in the brain were reduced to normal in EPO-treated mice. Also caspase and calpain activity was reduced markedly in EPO-treated mice.
Collapse
Affiliation(s)
- Casper Hempel
- Centre for Medical Parasitology, Department of Clinical Microbiology, Copenhagen University Hospital , Copenhagen , Denmark ; Department of International Health, Immunology and Microbiology, University of Copenhagen , Copenhagen , Denmark
| | - Nils Hoyer
- Centre for Medical Parasitology, Department of Clinical Microbiology, Copenhagen University Hospital , Copenhagen , Denmark ; Department of International Health, Immunology and Microbiology, University of Copenhagen , Copenhagen , Denmark
| | - Anna Kildemoes
- Centre for Medical Parasitology, Department of Clinical Microbiology, Copenhagen University Hospital , Copenhagen , Denmark ; Department of International Health, Immunology and Microbiology, University of Copenhagen , Copenhagen , Denmark
| | - Charlotte Bille Jendresen
- Centre for Medical Parasitology, Department of Clinical Microbiology, Copenhagen University Hospital , Copenhagen , Denmark ; Department of International Health, Immunology and Microbiology, University of Copenhagen , Copenhagen , Denmark
| | - Jørgen Anders Lindholm Kurtzhals
- Centre for Medical Parasitology, Department of Clinical Microbiology, Copenhagen University Hospital , Copenhagen , Denmark ; Department of International Health, Immunology and Microbiology, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
27
|
Protein profiling of Helicobacter pylori-associated gastric cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1343-54. [PMID: 24589339 DOI: 10.1016/j.ajpath.2014.01.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 12/18/2013] [Accepted: 01/06/2014] [Indexed: 02/02/2023]
Abstract
Helicobacter pylori infection is an initiating factor in the development of gastric cancer. Gastric cancer can be divided into two groups on the basis of H. pylori serological status; seropositive H. pylori status predicts favorable prognosis in patients with gastric cancer. By using the protein pathway array, we identified 20 differentially expressed proteins in primary gastric cancer tissues between the H. pylori-seropositive and H. pylori-seronegative groups. Our results indicate that both brassinosteroid insensitive 1-associated kinase 1 and calpastatin are favorable prognostic factors in H. pylori-seropositive gastric cancer patients. In contrast, dachshund homolog 1 is a favorable prognostic factor in H. pylori-seronegative gastric cancer patients. Different signaling pathways were found to be altered between H. pylori-seropositive and H. pylori-seronegative gastric cancer, which may account for the different tumorigenesis and outcomes between these two subsets of patients.
Collapse
|
28
|
Abstract
Calpain is a conserved family of calcium-dependent, cytosolic, neutral cysteine proteases. The best characterized members of the family are the ubiquitously expressed calpain 1 and calpain 2. They perform controlled proteolysis of their target proteins. The regulation of these enzymes includes autolysis, calcium, phosphorylation as a posttranslational modification, and binding of calpastatin, phospholipids or activator proteins, respectively. Calpain are implicated in many physiological and pathological processes. They have significant role in the cell proliferation, differentiation and migration in a variety of mammalian cell types, contributing to the development of angiogenesis, vascular remodeling, and cancer. Therefore the knowledge of the precise mechanism of calpain signaling could provide therapeutic approaches in these processes.
Collapse
Affiliation(s)
- Laszlo Kovacs
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| |
Collapse
|
29
|
Chillà A, Magherini F, Margheri F, Laurenzana A, Gamberi T, Bini L, Bianchi L, Danza G, Mazzanti B, Serratì S, Modesti A, Del Rosso M, Fibbi G. Proteomic identification of VEGF-dependent protein enrichment to membrane caveolar-raft microdomains in endothelial progenitor cells. Mol Cell Proteomics 2013; 12:1926-38. [PMID: 23572564 DOI: 10.1074/mcp.m112.024638] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Endothelial cell caveolar-rafts are considered functional platforms that recruit several pro-angiogenic molecules to realize an efficient angiogenic program. Here we studied the differential caveolar-raft protein composition of endothelial colony-forming cells following stimulation with VEGF, which localizes in caveolae on interaction with its type-2 receptor. Endothelial colony-forming cells are a cell population identified in human umbilical blood that show all the properties of an endothelial progenitor cell and a high proliferative rate. Two-dimensional gel electrophoresis analysis was coupled with mass spectrometry to identify candidate proteins. The twenty-eight differentially expressed protein spots were grouped according to their function using Gene Ontology classification. In particular, functional categories relative to cell death inhibition and hydrogen peroxide metabolic processes resulted enriched. In these categories, Peroxiredoxin-2 and 6, that control hydrogen peroxide metabolic processes, are the main enriched molecules together with the anti-apoptotic 78 kDa glucose regulated protein. Some of the proteins we identified had never before identified as caveolar-raft components. Other identified proteins include calpain small subunit-1, known to mediates angiogenic response to VEGF, gelsolin, which regulates stress fiber assembly, and annexin A3, an angiogenic mediator that induces VEGF production. We validated the functional activity of the above proteins, showing that the siRNA silencing of these resulted in the inhibition of capillary morphogenesis. Overall, our data show that VEGF stimulation triggers the caveolar-raft recruitment of proteins that warrant a physiological amount of reactive oxygen species to maintain a proper angiogenic function of endothelial colony-forming cells and preserve the integrity of the actin cytoskeleton.
Collapse
Affiliation(s)
- Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Raimbourg Q, Perez J, Vandermeersch S, Prignon A, Hanouna G, Haymann JP, Baud L, Letavernier E. The calpain/calpastatin system has opposing roles in growth and metastatic dissemination of melanoma. PLoS One 2013; 8:e60469. [PMID: 23565252 PMCID: PMC3614974 DOI: 10.1371/journal.pone.0060469] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 02/26/2013] [Indexed: 11/18/2022] Open
Abstract
Conventional calpains are ubiquitous cysteine proteases whose activity is promoted by calcium signaling and specifically limited by calpastatin. Calpain expression has been shown to be increased in human malignant cells, but the contribution of the calpain/calpastatin system in tumorigenesis remains unclear. It may play an important role in tumor cells themselves (cell growth, migration, and a contrario cell death) and/or in tumor niche (tissue infiltration by immune cells, neo-angiogenesis). In this study, we have used a mouse model of melanoma as a tool to gain further understanding of the role of calpains in tumor progression. To determine the respective importance of each target, we overexpressed calpastatin in tumor and/or host in isolation. Our data demonstrate that calpain inhibition in both tumor and host blunts tumor growth, while paradoxically increasing metastatic dissemination to regional lymph nodes. Specifically, calpain inhibition in melanoma cells limits tumor growth in vitro and in vivo but increases dissemination by amplifying cell resistance to apoptosis and accelerating migration process. Meanwhile, calpain inhibition restricted to host cells blunts tumor infiltration by immune cells and angiogenesis required for antitumor immunity, allowing tumor cells to escape tumor niche and disseminate. The development of highly specific calpain inhibitors with potential medical applications in cancer should take into account the opposing roles of the calpain/calpastatin system in initial tumor growth and subsequent metastatic dissemination.
Collapse
Affiliation(s)
- Quentin Raimbourg
- Unité Mixte de Recherche 702 (UMR S 702), Université Pierre-et-Marie-Curie Paris VI et Institut National de la Santé et de la Recherche Médicale, Hôpital Tenon, Paris, France
| | - Joëlle Perez
- Unité Mixte de Recherche 702 (UMR S 702), Université Pierre-et-Marie-Curie Paris VI et Institut National de la Santé et de la Recherche Médicale, Hôpital Tenon, Paris, France
| | - Sophie Vandermeersch
- Unité Mixte de Recherche 702 (UMR S 702), Université Pierre-et-Marie-Curie Paris VI et Institut National de la Santé et de la Recherche Médicale, Hôpital Tenon, Paris, France
| | - Aurélie Prignon
- Département de Médecine Nucléaire et Université Pierre-et-Marie-Curie Paris VI, Hôpital Tenon, Paris, France
| | - Guillaume Hanouna
- Unité Mixte de Recherche 702 (UMR S 702), Université Pierre-et-Marie-Curie Paris VI et Institut National de la Santé et de la Recherche Médicale, Hôpital Tenon, Paris, France
| | - Jean-Philippe Haymann
- Unité Mixte de Recherche 702 (UMR S 702), Université Pierre-et-Marie-Curie Paris VI, Institut National de la Santé et de la Recherche Médicale et Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Laurent Baud
- Unité Mixte de Recherche 702 (UMR S 702), Université Pierre-et-Marie-Curie Paris VI, Institut National de la Santé et de la Recherche Médicale et Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Emmanuel Letavernier
- Unité Mixte de Recherche 702 (UMR S 702), Université Pierre-et-Marie-Curie Paris VI, Institut National de la Santé et de la Recherche Médicale et Assistance Publique des Hôpitaux de Paris, Hôpital Tenon, Paris, France
- * E-mail:
| |
Collapse
|
31
|
Trougakos IP, Sesti F, Tsakiri E, Gorgoulis VG. Non-enzymatic post-translational protein modifications and proteostasis network deregulation in carcinogenesis. J Proteomics 2013; 92:274-98. [PMID: 23500136 DOI: 10.1016/j.jprot.2013.02.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 02/19/2013] [Accepted: 02/27/2013] [Indexed: 12/25/2022]
Abstract
Organisms are constantly challenged by stressors and thus the maintenance of biomolecules functionality is essential for the assurance of cellular homeostasis. Proteins carry out the vast majority of cellular functions by mostly participating in multimeric protein assemblies that operate as protein machines. Cells have evolved a complex proteome quality control network for the rescue, when possible, or the degradation of damaged polypeptides. Nevertheless, despite these proteostasis ensuring mechanisms, new protein synthesis, and the replication-mediated dilution of proteome damage in mitotic cells, the gradual accumulation of stressors during aging (or due to lifestyle) results in increasingly damaged proteome. Non-enzymatic post-translational protein modifications mostly arise by unbalanced redox homeostasis and/or high glucose levels and may cause disruption of proteostasis as they can alter protein function. This outcome may then increase genomic instability due to reduced fidelity in processes like DNA replication or repair. Herein, we present a synopsis of the major non-enzymatic post-translation protein modifications and of the proteostasis network deregulation in carcinogenesis. We propose that activation of the proteostasis ensuring mechanisms in premalignant cells has tumor-preventive effects, whereas considering that over-activation of these mechanisms represents a hallmark of advanced tumors, their inhibition provides a strategy for the development of anti-tumor therapies. This article is part of a Special Issue entitled: Posttranslational Protein modifications in biology and Medicine.
Collapse
Affiliation(s)
- Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece.
| | | | | | | |
Collapse
|
32
|
Calpain 1 and -2 play opposite roles in cord formation of lymphatic endothelial cells via eNOS regulation. Hum Cell 2013; 25:36-44. [PMID: 22315009 DOI: 10.1007/s13577-012-0042-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 01/16/2012] [Indexed: 01/05/2023]
Abstract
Calpains are a family of calcium-dependent proteases. Two isoforms, calpain 1 and 2, have been implicated in angiogenesis and endothelial cell adhesion and migration. Calpains regulate the function of eNOS;however, the relation of calpains and eNOS to lymphangiogenesisis still unclear. In the present study, we evaluated the role of calpain and eNOS in the formation of cords by lymphatic endothelial cells on Matrigel. Human lymphatic microvascular dermal-derived endothelial cells were transfected with siRNA against calpain 1 or 2. Calpain 2 knockdown, but not calpain 1 knockdown, significantly reduced cord formation, adhesion, and migration on Matrigel. These decreases correlated with a reduction in eNOS, and phosphorylated eNOS and Hsp90 levels, as assayed by immunoprecipitation and western blotting. In contrast, the knockdown of calpain 1, but not calpain 2,increased cell adhesion, enhanced migration, and stabilized late-stage cord formation by increasing cord length compared to the control. These differences correlated with an increase in the level of phosphorylated eNOS. The results indicated that the functions of calpains and eNOS are important for cord formation by lymphatic endothelial cells. For the first time, we have found different functions of calpain 1 and 2. Calpain 1 is involved in the degradation of eNOS and Hsp90 and the phosphorylation of eNOS,while calpain 2 regulates eNOS phosphorylation during cord formation by lymphatic endothelial cells on Matrigel.
Collapse
|
33
|
Nassar D, Letavernier E, Baud L, Aractingi S, Khosrotehrani K. Calpain activity is essential in skin wound healing and contributes to scar formation. PLoS One 2012; 7:e37084. [PMID: 22615899 PMCID: PMC3353912 DOI: 10.1371/journal.pone.0037084] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 04/18/2012] [Indexed: 12/23/2022] Open
Abstract
Wound healing is a multistep phenomenon that relies on complex interactions between various cell types. Calpains are ubiquitously expressed proteases regulating several processes including cellular adhesion and motility as well as inflammation and angiogenesis. Calpains can be targeted by inhibitors, and their inhibition was shown to reduce organ damage in various disease models. We aimed to assess the role of calpains in skin healing and the potential benefit of calpain inhibition on scar formation. We used a pertinent model where calpain activity is inhibited only in lesional organs, namely transgenic mice overexpressing calpastatin (CPST), a specific natural calpain inhibitor. CPST mice showed a striking delay in wound healing particularly in the initial steps compared to wild types (WT). CPST wounds displayed reduced proliferation in the epidermis and delayed re-epithelization. Granulation tissue formation was impaired in CPST mice, with a reduction in CD45+ leukocyte infiltrate and in CD31+ blood vessel density. Interestingly, wounds on WT skin grafted on CPST mice (WT/CPST) showed a similar delayed healing with reduced angiogenesis and inflammation compared to wounds on WT/WT mice demonstrating the implication of calpain activity in distant extra-cutaneous cells during wound healing. CPST wounds showed a reduction in alpha-smooth muscle actin (αSMA) expressing myofibroblasts as well as αSMA RNA expression suggesting a defect in granulation tissue contraction. At later stages of skin healing, calpain inhibition proved beneficial by reducing collagen production and wound fibrosis. In vitro, human fibroblasts exposed to calpeptin, a pan-calpain inhibitor, showed reduced collagen synthesis, impaired TGFβ-induced differentiation into αSMA-expressing myofibroblasts, and were less efficient in a collagen gel contraction assay. In conclusion, calpains are major players in granulation tissue formation. In view of their specific effects on fibroblasts a late inhibition of calpains should be considered for scar reduction.
Collapse
Affiliation(s)
- Dany Nassar
- Université Pierre et Marie Curie-Paris 6, UMRS938, Paris, France.
| | | | | | | | | |
Collapse
|
34
|
Kwak HI, Kang H, Dave JM, Mendoza EA, Su SC, Maxwell SA, Bayless KJ. Calpain-mediated vimentin cleavage occurs upstream of MT1-MMP membrane translocation to facilitate endothelial sprout initiation. Angiogenesis 2012; 15:287-303. [PMID: 22407449 PMCID: PMC3338915 DOI: 10.1007/s10456-012-9262-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 02/20/2012] [Indexed: 12/19/2022]
Abstract
Endothelial cells normally line the vasculature and remain quiescent. However, these cells can be rapidly stimulated to undergo morphogenesis and initiate new blood vessel formation given the proper cues. This study reports a new mechanism for initiating angiogenic sprout formation that involves vimentin, the major intermediate filament protein in endothelial cells. Initial studies confirmed vimentin was required for sphingosine 1-phosphate (S1P)- and growth factor (GF)-induced endothelial cell invasion, and vimentin was cleaved by calpains during invasion. Calpains were predominantly activated by GF and were required for sprout initiation. Because others have reported membrane type 1-matrix metalloproteinase (MT1-MMP) is required for endothelial sprouting responses, we tested whether vimentin and calpain acted upstream of MT1-MMP. Both calpain and vimentin were required for successful MT1-MMP membrane translocation, which was stimulated by S1P. In addition, vimentin complexed with MT1-MMP in a manner that required both the cytoplasmic domain of MT1-MMP and calpain activation, which increased the soluble pool of vimentin in endothelial cells. Altogether, these data indicate that pro-angiogenic signals converge to activate calpain-dependent vimentin cleavage and increase vimentin solubility, which act upstream to facilitate MT1-MMP membrane translocation, resulting in successful endothelial sprout formation in three-dimensional collagen matrices. These findings help explain why S1P and GF synergize to stimulate robust sprouting in 3D collagen matrices.
Collapse
Affiliation(s)
- Hyeong-Il Kwak
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114 USA
| | - Hojin Kang
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114 USA
| | - Jui M. Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114 USA
| | - E. Adriana Mendoza
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114 USA
| | - Shih-Chi Su
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114 USA
| | - Steve A. Maxwell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114 USA
| | - Kayla J. Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114 USA
| |
Collapse
|
35
|
Letavernier B, Zafrani L, Nassar D, Perez J, Levi C, Bellocq A, Mesnard L, Sachon E, Haymann JP, Aractingi S, Faussat AM, Baud L, Letavernier E. Calpains Contribute to Vascular Repair in Rapidly Progressive Form of Glomerulonephritis: Potential Role of Their Externalization. Arterioscler Thromb Vasc Biol 2012; 32:335-42. [DOI: 10.1161/atvbaha.111.240242] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Béatrice Letavernier
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Lara Zafrani
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Dany Nassar
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Joëlle Perez
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Charlène Levi
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Agnès Bellocq
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Laurent Mesnard
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Emmanuelle Sachon
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Jean-Philippe Haymann
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Selim Aractingi
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Anne-Marie Faussat
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Laurent Baud
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| | - Emmanuel Letavernier
- From the INSERM (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), U702, Paris, France; Université Pierre et Marie Curie-Paris 6 (B.L., L.Z., J.P., C.L., A.B., L.M., J.-P.H., L.B., E.L.), UMRS702, Paris, France; AP HP (A.B., J.-P.H., L.B., E.L.), Tenon Hospital, Department of Physiology, Paris, France; Université Pierre et Marie Curie-Paris 6 and INSERM UMR_S938 (D.N., S.A.), Saint-Antoine Research Centre, Paris, France; Université Pierre et Marie Curie-Paris 6 (E.S.), UMR7203 CNRS, Paris,
| |
Collapse
|
36
|
Nagai A, Sado T, Naruse K, Noguchi T, Haruta S, Yoshida S, Tanase Y, Tsunemi T, Kobayashi H. Antiangiogenic-Induced Hypertension: The Molecular Basis of Signaling Network. Gynecol Obstet Invest 2012; 73:89-98. [DOI: 10.1159/000334458] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 10/16/2011] [Indexed: 01/09/2023]
|
37
|
Lal S, La Du J, Tanguay RL, Greenwood JA. Calpain 2 is required for the invasion of glioblastoma cells in the zebrafish brain microenvironment. J Neurosci Res 2011; 90:769-81. [PMID: 22183788 DOI: 10.1002/jnr.22794] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 08/12/2011] [Accepted: 08/18/2011] [Indexed: 01/21/2023]
Abstract
Glioblastoma is an aggressive primary brain tumor with a 5-year survival rate of less than 5%. The ability of glioblastoma cells to invade surrounding brain tissue presents the primary challenge for the success of focal therapeutic approaches. We previously reported that the calcium-activated protease calpain 2 is critical for glioblastoma cell invasion in vitro. Here, we show that expression of calpain 2 is required for the dispersal of glioblastoma cells in a living brain microenvironment. Knockdown of calpain 2 resulted in a 2.9-fold decrease in the invasion of human glioblastoma cells in zebrafish brain. Control cells diffusely migrated up to 450 μm from the site of injection, whereas knockdown cells remained confined in clusters. The invasion study was repeated in organotypic mouse brain tissues, and calpain 2 knockdown cells demonstrated a 2.3-fold lower area of dispersal compared with control cells. In zebrafish brain, glioblastoma cells appeared to migrate in part along the blood vessels of the host. Furthermore, angiogenesis was detected in 27% of zebrafish injected with control cells, whereas only 12.5% of fish receiving knockdown cells showed the formation of new vessels, suggesting a role for calpain 2 in tumor cell angiogenesis. Consistent with the progression of glioblastoma in humans, transplanted tumor cells were not observed to metastasize outside the brain of zebrafish. This study demonstrates that calpain 2 expression is required for the dispersal of glioblastoma cells within the dynamic microenvironment of the brain, identifying zebrafish as a valuable orthotopic system for studying glioblastoma cell invasion.
Collapse
Affiliation(s)
- Sangeet Lal
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | | | | | | |
Collapse
|
38
|
Kang H, Kwak HI, Kaunas R, Bayless KJ. Fluid shear stress and sphingosine 1-phosphate activate calpain to promote membrane type 1 matrix metalloproteinase (MT1-MMP) membrane translocation and endothelial invasion into three-dimensional collagen matrices. J Biol Chem 2011; 286:42017-42026. [PMID: 22002053 PMCID: PMC3234924 DOI: 10.1074/jbc.m111.290841] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 10/10/2011] [Indexed: 12/31/2022] Open
Abstract
The vascular endothelium continually senses and responds to biochemical and mechanical stimuli to appropriately initiate angiogenesis. We have shown previously that fluid wall shear stress (WSS) and sphingosine 1-phosphate (S1P) cooperatively initiate the invasion of human umbilical vein endothelial cells into collagen matrices (Kang, H., Bayless, K. J., and Kaunas, R. (2008) Am. J. Physiol. Heart Circ. Physiol. 295, H2087-2097). Here, we investigated the role of calpains in the regulation of endothelial cell invasion in response to WSS and S1P. Calpain inhibition significantly decreased S1P- and WSS-induced invasion. Short hairpin RNA-mediated gene silencing demonstrated that calpain 1 and 2 were required for WSS and S1P-induced invasion. Also, S1P synergized with WSS to induce invasion and to activate calpains and promote calpain membrane localization. Calpain inhibition results in a cell morphology consistent with reduced matrix proteolysis. Membrane type 1-matrix metalloproteinase (MT1-MMP) has been shown by others to regulate endothelial cell invasion, prompting us to test whether calpain acted upstream of MT1-MMP. S1P and WSS synergistically activated MT1-MMP and induced cell membrane localization of MT1-MMP in a calpain-dependent manner. Calpain activation, MT1-MMP activation and MT1-MMP membrane localization were all maximal with 5.3 dynes/cm(2) WSS and S1P treatment, which correlated with maximal invasion responses. Our data show for the first time that 5.3 dynes/cm(2) WSS in the presence of S1P combine to activate calpains, which direct MT1-MMP membrane localization to initiate endothelial sprouting into three-dimensional collagen matrices.
Collapse
Affiliation(s)
- Hojin Kang
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843
| | - Hyeong-Il Kwak
- Department of Molecular & Cellular Medicine, Texas A&M Health Science Center, College Station, Texas 77843-1114
| | - Roland Kaunas
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843
| | - Kayla J Bayless
- Department of Molecular & Cellular Medicine, Texas A&M Health Science Center, College Station, Texas 77843-1114.
| |
Collapse
|
39
|
Ma W, Han W, Greer PA, Tuder RM, Toque HA, Wang KKW, Caldwell RW, Su Y. Calpain mediates pulmonary vascular remodeling in rodent models of pulmonary hypertension, and its inhibition attenuates pathologic features of disease. J Clin Invest 2011; 121:4548-66. [PMID: 22005303 DOI: 10.1172/jci57734] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 08/25/2011] [Indexed: 11/17/2022] Open
Abstract
Pulmonary hypertension is a severe and progressive disease, a key feature of which is pulmonary vascular remodeling. Several growth factors, including EGF, PDGF, and TGF-β1, are involved in pulmonary vascular remodeling during pulmonary hypertension. However, increased knowledge of the downstream signaling cascades is needed if effective clinical interventions are to be developed. In this context, calpain provides an interesting candidate therapeutic target, since it is activated by EGF and PDGF and has been reported to activate TGF-β1. Thus, in this study, we examined the role of calpain in pulmonary vascular remodeling in two rodent models of pulmonary hypertension. These data showed that attenuated calpain activity in calpain-knockout mice or rats treated with a calpain inhibitor resulted in prevention of increased right ventricular systolic pressure, right ventricular hypertrophy, as well as collagen deposition and thickening of pulmonary arterioles in models of hypoxia- and monocrotaline-induced pulmonary hypertension. Additionally, inhibition of calpain in vitro blocked intracellular activation of TGF-β1, which led to attenuated Smad2/3 phosphorylation and collagen synthesis. Finally, smooth muscle cells of pulmonary arterioles from patients with pulmonary arterial hypertension showed higher levels of calpain activation and intracellular active TGF-β. Our data provide evidence that calpain mediates EGF- and PDGF-induced collagen synthesis and proliferation of pulmonary artery smooth muscle cells via an intracrine TGF-β1 pathway in pulmonary hypertension.
Collapse
Affiliation(s)
- Wanli Ma
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Hoang MV, Smith LEH, Senger DR. Calpain inhibitors reduce retinal hypoxia in ischemic retinopathy by improving neovascular architecture and functional perfusion. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1812:549-57. [PMID: 20804843 PMCID: PMC3005970 DOI: 10.1016/j.bbadis.2010.08.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 08/19/2010] [Accepted: 08/24/2010] [Indexed: 12/21/2022]
Abstract
In ischemic retinopathies, underlying hypoxia drives abnormal neovascularization that damages retina and causes blindness. The abnormal neovasculature is tortuous and leaky and fails to alleviate hypoxia, resulting in more pathological neovascularization and retinal damage. With an established model of ischemic retinopathy we found that calpain inhibitors, when administered in moderation, reduced architectural abnormalities, reduced vascular leakage, and most importantly reduced retinal hypoxia. Mechanistically, these calpain inhibitors improved stability and organization of the actin cytoskeleton in retinal endothelial cells undergoing capillary morphogenesis in vitro, and they similarly improved organization of actin cables within new blood vessels in vivo. Hypoxia induced calpain activity in retinal endothelial cells and severely disrupted the actin cytoskeleton, whereas calpain inhibitors preserved actin cables under hypoxic conditions. Collectively, these findings support the hypothesis that hyper-activation of calpains by hypoxia contributes to disruption of the retinal endothelial cell cytoskeleton, resulting in formation of neovessels that are defective both architecturally and functionally. Modest suppression of calpain activity with calpain inhibitors restores cytoskeletal architecture and promotes formation of a functional neovasculature, thereby reducing underlying hypoxia. In sharp contrast to "anti-angiogenesis" strategies that cannot restore normoxia and may aggravate hypoxia, the therapeutic strategy described here does not inhibit neovascularization. Instead, by improving the function of neovascularization to reduce underlying hypoxia, moderate calpain inhibition offers a method for alleviating retinal ischemia, thereby suggesting a new treatment paradigm based on improvement rather than inhibition of new blood vessel growth.
Collapse
Affiliation(s)
- Mien V. Hoang
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston MA 02215
| | - Lois E. H. Smith
- Department of Ophthalmology, Children’s Hospital Boston and Harvard Medical School, Boston MA 02115
| | - Donald R. Senger
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston MA 02215
| |
Collapse
|
41
|
Abstract
INTRODUCTION The intracellular signaling cysteine proteases, calpains (specifically the ubiquitous calpains 1 and 2), are involved in numerous physiological and pathological phenomena. Several works have highlighted the implication of calpains in processes crucial for cancer development and progression. For these reasons, calpains are considered by several authors as potential anti-cancer targets. AREAS COVERED How calpains are implicated in cancer formation and development, how these enzymes are deregulated in cancer cells and how these proteases could be targeted by anti-cancer drugs. Studies published in the last 10 years are focused on. EXPERT OPINION Targeting calpain activity with specific inhibitors could be a novel approach to limiting development of primary tumors and formation of metastases, by inhibiting tumor cell migration and invasion, which allows dissemination as well as tumor neovascularization, which in turn allows expansion. However, such drugs could interfere with anti-cancer treatments, as ubiquitous calpains play crucial roles in chemotherapy-induced apoptosis. For these reasons, drugs targeting calpains would have to be used selectively to avoid interference with other treatments and physiological processes. Further studies will be required concerning the other members of the calpain family and their potential implication in cancer development before considering treatments targeting their activity.
Collapse
Affiliation(s)
- Ludovic Leloup
- INSERM UMR 911 (CRO2), Aix-Marseille Université, Faculté de Pharmacie, 13385 Marseille cedex 5, France
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
42
|
Wang H, Su Y. Collagen IV contributes to nitric oxide-induced angiogenesis of lung endothelial cells. Am J Physiol Cell Physiol 2011; 300:C979-88. [PMID: 21307347 DOI: 10.1152/ajpcell.00368.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Nitric oxide (NO) mediates endothelial angiogenesis via inducing the expression of integrin α(v)β(3). During angiogenesis, endothelial cells adhere to and migrate into the extracellular matrix through integrins. Collagen IV binds to integrin α(v)β(3), leading to integrin activation, which affects a number of signaling processes in endothelial cells. In the present study, we evaluated the role of collagen IV in NO-induced angiogenesis. We found that NO donor 2,2'-(hydroxynitrosohydrazino)bis-ethanamine (NOC-18) causes increases in collagen IV mRNA and protein in lung endothelial cells and collagen IV release into the medium. Addition of collagen IV into the coating of endothelial culture increases endothelial monolayer wound repair, proliferation, and tube formation. Inhibition of collagen IV synthesis using gene silencing attenuates NOC-18-induced increases in monolayer wound repair, cell proliferation, and tube formation as well as in the phosphorylation of focal adhesion kinase (FAK). Integrin blocking antibody LM609 prevents NOC-18-induced increase in endothelial monolayer wound repair. Inhibition of protein kinase G (PKG) using the specific PKG inhibitor KT5823 or PKG small interfering RNA prevents NOC-18-induced increases in collagen IV protein and mRNA and endothelial angiogenesis. Together, these results indicate that NO promotes collagen IV synthesis via a PKG signaling pathway and that the increase in collagen IV synthesis contributes to NO-induced angiogenesis of lung endothelial cells through integrin-FAK signaling. Manipulation of collagen IV could be a novel approach for the prevention and treatment of diseases such as alveolar capillary dysplasia, severe pulmonary arterial hypertension, and tumor invasion.
Collapse
Affiliation(s)
- Huafang Wang
- Dept. of Pharmacology and Toxicology, Medical College of Georgia, Georgia Health Sciences University, 1120 15th St., Augusta, GA 30912, USA
| | | |
Collapse
|
43
|
Liu M, Hou J, Huang L, Huang X, Heibeck TH, Zhao R, Pasa-Tolic L, Smith RD, Li Y, Fu K, Zhang Z, Hinrichs SH, Ding SJ. Site-specific proteomics approach for study protein S-nitrosylation. Anal Chem 2011; 82:7160-8. [PMID: 20687582 DOI: 10.1021/ac100569d] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Here we present a novel and robust method for the identification of protein S-nitrosylation sites in complex protein mixtures. The approach utilizes the cysteinyl affinity resin to selectively enrich S-nitrosylated peptides reduced by ascorbate followed by nanoscale liquid chromatography tandem mass spectrometry. Two alkylation agents with different added masses were employed to differentiate the S-nitrosylation sites from the non-S-nitrosylation sites. We applied this approach to MDA-MB-231 cells treated with Angeli's salt, a nitric oxide donor that has been shown to inhibit breast tumor growth and angiogenesis. A total of 162 S-nitrosylation sites were identified and an S-nitrosylation motif was revealed in our study. The 162 sites are significantly more than the number reported by previous methods, demonstrating the efficiency of our approach. Our approach will further facilitate the functional study of protein S-nitrosylation in cellular processes and may reveal new therapeutic targets.
Collapse
Affiliation(s)
- Miao Liu
- Department of Pathology and Microbiology, Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Formononetin accelerates wound repair by the regulation of early growth response factor-1 transcription factor through the phosphorylation of the ERK and p38 MAPK pathways. Int Immunopharmacol 2011; 11:46-54. [DOI: 10.1016/j.intimp.2010.10.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 09/30/2010] [Accepted: 10/01/2010] [Indexed: 02/02/2023]
|
45
|
Mo XG, Chen QW, Li XS, Zheng MM, Ke DZ, Deng W, Li GQ, Jiang J, Wu ZQ, Wang L, Wang P, Yang Y, Cao GY. Suppression of NHE1 by small interfering RNA inhibits HIF-1α-induced angiogenesis in vitro via modulation of calpain activity. Microvasc Res 2010; 81:160-8. [PMID: 21185840 DOI: 10.1016/j.mvr.2010.12.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 11/14/2010] [Accepted: 12/13/2010] [Indexed: 12/26/2022]
Abstract
Hypoxia-inducible factor-1 (HIF-1) orchestrates angiogenesis under hypoxic conditions mainly due to increased expression of such target genes as vascular endothelial growth factor (VEGF). Na+/H+exchanger-1 (NHE1), a potential HIF target gene product, plays a pivotal role in proliferation, survival, migration, adhesion and so on. However, it is unknown whether NHE1 is involved in HIF-1α-induced angiogenesis. This present study demonstrated that the expression of NHE1 was much higher in human umbilical vein endothelial cells (HUVECs) infected with adenovirus encoding HIF-1α (rAd-HIF) than with vacuum adenovirus (vAd). HIF-1α also increased the expression of VEGF, the expression and activity of calpains, and the intracellular pH. Moreover, small interfering RNA targeting NHE1 (NHE1 siRNA) dramatically decreased the expression of NHE1 and thus lowered the intracellular pH, and it also attenuated the protein expression of calpain-2 but not calpain-1, resulting in the lower calpain activity. Furthermore, HIF-1α enhanced the proliferation, migration and Matrigel tube formation, which were inhibited by NHE1 siRNA. Finally, the inhibitory effect of NHE1 siRNA was reversed by VEGF and the reversibility of the later was abrogated by the calpain inhibitor ALLM. In conclusion, the findings have revealed that NHE1 might participate in HIF-1-induced angiogenesis due, at least in part, to the alteration of the calpain activity, suggesting that NHE1 as well as calpains might represent a potential target of controlling angiogenesis in response to the hypoxic stress under various pathological conditions.
Collapse
Affiliation(s)
- Xian-Gang Mo
- Department of Geriatrics Cardiology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Chaitanya GV, Alexander JS, Babu PP. PARP-1 cleavage fragments: signatures of cell-death proteases in neurodegeneration. Cell Commun Signal 2010; 8:31. [PMID: 21176168 PMCID: PMC3022541 DOI: 10.1186/1478-811x-8-31] [Citation(s) in RCA: 703] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Accepted: 12/22/2010] [Indexed: 11/16/2022] Open
Abstract
The normal function of poly (ADP-ribose) polymerase-1 (PARP-1) is the routine repair of DNA damage by adding poly (ADP ribose) polymers in response to a variety of cellular stresses. Recently, it has become widely appreciated that PARP-1 also participates in diverse physiological and pathological functions from cell survival to several forms of cell death and has been implicated in gene transcription, immune responses, inflammation, learning, memory, synaptic functions, angiogenesis and aging. In the CNS, PARP inhibition attenuates injury in pathologies like cerebral ischemia, trauma and excitotoxicity demonstrating a central role of PARP-1 in these pathologies. PARP-1 is also a preferred substrate for several 'suicidal' proteases and the proteolytic action of suicidal proteases (caspases, calpains, cathepsins, granzymes and matrix metalloproteinases (MMPs)) on PARP-1 produces several specific proteolytic cleavage fragments with different molecular weights. These PARP-1 signature fragments are recognized biomarkers for specific patterns of protease activity in unique cell death programs. This review focuses on specific suicidal proteases active towards PARP-1 to generate signature PARP-1 fragments that can identify key proteases and particular forms of cell death involved in pathophysiology. The roles played by some of the PARP-1 fragments and their associated binding partners in the control of different forms of cell death are also discussed.
Collapse
Affiliation(s)
- Ganta Vijay Chaitanya
- Department of Biotechnology, School of Life Sciences, University of Hyderabad, Hyderabad, India
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Louisiana-USA
| | - Jonathan S Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Louisiana-USA
| | - Phanithi Prakash Babu
- Department of Biotechnology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
47
|
Hoang MV, Nagy JA, Fox JEB, Senger DR. Moderation of calpain activity promotes neovascular integration and lumen formation during VEGF-induced pathological angiogenesis. PLoS One 2010; 5:e13612. [PMID: 21049044 PMCID: PMC2963609 DOI: 10.1371/journal.pone.0013612] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Accepted: 09/24/2010] [Indexed: 12/16/2022] Open
Abstract
Background Successful neovascularization requires that sprouting endothelial cells (ECs) integrate to form new vascular networks. However, architecturally defective, poorly integrated vessels with blind ends are typical of pathological angiogenesis induced by vascular endothelial growth factor-A (VEGF), thereby limiting the utility of VEGF for therapeutic angiogenesis and aggravating ischemia-related pathologies. Here we investigated the possibility that over-exuberant calpain activity is responsible for aberrant VEGF neovessel architecture and integration. Calpains are a family of intracellular calcium-dependent, non-lysosomal cysteine proteases that regulate cellular functions through proteolysis of numerous substrates. Methodology/Principal Findings In a mouse skin model of VEGF-driven angiogenesis, retroviral transduction with dominant-negative (DN) calpain-I promoted neovessel integration and lumen formation, reduced blind ends, and improved vascular perfusion. Moderate doses of calpain inhibitor-I improved VEGF-driven angiogenesis similarly to DN calpain-I. Conversely, retroviral transduction with wild-type (WT) calpain-I abolished neovessel integration and lumen formation. In vitro, moderate suppression of calpain activity with DN calpain-I or calpain inhibitor-I increased the microtubule-stabilizing protein tau in endothelial cells (ECs), increased the average length of microtubules, increased actin cable length, and increased the interconnectivity of vascular cords. Conversely, WT calpain-I diminished tau, collapsed microtubules, disrupted actin cables, and inhibited integration of cord networks. Consistent with the critical importance of microtubules for vascular network integration, the microtubule-stabilizing agent taxol supported vascular cord integration whereas microtubule dissolution with nocodazole collapsed cord networks. Conclusions/Significance These findings implicate VEGF-induction of calpain activity and impairment of cytoskeletal dynamics in the failure of VEGF-induced neovessels to form and integrate properly. Accordingly, calpain represents an important target for rectifying key vascular defects associated with pathological angiogenesis and for improving therapeutic angiogenesis with VEGF.
Collapse
Affiliation(s)
- Mien V Hoang
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | | | | | | |
Collapse
|
48
|
Chakrabarti S, Rizvi M, Morin K, Garg R, Freedman JE. The role of CD40L and VEGF in the modulation of angiogenesis and inflammation. Vascul Pharmacol 2010; 53:130-7. [PMID: 20546942 DOI: 10.1016/j.vph.2010.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 03/30/2010] [Accepted: 05/07/2010] [Indexed: 12/20/2022]
Abstract
Recently, there has been growing interest in deciphering the role of angiogenesis in the progression of atherogenesis. Importantly, CD40-CD40L interactions are of significant relevance because of their involvement in both angiogenesis and atherosclerotic development. Previously, we have shown that recombinant soluble CD40 ligand (rsCD40L) stimulates auto-inflammatory CD40L synthesis and reactive oxygen species (ROS) generation in vascular cells. In the current study, we demonstrate that redox-mediated CD40-CD40L interaction can enhance vascular endothelial growth factor (VEGF)-induced angiogenesis, endothelial migration, and actin polymerization processes. Interestingly, the addition of exogenous VEGF leads to cleavage of de novo CD40L produced in endothelial cells following rsCD40L treatment. Using inhibitor and silencing RNA-based experiments, it was observed that VEGF-induced protease, calpain 2, was responsible for the cleavage of de novo CD40L. While our in vivo experiments using a matrigel plug assay indicate a VEGF and CD40L induced enhancement of angiogenesis, our studies also identify a novel mechanism by which VEGF can abrogate CD40L-mediated inflammation. Together, these studies reveal a new pathway by which VEGF-CD40L interactions can regulate the angiogenic and inflammatory process depending on the specific environment.
Collapse
Affiliation(s)
- Subrata Chakrabarti
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA.
| | | | | | | | | |
Collapse
|
49
|
Kondrikov D, Fonseca FV, Elms S, Fulton D, Black SM, Block ER, Su Y. Beta-actin association with endothelial nitric-oxide synthase modulates nitric oxide and superoxide generation from the enzyme. J Biol Chem 2010; 285:4319-27. [PMID: 19946124 PMCID: PMC2836036 DOI: 10.1074/jbc.m109.063172] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 11/20/2009] [Indexed: 11/06/2022] Open
Abstract
Protein-protein interactions represent an important post-translational mechanism for endothelial nitric-oxide synthase (eNOS) regulation. We have previously reported that beta-actin is associated with eNOS oxygenase domain and that association of eNOS with beta-actin increases eNOS activity and nitric oxide (NO) production. In the present study, we found that beta-actin-induced increase in NO production was accompanied by decrease in superoxide formation. A synthetic actin-binding sequence (ABS) peptide 326 with amino acid sequence corresponding to residues 326-333 of human eNOS, one of the putative ABSs, specifically bound to beta-actin and prevented eNOS association with beta-actin in vitro. Peptide 326 also prevented beta-actin-induced decrease in superoxide formation and increase in NO and L-citrulline production. A modified peptide 326 replacing hydrophobic amino acids leucine and tryptophan with neutral alanine was unable to interfere with eNOS-beta-actin binding and to prevent beta-actin-induced changes in NO and superoxide formation. Site-directed mutagenesis of the actin-binding domain of eNOS replacing leucine and tryptophan with alanine yielded an eNOS mutant that exhibited reduced eNOS-beta-actin association, decreased NO production, and increased superoxide formation in COS-7 cells. Disruption of eNOS-beta-actin interaction in endothelial cells using ABS peptide 326 resulted in decreased NO production, increased superoxide formation, and decreased endothelial monolayer wound repair, which was prevented by PEG-SOD and NO donor NOC-18. Taken together, this novel finding indicates that beta-actin binding to eNOS through residues 326-333 in the eNOS protein results in shifting the enzymatic activity from superoxide formation toward NO production. Modulation of NO and superoxide formation from eNOS by beta-actin plays an important role in endothelial function.
Collapse
Affiliation(s)
| | | | | | - David Fulton
- From the Department of Pharmacology and Toxicology
- Vascular Biology Center, and
| | | | - Edward R. Block
- the Department of Medicine, University of Florida College of Medicine, Gainesville, Florida 32610
| | - Yunchao Su
- From the Department of Pharmacology and Toxicology
- Department of Medicine
- Vascular Biology Center, and
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta, Georgia 30912 and
| |
Collapse
|
50
|
Chaudhuri P, Harfouche R, Soni S, Hentschel DM, Sengupta S. Shape effect of carbon nanovectors on angiogenesis. ACS NANO 2010; 4:574-582. [PMID: 20043662 DOI: 10.1021/nn901465h] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Physically diverse carbon nanostructures are increasingly being studied for potential applications in cancer chemotherapy. However, limited knowledge exists on the effect of their shape in tuning the biological outcomes when used as nanovectors for drug delivery. In this study, we evaluated the effect of doxorubicin-conjugated single walled carbon nanotubes (CNT-Dox) and doxorubicin-conjugated spherical polyhydroxylated fullerenes or fullerenols (Ful-Dox) on angiogenesis. We report that CNTs exert a pro-angiogenic effect in vitro and in vivo. In contrast, the fullerenols or doxorubicin-conjugated fullerenols exerted a dramatically opposite antiangiogenic activity in zebrafish and murine tumor angiogenesis models. Dissecting the angiogenic phenotype into discrete cellular steps revealed that fullerenols inhibited endothelial cell proliferation, while CNTs attenuated the cytotoxic effect of doxorubicin on the endothelial cells. Interestingly, CNT promoted endothelial tubulogenesis, a late step during angiogenesis. Further, mechanistic studies revealed that CNTs, but not fullerenols, induced integrin clustering and activated focal adhesion kinase and downstream phosphoinositide-3-kinase (PI3K) signaling in endothelial cells, which can explain the distinct angiogenic outcomes. The results of the study highlight the function of physical parameters of nanoparticles in determining their activity in biological settings.
Collapse
Affiliation(s)
- Padmaparna Chaudhuri
- Department of Medicine, Laboratory of Nanomedicine, BWH-HST Center for Biomedical Engineering, Brigham and Women's Hospital, Harvard MedicalSchool, USA
| | | | | | | | | |
Collapse
|