1
|
Fletcher EK, Ngwenyama N, Nguyen N, Turner SE, Covic L, Alcaide P, Kuliopulos A. Suppression of Heart Failure With PAR1 Pepducin Technology in a Pressure Overload Model in Mice. Circ Heart Fail 2023; 16:e010621. [PMID: 37477012 PMCID: PMC10592519 DOI: 10.1161/circheartfailure.123.010621] [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: 02/16/2023] [Accepted: 06/26/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND PAR1 (protease-activated receptor-1) contributes to acute thrombosis, but it is not clear whether the receptor is involved in deleterious inflammatory and profibrotic processes in heart failure. Here, we employ the pepducin technology to determine the effects of targeting PAR1 in a mouse heart failure with reduced ejection fraction model. METHODS After undergoing transverse aortic constriction pressure overload or sham surgery, C57BL/6J mice were randomized to daily sc PZ-128 pepducin or vehicle, and cardiac function, inflammation, fibrosis, and molecular analyses conducted at 7 weeks RESULTS: After 7 weeks of transverse aortic constriction, vehicle mice had marked increases in macrophage/monocyte infiltration and fibrosis of the left ventricle as compared with Sham mice. PZ-128 treatment significantly suppressed the inflammatory cell infiltration and cardiac fibrosis. Despite no effect on myocyte cell hypertrophy, PZ-128 afforded a significant reduction in overall left ventricle weight and completely protected against the transverse aortic constriction-induced impairments in left ventricle ejection fraction. PZ-128 significantly suppressed transverse aortic constriction-induced increases in an array of genes involved in myocardial stress, fibrosis, and inflammation. CONCLUSIONS The PZ-128 pepducin is highly effective in protecting against cardiac inflammation, fibrosis, and loss of left ventricle function in a mouse model.
Collapse
Affiliation(s)
- Elizabeth K Fletcher
- Center for Hemostasis and Thrombosis Research, Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center (E.K.F., N. Nguyen, S.E.T., L.C., A.K.)
| | - Njabulo Ngwenyama
- Department of Immunology, Tufts University School of Medicine, Boston, MA (N. Ngwenyama, P.A.)
| | - Nga Nguyen
- Center for Hemostasis and Thrombosis Research, Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center (E.K.F., N. Nguyen, S.E.T., L.C., A.K.)
| | - Susan E Turner
- Center for Hemostasis and Thrombosis Research, Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center (E.K.F., N. Nguyen, S.E.T., L.C., A.K.)
| | - Lidija Covic
- Center for Hemostasis and Thrombosis Research, Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center (E.K.F., N. Nguyen, S.E.T., L.C., A.K.)
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, MA (N. Ngwenyama, P.A.)
| | - Athan Kuliopulos
- Center for Hemostasis and Thrombosis Research, Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center (E.K.F., N. Nguyen, S.E.T., L.C., A.K.)
| |
Collapse
|
2
|
Govatati S, Pichavaram P, Kumar R, Rao GN. Blockade of CD47 function attenuates restenosis by promoting smooth muscle cell efferocytosis and inhibiting their migration and proliferation. J Biol Chem 2023; 299:104594. [PMID: 36898577 PMCID: PMC10124914 DOI: 10.1016/j.jbc.2023.104594] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/10/2023] Open
Abstract
Cluster of differentiation 47 (CD47) plays an important role in the pathophysiology of various diseases including atherosclerosis, but its role in neointimal hyperplasia which contributes to restenosis, has not been studied. Using molecular approaches in combination with a mouse vascular endothelial denudation model, we studied the role of CD47 in injury-induced neointimal hyperplasia. We determined that thrombin induced CD47 expression both in human and mouse aortic smooth muscle cells (HASMCs and MASMCs). In exploring the mechanisms, we found that the protease-activated receptor 1 (PAR1)-Gα protein q/11 (Gαq/11)-phospholipase Cβ3 (PLCβ3)-nuclear factor of activated T cells c1 (NFATc1) signaling axis regulates thrombin-induced CD47 expression in HASMCs. Depletion of CD47 levels using its siRNA or interference of its function by its blocking antibody (bAb) blunted thrombin-induced migration and proliferation of HASMCs and MASMCs. In addition, we found that thrombin-induced HASMC migration requires CD47 interaction with integrin β3. On the other hand, thrombin-induced HASMC proliferation was dependent on CD47's role in nuclear export and degradation of CDK-interacting protein 1 (p21Cip1). In addition, suppression of CD47 function by its bAb rescued HASMC efferocytosis from inhibition by thrombin. We also found that vascular injury induces CD47 expression in intimal SMCs and that inhibition of CD47 function by its bAb, while alleviating injury-induced inhibition of SMC efferocytosis, attenuated SMC migration and proliferation resulting in reduced neointima formation. Thus, these findings reveal a pathological role for CD47 in neointimal hyperplasia.
Collapse
Affiliation(s)
- Suresh Govatati
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Prahalathan Pichavaram
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Raj Kumar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
3
|
Li C, Pan Y, Tan Y, Wang Y, Sun X. PINK1-Dependent Mitophagy Reduced Endothelial Hyperpermeability and Cell Migration Capacity Under Simulated Microgravity. Front Cell Dev Biol 2022; 10:896014. [PMID: 35874841 PMCID: PMC9300855 DOI: 10.3389/fcell.2022.896014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
The effect of cardiovascular dysfunction including orthostatic intolerance and disability on physical exercise is one of the health problems induced by long-term spaceflight astronauts face. As an important part of vascular structure, the vascular endothelium, uniquely sensitive to mechanical force, plays a pivotal role in coordinating vascular functions. Our study found that simulated microgravity induced PINK1-dependent mitophagy in human umbilical vein endothelial cells (HUVECs). Here, we explored the underlying mechanism of mitophagy induction. The ER stress induced by proteostasis failure in HUVECs promoted the Ca2+ transfer from ER to mitochondria, resulting in mitochondria Ca2+ overload, decreased mitochondrial membrane potential, mitochondria fission, and accumulation of Parkin and p62 in mitochondria and mitophagy under simulated microgravity. Moreover, we assumed that mitophagy played a vital role in functional changes in endothelial cells under simulated microgravity. Using mdivi-1 and PINK1 knockdown, we found that NLRP3 inflammasome activation was enhanced after mitophagy was inhibited. The NLRP3 inflammasome contributed to endothelial hyperpermeability and cellular migration by releasing IL-1β. Thus, mitophagy inhibited cell migration ability and hyperpermeability in HUVECs exposed to clinostat-simulated microgravity. Collectively, we here clarify the mechanism of mitophagy induction by simulated microgravity in vitro and demonstrate the relationship between mitophagy and vascular endothelial functional changes including cellular migration and permeability. This study deepens the understanding of vascular functional changes under microgravity.
Collapse
Affiliation(s)
- Chengfei Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yikai Pan
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Yingjun Tan
- China Astronaut Research and Training Center, Beijing, China
| | - Yongchun Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiqing Sun, , Yongchun Wang,
| | - Xiqing Sun
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
- *Correspondence: Xiqing Sun, , Yongchun Wang,
| |
Collapse
|
4
|
Wang X, Wu YL, Zhang YY, Ke J, Wang ZW, Zhang BY, Ma Y, Yang LY, Zhao D. AK098656: a new biomarker of coronary stenosis severity in hypertensive and coronary heart disease patients. Diabetol Metab Syndr 2022; 14:10. [PMID: 35033179 PMCID: PMC8761339 DOI: 10.1186/s13098-022-00783-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AK098656 may be an adverse factor for coronary heart disease (CHD), especially in patients with hypertension. This study aimed to analyze the effect of AK098656 on CHD and CHD with various complications. METHODS A total of 117 CHD patients and 27 healthy control subjects were enrolled in the study. Plasma AK098656 expression was determined using the quantitative real-time polymerase chain reaction. Student's t-test was used to compare AK098656 expression levels in different groups. Receiver operating characteristic (ROC) curve and area under the curve (AUC) were used to quantify the discrimination ability between CHD patients and health controls and between CHD and CHD + complications patients. The relationship between AK098656 and coronary stenosis was analyzed using Spearman's correlation. RESULTS AK098656 expression was remarkably higher in CHD patients than in healthy controls (P = 0.03). The ROC curve revealed an effective predictive AK098656 expression value for CHD risk, with an AUC of 0.656 (95% CI 0.501-0.809). Moreover, AK098656 expression was increased in CHD + complications patients compared to CHD patients alone (P = 0.005), especially in patients with hypertension (CHD + hHTN, P = 0.030). The ROC curve revealed a predictive AK098656 prognostic value for discriminating between CHD and CHD + hHTN patients, with an AUC of 0.666 (95% CI 0.528-0.805). There was no significant difference in AK098656 expression in CHD patients with diabetes mellitus compared to CHD patients alone. In addition, AK098656 expression in CHD patients was positively correlated with stenosis severity (R = 0.261, P = 0.006). CONCLUSION AK098656 expression was significantly increased in patients with CHD, especially those with hypertension, and its expression level was positively correlated with the degree of coronary stenosis. This implied that AK098656 may be a risk factor for CHD and can potentially be applied in clinical diagnosis or provide a novel target for treatment.
Collapse
Affiliation(s)
- Xin Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Ya-Li Wu
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Yuan-Yuan Zhang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Zong-Wei Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Bao-Yu Zhang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Yan Ma
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Long-Yan Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China.
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China.
| |
Collapse
|
5
|
Lei D, Zhang X, Rouf MA, Mahendra Y, Wen L, Li Y, Zhang X, Li L, Wang L, Zhang T, Wang G, Wang Y. Noncanonical protease-activated receptor 1 regulates lymphatic differentiation in zebrafish. iScience 2021; 24:103386. [PMID: 34816109 PMCID: PMC8593614 DOI: 10.1016/j.isci.2021.103386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/26/2021] [Accepted: 10/26/2021] [Indexed: 10/26/2022] Open
Abstract
The differentiation of lymphatic progenitors is a crucial step in lymphangiogenesis. However, its underlying mechanism remains unclear. Here, we found that noncanonical protease-activated receptor 1 (par1) regulates the differentiation of lymphatic progenitors in zebrafish embryos. Loss of par1 function impaired lymphatic differentiation by downregulating prox1a expression in parachordal lymphangioblasts and caused compromised thoracic duct formation in zebrafish. Meanwhile, the G protein gnai2a, a par1 downstream effector, was selectively required for lymphatic development in zebrafish, and its mutation mimicked the lymphatic phenotype observed in par1 mutants. Interestingly, mmp13, but not thrombin, was required for lymphatic development in zebrafish. Furthermore, analyses of genetic interactions confirmed that mmp13b serves as a par1 upstream protease to regulate lymphatic development in zebrafish embryos. Mechanistically, par1 promotes flt4 expression and phospho-Erk1/2 activity in the posterior cardinal vein. Taken together, our findings highlight a function of par1 in the regulation of lymphatic differentiation in zebrafish embryos.
Collapse
Affiliation(s)
- Daoxi Lei
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.,Department of Ophthalmology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China
| | - Xiuru Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Muhammad Abdul Rouf
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yoga Mahendra
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Lin Wen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yan Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Xiaojuan Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Li Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Luming Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Tao Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yeqi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| |
Collapse
|
6
|
Seo Y, Heo Y, Jo S, Park SH, Lee C, Chang J, Jeon DK, Kim TG, Han G, Namkung W. Novel positive allosteric modulator of protease-activated receptor 1 promotes skin wound healing in hairless mice. Br J Pharmacol 2021; 178:3414-3427. [PMID: 33837955 DOI: 10.1111/bph.15489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 02/04/2021] [Accepted: 03/29/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Protease-activated receptor 1 (PAR1) is a GPCR expressed in several skin cell types, including keratinocyte and dermal fibroblast. PAR1 activation plays a crucial role in the process of skin wound healing such as thrombosis, inflammation, proliferation and tissue repair. In the present study, we identified a novel positive allosteric modulator of PAR1, GB83, and investigated its effect on skin wound healing. EXPERIMENTAL APPROACH The enhancement of PAR1 activity by GB83 was measured using Fluo-4 calcium assay. In silico docking analysis of GB83 in PAR1 was performed using dock ligands method (CDOCKER) with CHARMm force field. Effects of GB83 on cell viability and gene expression were observed using MTS assay and quantitative real-time PCRs, respectively. SKH-1 hairless mice were used to investigate the wound healing effect of GB83. KEY RESULTS We demonstrated that GB83 did not activate PAR1 by itself but strongly enhanced PAR1 activation by thrombin and PAR1-activating peptide (AP). In silico docking analysis revealed that GB83 can bind to the PAR1 binding site of vorapaxar. GB83 significantly promoted PAR1-mediated cell viability and migration. In addition, the enhancement of PAR1 activity by GB83 strongly increased gene expression of TGF-β, fibronectin and type I collagen in vitro and promoted skin wound healing in vivo. CONCLUSION AND IMPLICATIONS Our results revealed that GB83 is the first positive allosteric modulator of PAR1 and it can be a useful pharmacological tool for studying PAR1 and a potential therapeutic agent for skin wound healing.
Collapse
Affiliation(s)
- Yohan Seo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea.,Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul, Republic of Korea.,New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Yunkyung Heo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea
| | - Sungwoo Jo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea
| | - So-Hyeon Park
- Graduate Program of Industrial Pharmaceutical Science, Yonsei University, Incheon, Republic of Korea
| | - Chulho Lee
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jiwon Chang
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea
| | - Dong-Kyu Jeon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea
| | - Tae Gun Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Gyoonhee Han
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Wan Namkung
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea.,Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Fletcher EK, Wang Y, Flynn LK, Turner SE, Rade JJ, Kimmelstiel CD, Gurbel PA, Bliden KP, Covic L, Kuliopulos A. Deficiency of MMP1a (Matrix Metalloprotease 1a) Collagenase Suppresses Development of Atherosclerosis in Mice: Translational Implications for Human Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2021; 41:e265-e279. [PMID: 33761760 PMCID: PMC8062306 DOI: 10.1161/atvbaha.120.315837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Elizabeth K Fletcher
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| | - Yanling Wang
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| | - Laura K Flynn
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| | - Susan E Turner
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| | - Jeffrey J Rade
- Interventional Cardiology, Division of Cardiology, University of Massachusetts Memorial Medical Center, University of Massachusetts Medical School, Worcester (J.J.R.)
| | - Carey D Kimmelstiel
- Adult Interventional Cardiology, Division of Cardiology, Tufts Medical Center, Boston, MA (C.D.K.)
| | - Paul A Gurbel
- Inova Center for Thrombosis Research and Translational Medicine, Inova Fairfax Hospital, Falls Church, VA (P.A.G., K.P.B.)
- Sinai Hospital of Baltimore, MD (P.A.G., K.P.B.)
| | - Kevin P Bliden
- Inova Center for Thrombosis Research and Translational Medicine, Inova Fairfax Hospital, Falls Church, VA (P.A.G., K.P.B.)
- Sinai Hospital of Baltimore, MD (P.A.G., K.P.B.)
| | - Lidija Covic
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| | - Athan Kuliopulos
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| |
Collapse
|
8
|
Chandrabalan A, Ramachandran R. Molecular mechanisms regulating Proteinase‐Activated Receptors (PARs). FEBS J 2021; 288:2697-2726. [DOI: 10.1111/febs.15829] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Arundhasa Chandrabalan
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| |
Collapse
|
9
|
Sarver DC, Lei X, Wong GW. FAM19A (TAFA): An Emerging Family of Neurokines with Diverse Functions in the Central and Peripheral Nervous System. ACS Chem Neurosci 2021; 12:945-958. [PMID: 33621067 DOI: 10.1021/acschemneuro.0c00757] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cytokines and chemokines have diverse and pleiotropic functions in peripheral tissues and in the brain. Recent studies uncovered a novel family of neuron-derived secretory proteins, or neurokines, distantly related to chemokines. The FAM19A family comprises five ∼12-15 kDa secretory proteins (FAM19A1-5), also known as TAFA1-5, that are predominantly detected in the central and peripheral nervous system. FAM19A expression in the central nervous system is dynamically regulated during development and in the postnatal brain. As secreted ligands, FAM19A proteins appear to bind to different classes of cell surface receptors (e.g., GPCRs and neurexins). Functional studies using gain- and loss-of-function mouse models established nonredundant roles for each FAM19A family member in regulating diverse physiological processes ranging from locomotor activity and food intake to learning and memory, anxiety- and depressive-like behaviors, social communication, repetitive behaviors, and somatosensory functions. This review summarizes major advances as well as the limitations and knowledge gaps in understanding the regulation and diverse biological functions of this conserved family of neurokines.
Collapse
Affiliation(s)
- Dylan C. Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Xia Lei
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - G. William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
10
|
Abdel Rahman F, d'Almeida S, Zhang T, Asadi M, Bozoglu T, Bongiovanni D, von Scheidt M, Dietzel S, Schwedhelm E, Hinkel R, Laugwitz KL, Kupatt C, Ziegler T. Sphingosine-1-Phosphate Attenuates Lipopolysaccharide-Induced Pericyte Loss via Activation of Rho-A and MRTF-A. Thromb Haemost 2021; 121:341-350. [PMID: 33011963 DOI: 10.1055/s-0040-1716844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The high mortality seen in sepsis is caused by a systemic hypotension in part owing to a drastic increase in vascular permeability accompanied by a loss of pericytes. As has been shown previously, pericyte retention in the perivascular niche during sepsis can enhance the integrity of the vasculature and promote survival via recruitment of adhesion proteins such as VE-cadherin and N-cadherin. Sphingosine-1-phosphate (S1P) represents a lipid mediator regulating the deposition of the crucial adhesion molecule VE-cadherin at sites of interendothelial adherens junctions and of N-cadherin at endothelial-pericyte adherens junctions. Furthermore, in septic patients, S1P plasma levels are decreased and correlate with mortality in an indirectly proportional way. In the present study, we investigated the potential of S1P to ameliorate a lipopolysaccharide-induced septic hypercirculation in mice. Here we establish S1P as an antagonist of pericyte loss, vascular hyperpermeability, and systemic hypotension, resulting in an increased survival in mice. During sepsis S1P preserved VE-cadherin and N-cadherin deposition, mediated by a reduction of Src and cadherin phosphorylation. At least in part, this effect is mediated by a reduction of globular actin and a subsequent increase in nuclear translocation of MRTF-A (myocardin-related transcription factor A). These findings indicate that S1P may counteract pericyte loss and microvessel disassembly during sepsis and additionally emphasize the importance of pericyte-endothelial interactions to stabilize the vasculature.
Collapse
Affiliation(s)
- Farah Abdel Rahman
- Klinik & Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Sascha d'Almeida
- Klinik & Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | - Tina Zhang
- Klinik & Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | - Morad Asadi
- Klinik & Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | - Tarik Bozoglu
- Klinik & Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | - Dario Bongiovanni
- Klinik & Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Moritz von Scheidt
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Steffen Dietzel
- Walter-Brendl-Center for Experimental Medicine, LMU Munich, Munich, Germany
| | - Edzard Schwedhelm
- Center for Experimental Medicine, Institute of Clinical Pharmacology and Toxicology, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rabea Hinkel
- Klinik & Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention, LMU Munich, Munich, Germany
| | - Karl Ludwig Laugwitz
- Klinik & Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Kupatt
- Klinik & Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Tilman Ziegler
- Klinik & Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
11
|
Liu J, Xie X, Yan D, Wang Y, Yuan H, Cai Y, Luo J, Xu A, Huang Y, Cheung CW, Irwin MG, Xia Z. Up-regulation of FoxO1 contributes to adverse vascular remodelling in type 1 diabetic rats. J Cell Mol Med 2020; 24:13727-13738. [PMID: 33108705 PMCID: PMC7754018 DOI: 10.1111/jcmm.15935] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Vascular complications from diabetes often result in poor outcomes for patients, even after optimized interventions. Forkhead box protein O1 (FoxO1) is a key regulator of cellular metabolism and plays an important role in vessel formation and maturation. Alterations of FoxO1 occur in the cardiovascular system in diabetes, yet the role of FoxO1 in diabetic vascular complications is poorly understood. In Streptozotocin (STZ)‐induced type 1 diabetic rats, FoxO1 expression was up‐regulated in carotid arteries at 8 weeks of diabetes that was accompanied with adverse vascular remodelling characterized as increased wall thickness, carotid medial cross‐sectional area, media‐to‐lumen ratio and decreased carotid artery lumen area. This adverse vascular remodelling induced by hyperglycaemia in diabetic rats required FoxO1 activation as pharmacological inhibition of FoxO1 with 50mg/kg AS1842856 (AS) reversed vascular remodelling in type 1 diabetic rats. The adverse vascular remodelling in type 1 diabetes mellitus (T1DM) occurred concomitantly with increases in pro‐inflammatory factors, adhesion factors, apoptosis, NOD‐like receptor family protein‐3 inflammasome activation and the phenotypic switch of arterial smooth muscle cells, which were all reversed by AS. In addition, FoxO1 inhibition counteracted the down‐regulation of its upstream mediator PDK1 in T1DM. PDK1 activator reduced FoxO1 nuclear translocation, which serves as the basis for subsequent transcriptional regulation during hyperglycaemia. Taken together, our data suggest that FoxO1 is a critical trigger for type 1 diabetes‐induced vascular remodelling in rats, and inhibition of FoxO1 thus offers a potential therapeutic option for diabetes‐associated cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjin Liu
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Xiang Xie
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China.,Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dan Yan
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Yongshun Wang
- Department of Biomedical Science, University of Hong Kong, Hong Kong, China
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yin Cai
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Jierong Luo
- Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Heart and Vascular Institute and School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi Wai Cheung
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Michael G Irwin
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China.,State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| |
Collapse
|
12
|
Targeting perivascular and epicardial adipose tissue inflammation: therapeutic opportunities for cardiovascular disease. Clin Sci (Lond) 2020; 134:827-851. [PMID: 32271386 DOI: 10.1042/cs20190227] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Major shifts in human lifestyle and dietary habits toward sedentary behavior and refined food intake triggered steep increase in the incidence of metabolic disorders including obesity and Type 2 diabetes. Patients with metabolic disease are at a high risk of cardiovascular complications ranging from microvascular dysfunction to cardiometabolic syndromes including heart failure. Despite significant advances in the standards of care for obese and diabetic patients, current therapeutic approaches are not always successful in averting the accompanying cardiovascular deterioration. There is a strong relationship between adipose inflammation seen in metabolic disorders and detrimental changes in cardiovascular structure and function. The particular importance of epicardial and perivascular adipose pools emerged as main modulators of the physiology or pathology of heart and blood vessels. Here, we review the peculiarities of these two fat depots in terms of their origin, function, and pathological changes during metabolic deterioration. We highlight the rationale for pharmacological targeting of the perivascular and epicardial adipose tissue or associated signaling pathways as potential disease modifying approaches in cardiometabolic syndromes.
Collapse
|
13
|
Willis Fox O, Preston RJS. Molecular basis of protease-activated receptor 1 signaling diversity. J Thromb Haemost 2020; 18:6-16. [PMID: 31549766 DOI: 10.1111/jth.14643] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/13/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022]
Abstract
Protease-activated receptors (PARs) are a family of highly conserved G protein-coupled receptors (GPCRs) that respond to extracellular proteases via a unique proteolysis-dependent activation mechanism. Protease-activated receptor 1 (PAR1) was the first identified member of the receptor family and plays important roles in hemostasis, inflammation and malignancy. The biology underlying PAR1 signaling by its canonical agonist thrombin is well characterized; however, definition of the mechanistic basis of PAR1 signaling by other proteases, including matrix metalloproteases, activated protein C, plasmin, and activated factors VII and X, remains incompletely understood. In this review, we discuss emerging insights into the molecular bases for "biased" PAR1 signaling, including atypical PAR1 proteolysis, PAR1 heterodimer and coreceptor interactions, PAR1 translocation on the membrane surface, and interactions with different G-proteins and β-arrestins upon receptor activation. Moreover, we consider how these new insights into PAR1 signaling have acted to spur development of novel PAR1-targeted therapeutics that act to inhibit, redirect, or fine-tune PAR1 signaling output to treat cardiovascular and inflammatory disease. Finally, we discuss some of the key unanswered questions relating to PAR1 biology, in particular how differences in PAR1 proteolysis, signaling intermediate coupling, and engagement with coreceptors and GPCRs combine to mediate the diversity of identified PAR1 signaling outputs.
Collapse
Affiliation(s)
- Orla Willis Fox
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| |
Collapse
|
14
|
Atherosclerosis and the Capillary Network; Pathophysiology and Potential Therapeutic Strategies. Cells 2019; 9:cells9010050. [PMID: 31878229 PMCID: PMC7016600 DOI: 10.3390/cells9010050] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis and associated ischemic organ dysfunction represent the number one cause of mortality worldwide. While the key drivers of atherosclerosis, arterial hypertension, hypercholesterolemia and diabetes mellitus, are well known disease entities and their contribution to the formation of atherosclerotic plaques are intensively studied and well understood, less effort is put on the effect of these disease states on microvascular structure an integrity. In this review we summarize the pathological changes occurring in the vascular system in response to prolonged exposure to these major risk factors, with a particular focus on the differences between these pathological alterations of the vessel wall in larger arteries as compared to the microcirculation. Furthermore, we intend to highlight potential therapeutic strategies to improve microvascular function during atherosclerotic vessel disease.
Collapse
|
15
|
Zhang X, Huang T, Zhai H, Peng W, Zhou Y, Li Q, Yang H. Inhibition of lysine-specific demethylase 1A suppresses neointimal hyperplasia by targeting bone morphogenetic protein 2 and mediating vascular smooth muscle cell phenotype. Cell Prolif 2019; 53:e12711. [PMID: 31737960 PMCID: PMC6985674 DOI: 10.1111/cpr.12711] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/17/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Vascular disorders are associated with phenotypical switching of vascular smooth muscle cells (VSMCs). We investigated the effect of bone morphogenetic protein (BMP)-2 in controlling VSMC phenotype and vascular disorder progression. Lysine (K)-specific demethylase 1A (KDM1A) has been identified to target BMP-2 and is employed as a therapeutic means of regulating BMP-2 expression in VSMCs. MATERIALS AND METHODS VSMCs were stimulated with angiotensin II, and the expression of KDM1A and BMP-2 was detected. VSMC proliferation, apoptosis, and phenotype were evaluated. An in vivo aortic injury model was established, and VSMC behaviour was evaluated by the expression of key markers. The activation of BMP-2-associated signalling pathways was examined. RESULTS We confirmed the inhibitory effect of KDM1A on BMP-2 activity and demonstrated that KDM1A inhibition prevented VSMC transformation from a contractile to synthetic phenotype. In angiotensin II-treated VSMCs, KDM1A inhibition triggered a decrease in cell proliferation and inflammatory response. In vivo, KDM1A inhibition alleviated post-surgery neointimal formation and collagen deposition, preventing VSMCs from switching into a synthetic phenotype and suppressing disease onset. These processes were mediated by BMP-2 through canonical small mothers against decapentaplegic signalling, which was associated with the activation of BMP receptors 1A and 1B. CONCLUSIONS The regulatory correlation between KDM1A and BMP-2 offers insights into vascular remodelling and VSMC phenotypic modulation. The reported findings contribute to the development of innovative strategies against vascular disorders.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Huang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Zhai
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenpeng Peng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhou
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Li
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haifeng Yang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Slack MA, Gordon SM. Protease Activity in Vascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:e210-e218. [PMID: 31553665 PMCID: PMC6764587 DOI: 10.1161/atvbaha.119.312413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 01/13/2023]
Affiliation(s)
- Megan A. Slack
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Scott M. Gordon
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
17
|
Chen Y, Peng S, Cen H, Lin Y, Huang C, Chen Y, Shan H, Su Y, Zeng L. MicroRNA hsa-miR-623 directly suppresses MMP1 and attenuates IL-8-induced metastasis in pancreatic cancer. Int J Oncol 2019; 55:142-156. [PMID: 31115512 PMCID: PMC6561617 DOI: 10.3892/ijo.2019.4803] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 05/14/2019] [Indexed: 12/18/2022] Open
Abstract
Matrix metalloproteinase-1 (MMP1) participates in the metastasis of pancreatic cancer, and its expression can be regulated by endogenous microRNAs (miRs/miRNAs) and exogenous inflammatory factors. Whether miRNAs that potentially modulate MMP1 expression can also attenuate the pro-metastatic effects of its inducer on pancreatic cancer is yet to be completely elucidated. In the present study, a systematic analysis including in silico and bioinformatics analyses, a luciferase reporter assay and an RNA electrophoretic mobility shift assay (EMSA), were used to investigate the interaction between miRNAs and MMP1 mRNA. In addition, wound-healing assays, Transwell assays and xenograft nude mouse models were implemented to investigate the antitumor activities exerted by candidate miRNAs. As a result, hsa-miR-623 was screened as a candidate miRNA that interacts with the MMP1 transcript, and an inverse correlation between the expression of hsa-miR-623 and MMP1 was observed in human pancreatic cancer tissue samples. The EMSA confirmed that hsa-miR-623 was able to directly bind to its cognate target within the 3′-untranslated region of the MMP1 transcript. In addition, transfection of hsa-miR-623 mimics into PANC-1 and BXPC-3 cell lines markedly inhibited the expression of MMP1 at the mRNA and protein levels, and attenuated IL-8-induced MMP1 expression. hsa-miR-623 also decreased IL-8-induced epithelial-mesenchymal transition in PANC-1 and BXPC-3 cells via the underlying mechanism of inhibition of ERK phosphorylation. Consequently, hsa-miR-623 inhibited pancreatic cancer cell migration and invasion in vitro and metastasis in vivo. The results of the present study suggest that hsa-miR-623 represents a novel adjuvant therapeutic target to prevent metastasis in pancreatic cancer.
Collapse
Affiliation(s)
- Yutong Chen
- Department of Abdominal Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Siqi Peng
- Department of Abdominal Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Hong Cen
- Department of General Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Yujing Lin
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Chumei Huang
- Department of Gastroenterology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Yinting Chen
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Hong Shan
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Yonghui Su
- Department of General Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, P.R. China
| | - Linjuan Zeng
- Department of Abdominal Oncology, The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, P.R. China
| |
Collapse
|
18
|
Heuberger DM, Schuepbach RA. Protease-activated receptors (PARs): mechanisms of action and potential therapeutic modulators in PAR-driven inflammatory diseases. Thromb J 2019; 17:4. [PMID: 30976204 PMCID: PMC6440139 DOI: 10.1186/s12959-019-0194-8] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/08/2019] [Indexed: 12/29/2022] Open
Abstract
Inflammatory diseases have become increasingly prevalent with industrialization. To address this, numerous anti-inflammatory agents and molecular targets have been considered in clinical trials. Among molecular targets, protease-activated receptors (PARs) are abundantly recognized for their roles in the development of chronic inflammatory diseases. In particular, several inflammatory effects are directly mediated by the sensing of proteolytic activity by PARs. PARs belong to the seven transmembrane domain G protein-coupled receptor family, but are unique in their lack of physiologically soluble ligands. In contrast with classical receptors, PARs are activated by N-terminal proteolytic cleavage. Upon removal of specific N-terminal peptides, the resulting N-termini serve as tethered activation ligands that interact with the extracellular loop 2 domain and initiate receptor signaling. In the classical pathway, activated receptors mediate signaling by recruiting G proteins. However, activation of PARs alternatively lead to the transactivation of and signaling through receptors such as co-localized PARs, ion channels, and toll-like receptors. In this review we consider PARs and their modulators as potential therapeutic agents, and summarize the current understanding of PAR functions from clinical and in vitro studies of PAR-related inflammation.
Collapse
Affiliation(s)
- Dorothea M Heuberger
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Surgical Research Division, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reto A Schuepbach
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Covic L, Kuliopulos A. Protease-Activated Receptor 1 as Therapeutic Target in Breast, Lung, and Ovarian Cancer: Pepducin Approach. Int J Mol Sci 2018; 19:ijms19082237. [PMID: 30065181 PMCID: PMC6121574 DOI: 10.3390/ijms19082237] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/19/2018] [Accepted: 07/25/2018] [Indexed: 12/20/2022] Open
Abstract
The G-protein coupled receptors (GPCRs) belong to a large family of diverse receptors that are well recognized as pharmacological targets. However, very few of these receptors have been pursued as oncology drug targets. The Protease-activated receptor 1 (PAR1), which is a G-protein coupled receptor, has been shown to act as an oncogene and is an emerging anti-cancer drug target. In this paper, we provide an overview of PAR1’s biased signaling role in metastatic cancers of the breast, lungs, and ovaries and describe the development of PAR1 inhibitors that are currently in clinical use to treat acute coronary syndromes. PAR1 inhibitor PZ-128 is in a Phase II clinical trial and is being developed to prevent ischemic and thrombotic complication of patients undergoing cardiac catheterization. PZ-128 belongs to a new class of cell-penetrating, membrane-tethered peptides named pepducins that are based on the intracellular loops of receptors targeting the receptor G-protein interface. Application of PZ-128 as an anti-metastatic and anti-angiogenic therapeutic agent in breast, lung, and ovarian cancer is being reviewed.
Collapse
Affiliation(s)
- Lidija Covic
- Division of Hematology/Oncology, Tufts Medical Center, Boston, MA 02111, USA.
- Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA.
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, MA 02111, USA.
| | - Athan Kuliopulos
- Division of Hematology/Oncology, Tufts Medical Center, Boston, MA 02111, USA.
- Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA.
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Boston, MA 02111, USA.
| |
Collapse
|
20
|
Wang Y, Chen D, Zhang Y, Wang P, Zheng C, Zhang S, Yu B, Zhang L, Zhao G, Ma B, Cai Z, Xie N, Huang S, Liu Z, Mo X, Guan Y, Wang X, Fu Y, Ma D, Wang Y, Kong W. Novel Adipokine, FAM19A5, Inhibits Neointima Formation After Injury Through Sphingosine-1-Phosphate Receptor 2. Circulation 2018; 138:48-63. [DOI: 10.1161/circulationaha.117.032398] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 02/01/2018] [Indexed: 01/10/2023]
Abstract
Background:
Obesity plays crucial roles in the development of cardiovascular diseases. However, the mechanisms that link obesity and cardiovascular diseases remain elusive. Compelling evidence indicates that adipokines play an important role in obesity-related cardiovascular diseases. Here, we found a new adipokine-named family with sequence similarity 19, member A5 (FAM19A5), a protein with unknown function that was predicted to be distantly related to the CC-chemokine family. We aimed to test whether adipose-derived FAM19A5 regulates vascular pathology on injury.
Methods:
DNA cloning, protein expression, purification, and N-terminal sequencing were applied to characterize FAM19A5. Adenovirus infection and siRNA transfection were performed to regulate FAM19A5 expression. Balloon and wire injury were performed in vivo on the rat carotid arteries and mouse femoral arteries, respectively. Bioinformatics analysis, radioactive ligand-receptor binding assays, receptor internalization, and calcium mobilization assays were used to identify the functional receptor for FAM19A5.
Results:
We first characterized FAM19A5 as a secreted protein, and the first 43 N-terminal amino acids were the signal peptides. Both FAM19A5 mRNA and protein were abundantly expressed in the adipose tissue but were downregulated in obese mice. Overexpression of FAM19A5 markedly inhibited vascular smooth muscle cell proliferation and migration and neointima formation in the carotid arteries of balloon-injured rats. Accordingly, FAM19A5 silencing in adipocytes significantly promoted vascular smooth muscle cell activation. Adipose-specific FAM19A5 transgenic mice showed greater attenuation of neointima formation compared with wild-type littermates fed with or without Western-style diet. We further revealed that sphingosine-1-phosphate receptor 2 was the functional receptor for FAM19A5, with a dissociation constant (
K
d
) of 0.634 nmol/L. Inhibition of sphingosine-1-phosphate receptor 2 or its downstream G12/13-RhoA signaling circumvented the suppressive effects of FAM19A5 on vascular smooth muscle cell proliferation and migration.
Conclusions:
We revealed that a novel adipokine, FAM19A5, was capable of inhibiting postinjury neointima formation via sphingosine-1-phosphate receptor 2-G12/13-RhoA signaling. Downregulation of FAM19A5 during obesity may trigger cardiometabolic diseases.
Collapse
Affiliation(s)
- Yingbao Wang
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Dixin Chen
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yan Zhang
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Pingzhang Wang
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China (P.W., X.M., D.M., Y.W.)
| | - Can Zheng
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Songyang Zhang
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Bing Yu
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Lu Zhang
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Guizhen Zhao
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Baihui Ma
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Zeyu Cai
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Nan Xie
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Shiyang Huang
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ziyi Liu
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Xiaoning Mo
- Center for Human Disease Genomics, Peking University, Beijing, China (P.W., X.M., D.M., Y.W.)
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Liaoning, China (Y.G.)
| | - Xian Wang
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Yi Fu
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Dalong Ma
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China (P.W., X.M., D.M., Y.W.)
| | - Ying Wang
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China (P.W., X.M., D.M., Y.W.)
| | - Wei Kong
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| |
Collapse
|
21
|
Raghavan S, Singh NK, Mani AM, Rao GN. Protease-activated receptor 1 inhibits cholesterol efflux and promotes atherogenesis via cullin 3-mediated degradation of the ABCA1 transporter. J Biol Chem 2018; 293:10574-10589. [PMID: 29777060 DOI: 10.1074/jbc.ra118.003491] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/10/2018] [Indexed: 12/25/2022] Open
Abstract
Although signaling of thrombin via its receptor protease-activated receptor 1 (Par1) is known to occur in atherothrombosis, its link to the actual pathogenesis of this condition is less clear. To better understand the role of thrombin-Par1 signaling in atherosclerosis, here we have studied their effects on cellular cholesterol efflux in mice. We found that by activating Par1 and cullin 3-mediated ubiquitination and degradation of ABC subfamily A member 1 (ABCA1), thrombin inhibits cholesterol efflux in both murine macrophages and smooth muscle cells. Moreover, disruption of the Par1 gene rescued ABCA1 from Western diet-induced ubiquitination and degradation and restored cholesterol efflux in apolipoprotein E-deficient (ApoE-/-) mice. Similarly, the Par1 deletion diminished diet-induced atherosclerotic lesions in the ApoE-/- mice. These observations for the first time indicate a role for thrombin-Par1 signaling in the pathogenesis of diet-induced atherosclerosis. We identify cullin 3 as a cullin-RING ubiquitin E3 ligase that mediates ABCA1 ubiquitination and degradation and thereby inhibits cholesterol efflux. Furthermore, compared with peripheral blood mononuclear cells (PBMCs) from ApoE-/- mice, the PBMCs from ApoE-/-:Par1-/- mice exhibited decreased trafficking to inflamed arteries of Western diet-fed ApoE-/- mice. This finding suggested that besides inhibiting cholesterol efflux, thrombin-Par1 signaling also plays a role in the recruitment of leukocytes during diet-induced atherogenesis. Based on these findings, we conclude that thrombin-Par1 signaling appears to contribute to the pathogenesis of atherosclerosis by impairing cholesterol efflux from cells and by recruiting leukocytes to arteries.
Collapse
Affiliation(s)
- Somasundaram Raghavan
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Nikhlesh K Singh
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Arul M Mani
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Gadiparthi N Rao
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| |
Collapse
|
22
|
Rana R, Huang T, Koukos G, Fletcher EK, Turner SE, Shearer A, Gurbel PA, Rade JJ, Kimmelstiel CD, Bliden KP, Covic L, Kuliopulos A. Noncanonical Matrix Metalloprotease 1-Protease-Activated Receptor 1 Signaling Drives Progression of Atherosclerosis. Arterioscler Thromb Vasc Biol 2018; 38:1368-1380. [PMID: 29622563 DOI: 10.1161/atvbaha.118.310967] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/22/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Protease-activated receptor-1 (PAR1) is classically activated by thrombin and is critical in controlling the balance of hemostasis and thrombosis. More recently, it has been shown that noncanonical activation of PAR1 by matrix metalloprotease-1 (MMP1) contributes to arterial thrombosis. However, the role of PAR1 in long-term development of atherosclerosis is unknown, regardless of the protease agonist. APPROACH AND RESULTS We found that plasma MMP1 was significantly correlated (R=0.33; P=0.0015) with coronary atherosclerotic burden as determined by angiography in 91 patients with coronary artery disease and acute coronary syndrome undergoing cardiac catheterization or percutaneous coronary intervention. A cell-penetrating PAR1 pepducin, PZ-128, currently being tested as an antithrombotic agent in the acute setting in the TRIP-PCI study (Thrombin Receptor Inhibitory Pepducin-Percutaneous Coronary Intervention), caused a significant decrease in total atherosclerotic burden by 58% to 70% (P<0.05) and reduced plaque macrophage content by 54% (P<0.05) in apolipoprotein E-deficient mice. An MMP1 inhibitor gave similar beneficial effects, in contrast to the thrombin inhibitor bivalirudin that gave no improvement on atherosclerosis end points. Mechanistic studies revealed that inflammatory signaling mediated by MMP1-PAR1 plays a critical role in amplifying tumor necrosis factor α signaling in endothelial cells. CONCLUSIONS These data suggest that targeting the MMP1-PAR1 system may be effective in tamping down chronic inflammatory signaling in plaques and halting the progression of atherosclerosis.
Collapse
Affiliation(s)
- Rajashree Rana
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Tianfang Huang
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Georgios Koukos
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Elizabeth K Fletcher
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Susan E Turner
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Andrew Shearer
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Paul A Gurbel
- Inova Center for Thrombosis Research and Translational Medicine, Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, VA (P.A.G., K.P.B.)
| | - Jeffrey J Rade
- Department of Medicine, Division of Cardiology, University of Massachusetts Memorial Medical Center, University of Massachusetts Medical School, Worcester (J.J.R.)
| | - Carey D Kimmelstiel
- Department of Medicine, Division of Cardiology, Tufts Medical Center, Boston, MA (C.D.K.)
| | - Kevin P Bliden
- Inova Center for Thrombosis Research and Translational Medicine, Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, VA (P.A.G., K.P.B.)
| | - Lidija Covic
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Athan Kuliopulos
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| |
Collapse
|
23
|
Wei SD, Cao D, Xu XS, Bai H, Wu H, Wang MH. Investigation of the expressions of MMPs and TIMPs between isogeneic and allogeneic rat aortic transplantation. Mol Med Rep 2018; 17:6909-6913. [PMID: 29512764 DOI: 10.3892/mmr.2018.8684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/13/2018] [Indexed: 11/05/2022] Open
Abstract
The present study investigated the effects of matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs) in transplantation-associated arteriosclerosis by observing their expression in transplanted aortas in rats. Allogenic and isogenic abdominal aortic transplantations were performed and grafts were removed from the recipients at the designated time points (day 7, 14, 28 and 56 post transplantation). Hematoxylin and eosin staining, immunohistochemistry, immunofluorescence and western blot analysis were used to evaluate the grafts. Significant proliferation of the intima was observed in the allogenic transplantation groups (P<0.05). The expressions of MMPs and TIMPs in the allografts were significantly increased compared with the isografts, and the suppression of MMP2 in allografts reduced injury after transplantation. The present study concluded that the imbalance of MMPs and TIMPs led to the disturbance of synthesis and the degradation of the extracellular matrix and it may represent a key cause of chronic rejection.
Collapse
Affiliation(s)
- Si-Dong Wei
- Department of Hepatobiliary Surgery, People's Hospital of Zhengzhou, Southern Medical University, Zhengzhou, Henan 450003, P.R. China
| | - Ding Cao
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Xue-Song Xu
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - He Bai
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Hao Wu
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Meng-Hao Wang
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
24
|
Janjanam J, Zhang B, Mani AM, Singh NK, Traylor JG, Orr AW, Rao GN. LIM and cysteine-rich domains 1 is required for thrombin-induced smooth muscle cell proliferation and promotes atherogenesis. J Biol Chem 2018; 293:3088-3103. [PMID: 29326163 DOI: 10.1074/jbc.ra117.000866] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/09/2018] [Indexed: 11/06/2022] Open
Abstract
Restenosis arises after vascular injury and is characterized by arterial wall thickening and decreased arterial lumen space. Vascular injury induces the production of thrombin, which in addition to its role in blood clotting acts as a mitogenic and chemotactic factor. In exploring the molecular mechanisms underlying restenosis, here we identified LMCD1 (LIM and cysteine-rich domains 1) as a gene highly responsive to thrombin in human aortic smooth muscle cells (HASMCs). Of note, LMCD1 depletion inhibited proliferation of human but not murine vascular smooth muscle cells. We also found that by physically interacting with E2F transcription factor 1, LMCD1 mediates thrombin-induced expression of the CDC6 (cell division cycle 6) gene in the stimulation of HASMC proliferation. Thrombin-induced LMCD1 and CDC6 expression exhibited a requirement for protease-activated receptor 1-mediated Gαq/11-dependent activation of phospholipase C β3. Moreover, the expression of LMCD1 was highly induced in smooth muscle cells located at human atherosclerotic lesions and correlated with CDC6 expression and that of the proliferation marker Ki67. Furthermore, the LMCD1- and SMCαactin-positive cells had higher cholesterol levels in the atherosclerotic lesions. In conclusion, these findings indicate that by acting as a co-activator with E2F transcription factor 1 in CDC6 expression, LMCD1 stimulates HASMC proliferation and thereby promotes human atherogenesis, suggesting an involvement of LMCD1 in restenosis.
Collapse
Affiliation(s)
- Jagadeesh Janjanam
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Baolin Zhang
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Arul M Mani
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Nikhlesh K Singh
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - James G Traylor
- the Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71103
| | - A Wayne Orr
- the Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71103
| | - Gadiparthi N Rao
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| |
Collapse
|
25
|
Downregulation of MMP1 in MDS-derived mesenchymal stromal cells reduces the capacity to restrict MDS cell proliferation. Sci Rep 2017; 7:43849. [PMID: 28262842 PMCID: PMC5338350 DOI: 10.1038/srep43849] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/27/2017] [Indexed: 12/12/2022] Open
Abstract
The role of mesenchymal stromal cells (MSCs) in the pathogenesis of myelodysplastic syndromes (MDS) has been increasingly addressed, but has yet to be clearly elucidated. In this investigation, we found that MDS cells proliferated to a greater extent on MDS-derived MSCs compared to normal MSCs. Matrix metalloproteinase 1(MMP1), which was downregulated in MDS-MSCs, was identified as an inhibitory factor of MDS cell proliferation, given that treatment with an MMP1 inhibitor or knock-down of MMP1 in normal MSCs resulted in increased MDS cell proliferation. Further investigations indicated that MMP1 induced apoptosis of MDS cells by interacting with PAR1 and further activating the p38 MAPK pathway. Inhibition of either PAR1 or p38 MAPK can reverse the apoptosis-inducing effect of MMP1. Taken together, these data indicate that downregulation of MMP1 in MSCs of MDS patients may contribute to the reduced capacity of MSCs to restrict MDS cell proliferation, which may account for the malignant proliferation of MDS cells.
Collapse
|
26
|
Nguyen CH, Senfter D, Basilio J, Holzner S, Stadler S, Krieger S, Huttary N, Milovanovic D, Viola K, Simonitsch-Klupp I, Jäger W, de Martin R, Krupitza G. NF-κB contributes to MMP1 expression in breast cancer spheroids causing paracrine PAR1 activation and disintegrations in the lymph endothelial barrier in vitro. Oncotarget 2016; 6:39262-75. [PMID: 26513020 PMCID: PMC4770771 DOI: 10.18632/oncotarget.5741] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/05/2015] [Indexed: 12/31/2022] Open
Abstract
RELA, RELB, CREL, NFKB1 and NFKB2, and the upstream regulators NEMO and NIK were knocked-down in lymph endothelial cells (LECs) and in MDA-MB231 breast cancer spheroids to study the contribution of NF-κB in vascular barrier breaching. Suppression of RELA, NFKB1 and NEMO inhibited “circular chemo-repellent induced defects” (CCIDs), which form when cancer cells cross the lymphatic vasculature, by ~20–30%. Suppression of RELB, NFKB2 and NIK inhibited CCIDs by only ~10–15%. In MDA-MB231 cells RELA and NFKB1 constituted MMP1 expression, which caused the activation of PAR1 in adjacent LECs. The knock-down of MMP1 in MDA-MB231 spheroids and pharmacological inhibition of PAR1 in LECs inhibited CCID formation by ~30%. Intracellular Ca2+ release in LECs, which was induced by recombinant MMP1, was suppressed by the PAR1 inhibitor SCH79797, thereby confirming a functional intercellular axis: RELA/NFKB1 – MMP1 (MDA-MB231) – PAR1 (LEC). Recombinant MMP1 induced PAR1-dependent phosphorylation of MLC2 and FAK in LECs, which is indicative for their activity and for directional cell migration such as observed during CCID formation. The combined knock-down of the NF-κB pathways in LECs and MDA-MB231 spheroids inhibited CCIDs significantly stronger than knock-down in either cell type alone. Also the knock-down of ICAM-1 in LECs (a NF-κB endpoint with relevance for CCID formation) and knock-down of MMP1 in MDA-MB231 augmented CCID inhibition. This evidences that in both cell types NF-κB significantly and independently contributes to tumour-mediated breaching of the lymphatic barrier. Hence, inflamed tumour tissue and/or vasculature pose an additional threat to cancer progression.
Collapse
Affiliation(s)
- Chi Huu Nguyen
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria.,Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Daniel Senfter
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Jose Basilio
- Department of Vascular Biology and Thrombosis Research, Center of Biomolecular Medicine and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Silvio Holzner
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Serena Stadler
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Sigurd Krieger
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Nicole Huttary
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Daniela Milovanovic
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Katharina Viola
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | | | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Rainer de Martin
- Department of Vascular Biology and Thrombosis Research, Center of Biomolecular Medicine and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Georg Krupitza
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
27
|
Palygin O, Ilatovskaya DV, Staruschenko A. Protease-activated receptors in kidney disease progression. Am J Physiol Renal Physiol 2016; 311:F1140-F1144. [PMID: 27733370 DOI: 10.1152/ajprenal.00460.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/07/2016] [Indexed: 01/05/2023] Open
Abstract
Protease-activated receptors (PARs) are members of a well-known family of transmembrane G protein-coupled receptors (GPCRs). Four PARs have been identified to date, of which PAR1 and PAR2 are the most abundant receptors, and have been shown to be expressed in the kidney vascular and tubular cells. PAR signaling is mediated by an N-terminus tethered ligand that can be unmasked by serine protease cleavage. The receptors are activated by endogenous serine proteases, such as thrombin (acts on PARs 1, 3, and 4) and trypsin (PAR2). PARs can be involved in glomerular, microvascular, and inflammatory regulation of renal function in both normal and pathological conditions. As an example, it was shown that human glomerular epithelial and mesangial cells express PARs, and these receptors are involved in the pathogenesis of crescentic glomerulonephritis, glomerular fibrin deposition, and macrophage infiltration. Activation of these receptors in the kidney also modulates renal hemodynamics and glomerular filtration rate. Clinical studies further demonstrated that the concentration of urinary thrombin is associated with glomerulonephritis and type 2 diabetic nephropathy; thus, molecular and functional mechanisms of PARs activation can be directly involved in renal disease progression. We briefly discuss here the recent literature related to activation of PAR signaling in glomeruli and the kidney in general and provide some examples of PAR1 signaling in glomeruli podocytes.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | |
Collapse
|
28
|
Zeng L, Li Y, Yang J, Wang G, Margariti A, Xiao Q, Zampetaki A, Yin X, Mayr M, Mori K, Wang W, Hu Y, Xu Q. XBP 1-Deficiency Abrogates Neointimal Lesion of Injured Vessels Via Cross Talk With the PDGF Signaling. Arterioscler Thromb Vasc Biol 2015; 35:2134-44. [PMID: 26315405 DOI: 10.1161/atvbaha.115.305420] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 08/16/2015] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Smooth muscle cell (SMC) migration and proliferation play an essential role in neointimal formation after vascular injury. In this study, we intended to investigate whether the X-box-binding protein 1 (XBP1) was involved in these processes. APPROACH AND RESULTS In vivo studies on femoral artery injury models revealed that vascular injury triggered an immediate upregulation of XBP1 expression and splicing in vascular SMCs and that XBP1 deficiency in SMCs significantly abrogated neointimal formation in the injured vessels. In vitro studies indicated that platelet-derived growth factor-BB triggered XBP1 splicing in SMCs via the interaction between platelet-derived growth factor receptor β and the inositol-requiring enzyme 1α. The spliced XBP1 (XBP1s) increased SMC migration via PI3K/Akt activation and proliferation via downregulating calponin h1 (CNN1). XBP1s directed the transcription of mir-1274B that targeted CNN1 mRNA degradation. Proteomic analysis of culture media revealed that XBP1s decreased transforming growth factor (TGF)-β family proteins secretion via transcriptional suppression. TGF-β3 but not TGF-β1 or TGF-β2 attenuated XBP1s-induced CNN1 decrease and SMC proliferation. CONCLUSIONS This study demonstrates for the first time that XBP1 is crucial for SMC proliferation via modulating the platelet-derived growth factor/TGF-β pathways, leading to neointimal formation.
Collapse
Affiliation(s)
- Lingfang Zeng
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.).
| | - Yi Li
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Juanyao Yang
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Gang Wang
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Andriana Margariti
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Qingzhong Xiao
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Anna Zampetaki
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Xiaoke Yin
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Manuel Mayr
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Kazutoshi Mori
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Wen Wang
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Yanhua Hu
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.)
| | - Qingbo Xu
- From the Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (L.Z., Y.L., J.Y., A.Z., X.Y., M.M., Y.H., Q.X.); Institute of Bioengineering (J.Y., W.W.) and Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry (Q.X.), Queen Mary University of London, London, United Kingdom; Department of Emergency Medicine, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China (G.W.); Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom (A.M.); and Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan (K.M.).
| |
Collapse
|
29
|
Hegedűs P, Korkmaz S, Radovits T, Schmidt H, Li S, Yoshikawa Y, Yasui H, Merkely B, Karck M, Szabó G. Bis (aspirinato) zinc (II) complex successfully inhibits carotid arterial neointima formation after balloon-injury in rats. Cardiovasc Drugs Ther 2015; 28:533-9. [PMID: 25129612 DOI: 10.1007/s10557-014-6549-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Neointima formation following angioplasty is a serious consequence of endothelial damage in arteries. Inflammatory mediators and lack of endothelial regulatory mechanisms lead to migration and proliferation of smooth-muscle cells and thus to restenosis. This study examines the effect of the novel bis (aspirinato) zinc (II) complex on neointima formation in a rat model of carotid balloon-injury. METHODS Rats underwent balloon-injury of the right common carotid artery, then received PEG400 vehicle (untreated-group), acetylsalicylic-acid (ASA-group), zinc-chloride (Zn-group) and bis (aspirinato) zinc (II) complex (Zn(ASA) 2-group) orally for 18 consecutive days. From harvested carotid arteries, histology, immunohistochemistry and mRNA expression analysis were performed. RESULTS Compared to the untreated-group, Zn (ASA) 2-treatment significantly lowered stenosis ratio (54.0 ± 5.8% to 25.5 ± 3.9%) and reduced neointima/media ratio (1.5 ± 0.2 to 0.5 ± 0.1). Significantly higher alpha smooth muscle actin mRNA and protein expression were measured after Zn (ASA)2 and Zn-treatment in comparison with the untreated and ASA-groups while the expression of matrix-metalloproteinase-9 was significantly higher in these groups compared to Zn (ASA)2. The presence of collagen in media was significantly decreased in all treated groups. mRNA expressions of nuclear factor kappa-b, transforming growth-factor-β and proliferating cell nuclear antigen were significantly down-regulated, whereas a20 was up-regulated by Zn (ASA)2 treatment compared to the untreated and ASA-groups. CONCLUSION This study proves the effectivity of the novel bis (aspirinato) zinc complex in reducing neointima formation and restenosis after balloon-injury and supports the hypothesis that inhibition of smooth-muscle transformation/proliferation plays a key role in the prevention of restenosis.
Collapse
Affiliation(s)
- Péter Hegedűs
- Department of Cardiac Surgery, University of Heidelberg, INF 326, Heidelberg, 69120, Germany,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The endothelium provides an essential and selective membrane barrier that regulates the movement of water, solutes, gases, macromolecules and the cellular elements of the blood from the tissue compartment in health and disease. Its structure and continuous function is essential for life for all vertebrate organisms. Recent evidence indicates that the endothelial surface does not have a passive role in systemic inflammatory states such as septic shock. In fact, endothelial cells are in dynamic equilibrium with a myriad of inflammatory mediators and elements of the innate immune and coagulation systems to orchestrate the host response in sepsis. The barrier function of the endothelial surface is almost uniformly impaired in septic shock, and it is likely that this contributes to adverse outcomes. In this review, we will highlight recent advances in the understanding of the signalling events that regulate endothelial function and molecular events that induce endothelial dysfunction in sepsis. Endothelial barrier repair strategies as a treatment for sepsis include modulation of C5a, high-mobility group box 1 and VEGF receptor 2; stimulation of angiopoietin-1, sphingosine 1 phosphate receptor 1 and Slit; and a number of other innovative approaches.
Collapse
Affiliation(s)
- S M Opal
- Infectious Disease Division, Alpert Medical School of Brown University, Pawtucket, RI, USA
| | - T van der Poll
- Academic Medical Center, Division of Infectious Diseases & The Center of Experimental and Molecular Medicine, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
31
|
Wang M, Kim SH, Monticone RE, Lakatta EG. Matrix metalloproteinases promote arterial remodeling in aging, hypertension, and atherosclerosis. Hypertension 2015; 65:698-703. [PMID: 25667214 DOI: 10.1161/hypertensionaha.114.03618] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Mingyi Wang
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Biomedical Research Center (BRC), Baltimore, MD.
| | - Soo Hyuk Kim
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Biomedical Research Center (BRC), Baltimore, MD
| | - Robert E Monticone
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Biomedical Research Center (BRC), Baltimore, MD
| | - Edward G Lakatta
- From the Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Biomedical Research Center (BRC), Baltimore, MD.
| |
Collapse
|
32
|
Foley CJ, Kuliopulos A. Mouse matrix metalloprotease-1a (Mmp1a) gives new insight into MMP function. J Cell Physiol 2014; 229:1875-80. [PMID: 24737602 DOI: 10.1002/jcp.24650] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 04/14/2014] [Indexed: 01/18/2023]
Abstract
Matrix metalloprotease-1 (MMP1) has been implicated in many human disease processes, however the lack of a well characterized murine homologue has significantly limited the study of MMP1 and the development of MMP-targeted therapeutics. The discovery of murine Mmp1a in 2001, the functional mouse homologue of MMP1, offers a valuable tool for modeling MMP1-mediated processes in mice. Variation in physiologic expression levels of Mmp1a in mice as compared to MMP1 in humans highlights the importance of understanding the similarities and differences between the homologues. Recent studies have demonstrated tumor growth-, invasion-, and angiogenesis-promoting functions of Mmp1a in lung cancer models, consistent with the analogous functions observed for human MMP1. Biochemical investigations have shown that point mutations in the pro-domain of mouse Mmp1a weaken docking between the pro- and catalytic domains, generating an unstable zymogen primed for activation. The difficulty to effectively maintain Mmp1a in the zymogen form may account for the tight control of Mmp1a expression and reduced expression in normal tissue as compared to inflammatory states or cancer. This discovery raises important questions about the activation mechanisms and regulation of the MMP family in general.
Collapse
Affiliation(s)
- Caitlin J Foley
- Molecular Oncology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts; Program in Genetics, Tufts University School of Medicine, Boston, Massachusetts
| | | |
Collapse
|
33
|
Zhao C, Ju J. Molecular cloning, expression, and anti-tumor activity of a novel serine protease from Arenicola cristata. Acta Biochim Biophys Sin (Shanghai) 2014; 46:450-9. [PMID: 24709333 DOI: 10.1093/abbs/gmu020] [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] [Indexed: 11/14/2022] Open
Abstract
Arenicola cristata, a marine annelid, is a well-known and prized traditional Chinese medicine. However, the serine protease gene of A. cristata has not been cloned yet. In this study, a novel protease of A. cristata was cloned, sequenced, and expressed in Escherichia coli, and the functions of this recombinant protease were also investigated. The whole complementary DNA (cDNA) of this novel protease was of 980 bp in length and consisted of an open reading frame of 861 bp encoding 286 aa. Sequence analysis of the deduced amino acid sequence revealed that the protease belongs to the serine protease family. The active enzyme of the proposed A. cristata protease is composed of a signal peptide, a propeptide, and a mature polypeptide. The molecular weight of the recombinant mature protein was ~26 kDa after over-expression in E. coli. The recombinant protein significantly inhibited cell growth and induced cell apoptosis of esophageal squamous cell carcinoma (ESCC) in vitro, and reduced tumorigenicity in vivo. Furthermore, administration of the recombinant protein led to the activation of caspase-9 as well as down-regulation of Mcl-1 and Bcl-2. Taken together, our findings indicated that the recombinant serine protease of A. cristata could inhibit ESCC cell growth by mitochondrial apoptotic pathway and might act as a potential pharmacological agent for ESCC therapy.
Collapse
Affiliation(s)
- Chunling Zhao
- College of Pharmacy and Biological Science, Weifang Medical University, Weifang 261053, China
| | - Jiyu Ju
- College of Basic Medicine, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
34
|
Zhao P, Metcalf M, Bunnett NW. Biased signaling of protease-activated receptors. Front Endocrinol (Lausanne) 2014; 5:67. [PMID: 24860547 PMCID: PMC4026716 DOI: 10.3389/fendo.2014.00067] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/22/2014] [Indexed: 01/06/2023] Open
Abstract
In addition to their role in protein degradation and digestion, proteases can also function as hormone-like signaling molecules that regulate vital patho-physiological processes, including inflammation, hemostasis, pain, and repair mechanisms. Certain proteases can signal to cells by cleaving protease-activated receptors (PARs), a family of four G protein-coupled receptors. PARs are expressed by almost all cell types, control important physiological and disease-relevant processes, and are an emerging therapeutic target for major diseases. Most information about PAR activation and function derives from studies of a few proteases, for example thrombin in the case of PAR1, PAR3, and PAR4, and trypsin in the case of PAR2 and PAR4. These proteases cleave PARs at established sites with the extracellular N-terminal domains, and expose tethered ligands that stabilize conformations of the cleaved receptors that activate the canonical pathways of G protein- and/or β-arrestin-dependent signaling. However, a growing number of proteases have been identified that cleave PARs at divergent sites to activate distinct patterns of receptor signaling and trafficking. The capacity of these proteases to trigger distinct signaling pathways is referred to as biased signaling, and can lead to unique patho-physiological outcomes. Given that a different repertoire of proteases are activated in various patho-physiological conditions that may activate PARs by different mechanisms, signaling bias may account for the divergent actions of proteases and PARs. Moreover, therapies that target disease-relevant biased signaling pathways may be more effective and selective approaches for the treatment of protease- and PAR-driven diseases. Thus, rather than mediating the actions of a few proteases, PARs may integrate the biological actions of a wide spectrum of proteases in different patho-physiological conditions.
Collapse
Affiliation(s)
- Peishen Zhao
- Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
| | - Matthew Metcalf
- Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
| | - Nigel W. Bunnett
- Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
- Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia
- *Correspondence: Nigel W. Bunnett, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, VIC 3052, Australia e-mail:
| |
Collapse
|