1
|
Jiang S, Zhu G, Tan Y, Zhou T, Zheng S, Wang F, Lei W, Liu X, Du J, Tian M. Identification of VEGFs-related gene signature for predicting microangiogenesis and hepatocellular carcinoma prognosis. Aging (Albany NY) 2024; 16:10321-10347. [PMID: 38874512 PMCID: PMC11236318 DOI: 10.18632/aging.205931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 04/08/2024] [Indexed: 06/15/2024]
Abstract
Microangiogenesis is an important prognostic factor in various cancers, including hepatocellular carcinoma (HCC). The Vascular Endothelial Growth Factor (VEGF) has been shown to contribute to tumor angiogenesis. Recently, several studies have investigated the regulation of VEGF production by a single gene, with few researchers exploring all genes that affect VEGF production. In this study, we comprehensively analyzed all genes affecting VEGF production in HCC and developed a risk model and gene-based risk score based on VEGF production. Moreover, the model's predictive capacity on prognosis of HCCs was verified using training and validation datasets. The developed model showed good prediction of the overall survival rate. Patients with a higher risk score experienced poor outcomes compared to those with a lower risk score. Furthermore, we identified the immunological causes of the poor prognosis of patients with high-risk scores comparing with those with low-risk scores.
Collapse
Affiliation(s)
- Shengpan Jiang
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Guoting Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yiqing Tan
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Tao Zhou
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Shilin Zheng
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Fuhua Wang
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Wenfeng Lei
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Xuan Liu
- Department of Interventional Medicine, Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei Province, China
| | - Jinjun Du
- Department of Hepatology and Gastroenterology, Wuhan Hospital of Traditional Chinese Medicine (The Third Clinical College of Hubei University of Chinese Medicine), Wuhan, Hubei Province, China
| | - Manman Tian
- Department of Hepatology and Gastroenterology, Wuhan Hospital of Traditional Chinese Medicine (The Third Clinical College of Hubei University of Chinese Medicine), Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Potz BA, Sabe SA, Scrimgeour LA, Sabe AA, Harris DD, Abid MR, Clements RT, Sellke FW. Calpain inhibition decreases oxidative stress via mitochondrial regulation in a swine model of chronic myocardial ischemia. Free Radic Biol Med 2023; 208:700-707. [PMID: 37748718 PMCID: PMC10598262 DOI: 10.1016/j.freeradbiomed.2023.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/18/2023] [Accepted: 09/23/2023] [Indexed: 09/27/2023]
Abstract
INTRODUCTION Calpain overexpression is implicated in mitochondrial damage leading to tissue oxidative stress and myocardial ischemic injury. The aim of this study was to determine the effects of calpain inhibition (CI) on mitochondrial impairment and oxidative stress in a swine model of chronic myocardial ischemia and metabolic syndrome. METHODS Yorkshire swine were fed a high-fat diet for 4 weeks to induce metabolic syndrome then underwent placement of an ameroid constrictor to the left circumflex artery. Three weeks later, animals received: no drug (control, "CON"; n= 7); a low-dose calpain inhibitor (0.12 mg/kg; "LCI", n= 7); or high-dose calpain inhibitor (0.25 mg/kg; "HCI", n=7). Treatment continued for 5 weeks, followed by tissue harvest. Cardiac tissue was assayed for protein carbonyl content, as well as antioxidant and mitochondrial protein expression. Reactive oxygen species (ROS) and mitochondrial respiration was measured in H9c2 cells following exposure to normoxia or hypoxia (1%) for 24 h with or without CI. RESULTS In ischemic myocardial tissue, CI was associated with decreased total oxidative stress compared to control. CI was also associated with increased expression of mitochondrial proteins superoxide dismutase 1, SDHA, and pyruvate dehydrogenase compared to control. 100 nM of calpain inhibitor decreased ROS levels and respiration in both normoxic and hypoxic H9c2 cardiomyoblasts. CONCLUSIONS In the setting of metabolic syndrome, CI improves oxidative stress in chronically ischemic myocardial tissue. Decreased oxidative stress may be via modulation of mitochondrial proteins involved in free radical scavenging and production.
Collapse
Affiliation(s)
- Brittany A Potz
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Sharif A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Laura A Scrimgeour
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Ashraf A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Dwight D Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Richard T Clements
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, USA.
| |
Collapse
|
3
|
Potz BA, Sabe AA, Sabe SA, Lawandy IJ, Abid MR, Clements RT, Sellke FW. Calpain inhibition decreases myocardial fibrosis in chronically ischemic hypercholesterolemic swine. J Thorac Cardiovasc Surg 2022; 163:e11-e27. [PMID: 32359903 PMCID: PMC7529741 DOI: 10.1016/j.jtcvs.2019.11.150] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 11/08/2019] [Accepted: 11/29/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Calpain activation during ischemia is known to play critical roles in myocardial remodeling. We hypothesize that calpain inhibition (CI) may serve to reverse and/or prevent fibrosis in chronically ischemic myocardium. METHODS Yorkshire swine were fed a high-cholesterol diet for 4 weeks followed by placement of an ameroid constrictor on the left circumflex artery to induce myocardial ischemia. 3 weeks later, animals received either: no drug; high-cholesterol control group (CON; n = 8); low-dose CI (0.12 mg/kg; LCI, n = 9); or high-dose CI (0.25 mg/kg; HCI, n = 8). The high-cholesterol diet and CI were continued for 5 weeks, after which myocardial tissue was harvested. Tissue samples were analyzed by western blot for changes in protein content. RESULTS In the setting of hypercholesterolemia and chronic myocardial ischemia, CI decreased the expression of collagen in ischemic and nonischemic myocardial tissue. This reduced collagen content was associated with a corresponding decrease in Jak/STAT/MCP-1 signaling pathway, suggesting a role for Jak 2 signaling in calpain activity. CI also decreases the expression of focal adhesion proteins (vinculin) and stabilizes the expression of cytoskeletal and structural proteins (N-cadherin, α-fodrin, desmin, vimentin, filamin, troponin-I). CI had no significant effect on metabolic and hemodynamic parameters. CONCLUSIONS Calpain inhibition may be a beneficial medical therapy to decrease collagen formation in patients with coronary artery disease and associated comorbidities.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Frank W. Sellke
- Dr. Frank W. Sellke, 2 Dudley Street, MOC 360, Division of Cardiothoracic Surgery, Providence, RI 02905, Phone: (401) 444-2732, Fax: (401) 444-2380,
| |
Collapse
|
4
|
Peng H, Hulleman JD. Prospective Application of Activity-Based Proteomic Profiling in Vision Research-Potential Unique Insights into Ocular Protease Biology and Pathology. Int J Mol Sci 2019; 20:ijms20163855. [PMID: 31398819 PMCID: PMC6720450 DOI: 10.3390/ijms20163855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 12/12/2022] Open
Abstract
Activity-based proteomic profiling (ABPP) is a powerful tool to specifically target and measure the activity of a family of enzymes with the same function and reactivity, which provides a significant advantage over conventional proteomic strategies that simply provide abundance information. A number of inherited and age-related eye diseases are caused by polymorphisms/mutations or abnormal expression of proteases including serine proteases, cysteine proteases, and matrix metalloproteinases, amongst others. However, neither conventional genomic, transcriptomic, nor traditional proteomic profiling directly interrogate protease activities. Thus, leveraging ABPP to probe the activity of these enzyme classes as they relate to normal function and pathophysiology of the eye represents a unique potential opportunity for disease interrogation and possibly intervention.
Collapse
Affiliation(s)
- Hui Peng
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9057, USA
| | - John D Hulleman
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9057, USA.
- Department of Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA.
| |
Collapse
|
5
|
Betrie AH, Ayton S, Bush AI, Angus JA, Lei P, Wright CE. Evidence of a Cardiovascular Function for Microtubule-Associated Protein Tau. J Alzheimers Dis 2018; 56:849-860. [PMID: 28059795 DOI: 10.3233/jad-161093] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Aggregation of tau protein into intracellular deposits is a pathognomonic feature of tauopathies such as Alzheimer's disease (AD) and lowering tau is a prominent therapeutic strategy under development. However, the physiological function of tau protein is not well known, particularly in the periphery. Lowering tau protein risks disrupting its physiological role leading to unwanted effects. In this study, the presence of tau protein in cardiac tissue is confirmed and the functional role in the cardiovascular system and the consequences of its loss were explored. Isolated right and left atria and small mesenteric arteries from wild type and tau deficient (KO) mice of two age groups (13 and 23 months old) were used to assess cardiovascular phenotypes. Tau KO mice showed an increased systolic blood pressure and cardiac hypertrophy at 13 months, which was accompanied by a significantly lower right atrial rate and a subtle decrease in the maximum contractility to calcium, isoprenaline, and electrical sympathetic nerve stimulation. Aging tau KO mice to 23 months resulted in cardiac hypertrophy with significantly attenuated left atrial contractility, increased blood pressure, and sensitivity of isolated mesenteric arteries to angiotensin II contraction and isoprenaline relaxation compared to their younger counterparts. This study supports a functional role of tau in the heart and loss of this protein leads to a deterioration in cardiovascular performance which worsens with age. Taken together, these results provide insight into the peripheral function of tau protein, and give caution to the therapeutic strategy of lowering tau protein.
Collapse
Affiliation(s)
- Ashenafi H Betrie
- Department of Pharmacology and Therapeutics, Cardiovascular Therapeutics Unit, The University of Melbourne, VIC, Australia
| | - Scott Ayton
- The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Ashley I Bush
- The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - James A Angus
- Department of Pharmacology and Therapeutics, Cardiovascular Therapeutics Unit, The University of Melbourne, VIC, Australia
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, China
| | - Christine E Wright
- Department of Pharmacology and Therapeutics, Cardiovascular Therapeutics Unit, The University of Melbourne, VIC, Australia
| |
Collapse
|
6
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
7
|
Potz BA, Parulkar AB, Abid RM, Sodha NR, Sellke FW. Novel molecular targets for coronary angiogenesis and ischemic heart disease. Coron Artery Dis 2017; 28:605-613. [PMID: 28678145 PMCID: PMC5624824 DOI: 10.1097/mca.0000000000000516] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Coronary artery disease (CAD) is the number one cause of death among men and women in the USA. Genetic predisposition and environmental factors lead to the development of atherosclerotic plaques in the vessel walls of the coronary arteries, resulting in decreased myocardial perfusion. Treatment includes a combination of revascularization procedures and medical therapy. Because of the high surgical risk of many of the patients undergoing revascularization procedures, medical therapies to reduce ischemic disease are an area of active research. Small molecule, cytokine, endothelial progenitor cell, stem cell, gene, and mechanical therapies show promise in increasing the collateral growth of blood vessels, thereby reducing myocardial ischemia.
Collapse
Affiliation(s)
- Brittany A Potz
- Department of Cardiothoracic Surgery, Research Division, Institution of Warren Alpert Medical School Brown University, Providence, Rhode Island, USA
| | | | | | | | | |
Collapse
|
8
|
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: 3.4] [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
|
9
|
Potz BA, Sabe AA, Elmadhun NY, Sabe SA, Braun BJV, Clements RT, Usheva A, Sellke FW. Calpain inhibition decreases inflammatory protein expression in vessel walls in a model of chronic myocardial ischemia. Surgery 2016; 161:1394-1404. [PMID: 28024857 DOI: 10.1016/j.surg.2016.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/03/2016] [Accepted: 11/05/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Emerging data suggest a link between calpain activation and the enhanced inflammatory response of the cardiovascular system. We hypothesize that calpain activation associates with altered inflammatory protein expression in correlation with the proinflammatory profile of the myocardium. Our pig hypercholesterolemic model with chronic myocardial ischemia was treated with calpain inhibitors to establish their potential to improve cardiac function. METHODS Yorkshire swine, fed a high cholesterol diet for 4 weeks then underwent placement of an ameroid constrictor on the left circumflex artery. Two weeks later, animals received either no drug (high-cholesterol control group, n = 8), a low dose of calpain inhibitors (0.12 mg/kg, n = 9), or a high dose of calpain inhibitors (0.25 mg/kg; n = 8). The high-cholesterol diet and calpain inhibitors were continued for 5 weeks, after which the pig was euthanized. The left ventricular myocardial tissue (ischemic and nonischemic) was harvested and analyzed for inflammatory protein expression. Data were statistically analyzed via the Kruskal-Wallis and Dunn post hoc test. RESULTS Calpain inhibitor treatment coincides with increased expression of IKB-α and decreased expression of macrophages, NFkB, IL-1, and tumor necrosis factor (TNF)-α in the ischemic myocardial tissue as compared with the control group. An NFkB array revealed decreased expression of IRF5, JNK1/2, JNK2, CD18, NFkB p65, c-Rel, Sharpin, TNF R1, TNF R2, and DR5 in the ischemic myocardium of the group treated with a high dose of calpain inhibitors compared with the control. CONCLUSION Calpain activation in metabolic syndrome is a potential contributor to cardiac dysfunction in metabolic disorders with ischemic background. We suggest that calpain inhibition downregulates NFkB signaling in the vessel walls, which might be useful for improving myocardial blood flow in ischemic conditions.
Collapse
Affiliation(s)
- Brittany A Potz
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Ashraf A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Nassrene Y Elmadhun
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Sharif A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Benedikt J V Braun
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Richard T Clements
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Anny Usheva
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI.
| |
Collapse
|
10
|
Potz BA, Sabe AA, Elmadhun NY, Clements RT, Abid MR, Sodha NR, Sellke FW. Calpain inhibition modulates glycogen synthase kinase 3β pathways in ischemic myocardium: A proteomic and mechanistic analysis. J Thorac Cardiovasc Surg 2016; 153:342-357. [PMID: 27986275 DOI: 10.1016/j.jtcvs.2016.09.087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 09/15/2016] [Accepted: 09/26/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Calpain inhibition has an enhancing effect on myocardial perfusion and improves myocardial density by inhibiting glycogen synthase kinase 3β (GSK-3β) and up-regulating downstream signaling pathways, including the insulin/PI3K and WNT/β-catenin pathways, in a pig model of chronic myocardial ischemia in the setting of metabolic syndrome. METHODS Pigs were fed a high-fat diet for 4 weeks, then underwent placement of an ameroid constrictor to the left circumflex artery. Three weeks later, the animals received no drug (high-cholesterol controls [HCC]), a high-dose calpain inhibitor (HCI), a low-dose calpain inhibitor (LCI), or a GSK-3β inhibitor (GSK-3βI). The diets and drug regimens were continued for 5 weeks and the myocardial tissue was harvested. RESULTS Calpain and GSK-3β inhibition caused an increase in myocardial perfusion ratios at rest and during pacing compared with controls. Pigs in the LCI and HCI groups had increased vessel density in the ischemic myocardium, and pigs in the GSK-3βI group had increased vessel density in the ischemic and nonischemic myocardium compared with the HCC group. Calpain inhibition modulates proteins involved in the insulin/PI3K and WNT/β-catenin pathways. Quantitative proteomics revealed that calpain and GSK-3β inhibition significantly modulated the expression of proteins enriched in cytoskeletal regulation, metabolism, respiration, and calcium-binding pathways. CONCLUSIONS In the setting of metabolic syndrome, calpain or GSK-3β inhibition increases vessel density in both ischemic and nonischemic myocardial tissue. Calpain inhibition may exert these effects through the inhibition of GSK-3β and up-regulation of downstream signaling pathways, including the insulin/PI3K and WNT/β-catenin pathways.
Collapse
Affiliation(s)
- Brittany A Potz
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Ashraf A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Nassrene Y Elmadhun
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Richard T Clements
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Neel R Sodha
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI.
| |
Collapse
|
11
|
Potz BA, Abid MR, Sellke FW. Role of Calpain in Pathogenesis of Human Disease Processes. JOURNAL OF NATURE AND SCIENCE 2016; 2:e218. [PMID: 27747292 PMCID: PMC5065022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Calpains are a 15-member class of calcium activated nonlysosomal neutral proteases which are involved in a broad range of cellular function. Calpains are usually localized to the cytosol and within mitochondria. Calpastatin is an endogenous protein that specifically binds to and inhibits calpain. Overactivation of calpain has been implicated in a number of disease processes of the brain, eyes, heart, lungs, pancreas, kidneys, vascular system and skeletal muscle. Therefore, calpain may serve as a potential therapeutic target for a wide variety of disease processes. This review briefly outlines the current literature regarding the involvement of calpain overactivation in the pathogenesis of almost every organ in the body.
Collapse
Affiliation(s)
| | | | - Frank W. Sellke
- Corresponding Author. Frank W Sellke, M.D., Division of Cardiothoracic Surgery, Cardiovascular Research Center Warren Alpert Medical School Brown University, 2 Dudley Street MOC 360, Providence, RI 02905, USA.
| |
Collapse
|
12
|
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
|
13
|
Abstract
Despite many advances in percutaneous and surgical interventions in the treatment of coronary artery disease (CAD), up to one-third of patients are still either not candidates or receive suboptimal revascularization. Calpains are a class of calcium-activated non-lysosomal cysteine proteases that serve as a proteolytic unit for cellular homeostasis. Uncontrolled activation of calpain has been found to be involved in the pathogenesis of myocardial reperfusion injury, cardiac hypertrophy, myocardial stunning and cardiac ischemia. Inhibition of calpains has been shown to significantly attenuate myocardial stunning and reduced infarct size after ischemia-reperfusion. Calpain inhibition therefore serves as a potential medical therapy for patients suffering from a number of diseases, including CAD.
Collapse
Affiliation(s)
- Brittany A Potz
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Warren Alpert Medical School Brown University
| | | | | | | |
Collapse
|
14
|
Lawton JS. What is the significance of "collateral dependent perfusion"? J Thorac Cardiovasc Surg 2015; 151:253-4. [PMID: 26456814 DOI: 10.1016/j.jtcvs.2015.09.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
Affiliation(s)
- Jennifer S Lawton
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St Louis, Mo.
| |
Collapse
|
15
|
Calpain inhibition improves collateral-dependent perfusion in a hypercholesterolemic swine model of chronic myocardial ischemia. J Thorac Cardiovasc Surg 2015; 151:245-52. [PMID: 26478238 DOI: 10.1016/j.jtcvs.2015.08.101] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/14/2015] [Accepted: 08/26/2015] [Indexed: 12/24/2022]
Abstract
PURPOSE Calpain overexpression is implicated in aberrant angiogenesis. We hypothesized that calpain inhibition (MDL28170) would improve collateral perfusion in a swine model with hypercholesterolemia and chronic myocardial ischemia. METHODS Yorkshire swine fed a high cholesterol diet for 4 weeks underwent surgical placement of an ameroid constrictor to their left circumflex coronary artery. Three weeks later, animals received no drug, high cholesterol control group (n = 8); low-dose calpain inhibition (0.12 mg/kg; n = 9); or high-dose calpain inhibition (0.25 mg/kg; n = 8). The heart was harvested after 5 weeks. RESULTS Myocardial perfusion in ischemic myocardium significantly improved with high-dose calpain inhibition at rest and with demand pacing (P = .016 and .011). Endothelium-dependent microvessel relaxation was significantly improved with low-dose calpain inhibition (P = .001). There was a significant increase in capillary density, with low-dose calpain inhibition and high-dose calpain inhibition (P = .01 and .01), and arteriolar density with low-dose calpain inhibition (P = .001). Calpain inhibition significantly increased several proangiogenic proteins, including vascular endothelial growth factor (P = .02), vascular endothelial growth factor receptor 1 (P = .003), vascular endothelial growth factor receptor 2 (P = .003), and talin, a microvascular structural protein (P = .0002). There was a slight increase in proteins implicated in endothelial-dependent (nitric oxide mediated) relaxation, including extracellular signal-regulated kinase, phosphorylated extracellular signal-regulated kinase, and inducible nitric oxide synthase with calpain inhibition. CONCLUSIONS In the setting of hypercholesterolemia, calpain inhibition improved perfusion, with a trend toward increased collateralization on angiography and increased capillary and arteriolar densities in ischemic myocardium. Calpain inhibition also improved endothelium-dependent microvessel relaxation and increased expression of proteins implicated in angiogenesis and vasodilatation.
Collapse
|
16
|
Potz BA, Sabe AA, Elmadhun NY, Feng J, Liu Y, Mitchell H, Quesenberry P, Abid MR, Sellke FW. Calpain inhibition decreases myocardial apoptosis in a swine model of chronic myocardial ischemia. Surgery 2015; 158:445-52. [PMID: 25991048 DOI: 10.1016/j.surg.2015.03.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/27/2015] [Accepted: 03/18/2015] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Calpain is a family of cysteine proteases that has an important role in the initiation, regulation, and execution of cell death. Our recent studies using a hypercholesterolemic swine model demonstrated that in the setting of the metabolic syndrome, calpain inhibition (CI) improved collateral-dependent perfusion and increased expression of proteins implicated in angiogenesis and vasodilation. In this study, we hypothesized that CI (by MLD28170) would decrease myocardial apoptosis in the same model. METHODS Yorkshire swine, all fed a high-cholesterol diet for 4 weeks underwent placement of an ameroid constrictor on the left circumflex coronary artery. Three weeks later, animals received either no drug, termed the high-cholesterol control group (HCC; n = 8); low-dose CI (0.12 mg/kg; LCI, n = 9); or high-dose CI (0.25 mg/kg; HCI, n = 8). The high-cholesterol diet and the CI were continued for 5 weeks, after which the pig was humanely killed and the left ventricular myocardium was harvested and analyzed via terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, oxyblot analysis, and Western blots. Data were analyzed using the Kruskal-Wallis test. RESULTS The percentage of apoptotic cells to total cells in ischemic myocardial territory was decreased in the LCI and HCI groups compared with the HCC group as shown by TUNEL staining (P = .018). There was a decrease in proapoptotic proteins, including cleaved caspase 3, caspase 9, cleaved caspase 9, Bax, BAD, p-BAD, and Erk 1/2 (P ≤ .049 each), but no decrease in caspase 3 (P = .737). There was also an increase in antiapoptotic proteins, including BCL-2 and p-BCL2 (P ≤ .025 each). In the ischemic myocardium, several proangiogenic proteins were increased in the LCI and HCI groups compared with the HCC group, including p-AKT, p-eNOS, and eNOS (P ≤ .006 each) but there was no increase in AKT (P = .311). CI decreased tissue oxidative stress in both the LCI and HCI groups compared to the HCC group as shown by oxyblot analysis (P = .021). CONCLUSION In the setting of hypercholesterolemia, CI decreases apoptosis and the expression of proteins in proapoptotic signaling pathways. CI also increased expression of proteins implicated in anti apoptotic pathways and improves oxidative stress in ischemic myocardial tissue.
Collapse
Affiliation(s)
- Brittany A Potz
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Ashraf A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Nassrene Y Elmadhun
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Jun Feng
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Yuhong Liu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Hunter Mitchell
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Peter Quesenberry
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI.
| |
Collapse
|
17
|
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: 116] [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
|
18
|
During tangential migration, SDF1 lends the cytoskeleton a guiding hand. J Neurosci 2014; 34:11868-9. [PMID: 25186735 DOI: 10.1523/jneurosci.2245-14.2014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
19
|
Sun FL, Wang W, Cheng H, Wang Y, Li L, Xue JL, Wang XF, Ai HX, Zhang L, Xu JD, Wang XM, Ji XM. Morroniside improves microvascular functional integrity of the neurovascular unit after cerebral ischemia. PLoS One 2014; 9:e101194. [PMID: 24979385 PMCID: PMC4076313 DOI: 10.1371/journal.pone.0101194] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 06/04/2014] [Indexed: 12/20/2022] Open
Abstract
Treating the vascular elements within the neurovascular unit is essential for protecting and repairing the brain after stroke. Acute injury on endothelial systems results in the disruption of blood-brain barrier (BBB), while post-ischemic angiogenesis plays an important role in delayed functional recovery. Here, we considered alterations in microvessel integrity to be targets for brain recovery, and tested the natural compound morroniside as a therapeutic approach to restore the vascular elements of injured tissue in a rat model of focal cerebral ischemia. Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) model, and morroniside was then administered intragastrically once a day at doses of 30, 90, and 270 mg/kg. BBB integrity and associated factors were analyzed to identify cerebrovascular permeability 3 days after MCAO. The recruitment of endothelial progenitor cells (EPCs), the expression of angiogenic factors and the new vessel formation in the peri-infarct cortex of rats were examined 7 days after MCAO to identify the angiogenesis. We demonstrated that at 3 days post-ischemia, morroniside preserved neurovascular unit function by ameliorating BBB injury. By 7 days post-ischemia, morroniside amplified angiogenesis, in part by enhancing endothelial progenitor cell proliferation and expression of angiogenic factors. Morever, the increase in the amount of vWF+ vessels induced by ischemia could be extended to 28 days after administration of morroniside, indicating the crucial role of morroniside in angiogenesis during the chronic phase. Taken together, our findings suggested that morroniside might offer a novel therapeutic approach for promoting microvascular integrity recovery and provide a thoroughly new direction for stroke therapy.
Collapse
Affiliation(s)
- Fang-Ling Sun
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wen Wang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
- * E-mail: (WW); (XMW); (XMJ)
| | - Hua Cheng
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ying Wang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lei Li
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Jin-Long Xue
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xiao-feng Wang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hou-Xi Ai
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Li Zhang
- Department of Pharmacology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Jing-dong Xu
- Department of Physiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao-Min Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- * E-mail: (WW); (XMW); (XMJ)
| | - Xun-Ming Ji
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
- * E-mail: (WW); (XMW); (XMJ)
| |
Collapse
|
20
|
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: 1.1] [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
|
21
|
Martin M, Geudens I, Bruyr J, Potente M, Bleuart A, Lebrun M, Simonis N, Deroanne C, Twizere JC, Soubeyran P, Peixoto P, Mottet D, Janssens V, Hofmann WK, Claes F, Carmeliet P, Kettmann R, Gerhardt H, Dequiedt F. PP2A regulatory subunit Bα controls endothelial contractility and vessel lumen integrity via regulation of HDAC7. EMBO J 2013; 32:2491-503. [PMID: 23955003 DOI: 10.1038/emboj.2013.187] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/19/2013] [Indexed: 01/04/2023] Open
Abstract
To supply tissues with nutrients and oxygen, the cardiovascular system forms a seamless, hierarchically branched, network of lumenized tubes. Here, we show that maintenance of patent vessel lumens requires the Bα regulatory subunit of protein phosphatase 2A (PP2A). Deficiency of Bα in zebrafish precludes vascular lumen stabilization resulting in perfusion defects. Similarly, inactivation of PP2A-Bα in cultured ECs induces tubulogenesis failure due to alteration of cytoskeleton dynamics, actomyosin contractility and maturation of cell-extracellular matrix (ECM) contacts. Mechanistically, we show that PP2A-Bα controls the activity of HDAC7, an essential transcriptional regulator of vascular stability. In the absence of PP2A-Bα, transcriptional repression by HDAC7 is abrogated leading to enhanced expression of the cytoskeleton adaptor protein ArgBP2. ArgBP2 hyperactivates RhoA causing inadequate rearrangements of the EC actomyosin cytoskeleton. This study unravels the first specific role for a PP2A holoenzyme in development: the PP2A-Bα/HDAC7/ArgBP2 axis maintains vascular lumens by balancing endothelial cytoskeletal dynamics and cell-matrix adhesion.
Collapse
Affiliation(s)
- Maud Martin
- Laboratory of Protein Signaling and Interactions, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, Sart-Tilman, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
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
|
23
|
EB1, p150Glued, and Clasp1 control endothelial tubulogenesis through microtubule assembly, acetylation, and apical polarization. Blood 2013; 121:3521-30. [PMID: 23444400 DOI: 10.1182/blood-2012-11-470179] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Vascular tube morphogenesis requires the establishment of endothelial cell (EC) apical-basal polarity in three-dimensional (3D) extracellular matrices. To date, there is little understanding of how EC polarity is controlled during these highly dynamic and rapid morphogenic events. We show that the microtubule tip complex proteins, end binding 1 (EB1), p150(Glued), and Clasp1, control human EC tube formation by (1) inducing microtubule assembly and asymmetric cytoskeletal polarization, whereby acetylated and detyrosinated tubulins distribute in a subapical membrane location and filamentous actin distributes basally; (2) increasing tubulin posttranslational modifications, including required acetylation events; and (3) regulating an EC lumen signaling cascade that involves membrane type 1 matrix metallopatrinase (MT1-MMP)-dependent proteolysis as well as Pak, Raf, and Erk kinases. Another regulator of this process is the microtubule stabilizing protein, tau, which binds p150(Glued) and similarly affects EC lumen formation by controlling the levels of acetylated and detyrosinated tubulins. Increased expression of the tubulin deacetylases, sirtuin 2, and histone deacetylase 6 (HDAC6), blocks EC tube formation and cytoskeletal polarization, while siRNA suppression of these deacetylases stimulates these events. Overall, this work reveals a fundamental role for microtubule tip complex proteins in coordinating microtubule assembly, posttranslational modifications including acetylation, and apical-basal cytoskeletal polarization to control the developing apical membrane surface during blood vessel tubulogenesis in 3D matrix environments.
Collapse
|
24
|
Miyazaki T, Koya T, Kigawa Y, Oguchi T, Lei XF, Kim-Kaneyama JR, Miyazaki A. Calpain and atherosclerosis. J Atheroscler Thromb 2012; 20:228-37. [PMID: 23171729 DOI: 10.5551/jat.14787] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
This review highlights the pro-atherogenic roles of Ca(2+)-sensitive intracellular protease calpains. Among more than ten species of calpain isozymes, µ- and m-calpains have been characterized most extensively. These two isozymes are ubiquitously expressed in mammalian tissues, including blood vessels, and tightly regulate functional molecules in the vascular component cells through limited proteolytic cleavage. Indeed, previous cell-based experiments showed that calpains play significant roles in nitric oxide production in vascular endothelial cells (ECs), maintenance of EC barrier function and angiogenesis for maintaining vascular homeostasis. Recently, we demonstrated that modified-low density lipoprotein (LDL)-induced m-calpain causes hyperpermeability in ECs, leading to the infiltration of monocytes/macrophages and plasma lipids into the intimal spaces (Miyazaki T. et al., Circulation. 2011; 124: 2522-2532). Calpains also mediate oxidized LDL-induced apoptotic death in ECs. In monocytes/macrophages, calpains induce proteolytic degradation of ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1), which results in impaired cholesterol efflux and subsequent macrophage foam cell formation. In vascular smooth muscle cells, calpains may be involved in the conversion from contractile phenotype to proliferative phenotype. In hepatocytes, calpains disrupt the biogenesis of high-density lipoprotein via proteolytic degradation of ABCA1. Thus, calpains may serve as novel candidate molecular targets for control of atherosclerosis.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, Tokyo 142-8555, Japan.
| | | | | | | | | | | | | |
Collapse
|
25
|
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: 46] [Impact Index Per Article: 3.8] [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
|
26
|
Abstract
Mitochondrial activity is critical for efficient function of the cardiovascular system. In response to cardiovascular injury, mitochondrial dysfunction occurs and can lead to apoptosis and necrosis. Calpains are a 15-member family of Ca(2+)-activated cysteine proteases localized to the cytosol and mitochondria, and several have been shown to regulate apoptosis and necrosis. For example, in endothelial cells, Ca(2+) overload causes mitochondrial calpain 1 cleavage of the Na(+)/Ca(2+) exchanger leading to mitochondrial Ca(2+) accumulation. Also, activated calpain 1 cleaves Bid, inducing cytochrome c release and apoptosis. In renal cells, calpains 1 and 2 promote apoptosis and necrosis by cleaving cytoskeletal proteins, which increases plasma membrane permeability and cleavage of caspases. Calpain 10 cleaves electron transport chain proteins, causing decreased mitochondrial respiration and excessive activation, or inhibition of calpain 10 activity induces mitochondrial dysfunction and apoptosis. In cardiomyocytes, calpain 1 activates caspase 3 and poly-ADP ribose polymerase during tumour necrosis factor-α-induced apoptosis, and calpain 1 cleaves apoptosis-inducing factor after Ca(2+) overload. Many of these observations have been elucidated with calpain inhibitors, but most calpain inhibitors are not specific for calpains or a specific calpain family member, creating more questions. The following review will discuss how calpains affect mitochondrial function and apoptosis within the cardiovascular system.
Collapse
Affiliation(s)
- Matthew A Smith
- Department of Pharmaceutical and Biomedical Sciences, Center for Cell Death, Injury, and Regeneration, Medical University of South Carolina, 280 Calhoun Street, MSC140, Charleston, SC 29425, USA
| | | |
Collapse
|
27
|
Ribeiro R, Monteiro C, Cunha V, Oliveira MJ, Freitas M, Fraga A, Príncipe P, Lobato C, Lobo F, Morais A, Silva V, Sanches-Magalhães J, Oliveira J, Pina F, Mota-Pinto A, Lopes C, Medeiros R. Human periprostatic adipose tissue promotes prostate cancer aggressiveness in vitro. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:32. [PMID: 22469146 PMCID: PMC3379940 DOI: 10.1186/1756-9966-31-32] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 04/02/2012] [Indexed: 12/13/2022]
Abstract
Background Obesity is associated with prostate cancer aggressiveness and mortality. The contribution of periprostatic adipose tissue, which is often infiltrated by malignant cells, to cancer progression is largely unknown. Thus, this study aimed to determine if periprostatic adipose tissue is linked with aggressive tumor biology in prostate cancer. Methods Supernatants of whole adipose tissue (explants) or stromal vascular fraction (SVF) from paired fat samples of periprostatic (PP) and pre-peritoneal visceral (VIS) anatomic origin from different donors were prepared and analyzed for matrix metalloproteinases (MMPs) 2 and 9 activity. The effects of those conditioned media (CM) on growth and migration of hormone-refractory (PC-3) and hormone-sensitive (LNCaP) prostate cancer cells were measured. Results We show here that PP adipose tissue of overweight men has higher MMP9 activity in comparison with normal subjects. The observed increased activities of both MMP2 and MMP9 in PP whole adipose tissue explants, likely reveal the contribution of adipocytes plus stromal-vascular fraction (SVF) as opposed to SVF alone. MMP2 activity was higher for PP when compared to VIS adipose tissue. When PC-3 cells were stimulated with CM from PP adipose tissue explants, increased proliferative and migratory capacities were observed, but not in the presence of SVF. Conversely, when LNCaP cells were stimulated with PP explants CM, we found enhanced motility despite the inhibition of proliferation, whereas CM derived from SVF increased both cell proliferation and motility. Explants culture and using adipose tissue of PP origin are most effective in promoting proliferation and migration of PC-3 cells, as respectively compared with SVF culture and using adipose tissue of VIS origin. In LNCaP cells, while explants CM cause increased migration compared to SVF, the use of PP adipose tissue to generate CM result in the increase of both cellular proliferation and migration. Conclusions Our findings suggest that the PP depot has the potential to modulate extra-prostatic tumor cells' microenvironment through increased MMPs activity and to promote prostate cancer cell survival and migration. Adipocyte-derived factors likely have a relevant proliferative and motile role.
Collapse
Affiliation(s)
- Ricardo Ribeiro
- Molecular Oncology Group-CI, Portuguese Institute of Oncology, Porto, Portugal.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
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: 64] [Impact Index Per Article: 5.3] [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
|
29
|
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: 2.2] [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
|
30
|
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: 37] [Impact Index Per Article: 2.8] [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
|