1
|
Proteomic Analysis of Synovial Fibroblasts and Articular Chondrocytes Co-Cultures Reveals Valuable VIP-Modulated Inflammatory and Degradative Proteins in Osteoarthritis. Int J Mol Sci 2021; 22:ijms22126441. [PMID: 34208590 PMCID: PMC8235106 DOI: 10.3390/ijms22126441] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is the most common musculoskeletal disorder causing a great disability and a reduction in the quality of life. In OA, articular chondrocytes (AC) and synovial fibroblasts (SF) release innate-derived immune mediators that initiate and perpetuate inflammation, inducing cartilage extracellular matrix (ECM) degradation. Given the lack of therapies for the treatment of OA, in this study, we explore biomarkers that enable the development of new therapeutical approaches. We analyze the set of secreted proteins in AC and SF co-cultures by stable isotope labeling with amino acids (SILAC). We describe, for the first time, 115 proteins detected in SF-AC co-cultures stimulated by fibronectin fragments (Fn-fs). We also study the role of the vasoactive intestinal peptide (VIP) in this secretome, providing new proteins involved in the main events of OA, confirmed by ELISA and multiplex analyses. VIP decreases proteins involved in the inflammatory process (CHI3L1, PTX3), complement activation (C1r, C3), and cartilage ECM degradation (DCN, CTSB and MMP2), key events in the initiation and progression of OA. Our results support the anti-inflammatory and anti-catabolic properties of VIP in rheumatic diseases and provide potential new targets for OA treatment.
Collapse
|
2
|
Martínez C, Juarranz Y, Gutiérrez-Cañas I, Carrión M, Pérez-García S, Villanueva-Romero R, Castro D, Lamana A, Mellado M, González-Álvaro I, Gomariz RP. A Clinical Approach for the Use of VIP Axis in Inflammatory and Autoimmune Diseases. Int J Mol Sci 2019; 21:E65. [PMID: 31861827 PMCID: PMC6982157 DOI: 10.3390/ijms21010065] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
The neuroendocrine and immune systems are coordinated to maintain the homeostasis of the organism, generating bidirectional communication through shared mediators and receptors. Vasoactive intestinal peptide (VIP) is the paradigm of an endogenous neuropeptide produced by neurons and endocrine and immune cells, involved in the control of both innate and adaptive immune responses. Exogenous administration of VIP exerts therapeutic effects in models of autoimmune/inflammatory diseases mediated by G-protein-coupled receptors (VPAC1 and VPAC2). Currently, there are no curative therapies for inflammatory and autoimmune diseases, and patients present complex diagnostic, therapeutic, and prognostic problems in daily clinical practice due to their heterogeneous nature. This review focuses on the biology of VIP and VIP receptor signaling, as well as its protective effects as an immunomodulatory factor. Recent progress in improving the stability, selectivity, and effectiveness of VIP/receptors analogues and new routes of administration are highlighted, as well as important advances in their use as biomarkers, contributing to their potential application in precision medicine. On the 50th anniversary of VIP's discovery, this review presents a spectrum of potential clinical benefits applied to inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Carmen Martínez
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Yasmina Juarranz
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Irene Gutiérrez-Cañas
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Mar Carrión
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Selene Pérez-García
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Raúl Villanueva-Romero
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - David Castro
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Amalia Lamana
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Mario Mellado
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología (CNB)/CSIC, 28049 Madrid, Spain;
| | - Isidoro González-Álvaro
- Servicio de Reumatología, Instituto de Investigación Médica, Hospital Universitario La Princesa, 28006 Madrid, Spain;
| | - Rosa P. Gomariz
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| |
Collapse
|
3
|
Tang B, Wu J, Zhu MX, Sun X, Liu J, Xie R, Dong TX, Xiao Y, Carethers JM, Yang S, Dong H. VPAC1 couples with TRPV4 channel to promote calcium-dependent gastric cancer progression via a novel autocrine mechanism. Oncogene 2019; 38:3946-3961. [PMID: 30692637 DOI: 10.1038/s41388-019-0709-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/12/2018] [Accepted: 01/04/2019] [Indexed: 01/20/2023]
Abstract
Although VPAC1 and its ligand vasoactive intestinal peptide (VIP) are important in gastrointestinal physiology, their involvements in progression of gastrointestinal tumor have not been explored. Here, we found that higher expression of VIP/VPAC1 was observed in gastric cancer compared to the adjacent normal tissues. The increased expression of VIP/VPAC1 in gastric cancer correlated positively with invasion, tumor stage, lymph node, distant metastases, and poor survival. Moreover, high expression of VIP and VPAC1, advanced tumor stage and distant metastasis were independent prognostic factors. VPAC1 activation by VIP markedly induced TRPV4-mediated Ca2+ entry, and eventually promoted gastric cancer progression in a Ca2+ signaling-dependent manner. Inhibition of VPAC1 and its signaling pathway could block the progressive responses. VPAC1/TRPV4/Ca2+ signaling in turn enhanced the expression and secretion of VIP in gastric cancer cells, enforcing a positive feedback regulation mechanism. Taken together, our study demonstrate that VPAC1 is significantly overexpressed in gastric cancer and VPAC1/TRPV4/Ca2+ signaling axis could enforce a positive feedback regulation in gastric cancer progression. VIP/VPAC1 may serve as potential prognostic markers and therapeutic targets for gastric cancer.
Collapse
Affiliation(s)
- Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Department of Medicine, School of Medicine, University of California, San Diego, CA, USA
| | - Jilin Wu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Michael X Zhu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xuemei Sun
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jingjing Liu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Rui Xie
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Department of Gastroenterology, Affiliated Hospital to Zunyi Medical College, Zunyi, China
| | - Tobias Xiao Dong
- Department of Medicine, School of Medicine, University of California, San Diego, CA, USA
| | - Yufeng Xiao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - John M Carethers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China. .,Department of Medicine, School of Medicine, University of California, San Diego, CA, USA.
| |
Collapse
|
4
|
Mirsaidi N, Burns MP, McClain SA, Forsyth E, Li J, Dukes B, Lin D, Nahvi R, Giraldo J, Patton M, Wang P, Lin K, Miller E, Ratliff T, Hamidi S, Crist S, Takemaru KI, Szema A. Enhanced Mortality to Metastatic Bladder Cancer Cell Line MB49 in Vasoactive Intestinal Peptide Gene Knockout Mice. Front Endocrinol (Lausanne) 2017; 8:162. [PMID: 28824540 PMCID: PMC5545686 DOI: 10.3389/fendo.2017.00162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 06/23/2017] [Indexed: 11/24/2022] Open
Abstract
To identify if the absence of the vasoactive intestinal peptide (VIP) gene enhances susceptibility to death from metastatic bladder cancer, two strains of mice were injected with MB49 murine bladder cancer cells. The growth and spread of the cancer was measured over a period of 4 weeks in C57BL/6 mice and 5 weeks in VIP knockout (KO) mice. A Kaplan-Meier plot was constructed to compare control C57BL/6 mice and C57BL/6 mice with MB49 vs. VIP KO controls and VIP KO mice with MB49. The wild-type (WT) strain (C57BL/6) contained the VIP gene, while the other strain, VIP knockout backcrossed to C57BL/6 (VIP KO) did not and was thus unable to endogenously produce VIP. VIP KO mice had increased mortality compared to C57BL/6 mice at 4 weeks. The number of ulcers between both groups was not statistically significant. In vitro studies indicated that the presence VIP in high doses reduced MB49 cell growth, as well as macrophage inhibitory factor (MIF), a growth factor in bladder cancer cells. These findings support the concept that VIP may attenuate susceptibility to death from bladder cancer, and that it exerts its effect via downregulation of MIF.
Collapse
Affiliation(s)
- Niely Mirsaidi
- Department of Technology and Society, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, United States
- Three Village Allergy & Asthma, PLLC, South Setauket, NY, United States
| | - Matthew P. Burns
- Department of Technology and Society, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, United States
- Three Village Allergy & Asthma, PLLC, South Setauket, NY, United States
| | | | - Edward Forsyth
- Department of Urology, Stony Brook University School of Medicine, Stony Brook, NY, United States
| | - Jonathan Li
- Department of Technology and Society, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, United States
- Three Village Allergy & Asthma, PLLC, South Setauket, NY, United States
| | - Brittany Dukes
- Department of Technology and Society, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, United States
- Three Village Allergy & Asthma, PLLC, South Setauket, NY, United States
| | - David Lin
- Department of Technology and Society, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, United States
- Three Village Allergy & Asthma, PLLC, South Setauket, NY, United States
| | - Roxanna Nahvi
- Department of Technology and Society, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, United States
- Three Village Allergy & Asthma, PLLC, South Setauket, NY, United States
| | - Jheison Giraldo
- Department of Technology and Society, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, United States
- Three Village Allergy & Asthma, PLLC, South Setauket, NY, United States
| | - Megan Patton
- Three Village Allergy & Asthma, PLLC, South Setauket, NY, United States
| | - Ping Wang
- The Feinstein Institute for Medical Research, Center for Heart and Lung Research, Manhasset, NY, United States
| | - Ke Lin
- The Feinstein Institute for Medical Research, Center for Heart and Lung Research, Manhasset, NY, United States
| | - Edmund Miller
- The Feinstein Institute for Medical Research, Center for Heart and Lung Research, Manhasset, NY, United States
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- Hofstra Northwell School of Medicine, Hempstead, NY, United States
| | - Timothy Ratliff
- Purdue University, Center for Cancer Research, West Lafayette, IN, United States
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Sayyed Hamidi
- James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Scott Crist
- Purdue University, Center for Cancer Research, West Lafayette, IN, United States
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Ken-Ichi Takemaru
- Department of Pharmacological Sciences, Stony Brook University School of Medicine, Stony Brook, NY, United States
| | - Anthony Szema
- Department of Technology and Society, College of Engineering and Applied Sciences, Stony Brook University, Stony Brook, NY, United States
- Three Village Allergy & Asthma, PLLC, South Setauket, NY, United States
- The Feinstein Institute for Medical Research, Center for Heart and Lung Research, Manhasset, NY, United States
- Department of Occupational Medicine, Epidemiology, and Prevention, Hofstra Northwell School of Medicine, Hempstead, NY, United States
- Northwell Health, Department of Medicine, Division of Pulmonary and Critical Care, Manhasset, NY, United States
- Northwell Health, Department of Medicine, Division of Allergy and Immunology, Manhasset, NY, United States
- *Correspondence: Anthony Szema,
| |
Collapse
|
5
|
Wu W, Xia Q, Luo RJ, Lin ZQ, Xue P. In vitro Study of the Antagonistic Effect of Low-dose Liquiritigenin on Gemcitabine-induced Capillary Leak Syndrome in Pancreatic Adenocarcinoma via Inhibiting ROS- Mediated Signalling Pathways. Asian Pac J Cancer Prev 2016; 16:4369-76. [PMID: 26028101 DOI: 10.7314/apjcp.2015.16.10.4369] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND To investigate in-vitro antagonistic effect of low-dose liquiritigenin on gemcitabine-induced capillary leak syndrome (CLS) in pancreatic adenocarcinoma via inhibiting reactive oxygen species (ROS)- mediated signalling pathways. MATERIALS AND METHODS Human pancreatic adenocarcinoma Panc-1 cells and human umbilical vein endothelial cells (HUVECs) were pre-treated using low-dose liquiritigenin for 24 h, then added into gemcitabine and incubated for 48 h. Cell viability, apoptosis rate and ROS levels of Panc-1 cells and HUVECs were respectively detected through methylthiazolyldiphenyl-tetrazoliumbromide (MTT) and flow cytometry. For HUVECs, transendothelial electrical resistance (TEER) and transcellular and paracellular leak were measured using transwell assays, then poly (ADP-ribose) polymerase 1 (PARP-1) and metal matrix proteinase-9 (MMP9) activity were assayed via kits, mRNA expressions of p53 and Rac-1 were determined through quantitative polymerase chain reaction (qPCR); The expressions of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and PARP-1 were measured via western blotting. RESULTS Low-dose liquiritigenin exerted no effect on gemcitabine-induced changes of cell viability, apoptosis rate and ROS levels in Panc-1 cells, but for HUVECs, liquiritigenin (3 μM) could remarkably elevate gemcitabine- induced decrease of cell viability, transepithelial electrical resistance (TEER), pro-MMP9 level and expression of ICAM-1 and VCAM-1 (p<0.01). Meanwhile, it could also significantly decrease gemcitabine-induced increase of transcellular and paracellular leak, ROS level, PARP-1 activity, Act-MMP9 level, mRNA expressions of p53 and Rac-1, expression of PARP-1 and apoptosis rate (p<0.01). CONCLUSIONS Low-dose liquiritigenin exerts an antagonistic effect on gemcitabine-induced leak across HUVECs via inhibiting ROS-mediated signalling pathways, but without affecting gemcitabine-induced Panc-1 cell apoptosis. Therefore, low-dose liquiritigenin might be beneficial to prevent the occurrence of gemcitabine-induced CLS in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Wei Wu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China E-mail :
| | | | | | | | | |
Collapse
|
6
|
Peptide-Based Treatment: A Promising Cancer Therapy. J Immunol Res 2015; 2015:761820. [PMID: 26568964 PMCID: PMC4629048 DOI: 10.1155/2015/761820] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/14/2014] [Indexed: 12/16/2022] Open
Abstract
Many new therapies are currently being used to treat cancer. Among these new methods, chemotherapy based on peptides has been of great interest due to the unique advantages of peptides, such as a low molecular weight, the ability to specifically target tumor cells, and low toxicity in normal tissues. In treating cancer, peptide-based chemotherapy can be mainly divided into three types, peptide-alone therapy, peptide vaccines, and peptide-conjugated nanomaterials. Peptide-alone therapy may specifically enhance the immune system's response to kill tumor cells. Peptide-based vaccines have been used in advanced cancers to improve patients' overall survival. Additionally, the combination of peptides with nanomaterials expands the therapeutic ability of peptides to treat cancer by enhancing drug delivery and sensitivity. In this review, we mainly focus on the new advances in the application of peptides in treating cancer in recent years, including diagnosis, treatment, and prognosis.
Collapse
|
7
|
CHEN LU, YUAN WEIJIE, CHEN ZHIKANG, WU SHAOBIN, GE JIE, CHEN JINXIANG, CHEN ZIHUA. Vasoactive intestinal peptide represses activation of tumor-associated macrophages in gastric cancer via regulation of TNFα, IL-6, IL-12 and iNOS. Int J Oncol 2015; 47:1361-70. [DOI: 10.3892/ijo.2015.3126] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 07/20/2015] [Indexed: 11/06/2022] Open
|
8
|
Salles TA, dos Santos L, Barauna VG, Girardi ACC. Potential role of dipeptidyl peptidase IV in the pathophysiology of heart failure. Int J Mol Sci 2015; 16:4226-49. [PMID: 25690036 PMCID: PMC4346954 DOI: 10.3390/ijms16024226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/05/2015] [Accepted: 02/09/2015] [Indexed: 12/14/2022] Open
Abstract
Dipeptidyl peptidase IV (DPPIV) is a widely expressed multifunctional serine peptidase that exists as a membrane-anchored cell surface protein or in a soluble form in the plasma and other body fluids. Numerous substrates are cleaved at the penultimate amino acid by DPPIV, including glucagon-like peptide-1 (GLP-1), brain natriuretic peptide (BNP) and stromal cell-derived factor-1 (SDF-α), all of which play important roles in the cardiovascular system. In this regard, recent reports have documented that circulating DPPIV activity correlates with poorer cardiovascular outcomes in human and experimental heart failure (HF). Moreover, emerging evidence indicates that DPPIV inhibitors exert cardioprotective and renoprotective actions in a variety of experimental models of cardiac dysfunction. On the other hand, conflicting results have been found when translating these promising findings from preclinical animal models to clinical therapy. In this review, we discuss how DPPIV might be involved in the cardio-renal axis in HF. In addition, the potential role for DPPIV inhibitors in ameliorating heart disease is revised, focusing on the effects of the main DPPIV substrates on cardiac remodeling and renal handling of salt and water.
Collapse
Affiliation(s)
- Thiago A Salles
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-000, SP, Brazil.
| | - Leonardo dos Santos
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitoria 29043-900, ES, Brazil.
| | - Valério G Barauna
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitoria 29043-900, ES, Brazil.
| | - Adriana C C Girardi
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-000, SP, Brazil.
| |
Collapse
|
9
|
Vacas E, Muñoz-Moreno L, Fernández-Martínez AB, Bajo AM, Sánchez-Chapado M, Prieto JC, Carmena MJ. Signalling pathways involved in antitumoral effects of VIP in human renal cell carcinoma A498 cells: VIP induction of p53 expression. Int J Biochem Cell Biol 2014; 53:295-301. [PMID: 24905957 DOI: 10.1016/j.biocel.2014.05.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 05/21/2014] [Accepted: 05/26/2014] [Indexed: 01/04/2023]
Abstract
Vasoactive intestinal peptide (VIP) decreases cell proliferation through PI3K signalling and prevents tumour progression in clear renal cell carcinoma (RCC). Here we analyzed the signalling pathways that mediate such VIP effects by using human RCC A498 cells. The effects of treatment with 1 μM VIP and/or specific protein kinase inhibitors such as H89, Wortmannin and PD98059 were studied by cell adhesion assay, ELISA of VEGF165 and ROS production assays. Semiquantitative RT-PCR and western blot were performed to study p53 expression. VIP increased cell adhesion and ROS production, and decreased VEGF165 secretion through PI3K signalling. Moreover, VIP increased nuclear expression of tumour suppressor p53. VIP effects could be blocked by cell incubation with a specific p53 inhibitor, cyclin pifithrin-α hydrobromide (CPFT-αH). In conclusion, this study provides a p53-dependent mechanism by which VIP regulates cell proliferation in RCC development. It supports a potential usefulness of VIP in new therapies of RCC.
Collapse
Affiliation(s)
- Eva Vacas
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | - Laura Muñoz-Moreno
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | - Ana B Fernández-Martínez
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | - Ana M Bajo
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | - Manuel Sánchez-Chapado
- Department of Surgery and Medical and Social Sciences, Unit of Surgery, University of Alcalá, 28871 Alcalá de Henares, Spain; Department of Urology, Príncipe de Asturias Hospital, 28871 Alcalá de Henares, Spain
| | - Juan C Prieto
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain.
| | - María J Carmena
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| |
Collapse
|
10
|
Guo CC, Zhang XL, Yang B, Geng J, Peng B, Zheng JH. Decreased expression of Dkk1 and Dkk3 in human clear cell renal cell carcinoma. Mol Med Rep 2014; 9:2367-73. [PMID: 24676838 DOI: 10.3892/mmr.2014.2077] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 03/06/2014] [Indexed: 11/06/2022] Open
Abstract
The expression patterns of the Dickkopf (Dkk) family of proteins varies in different cancers. In the present study, the expression levels of Dkk1 and Dkk3 were investigated in clear cell renal cell carcinoma (ccRCC) tissues. Dkk1 and Dkk3 serum levels were also examined in patients with ccRCC, and the association between clinicopathological features and Dkk levels was investigated. Serum Dkk1 and Dkk3 were quantified using ELISA in 64 patients with ccRCC and in 30 healthy individuals (controls). The expression levels of Dkk1 and Dkk3 were analyzed in tumor and adjacent normal tissues obtained from patients with ccRCC (n=20) using quantitative polymerase chain reaction (qPCR), western blot analysis and immunohistochemistry. The mean serum levels of Dkk1 and Dkk3 in the patients with ccRCC were significantly lower than those in the healthy controls. Furthermore, the serum Dkk1 levels were significantly lower at higher tumor‑node‑metastasis stages and tumor grades. qPCR, western blot analysis and immunohistochemistry revealed a significant decrease in the Dkk1 and Dkk3 mRNA and protein levels in the tumor tissues compared with the adjacent normal tissues. Consequently, Dkk1 and Dkk3 may present a novel molecular target for the diagnosis and therapeutic treatment of ccRCC.
Collapse
Affiliation(s)
- Chang-Cheng Guo
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Xiao-Long Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Bin Yang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Jiang Geng
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| | - Jun-Hua Zheng
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, P.R. China
| |
Collapse
|
11
|
Chéret J, Lebonvallet N, Buhé V, Carre JL, Misery L, Le Gall-Ianotto C. Influence of sensory neuropeptides on human cutaneous wound healing process. J Dermatol Sci 2014; 74:193-203. [PMID: 24630238 DOI: 10.1016/j.jdermsci.2014.02.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 01/15/2014] [Accepted: 02/05/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Close interactions exist between primary sensory neurons of the peripheral nervous system (PNS) and skin cells. The PNS may be implicated in the modulation of different skin functions as wound healing. OBJECTIVE Study the influence of sensory neurons in human cutaneous wound healing. METHODS We incubated injured human skin explants either with rat primary sensory neurons from dorsal root ganglia (DRG) or different neuropeptides (vasoactive intestinal peptide or VIP, calcitonin gene-related peptide or CGRP, substance P or SP) at various concentrations. Then we evaluated their effects on the proliferative and extracellular matrix (ECM) remodeling phases, dermal fibroblasts adhesion and differentiation into myofibroblasts. RESULTS Thus, DRG and all studied neuromediators increased fibroblasts and keratinocytes proliferation and act on the expression ratio between collagen type I and type III in favor of collagen I, particularly between the 3rd and 7th day of culture. Furthermore, the enzymatic activities of matrix metalloprotesases (MMP-2 and MMP-9) were increased in the first days of wound healing process. Finally, the adhesion of human dermal fibroblasts and their differentiation into myofibroblasts were promoted after incubation with neuromediators. Interestingly, the most potent concentrations for each tested molecules, were the lowest concentrations, corresponding to physiological concentrations. CONCLUSION Sensory neurons and their derived-neuropeptides are able to promote skin wound healing.
Collapse
Affiliation(s)
- J Chéret
- Laboratory of Neurosciences of Brest (EA4685), University of Western Brittany, Brest, France
| | - N Lebonvallet
- Laboratory of Neurosciences of Brest (EA4685), University of Western Brittany, Brest, France
| | - V Buhé
- Laboratory of Neurosciences of Brest (EA4685), University of Western Brittany, Brest, France
| | - J L Carre
- Laboratory of Neurosciences of Brest (EA4685), University of Western Brittany, Brest, France
| | - L Misery
- Laboratory of Neurosciences of Brest (EA4685), University of Western Brittany, Brest, France; Department of Dermatology, University Hospital of Brest, Brest, France.
| | - C Le Gall-Ianotto
- Laboratory of Neurosciences of Brest (EA4685), University of Western Brittany, Brest, France; Department of Dermatology, University Hospital of Brest, Brest, France
| |
Collapse
|
12
|
Pérez-García S, Carrión M, Jimeno R, Ortiz AM, González-Álvaro I, Fernández J, Gomariz RP, Juarranz Y. Urokinase plasminogen activator system in synovial fibroblasts from osteoarthritis patients: modulation by inflammatory mediators and neuropeptides. J Mol Neurosci 2013; 52:18-27. [PMID: 24318839 DOI: 10.1007/s12031-013-0189-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 11/18/2013] [Indexed: 02/08/2023]
Abstract
Plasminogen activators are specific proteolytic enzymes implicated in a variety of basic biological processes. The expression of the urokinase plasminogen activator system components is increased in some human diseases, including osteoarthritis. We sought to study the effect of two components of the inflamed synovial microenvironment on this system, IL-1β and fibronectin fragments, elucidating whether corticotropin-releasing factor (CRF) and vasoactive intestinal peptide (VIP) neuropeptides modulate it, and analyzing the physiological consequences in joint destruction by measuring matrix metalloproteinases-9 and metalloproteinases-13 levels in osteoarthritis fibroblast-like synoviocytes. We showed that IL-1β and fibronectin fragments stimulated urokinase system contributing to the perpetuation of the destructive cascade in joint. VIP modulated, even at constitutive level, this system, also counteracting the effect of both inflammatory stimuli. However, CRF seemed to be ineffective in controlling the production of these proteinases. Moreover, VIP was able to reduce the constitutive expression of matrix metalloproteinase-13 and the levels of both matrix metalloproteinases after stimulation with the pro-inflammatory stimuli. Our results suggest that the presence of early and later inflammatory mediators, such as IL-1β and fibronectin fragments, increases the urokinase system and the matrix metalloproteinases levels. Whereas CRF did not affect this system, VIP counteracts these actions supporting its therapeutic potential for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Selene Pérez-García
- Departamento de Biología Celular, Facultad de Biología, Universidad Complutense de Madrid, José Antonio Novais n°2, Ciudad Universitaria, 28040, Madrid, Spain,
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Vacas E, Arenas MI, Muñoz-Moreno L, Bajo AM, Sánchez-Chapado M, Prieto JC, Carmena MJ. Antitumoral effects of vasoactive intestinal peptide in human renal cell carcinoma xenografts in athymic nude mice. Cancer Lett 2013; 336:196-203. [PMID: 23664888 DOI: 10.1016/j.canlet.2013.04.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 04/16/2013] [Accepted: 04/29/2013] [Indexed: 10/26/2022]
Abstract
We studied antitumor effect of VIP in human renal cell carcinoma (RCC) (A498 cells xenografted in immunosuppressed mice). VIP-treated cells gave resulted in p53 upregulation and decreased nuclear β-catenin translocation and NFκB expression, MMP-2 and MMP-9 activities, VEGF levels and CD-34 expression. VIP led to a more differentiated tubular organization in tumours and less metastatic areas. Thus, VIP inhibits growth of A498-cell tumours acting on the major issues involved in RCC progression such as cell proliferation, microenvironment remodelling, tumour invasion, angiogenesis and metastatic ability. These antitumoral effects of VIP offer new therapeutical possibilities in RCC treatment.
Collapse
Affiliation(s)
- Eva Vacas
- Department of Systems Biology, Unit of Biochemistry and Molecular Biology, University of Alcalá, 28871 Alcalá de Henares, Spain
| | | | | | | | | | | | | |
Collapse
|