1
|
Cao X, Wang T, Mu G, Chen Y, Xiang B, Zhu J, Shen Z. Dysregulated homocysteine metabolism and cardiovascular disease and clinical treatments. Mol Cell Biochem 2025:10.1007/s11010-025-05284-1. [PMID: 40347210 DOI: 10.1007/s11010-025-05284-1] [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: 01/20/2025] [Accepted: 04/06/2025] [Indexed: 05/12/2025]
Abstract
Elevated homocysteine (Hcy) levels, known as hyperhomocysteinemia (HHcy), are recognized as a separate risk factor for cardiovascular disease. Mutations in methylenetetrahydrofolate reductase (MTHFR) and cystathionine beta synthase (CBS)-enzymes pivotal at the juncture of the trans-sulfuration and remethylation pathways-underlie the pathogenesis of HHcy. Although vitamin supplementation has been proven to effectively decrease Hcy levels, there is still uncertainty about whether this reduction translates to a decrease in the incidence rates from cardiovascular diseases (CVDs). This review seeks to explore the linking between Hcy and specific diseases, the role of Hcy in vascular homeostasis, and the research on the possible advantages of therapies designed to lower Hcy levels. Understanding the intricate mechanisms of their metabolism and interactions is essential for pharmacological treatments to mitigate the adverse effects associated with metabolic dysregulation of Hcy. Given the widespread availability and ease of use of Hcy test kits, we strongly advocate for the routine administration of rapid blood tests for individuals at high risk of CVDs, particularly among the elderly population.
Collapse
Affiliation(s)
- Xiangyu Cao
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University and Institute for Cardiovascular Science, Soochow University, Suzhou, 215000, People's Republic of China
| | - Tingyu Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University and Institute for Cardiovascular Science, Soochow University, Suzhou, 215000, People's Republic of China
| | - Gaohang Mu
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University and Institute for Cardiovascular Science, Soochow University, Suzhou, 215000, People's Republic of China
| | - Yupeng Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University and Institute for Cardiovascular Science, Soochow University, Suzhou, 215000, People's Republic of China
| | - Bo Xiang
- Cardiac Surgery Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Affiliated Hospital of University of Electronic Science and Technology, Chengdu, 610072, People's Republic of China
| | - Jingze Zhu
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University and Institute for Cardiovascular Science, Soochow University, Suzhou, 215000, People's Republic of China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University and Institute for Cardiovascular Science, Soochow University, Suzhou, 215000, People's Republic of China.
| |
Collapse
|
2
|
Marchant V, Trionfetti F, Tejedor-Santamaria L, Rayego-Mateos S, Rotili D, Bontempi G, Domenici A, Menè P, Mai A, Martín-Cleary C, Ortiz A, Ramos AM, Strippoli R, Ruiz-Ortega M. BET Protein Inhibitor JQ1 Ameliorates Experimental Peritoneal Damage by Inhibition of Inflammation and Oxidative Stress. Antioxidants (Basel) 2023; 12:2055. [PMID: 38136175 PMCID: PMC10740563 DOI: 10.3390/antiox12122055] [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: 10/21/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Peritoneal dialysis (PD) is a current replacement therapy for end-stage kidney diseases (ESKDs). However, long-term exposure to PD fluids may lead to damage of the peritoneal membrane (PM) through mechanisms involving the activation of the inflammatory response and mesothelial-to-mesenchymal transition (MMT), leading to filtration failure. Peritoneal damage depends on a complex interaction among external stimuli, intrinsic properties of the PM, and subsequent activities of the local innate-adaptive immune system. Epigenetic drugs targeting bromodomain and extra-terminal domain (BET) proteins have shown beneficial effects on different experimental preclinical diseases, mainly by inhibiting proliferative and inflammatory responses. However the effect of BET inhibition on peritoneal damage has not been studied. To this aim, we have evaluated the effects of treatment with the BET inhibitor JQ1 in a mouse model of peritoneal damage induced by chlorhexidine gluconate (CHX). We found that JQ1 ameliorated the CHX-induced PM thickness and inflammatory cell infiltration. Moreover, JQ1 decreased gene overexpression of proinflammatory and profibrotic markers, together with an inhibition of the nuclear factor-κB (NF-κB) pathway. Additionally, JQ1 blocked the activation of nuclear factor erythroid 2-related factor 2 (NRF2) and restored changes in the mRNA expression levels of NADPH oxidases (NOX1 and NOX4) and NRF2/target antioxidant response genes. To corroborate the in vivo findings, we evaluated the effects of the BET inhibitor JQ1 on PD patients' effluent-derived primary mesothelial cells and on the MeT-5A cell line. JQ1 inhibited tumor necrosis factor-α (TNF-α)-induced proinflammatory gene upregulation and restored MMT phenotype changes, together with the downmodulation of oxidative stress. Taken together, these results suggest that BET inhibitors may be a potential therapeutic option to ameliorate peritoneal damage.
Collapse
Affiliation(s)
- Vanessa Marchant
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (V.M.); (L.T.-S.); (S.R.-M.)
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Flavia Trionfetti
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, 00149 Rome, Italy; (F.T.); (G.B.); (R.S.)
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Lucia Tejedor-Santamaria
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (V.M.); (L.T.-S.); (S.R.-M.)
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Sandra Rayego-Mateos
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (V.M.); (L.T.-S.); (S.R.-M.)
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (D.R.); (A.M.)
| | - Giulio Bontempi
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, 00149 Rome, Italy; (F.T.); (G.B.); (R.S.)
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessandro Domenici
- Renal Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy; (A.D.); (P.M.)
| | - Paolo Menè
- Renal Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy; (A.D.); (P.M.)
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (D.R.); (A.M.)
| | - Catalina Martín-Cleary
- Laboratory of Nephrology, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain;
| | - Alberto Ortiz
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
- Laboratory of Nephrology, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain;
| | - Adrian M. Ramos
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
- Laboratory of Nephrology, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain;
| | - Raffaele Strippoli
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, 00149 Rome, Italy; (F.T.); (G.B.); (R.S.)
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Marta Ruiz-Ortega
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (V.M.); (L.T.-S.); (S.R.-M.)
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
| |
Collapse
|
3
|
Hu Y, Liu T, Zheng G, Zhou L, Ma K, Xiong X, Zheng C, Li J, Zhu Y, Bian W, Zheng X, Xiong Q, Lin J. Mechanism exploration of 6-Gingerol in the treatment of atherosclerosis based on network pharmacology, molecular docking and experimental validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 115:154835. [PMID: 37121058 DOI: 10.1016/j.phymed.2023.154835] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND The 6-Gingerol has significant anti-inflammatory, anti-oxidative and hypolipidemic activities and is widely used for treating cardiac-cerebral vascular diseases. However, the multi-target mechanism of 6-Gingerol in the treatment of atherosclerosis remains to be elucidated. METHODS Firstly, the therapeutic actions of 6-Gingerol anti-atherosclerosis were researched based on an atherosclerotic ApoE-deficient mice model induced by high-fat feed. Then, network pharmacology and molecular docking were employed to reveal the anti-atherogenic mechanism of 6-Gingerol. Finally, the target for these predictions was validated by target protein expression assay in vitro and in vivo experiments and further correlation analysis. RESULTS Firstly, 6-Gingerol possessed obvious anti-atherogenic activity, which was manifested by a significant reduction in the plaque area, decrease in the atherosclerosis index and vulnerability index. Secondly, based on network pharmacology, 14 predicted intersection target genes between the targets of 6-Gingerol and atherogenic-related targets were identified. The key core targets of 6-Gingerol anti-atherosclerosis were found to be TP53, RELA, BAX, BCL2, and CASP3. Lipid and atherosclerosis pathways might play a critical role in 6-Gingerol anti-atherosclerosis. Molecular docking results also further revealed that the 6-Gingerol bound well and stable to key core targets from network pharmacological predictions. Then, the experimental results in vivo and in vitro verified that the up-regulation of TP53, RELA, BAX, CASP3, and down-regulation of BCL2 from atherosclerotic ApoE-deficient mice model can be improved by 6-Gingerol intervention. Meanwhile, the correlation analysis further confirmed that 6-Gingerol anti-atherosclerosis was closely related to these targets. CONCLUSION The 6-Gingerol can markedly improve atherosclerosis by modulating key multi-targets TP53, RELA, BAX, CASP3, and BCL2 in lipid and atherosclerosis pathways. These novel findings shed light on the anti-atherosclerosis mechanism of 6-Gingerol from the perspective of multiple targets and pathways.
Collapse
Affiliation(s)
- Youdong Hu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Tingting Liu
- Department of Gynecology, Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, 223002, China
| | - Guangzhen Zheng
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China
| | - Li Zhou
- Department of Intensive Care Unit, Dazhou Central Hospital, Dazhou, 635000, Sichuan, China
| | - Ke Ma
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China
| | - Xiaolian Xiong
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China
| | - Cheng Zheng
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jin Li
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yong Zhu
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China
| | - Wenhui Bian
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China
| | - Xiangde Zheng
- Department of Intensive Care Unit, Dazhou Central Hospital, Dazhou, 635000, Sichuan, China.
| | - Qingping Xiong
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an, Jiangsu, 223003, China.
| | - Jiafeng Lin
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
4
|
Martinelli RP, Rayego-Mateos S, Alique M, Márquez-Expósito L, Tejedor-Santamaria L, Ortiz A, González-Parra E, Ruiz-Ortega M. Vitamin D, Cellular Senescence and Chronic Kidney Diseases: What Is Missing in the Equation? Nutrients 2023; 15:1349. [PMID: 36986078 PMCID: PMC10056834 DOI: 10.3390/nu15061349] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/12/2023] Open
Abstract
As life expectancy increases in many countries, the prevalence of age-related diseases also rises. Among these conditions, chronic kidney disease is predicted to become the second cause of death in some countries before the end of the century. An important problem with kidney diseases is the lack of biomarkers to detect early damage or to predict the progression to renal failure. In addition, current treatments only retard kidney disease progression, and better tools are needed. Preclinical research has shown the involvement of the activation of cellular senescence-related mechanisms in natural aging and kidney injury. Intensive research is searching for novel treatments for kidney diseases as well as for anti-aging therapies. In this sense, many experimental shreds of evidence support that treatment with vitamin D or its analogs can exert pleiotropic protective effects in kidney injury. Moreover, vitamin D deficiency has been described in patients with kidney diseases. Here, we review recent evidence about the relationship between vitamin D and kidney diseases, explaining the underlying mechanisms of the effect of vitamin D actions, with particular attention to the modulation of cellular senescence mechanisms.
Collapse
Affiliation(s)
- Romina P. Martinelli
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- Ricors2040, 28029 Madrid, Spain
| | - Matilde Alique
- Ricors2040, 28029 Madrid, Spain
- Departamento de Biología de Sistemas, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Laura Márquez-Expósito
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- Ricors2040, 28029 Madrid, Spain
| | - Lucia Tejedor-Santamaria
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- Ricors2040, 28029 Madrid, Spain
| | - Alberto Ortiz
- Ricors2040, 28029 Madrid, Spain
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Emilio González-Parra
- Ricors2040, 28029 Madrid, Spain
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- Ricors2040, 28029 Madrid, Spain
| |
Collapse
|
5
|
RELA∙8-Oxoguanine DNA Glycosylase1 Is an Epigenetic Regulatory Complex Coordinating the Hexosamine Biosynthetic Pathway in RSV Infection. Cells 2022; 11:cells11142210. [PMID: 35883652 PMCID: PMC9319012 DOI: 10.3390/cells11142210] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Respiratory syncytial virus (RSV), or human orthopneumovirus, is a negative-sense RNA virus that is the causative agent of severe lower respiratory tract infections in children and is associated with exacerbations of adult lung disease. The mechanisms how severe and/or repetitive virus infections cause declines in pulmonary capacity are not fully understood. We have recently discovered that viral replication triggers epithelial plasticity and metabolic reprogramming involving the hexosamine biosynthetic pathway (HBP). In this study, we examine the relationship between viral induced innate inflammation and the activation of hexosamine biosynthesis in small airway epithelial cells. We observe that RSV induces ~2-fold accumulation of intracellular UDP-GlcNAc, the end-product of the HBP and the obligate substrate of N glycosylation. Using two different silencing approaches, we observe that RSV replication activates the HBP pathway in a manner dependent on the RELA proto-oncogene (65 kDa subunit). To better understand the effect of RSV on the cellular N glycoproteome, and its RELA dependence, we conduct affinity enriched LC-MS profiling in wild-type and RELA-silenced cells. We find that RSV induces the accumulation of 171 N glycosylated peptides in a RELA-dependent manner; these proteins are functionally enriched in integrins and basal lamina formation. To elaborate this mechanism of HBP expression, we demonstrate that RSV infection coordinately induces the HBP pathway enzymes in a manner requiring RELA; these genes include Glutamine-Fructose-6-Phosphate Transaminase 1 (GFPT)-1/2, Glucosamine-Phosphate N-Acetyltransferase (GNPNAT)-1, phosphoglucomutase (PGM)-3 and UDP-N-Acetylglucosamine Pyrophosphorylase (UAP)-1. Using small-molecule inhibitor(s) of 8-oxoguanine DNA glycosylase1 (OGG1), we observe that OGG1 is also required for the expression of HBP pathway. In proximity ligation assays, RSV induces the formation of a nuclear and mitochondrial RELA∙OGG1 complex. In co-immunoprecipitaton (IP) experiments, we discover that RSV induces Ser 536-phosphorylated RELA to complex with OGG1. Chromatin IP experiments demonstrate a major role of OGG1 in supporting the recruitment of RELA and phosphorylated RNA Pol II to the HBP pathway genes. We conclude that the RELA∙OGG1 complex is an epigenetic regulator mediating metabolic reprogramming and N glycoprotein modifications of integrins in response to RSV. These findings have implications for viral-induced adaptive epithelial responses.
Collapse
|
6
|
Cyclosporine A and Tacrolimus Induce Functional Impairment and Inflammatory Reactions in Endothelial Progenitor Cells. Int J Mol Sci 2021; 22:ijms22189696. [PMID: 34575860 PMCID: PMC8472421 DOI: 10.3390/ijms22189696] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/29/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Immunosuppressants are a mandatory therapy for transplant patients to avoid rejection of the transplanted organ by the immune system. However, there are several known side effects, including alterations of the vasculature, which involve a higher occurrence of cardiovascular events. While the effects of the commonly applied immunosuppressive drugs cyclosporine A (CsA) and tacrolimus (Tac) on mature endothelial cells have been addressed in several studies, we focused our research on the unexplored effects of CsA and Tac on endothelial colony-forming cells (ECFCs), a subgroup of endothelial progenitor cells, which play an important role in vascular repair and angiogenesis. We hypothesized that CsA and Tac induce functional defects and activate an inflammatory cascade via NF-κB signaling in ECFCs. ECFCs were incubated with different doses (0.01 µM–10 µM) of CsA or Tac. ECFC function was determined using in vitro models. The expression of inflammatory cytokines and adhesion molecules was explored by quantitative real-time PCR and flow cytometry. NF-κB subunit modification was assessed by immunoblot and immunofluorescence. CsA and Tac significantly impaired ECFC function, including proliferation, migration, and tube formation. TNF-α, IL-6, VCAM, and ICAM mRNA expression, as well as PECAM and VCAM surface expression, were enhanced. Furthermore, CsA and Tac led to NF-κB p65 subunit phosphorylation and nuclear translocation. Pharmacological inhibition of NF-κB by parthenolide diminished CsA- and Tac-mediated proinflammatory effects. The data of functional impairment and activation of inflammatory signals provide new insight into mechanisms associated with CsA and Tac and cardiovascular risk in transplant patients.
Collapse
|
7
|
Russell JJ, Grisanti LA, Brown SM, Bailey CA, Bender SB, Chandrasekar B. Reversion inducing cysteine rich protein with Kazal motifs and cardiovascular diseases: The RECKlessness of adverse remodeling. Cell Signal 2021; 83:109993. [PMID: 33781845 DOI: 10.1016/j.cellsig.2021.109993] [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] [Received: 01/18/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/19/2022]
Abstract
The Reversion Inducing Cysteine Rich Protein With Kazal Motifs (RECK) is a glycosylphosphatidylinositol (GPI) anchored membrane-bound regulator of matrix metalloproteinases (MMPs). It is expressed throughout the body and plays a role in extracellular matrix (ECM) homeostasis and inflammation. In initial studies, RECK expression was found to be downregulated in various invasive cancers and associated with poor prognostic outcome. Restoring RECK, however, has been shown to reverse the metastatic phenotype. Downregulation of RECK expression is also reported in non-malignant diseases, such as periodontal disease, renal fibrosis, and myocardial fibrosis. As such, RECK induction has therapeutic potential in several chronic diseases. Mechanistically, RECK negatively regulates various matrixins involved in cell migration, proliferation, and adverse remodeling by targeting the expression and/or activation of multiple MMPs, A Disintegrin And Metalloproteinase Domain-Containing Proteins (ADAMs), and A Disintegrin And Metalloproteinase With Thrombospondin Motifs (ADAMTS). Outside of its role in remodeling, RECK has also been reported to exert anti-inflammatory effects. In cardiac diseases, for example, it has been shown to counteract several downstream effectors of Angiotensin II (Ang-II) that play a role in adverse cardiac and vascular remodeling, such as Interleukin-6 (IL-6)/IL-6 receptor (IL-6R)/glycoprotein 130 (IL-6 signal transducer) signaling and Epidermal Growth Factor Receptor (EGFR) transactivation. This review article focuses on the current understanding of the multifunctional effects of RECK and how its downregulation may contribute to adverse cardiovascular remodeling.
Collapse
Affiliation(s)
- Jacob J Russell
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Laurel A Grisanti
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America.
| | - Scott M Brown
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Chastidy A Bailey
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Shawn B Bender
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America; Dalton Cardiovascular Center, University of Missouri, Columbia, MO, United States of America.
| | - B Chandrasekar
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America; Medicine, University of Missouri School of Medicine, Columbia, MO, United States of America; Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States of America; Dalton Cardiovascular Center, University of Missouri, Columbia, MO, United States of America.
| |
Collapse
|
8
|
Li R, Zhou Y, Liu W, Li Y, Qin Y, Yu L, Chen Y, Xu Y. Rare earth element lanthanum protects against atherosclerosis induced by high-fat diet via down-regulating MAPK and NF-κB pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 207:111195. [PMID: 32891972 DOI: 10.1016/j.ecoenv.2020.111195] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/03/2020] [Accepted: 08/17/2020] [Indexed: 06/11/2023]
Abstract
Rare earth elements, which are extensively used in environmental protection, medicine, food, aerospace and other fields, have attracted widespread attention in recent years. However, the effect on atherosclerosis and its biological mechanism remains unclear. To elucidate these problems, here we performed a study that Apolipoprotein E-deficient mice were fed with high-fat diet to promote the development of atherosclerosis, meanwhile, mice were received 0.1, 0.2, 1.0, 2.0 mg/kg lanthanum nitrate (La(NO3)3) for 12 weeks. The results showed that La(NO3)3 prominently inhibited aorta morphological alternations by histopathological examination. Meanwhile, La(NO3)3 regulated serum lipids, including reducing total cholesterol and increasing high-density lipoprotein. Moreover, the oxidative stress was alleviated by La(NO3)3 intervention through enhancing superoxide dismutase and glutathione, and decreasing malondialdehyde levels. In addition, enzyme-linked immunosorbent assay analysis showed La(NO3)3 could ameliorate the dysfunction of vascular endothelium with declined endothelin-1 and increased prostacyclin. Furthermore, Western blot analysis indicated that La(NO3)3 significantly down-regulated inflammation-mediated proteins including phosphorylated p38 mitogen-activated protein kinases (p-p38 MAPK), monocyte chemo-attractant protein, intercellular adhesion molecule-1, nuclear factor-kappa B p65 (NF-κB p65), tumor necrosis factor-α, interleukin-6 and interleukin-1β, whereas up-regulated the inhibitor of NF-κB protein. In conclusion, La(NO3)3 ameliorates atherosclerosis by regulating lipid metabolism, oxidative stress, endothelial dysfunction and inflammatory response in mice. The potential mechanism associates with the inhibition of MAPK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Ruijun Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100083, China.
| | - Yalin Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100083, China.
| | - Wei Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100083, China.
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100083, China.
| | - Yong Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100083, China.
| | - Lanlan Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100083, China.
| | - Yuhan Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100083, China.
| | - Yajun Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, 100083, China; Toxicological Research and Risk Assessment for Food Safety, Beijing, 100083, China.
| |
Collapse
|
9
|
Gene Regulatory Network Analysis of Perivascular Adipose Tissue of Abdominal Aortic Aneurysm Identifies Master Regulators of Key Pathogenetic Pathways. Biomedicines 2020; 8:biomedicines8080288. [PMID: 32823940 PMCID: PMC7459520 DOI: 10.3390/biomedicines8080288] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/30/2020] [Accepted: 08/12/2020] [Indexed: 11/19/2022] Open
Abstract
The lack of medical therapy to treat abdominal aortic aneurysm (AAA) stems from our inadequate understanding of the mechanisms underlying AAA pathogenesis. To date, the only available treatment option relies on surgical intervention, which aims to prevent AAA rupture. Identifying specific regulators of pivotal pathogenetic mechanisms would allow the development of novel treatments. With this work, we sought to identify regulatory factors associated with co-expressed genes characterizing the diseased perivascular adipose tissue (PVAT) of AAA patients, which is crucially involved in AAA pathogenesis. We applied a reverse engineering approach to identify cis-regulatory elements of diseased PVAT genes, the associated transcription factors, and upstream regulators. Finally, by analyzing the topological properties of the reconstructed regulatory disease network, we prioritized putative targets for AAA interference treatment options. Overall, we identified NFKB1, SPIB, and TBP as the most relevant transcription factors, as well as MAPK1 and GSKB3 protein kinases and RXRA nuclear receptor as key upstream regulators. We showed that these factors could regulate different co-expressed gene subsets in AAA PVAT, specifically associated with both innate and antigen-driven immune response pathways. Inhibition of these factors may represent a novel option for the development of efficient immunomodulatory strategies to treat AAA.
Collapse
|
10
|
Rayego-Mateos S, Morgado-Pascual JL, Valdivielso JM, Sanz AB, Bosch-Panadero E, Rodrigues-Díez RR, Egido J, Ortiz A, González-Parra E, Ruiz-Ortega M. TRAF3 Modulation: Novel Mechanism for the Anti-inflammatory Effects of the Vitamin D Receptor Agonist Paricalcitol in Renal Disease. J Am Soc Nephrol 2020; 31:2026-2042. [PMID: 32631974 DOI: 10.1681/asn.2019111206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND CKD leads to vitamin D deficiency. Treatment with vitamin D receptor agonists (VDRAs) may have nephroprotective and anti-inflammatory actions, but their mechanisms of action are poorly understood. METHODS Modulation of the noncanonical NF-κB2 pathway and its component TNF receptor-associated factor 3 (TRAF3) by the VDRA paricalcitol was studied in PBMCs from patients with ESKD, cytokine-stimulated cells, and preclinical kidney injury models. RESULTS In PBMCs isolated from patients with ESKD, TRAF3 protein levels were lower than in healthy controls. This finding was associated with evidence of noncanonical NF-κB2 activation and a proinflammatory state. However, PBMCs from patients with ESKD treated with paricalcitol did not exhibit these features. Experiments in cultured cells confirmed the link between TRAF3 and NF-κB2/inflammation. Decreased TRAF3 ubiquitination in K48-linked chains and cIAP1-TRAF3 interaction mediated the mechanisms of paricalcitol action.TRAF3 overexpression by CRISPR/Cas9 technology mimicked VDRA's effects. In a preclinical model of kidney injury, paricalcitol inhibited renal NF-κB2 activation and decreased renal inflammation. In VDR knockout mice with renal injury, paricalcitol prevented TRAF3 downregulation and NF-κB2-dependent gene upregulation, suggesting a VDR-independent anti-inflammatory effect of paricalcitol. CONCLUSIONS These data suggest the anti-inflammatory actions of paricalcitol depend on TRAF3 modulation and subsequent inhibition of the noncanonical NF-κB2 pathway, identifying a novel mechanism for VDRA's effects. Circulating TRAF3 levels could be a biomarker of renal damage associated with the inflammatory state.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain.,Vascular and Renal Translational Research Group. Institut de Receca Biomedica de Lleida (IRBLleida), Lleida, Spain
| | - Jose Luis Morgado-Pascual
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain.,REDinREN (Red de Investigación Renal), Madrid, Spain
| | - José Manuel Valdivielso
- Vascular and Renal Translational Research Group. Institut de Receca Biomedica de Lleida (IRBLleida), Lleida, Spain.,REDinREN (Red de Investigación Renal), Madrid, Spain
| | - Ana Belén Sanz
- REDinREN (Red de Investigación Renal), Madrid, Spain.,Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Enrique Bosch-Panadero
- Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Raúl R Rodrigues-Díez
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz.Universidad Autónoma. 28040 Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM). 28029 Madrid, Spain
| | - Alberto Ortiz
- REDinREN (Red de Investigación Renal), Madrid, Spain.,Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Emilio González-Parra
- Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain .,REDinREN (Red de Investigación Renal), Madrid, Spain
| |
Collapse
|
11
|
Pons S, Fodil S, Azoulay E, Zafrani L. The vascular endothelium: the cornerstone of organ dysfunction in severe SARS-CoV-2 infection. Crit Care 2020; 24:353. [PMID: 32546188 PMCID: PMC7296907 DOI: 10.1186/s13054-020-03062-7] [Citation(s) in RCA: 343] [Impact Index Per Article: 68.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/04/2020] [Indexed: 12/18/2022] Open
Abstract
In severe SARS-CoV-2 infections, emerging data including recent histopathological studies have emphasized the crucial role of endothelial cells (ECs) in vascular dysfunction, immunothrombosis, and inflammation.Histopathological studies have evidenced direct viral infection of ECs, endotheliitis with diffuse endothelial inflammation, and micro- and macrovascular thrombosis both in the venous and arterial circulations. Venous thrombotic events, particularly pulmonary embolism, with elevated D-dimer and coagulation activation are highly prevalent in COVID-19 patients. The pro-inflammatory cytokine storm, with elevated levels of interleukin-6 (IL-6), IL-2 receptor, and tumor necrosis factor-α, could also participate in endothelial dysfunction and leukocyte recruitment in the microvasculature. COVID-19-induced endotheliitis may explain the systemic impaired microcirculatory function in different organs in COVID-19 patients. Ongoing trials directly and indirectly target COVID-19-related endothelial dysfunctions: i.e., a virus-cell entry using recombinant angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS-2) blockade, coagulation activation, and immunomodulatory therapies, such as anti-IL-6 strategies. Studies focusing on endothelial dysfunction in COVID-19 patients are warranted as to decipher their precise role in severe SARS-CoV-2 infection and organ dysfunction and to identify targets for further interventions.
Collapse
Affiliation(s)
- Stéphanie Pons
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Saint-Louis Teaching Hospital, Paris University, Paris, France
- Anesthesia and Critical Care Department, Avicenne Teaching Hospital, Assistance Publique des Hôpitaux de Paris, Bobigny, France
| | - Sofiane Fodil
- Department of Medical Intensive Care Unit, Saint-Louis Teaching Hospital, Assistance Publique des Hôpitaux de Paris, 1, Avenue Claude Vellefaux, 75010, Paris, France
| | - Elie Azoulay
- Department of Medical Intensive Care Unit, Saint-Louis Teaching Hospital, Assistance Publique des Hôpitaux de Paris, 1, Avenue Claude Vellefaux, 75010, Paris, France
| | - Lara Zafrani
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapy, Saint-Louis Teaching Hospital, Paris University, Paris, France.
- Department of Medical Intensive Care Unit, Saint-Louis Teaching Hospital, Assistance Publique des Hôpitaux de Paris, 1, Avenue Claude Vellefaux, 75010, Paris, France.
| |
Collapse
|
12
|
Li Z, Kong W. Cellular signaling in Abdominal Aortic Aneurysm. Cell Signal 2020; 70:109575. [PMID: 32088371 DOI: 10.1016/j.cellsig.2020.109575] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/31/2022]
Abstract
Abdominal aortic aneurysms (AAAs) are highly lethal cardiovascular diseases without effective medications. However, the molecular and signaling mechanisms remain unclear. A series of pathological cellular processes have been shown to contribute to AAA formation, including vascular extracellular matrix remodeling, inflammatory and immune responses, oxidative stress, and dysfunction of vascular smooth muscle cells. Each cellular process involves complex cellular signaling, such as NF-κB, MAPK, TGFβ, Notch and inflammasome signaling. In this review, we discuss how cellular signaling networks function in various cellular processes during the pathogenesis and progression of AAA. Understanding the interaction of cellular signaling networks with AAA pathogenesis as well as the crosstalk of different signaling pathways is essential for the development of novel therapeutic approaches to and personalized treatments of AAA diseases.
Collapse
Affiliation(s)
- Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| |
Collapse
|
13
|
Zhang S, Xu W, Wang H, Cao M, Li M, Zhao J, Hu Y, Wang Y, Li S, Xie Y, Chen G, Liu R, Cheng Y, Xu Z, Zou K, Gong S, Geng L. Inhibition of CREB-mediated ZO-1 and activation of NF-κB-induced IL-6 by colonic epithelial MCT4 destroys intestinal barrier function. Cell Prolif 2019; 52:e12673. [PMID: 31418947 PMCID: PMC6869122 DOI: 10.1111/cpr.12673] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 06/13/2019] [Accepted: 07/07/2019] [Indexed: 02/06/2023] Open
Abstract
Objective Inflammatory bowel disease (IBD) is a disorder intestinal inflammation and impaired barrier function, associated with increased epithelial expression of monocarboxylate transporter 4 (MCT4). However, the specific non‐metabolic function and clinical relevance of MCT4 in IBD remain to be fully elucidated. Methods Lentivirus‐mediated overexpression of MCT4 was used to assess the role of MCT4 in transcriptionally regulating ZO‐1 and IL‐6 expression by luciferase assays, WB and ChIP. IP was used to analyse the effect of MCT4 on the interaction NF‐κB‐CBP or CREB‐CBP, and these MCT4‐mediated effects were confirmed in vivo assay. Results We showed that ectopic expression of MCT4 inhibited ZO‐1 expression, while increased pro‐inflammatory factors expression, leading to destroy intestinal epithelial barrier function in vitro and in vivo. Mechanistically, MCT4 contributed NF‐κB p65 nuclear translocation and increased the binding of NF‐κB p65 to the promoter of IL‐6, which is attributed to MCT4 enhanced NF‐κB‐CBP interaction and dissolved CREB‐CBP complex, resulting in reduction of CREB activity and CREB‐mediated ZO‐1 expression. In addition, treatment of experimental colitis with MCT4 inhibitor α‐cyano‐4‐hydroxycinnamate (CHC) ameliorated mucosal intestinal barrier function, which was due to attenuation of pro‐inflammation factors expression and enhancement of ZO‐1 expression. Conclusion These findings suggested a novel role of MCT4 in controlling development of IBD and provided evidence for potential targets of IBD.
Collapse
Affiliation(s)
- Shunxian Zhang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wanfu Xu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hongli Wang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Meiwan Cao
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Musheng Li
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Junhong Zhao
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yan Hu
- Department of Anesthesiology, Hainan General Hospital, Haikou, China
| | - Yaodong Wang
- Department of Gastroenterology, Kunshan affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, China
| | - Songyu Li
- Department of Clinical Laboratory, Qionghai Hospital of Traditional Chinese Medicine, Qionghai, China
| | - Yuanwen Xie
- Department of Anorectal, Qionghai Hospital of Traditional Chinese Medicine, Qionghai, China
| | - Guanhua Chen
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ruitao Liu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yang Cheng
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhaohui Xu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Kejian Zou
- Department of General Surgery, Hainan General Hospital, Haikou, China
| | - Sitang Gong
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Lanlan Geng
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Zhang M, Xu Y, Qiu Z, Jiang L. Sulforaphane Attenuates Angiotensin II-Induced Vascular Smooth Muscle Cell Migration via Suppression of NOX4/ROS/Nrf2 Signaling. Int J Biol Sci 2019; 15:148-157. [PMID: 30662355 PMCID: PMC6329926 DOI: 10.7150/ijbs.28874] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022] Open
Abstract
Angiotensin II (Ang II) is involved in the pathogenic progress of cardiovascular diseases via the promotion of abnormal proliferation and migration of human vascular smooth muscle cells (HVSMCs). Sulforaphane (SFN) exerts potent anti-inflammatory effects both in vitro and in vivo. In the present study, we aimed to investigate the effects of SFN on Ang II-induced abnormal migration of HVSMCs as well as the underlying mechanisms of those effects. The results showed that Ang II-induced HVSMC proliferation and migration were inhibited by treatment with SFN. SFN also exhibited anti-inflammatory activity, as indicated by its reduction of monocyte adhesion to HVSMCs via the reduction of ICAM1 and VCAM1 levels. Moreover, SFN reduced the Ang II-induced upregulation of HVSMC migration; this effect was inhibited by pretreatment with inhibitors of NADPH oxidase and ROS or transfection with siNOX4. In addition, SFN reversed the Ang II-induced upregulation of HVSMC migration via elevation of Nrf2 activation and expression. Taken together, the results indicate that SFN reverses Ang II-induced HVSMC migration through suppression of the NOX4/ROS/Nrf2 pathway. Thus, SFN is a potential agent to reverse the pathological changes involved in various cardiovascular diseases.
Collapse
Affiliation(s)
- Min Zhang
- Division of Cardiology, TongRen Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yingjie Xu
- Division of Cardiology, TongRen Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Zhaohui Qiu
- Division of Cardiology, TongRen Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Li Jiang
- Division of Cardiology, TongRen Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| |
Collapse
|
15
|
Nsaibia MJ, Boulanger MC, Bouchareb R, Mkannez G, Le Quang K, Hadji F, Argaud D, Dahou A, Bossé Y, Koschinsky ML, Pibarot P, Arsenault BJ, Marette A, Mathieu P. OxLDL-derived lysophosphatidic acid promotes the progression of aortic valve stenosis through a LPAR1-RhoA-NF-κB pathway. Cardiovasc Res 2018; 113:1351-1363. [PMID: 28472283 DOI: 10.1093/cvr/cvx089] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 05/03/2017] [Indexed: 01/15/2023] Open
Abstract
Aims Oxidatively modified lipoproteins may promote the development/progression of calcific aortic valve stenosis (CAVS). Oxidative transformation of low-density lipoprotein (OxLDL) generates lysophosphatidic acid (LPA), a lipid mediator that accumulates in mineralized aortic valves. LPA activates at least six different G protein-coupled receptors, which may play a role in the pathophysiology of CAVS. We hypothesized that LPA derived from OxLDL may promote a NF-κB signature that drives osteogenesis in the aortic valve. Methods and results The role of OxLDL-LPA was examined in isolated valve interstitial cells (VICs) and the molecular pathway was validated in human explanted aortic valves and in a mouse model of CAVS. We found that OxLDL-LPA promoted the mineralization and osteogenic transition of VICs through LPAR1 and the activation of a RhoA-NF-κB pathway. Specifically, we identified that RhoA/ROCK activated IκB kinase alpha, which promoted the phosphorylation of p65 on serine 536 (p65 pS536). p65 pS536 was recruited to the BMP2 promoter and directed an osteogenic program not responsive to the control exerted by the inhibitor of kappa B. In LDLR-/-/ApoB100/100/IGFII transgenic mice (IGFII), which develop CAVS under a high-fat and high-sucrose diet the administration of Ki16425, a Lpar1 blocker, reduced by three-fold the progression rate of CAVS and also decreased the osteogenic activity as measured with a near-infrared fluorescent probe that recognizes hydroxyapatite of calcium. Conclusions OxLDL-LPA promotes an osteogenic program in the aortic valve through a LPAR1-RhoA/ROCK-p65 pS536 pathway. LPAR1 may represent a suitable target to prevent the progression of CAVS.
Collapse
Affiliation(s)
- Mohamed Jalloul Nsaibia
- Laboratory of Cardiovascular Pathobiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, Quebec, Quebec G1V-4G5, Canada
| | - Marie-Chloé Boulanger
- Laboratory of Cardiovascular Pathobiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, Quebec, Quebec G1V-4G5, Canada
| | - Rihab Bouchareb
- Laboratory of Cardiovascular Pathobiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, Quebec, Quebec G1V-4G5, Canada
| | - Ghada Mkannez
- Laboratory of Cardiovascular Pathobiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, Quebec, Quebec G1V-4G5, Canada
| | - Khai Le Quang
- Laboratory of Cardiovascular Pathobiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, Quebec, Quebec G1V-4G5, Canada
| | - Fayez Hadji
- Laboratory of Cardiovascular Pathobiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, Quebec, Quebec G1V-4G5, Canada
| | - Deborah Argaud
- Laboratory of Cardiovascular Pathobiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, Quebec, Quebec G1V-4G5, Canada
| | - Abdellaziz Dahou
- Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Laval University, Quebec, Canada
| | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Laval University, Quebec, Canada
| | | | - Philippe Pibarot
- Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Laval University, Quebec, Canada
| | - Benoit J Arsenault
- Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Laval University, Quebec, Canada
| | - André Marette
- Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Laval University, Quebec, Canada
| | - Patrick Mathieu
- Laboratory of Cardiovascular Pathobiology, Institut Universitaire de Cardiologie et de Pneumologie de Québec/Quebec Heart and Lung Institute, Research Center, Department of Surgery, Laval University, 2725 Chemin Ste-Foy, Quebec, Quebec G1V-4G5, Canada
| |
Collapse
|
16
|
Hirai K, Furusho H, Hirota K, Sasaki H. Activation of hypoxia-inducible factor 1 attenuates periapical inflammation and bone loss. Int J Oral Sci 2018; 10:12. [PMID: 29654284 PMCID: PMC5966812 DOI: 10.1038/s41368-018-0015-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/28/2017] [Accepted: 01/22/2018] [Indexed: 01/20/2023] Open
Abstract
Hypoxia (low oxygen level) is an important feature during infections and affects the host defence mechanisms. The host has evolved specific responses to address hypoxia, which are strongly dependent on the activation of hypoxia-inducible factor 1 (HIF-1). Hypoxia interferes degradation of HIF-1 alpha subunit (HIF-1α), leading to stabilisation of HIF-1α, heterodimerization with HIF-1 beta subunit (HIF-1β) and subsequent activation of HIF-1 pathway. Apical periodontitis (periapical lesion) is a consequence of endodontic infection and ultimately results in destruction of tooth-supporting tissue, including alveolar bone. Thus far, the role of HIF-1 in periapical lesions has not been systematically examined. In the present study, we determined the role of HIF-1 in a well-characterised mouse periapical lesion model using two HIF-1α-activating strategies, dimethyloxalylglycine (DMOG) and adenovirus-induced constitutively active HIF-1α (CA-HIF1A). Both DMOG and CA-HIF1A attenuated periapical inflammation and tissue destruction. The attenuation in vivo was associated with downregulation of nuclear factor-κappa B (NF-κB) and osteoclastic gene expressions. These two agents also suppressed NF-κB activation and subsequent production of proinflammatory cytokines by macrophages. Furthermore, activation of HIF-1α by DMOG specifically suppressed lipopolysaccharide-stimulated macrophage differentiation into M1 cells, increasing the ratio of M2 macrophages against M1 cells. Taken together, our data indicated that activation of HIF-1 plays a protective role in the development of apical periodontitis via downregulation of NF-κB, proinflammatory cytokines, M1 macrophages and osteoclastogenesis.
Collapse
Affiliation(s)
- Kimito Hirai
- Department of Cariology, Restorative Sciences & Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA
| | - Hisako Furusho
- Department of Oral and Maxillofacial Pathobiology, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| | - Hajime Sasaki
- Department of Cariology, Restorative Sciences & Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA.
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, MA, USA.
| |
Collapse
|
17
|
Ijaz T, Sun H, Pinchuk IV, Milewicz DM, Tilton RG, Brasier AR. Deletion of NF-κB/RelA in Angiotensin II-Sensitive Mesenchymal Cells Blocks Aortic Vascular Inflammation and Abdominal Aortic Aneurysm Formation. Arterioscler Thromb Vasc Biol 2017; 37:1881-1890. [PMID: 28818856 DOI: 10.1161/atvbaha.117.309863] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 08/07/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Infusion of angiotensin II (Ang II) induces extracellular matrix remodeling and inflammation resulting in abdominal aortic aneurysms (AAAs) in normolipidemic mice. Although Ang II activates mesenchymal cells in the media and adventitia to become fibrogenic, the sentinel role of this mesenchymal population in modulating the inflammatory response and aneurysms is not known. We test the hypothesis that these fibrogenic mesenchymal cells play a critical role in Ang II-induced aortic wall vascular inflammation and AAA formation. APPROACH AND RESULTS Ang II infusion increased phospho-Ser536-RelA and interleukin (IL)-6 immunostaining in the abdominal aorta. In addition, aortic mRNA transcripts of RelA-dependent cytokines IL-6 and IL-1β were significantly elevated suggesting that Ang II functionally activates RelA signaling. To test the role of mesenchymal RelA in AAA formation, we generated RelA-CKO mice by administering tamoxifen to double transgenic mice harboring RelA-flox alleles and tamoxifen-inducible Col1a2 promoter-driven Cre recombinase (Col1a2-CreERT). Tamoxifen administration to Col1a2-CreERT•mT/mG mice induced Cre expression and RelA depletion in aortic smooth muscle cells and fibroblasts but not in endothelial cells. Infusion of Ang II significantly increased abdominal aortic diameter and the incidence of AAA in RelA wild-type but not in RelA-CKO mice, independent of changes in systolic blood pressure. Furthermore, mesenchymal cell-specific RelA-CKO mice exhibited decreased expression of IL-6 and IL-1β cytokines and decreased recruitment of C68+ and F4/80lo•Ly6Chi monocytes during Ang II infusion. CONCLUSIONS Fibrogenic mesenchymal RelA plays a causal role in Ang II-induced vascular inflammation and AAA in normolipidemic mice.
Collapse
Affiliation(s)
- Talha Ijaz
- From the Department of Biochemistry and Molecular Biology (T.I.), MD-PhD Program (T.I.), Division of Gasteroenterology, Department of Internal Medicine (I.V.P.), Division of Endocrinology, Department of Internal Medicine (H.S., R.G.T., A.R.B.), Institute for Translational Sciences (R.G.T., A.R.B.), Sealy Center for Molecular Medicine (R.G.T., A.R.B.), University of Texas Medical Branch, Galveston; and Department of Internal Medicine, University of Texas Health Science Center at Houston (D.M.M.)
| | - Hong Sun
- From the Department of Biochemistry and Molecular Biology (T.I.), MD-PhD Program (T.I.), Division of Gasteroenterology, Department of Internal Medicine (I.V.P.), Division of Endocrinology, Department of Internal Medicine (H.S., R.G.T., A.R.B.), Institute for Translational Sciences (R.G.T., A.R.B.), Sealy Center for Molecular Medicine (R.G.T., A.R.B.), University of Texas Medical Branch, Galveston; and Department of Internal Medicine, University of Texas Health Science Center at Houston (D.M.M.)
| | - Irina V Pinchuk
- From the Department of Biochemistry and Molecular Biology (T.I.), MD-PhD Program (T.I.), Division of Gasteroenterology, Department of Internal Medicine (I.V.P.), Division of Endocrinology, Department of Internal Medicine (H.S., R.G.T., A.R.B.), Institute for Translational Sciences (R.G.T., A.R.B.), Sealy Center for Molecular Medicine (R.G.T., A.R.B.), University of Texas Medical Branch, Galveston; and Department of Internal Medicine, University of Texas Health Science Center at Houston (D.M.M.)
| | - Dianna M Milewicz
- From the Department of Biochemistry and Molecular Biology (T.I.), MD-PhD Program (T.I.), Division of Gasteroenterology, Department of Internal Medicine (I.V.P.), Division of Endocrinology, Department of Internal Medicine (H.S., R.G.T., A.R.B.), Institute for Translational Sciences (R.G.T., A.R.B.), Sealy Center for Molecular Medicine (R.G.T., A.R.B.), University of Texas Medical Branch, Galveston; and Department of Internal Medicine, University of Texas Health Science Center at Houston (D.M.M.)
| | - Ronald G Tilton
- From the Department of Biochemistry and Molecular Biology (T.I.), MD-PhD Program (T.I.), Division of Gasteroenterology, Department of Internal Medicine (I.V.P.), Division of Endocrinology, Department of Internal Medicine (H.S., R.G.T., A.R.B.), Institute for Translational Sciences (R.G.T., A.R.B.), Sealy Center for Molecular Medicine (R.G.T., A.R.B.), University of Texas Medical Branch, Galveston; and Department of Internal Medicine, University of Texas Health Science Center at Houston (D.M.M.)
| | - Allan R Brasier
- From the Department of Biochemistry and Molecular Biology (T.I.), MD-PhD Program (T.I.), Division of Gasteroenterology, Department of Internal Medicine (I.V.P.), Division of Endocrinology, Department of Internal Medicine (H.S., R.G.T., A.R.B.), Institute for Translational Sciences (R.G.T., A.R.B.), Sealy Center for Molecular Medicine (R.G.T., A.R.B.), University of Texas Medical Branch, Galveston; and Department of Internal Medicine, University of Texas Health Science Center at Houston (D.M.M.).
| |
Collapse
|
18
|
AT1 receptor signaling pathways in the cardiovascular system. Pharmacol Res 2017; 125:4-13. [PMID: 28527699 DOI: 10.1016/j.phrs.2017.05.008] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 01/14/2023]
Abstract
The importance of the renin angiotensin aldosterone system in cardiovascular physiology and pathophysiology has been well described whereas the detailed molecular mechanisms remain elusive. The angiotensin II type 1 receptor (AT1 receptor) is one of the key players in the renin angiotensin aldosterone system. The AT1 receptor promotes various intracellular signaling pathways resulting in hypertension, endothelial dysfunction, vascular remodeling and end organ damage. Accumulating evidence shows the complex picture of AT1 receptor-mediated signaling; AT1 receptor-mediated heterotrimeric G protein-dependent signaling, transactivation of growth factor receptors, NADPH oxidase and ROS signaling, G protein-independent signaling, including the β-arrestin signals and interaction with several AT1 receptor interacting proteins. In addition, there is functional cross-talk between the AT1 receptor signaling pathway and other signaling pathways. In this review, we will summarize an up to date overview of essential AT1 receptor signaling events and their functional significances in the cardiovascular system.
Collapse
|
19
|
Chen J, Peters A, Papke CL, Villamizar C, Ringuette LJ, Cao J, Wang S, Ma S, Gong L, Byanova KL, Xiong J, Zhu MX, Madonna R, Kee P, Geng YJ, Brasier AR, Davis EC, Prakash S, Kwartler CS, Milewicz DM. Loss of Smooth Muscle α-Actin Leads to NF-κB-Dependent Increased Sensitivity to Angiotensin II in Smooth Muscle Cells and Aortic Enlargement. Circ Res 2017; 120:1903-1915. [PMID: 28461455 DOI: 10.1161/circresaha.117.310563] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 04/24/2017] [Accepted: 05/01/2017] [Indexed: 11/16/2022]
Abstract
RATIONALE Mutations in ACTA2, encoding the smooth muscle isoform of α-actin, cause thoracic aortic aneurysms, acute aortic dissections, and occlusive vascular diseases. OBJECTIVE We sought to identify the mechanism by which loss of smooth muscle α-actin causes aortic disease. METHODS AND RESULTS Acta2-/- mice have an increased number of elastic lamellae in the ascending aorta and progressive aortic root dilation as assessed by echocardiography that can be attenuated by treatment with losartan, an angiotensin II (AngII) type 1 receptor blocker. AngII levels are not increased in Acta2-/- aortas or kidneys. Aortic tissue and explanted smooth muscle cells from Acta2-/- aortas show increased production of reactive oxygen species and increased basal nuclear factor κB signaling, leading to an increase in the expression of the AngII receptor type I a and activation of signaling at 100-fold lower levels of AngII in the mutant compared with wild-type cells. Furthermore, disruption of smooth muscle α-actin filaments in wild-type smooth muscle cells by various mechanisms activates nuclear factor κB signaling and increases expression of AngII receptor type I a. CONCLUSIONS These findings reveal that disruption of smooth muscle α-actin filaments in smooth muscle cells increases reactive oxygen species levels, activates nuclear factor κB signaling, and increases AngII receptor type I a expression, thus potentiating AngII signaling in vascular smooth muscle cells without an increase in the exogenous levels of AngII.
Collapse
Affiliation(s)
- Jiyuan Chen
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Andrew Peters
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Christina L Papke
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Carlos Villamizar
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Lea-Jeanne Ringuette
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Jiumei Cao
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Shanzhi Wang
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Shuangtao Ma
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Limin Gong
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Katerina L Byanova
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Jian Xiong
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Michael X Zhu
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Rosalinda Madonna
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Patrick Kee
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Yong-Jian Geng
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Allan R Brasier
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Elaine C Davis
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Siddharth Prakash
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Callie S Kwartler
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.)
| | - Dianna M Milewicz
- From the Departments of Internal Medicine (J.C., A.P., C.L.P., C.V., J.C., S.W., S.M., L.G., K.L.B., R.M., P.K., Y.-J.G., S.P., C.S.K., D.M.M.) and Integrative Biology and Pharmacology (J.X., M.X.Z.), The University of Texas Health Science Center at Houston; Anatomy and Cell Biology, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Quebec, Canada; and Internal Medicine, Institute for Translational Sciences, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston (A.R.B.).
| |
Collapse
|
20
|
Xu JY, Chang NB, Li T, Jiang R, Sun XL, He YZ, Jiang J. Endothelial Cells Inhibit the Angiotensin II Induced Phenotypic Modulation of Rat Vascular Adventitial Fibroblasts. J Cell Biochem 2017; 118:1921-1927. [PMID: 28218456 DOI: 10.1002/jcb.25941] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 02/17/2017] [Indexed: 11/12/2022]
Abstract
The phenotypic modulation of vascular adventitial fibroblasts plays an important role in vascular remodeling. Evidence have shown that endothelial cells and adventitial fibroblasts interact under certain conditions. In this study, we investigated the influence of endothelial cells on the phenotypic modulation of adventitial fibroblasts. Endothelial cells and adventitial fibroblasts from rat thoracic aorta were cultivated in a co-culture system and adventitial fibroblasts were induced with angiotensin II (Ang II). Collagen I and alpha smooth muscle actin (α-SMA) expression and migration of adventitial fibroblasts were analyzed. Ang II upregulated the expression of collagen I and α-SMA and the migration of adventitial fibroblasts. Adventitial fibroblasts-endothelial cells co-culturing attenuated the effects of Ang II. Homocysteine-treated endothelial cells, which are functionally impaired, were less inhibitory of the phenotypic modulation of adventitial fibroblasts. Supplementation of endothelial cells with L-arginine (L-Arg) or 8-bromoguanosine 3',5'-cyclic monophosphate (8-BrcGMP) enhanced the trends, while with L-NG-nitroarginine methyl ester (L-NAME) or 1H-[1,2,4]Oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) the opposite effect was observed. Under the influence of Ang II, adventitial fibroblasts were prone to undergo phenotypic modulation, which was closely related to vascular remodeling. Our study showed that endothelial cells influenced fibroblast phenotypic transformation and such effect would be mediated through the nitric oxide (NO)/cGMP signaling pathway. J. Cell. Biochem. 118: 1921-1927, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jia-Ying Xu
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Neng-Bin Chang
- Department of Anatomy, College of Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease/Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Rui Jiang
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiao-Lei Sun
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yan-Zheng He
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jun Jiang
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
21
|
Huante-Mendoza A, Silva-García O, Oviedo-Boyso J, Hancock REW, Baizabal-Aguirre VM. Peptide IDR-1002 Inhibits NF-κB Nuclear Translocation by Inhibition of IκBα Degradation and Activates p38/ERK1/2-MSK1-Dependent CREB Phosphorylation in Macrophages Stimulated with Lipopolysaccharide. Front Immunol 2016; 7:533. [PMID: 27933067 PMCID: PMC5122595 DOI: 10.3389/fimmu.2016.00533] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/11/2016] [Indexed: 01/06/2023] Open
Abstract
The inflammatory response is a critical molecular defense mechanism of the innate immune system that mediates the elimination of disease-causing bacteria. Repair of the damaged tissue, and the reestablishment of homeostasis, must be accomplished after elimination of the pathogen. The innate defense regulators (IDRs) are short cationic peptides that mimic natural host defense peptides and are effective in eliminating pathogens by enhancing the activity of the immune system while controlling the inflammatory response. Although the role of different IDRs as modulators of inflammation has been reported, there have been only limited studies of the signaling molecules regulated by this type of peptide. The present study investigated the effect of IDR-1002 on nuclear factor κB (NF-κB) and cAMP-response element-binding protein (CREB) transcription factors that are responsible for triggering and controlling inflammation, respectively, in macrophages. We found that TNF-α and COX-2 expression, IκBα phosphorylation, and NF-κB nuclear translocation were strongly inhibited in macrophages pre-incubated with IDR-1002 and then stimulated with lipopolysaccharide (LPS). IDR-1002 also increased CREB phosphorylation at Ser133 via activation of the p38/ERK1/2–MSK1 signaling pathways without detectable expression of the cytokines IL-4, IL-10, and IL-13 involved is suppressing inflammation or alternative activation. Transcriptional activation of NF-κB and CREB is known to require interaction with the transcriptional coactivator CREB-binding protein (CBP). To test for CBP–NF-κB and CBP–CREB complex formation, we performed co-immunoprecipitation assays. These assays showed that IDR-1002 inhibited the interaction between CBP and NF-κB in macrophages stimulated with LPS, which might explain the inhibition of TNF-α and COX-2 expression. Furthermore, the complex between CBP and CREB in macrophages stimulated with IDR-1002 was also inhibited, which might explain why IDR-1002 did not lead to expression of IL-4, IL-10, and IL-13, even though it induced an increase in phospho-CREB relative abundance. In conclusion, our results indicated that IDR-1002 has a dual effect. On one hand, it inhibited NF-κB nuclear translocation through a mechanism that involved inhibition of IκBα phosphorylation, and on the other, it activated a protein kinase signaling cascade that phosphorylated CREB to selectively influence cytokine gene expression. Based on these results, we think IDR-1002 could be a potential good biopharmaceutical candidate to control inflammation.
Collapse
Affiliation(s)
- Alejandro Huante-Mendoza
- Laboratory of Molecular Immunology and Signal Transduction, Facultad de Medicina Veterinaria y Zootecnia, Centro Multidisciplinario de Estudios en Biotecnología, Universidad Michoacana de San Nicolás de Hidalgo , Morelia, Michoacán , México
| | - Octavio Silva-García
- Laboratory of Molecular Immunology and Signal Transduction, Facultad de Medicina Veterinaria y Zootecnia, Centro Multidisciplinario de Estudios en Biotecnología, Universidad Michoacana de San Nicolás de Hidalgo , Morelia, Michoacán , México
| | - Javier Oviedo-Boyso
- Laboratory of Molecular Immunology and Signal Transduction, Facultad de Medicina Veterinaria y Zootecnia, Centro Multidisciplinario de Estudios en Biotecnología, Universidad Michoacana de San Nicolás de Hidalgo , Morelia, Michoacán , México
| | - Robert E W Hancock
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia , Vancouver, BC , Canada
| | - Víctor M Baizabal-Aguirre
- Laboratory of Molecular Immunology and Signal Transduction, Facultad de Medicina Veterinaria y Zootecnia, Centro Multidisciplinario de Estudios en Biotecnología, Universidad Michoacana de San Nicolás de Hidalgo , Morelia, Michoacán , México
| |
Collapse
|
22
|
Låg M, Øvrevik J, Totlandsdal AI, Lilleaas EM, Thormodsæter A, Holme JA, Schwarze PE, Refsnes M. Air pollution-related metals induce differential cytokine responses in bronchial epithelial cells. Toxicol In Vitro 2016; 36:53-65. [PMID: 27427241 DOI: 10.1016/j.tiv.2016.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/21/2016] [Accepted: 07/13/2016] [Indexed: 12/22/2022]
Abstract
Different transition metals have been shown to induce inflammatory responses in lung. We have compared eight different metal ions with regard to cytokine responses, cytotoxicity and signalling mechanisms in a human lung epithelial cell model (BEAS-2B). Among the metal ions tested, there were large differences with respect to pro-inflammatory potential. Exposure to Cd(2+), Zn(2+) and As(3+) induced CXCL8 and IL-6 release at concentrations below 100μM, and Mn(2+) and Ni(2+) at concentrations above 200μM. In contrast, VO4(3-), Cu(2+) and Fe(2+) did not induce any significant increase of these cytokines. An expression array of 20 inflammatory relevant genes also showed a marked up-regulation of CXCL10, IL-10, IL-13 and CSF2 by one or more of the metal ions. The most potent metals, Cd(2+), Zn(2+) and As(3+) induced highest levels of oxidative activity, and ROS appeared to be central in their CXCL8 and IL-6 responses. Activation of the MAPK p38 seemed to be a critical mediator. However, the NF-κB pathway appeared predominately to be involved only in Zn(2+)- and As(3+)-induced CXCL8 and IL-6 responses. Thus, the most potent metals Cd(2+), Zn(2+) and As(3+) seemed to induce a similar pattern for the cytokine responses, and with some exceptions, via similar signalling mechanisms.
Collapse
Affiliation(s)
- M Låg
- Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404, Nydalen, N-0403 Oslo, Norway.
| | - J Øvrevik
- Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404, Nydalen, N-0403 Oslo, Norway
| | - A I Totlandsdal
- Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404, Nydalen, N-0403 Oslo, Norway
| | - E M Lilleaas
- Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404, Nydalen, N-0403 Oslo, Norway
| | - A Thormodsæter
- Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404, Nydalen, N-0403 Oslo, Norway
| | - J A Holme
- Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404, Nydalen, N-0403 Oslo, Norway
| | - P E Schwarze
- Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404, Nydalen, N-0403 Oslo, Norway
| | - M Refsnes
- Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404, Nydalen, N-0403 Oslo, Norway
| |
Collapse
|
23
|
Ruef J, Browatzki M, Pfeiffer CAH, Schmidt J, Kranzhöfer R. Angiotensin II promotes the inflammatory response to CD40 ligation via TRAF-2. Vasc Med 2016; 12:23-7. [PMID: 17451090 DOI: 10.1177/1358863x07076766] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A plethora of evidence supports a link between inflammation and atherogenesis. Both the vasoactive peptide angiotensin II (ANG II) as well as the CD40/CD154 signaling pathway exhibit proinflammatory properties with a direct influence on atherogenesis. We therefore tested the hypothesis that ANG II interacts with CD40/CD154 in human vascular smooth muscle cells (SMC). ANG II did not increase expression of CD40 in human SMC. However, when SMC were prestimulated with ANG II and thereafter stimulated with CD154, the ligand for CD40, the release of IL-6 as a marker of inflammatory activation was augmented compared to cells not primed with ANG II. TNF receptor-associated factor 2 (TRAF-2), an important adaptor protein involved in CD40 signaling, but not TRAF-5 or -6, was increased by ANG II via activation of the angiotensin II type 1 (AT1) receptor subtype. These results suggest that a signaling pathway downstream of CD40 may be altered by ANG II prestimulation. Thus, ANG II can also indirectly cause inflammatory activation of vascular SMC. The data show a novel link between the proatherogenic vasoactive peptide ANG II and cell—cell contact-mediated inflammatory pathways and implicate options for the prevention and therapy of atherosclerotic disease.
Collapse
MESH Headings
- Angiotensin II/metabolism
- Angiotensin II/pharmacology
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Atherosclerosis/metabolism
- CD40 Antigens/metabolism
- CD40 Ligand/metabolism
- Cells, Cultured
- Dose-Response Relationship, Drug
- Humans
- Inflammation/metabolism
- Interleukin-6/metabolism
- Losartan/pharmacology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Receptor Cross-Talk
- Receptor, Angiotensin, Type 1/metabolism
- Saphenous Vein/metabolism
- TNF Receptor-Associated Factor 2/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Johannes Ruef
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
24
|
Yu OM, Brown JH. G Protein-Coupled Receptor and RhoA-Stimulated Transcriptional Responses: Links to Inflammation, Differentiation, and Cell Proliferation. Mol Pharmacol 2015; 88:171-80. [PMID: 25904553 PMCID: PMC4468647 DOI: 10.1124/mol.115.097857] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/22/2015] [Indexed: 01/06/2023] Open
Abstract
The low molecular weight G protein RhoA (rat sarcoma virus homolog family member A) serves as a node for transducing signals through G protein-coupled receptors (GPCRs). Activation of RhoA occurs through coupling of G proteins, most prominently, G12/13, to Rho guanine nucleotide exchange factors. The GPCR ligands that are most efficacious for RhoA activation include thrombin, lysophosphatidic acid, sphingosine-1-phosphate, and thromboxane A2. These ligands also stimulate proliferation, differentiation, and inflammation in a variety of cell and tissues types. The molecular events underlying these responses are the activation of transcription factors, transcriptional coactivators, and downstream gene programs. This review describes the pathways leading from GPCRs and RhoA to the regulation of activator protein-1, NFκB (nuclear factor κ-light-chain-enhancer of activated B cells), myocardin-related transcription factor A, and Yes-associated protein. We also focus on the importance of two prominent downstream transcriptional gene targets, the inflammatory mediator cyclooxygenase 2, and the matricellular protein cysteine-rich angiogenic inducer 61 (CCN1). Finally, we describe the importance of GPCR-induced activation of these pathways in the pathophysiology of cancer, fibrosis, and cardiovascular disease.
Collapse
Affiliation(s)
- Olivia M Yu
- Department of Pharmacology (O.Y., J.H.B.) and Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California (O.Y.)
| | - Joan Heller Brown
- Department of Pharmacology (O.Y., J.H.B.) and Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California (O.Y.)
| |
Collapse
|
25
|
Lavoz C, Alique M, Rodrigues-Diez R, Pato J, Keri G, Mezzano S, Egido J, Ruiz-Ortega M. Gremlin regulates renal inflammation via the vascular endothelial growth factor receptor 2 pathway. J Pathol 2015; 236:407-20. [PMID: 25810250 DOI: 10.1002/path.4537] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/17/2015] [Accepted: 03/18/2015] [Indexed: 01/03/2023]
Abstract
Inflammation is a main feature of progressive kidney disease. Gremlin binds to bone morphogenetic proteins (BMPs), acting as an antagonist and regulating nephrogenesis and fibrosis among other processes. Gremlin also binds to vascular endothelial growth factor receptor-2 (VEGFR2) in endothelial cells to induce angiogenesis. In renal cells, gremlin regulates proliferation and fibrosis, but there are no data about inflammatory-related events. We have investigated the direct effects of gremlin in the kidney, evaluating whether VEGFR2 is a functional gremlin receptor. Administration of recombinant gremlin to murine kidneys induced rapid and sustained activation of VEGFR2 signalling, located in proximal tubular epithelial cells. Gremlin bound to VEGFR2 in these cells in vitro, activating this signalling pathway independently of its action as an antagonist of BMPs. In vivo, gremlin caused early renal damage, characterized by activation of the nuclear factor (NF)-κB pathway linked to up-regulation of pro-inflammatory factors and infiltration of immune inflammatory cells. VEGFR2 blockade diminished gremlin-induced renal inflammatory responses. The link between gremlin/VEGFR2 and NF-κB/inflammation was confirmed in vitro. Gremlin overexpression was associated with VEGFR2 activation in human renal disease and in the unilateral ureteral obstruction experimental model, where VEGFR2 kinase inhibition diminished renal inflammation. Our data show that a gremlin/VEGFR2 axis participates in renal inflammation and could be a novel target for kidney disease.
Collapse
Affiliation(s)
- Carolina Lavoz
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| | - Matilde Alique
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| | - Raquel Rodrigues-Diez
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| | | | - Gyorgy Keri
- VichemChemie Ltd, Budapest, Hungary.,MTA-SE Pathobiochemistry Research Group, Department of Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral, Valdivia, Chile
| | - Jesús Egido
- Division of Nephrology and Hypertension. IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, CIBERDEM, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| |
Collapse
|
26
|
Rodrigues-Diez RR, Garcia-Redondo AB, Orejudo M, Rodrigues-Diez R, Briones AM, Bosch-Panadero E, Kery G, Pato J, Ortiz A, Salaices M, Egido J, Ruiz-Ortega M. The C-terminal module IV of connective tissue growth factor, through EGFR/Nox1 signaling, activates the NF-κB pathway and proinflammatory factors in vascular smooth muscle cells. Antioxid Redox Signal 2015; 22:29-47. [PMID: 25065408 PMCID: PMC4270131 DOI: 10.1089/ars.2013.5500] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS Connective tissue growth factor (CTGF/CCN2) is a developmental gene upregulated in pathological conditions, including cardiovascular diseases, whose product is a matricellular protein that can be degraded to biologically active fragments. Among them, the C-terminal module IV [CCN2(IV)] regulates many cellular functions, but there are no data about redox process. Therefore, we investigated whether CCN2(IV) through redox signaling regulates vascular responses. RESULTS CCN2(IV) increased superoxide anion (O2(•-)) production in murine aorta (ex vivo and in vivo) and in cultured vascular smooth muscle cells (VSMCs). In isolated murine aorta, CCN2(IV), via O2(•-), increased phenylephrine-induced vascular contraction. CCN2(IV) in vivo regulated several redox-related processes in mice aorta, including increased nonphagocytic NAD(P)H oxidases (Nox)1 activity, protein nitrosylation, endothelial dysfunction, and activation of the nuclear factor-κB (NF-κB) pathway and its related proinflammatory factors. The role of Nox1 in CCN2(IV)-mediated vascular responses in vivo was investigated by gene silencing. The administration of a Nox1 morpholino diminished aortic O2(•-) production, endothelial dysfunction, NF-κB activation, and overexpression of proinflammatory genes in CCN2(IV)-injected mice. The link CCN2(IV)/Nox1/NF-κB/inflammation was confirmed in cultured VSMCs. Epidermal growth factor receptor (EGFR) is a known CCN2 receptor. In VSMCs, CCN2(IV) activates EGFR signaling. Moreover, EGFR kinase inhibition blocked vascular responses in CCN2(IV)-injected mice. INNOVATION AND CONCLUSION CCN2(IV) is a novel prooxidant factor that in VSMCs induces O2(•-) production via EGFR/Nox1 activation. Our in vivo data demonstrate that CCN2(IV) through EGFR/Nox1 signaling pathway induces endothelial dysfunction and activation of the NF-κB inflammatory pathway. Therefore, CCN2(IV) could be considered a potential therapeutic target for redox-related cardiovascular diseases.
Collapse
Affiliation(s)
- Raúl R Rodrigues-Diez
- 1 Cellular Biology in Renal Diseases Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma Madrid , Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Alique M, Sánchez-López E, Rayego-Mateos S, Egido J, Ortiz A, Ruiz-Ortega M. Angiotensin II, via angiotensin receptor type 1/nuclear factor-κB activation, causes a synergistic effect on interleukin-1-β-induced inflammatory responses in cultured mesangial cells. J Renin Angiotensin Aldosterone Syst 2014; 16:23-32. [PMID: 25354522 DOI: 10.1177/1470320314551564] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 08/17/2014] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION The nuclear factor-κB (NF-κB) is an important regulator of the inflammatory response. Angiotensin II (Ang II) activates the NF-κB pathway linked to renal inflammation. Although both AT1 and AT2 receptors are involved in Ang II-mediated NF-κB activation, the biological processes mediated by each receptor are not fully characterized. Interleukin-1β (IL-1β) is an important macrophage-derived cytokine that regulates immune and inflammatory processes, activating intracellular pathways shared with Ang II, including the NF-κB. MATERIALS AND METHODS In vitro studies were done in primary cultured rat mesangial cells. NF-κB pathway was evaluated by phosphorylated levels of p65/IκB and DNA binding activity. The Ang II receptor subtype was determined by pretreatment with AT1 and AT2 antagonists. RESULTS In mesangial cells the simultaneous presence of Ang II and IL-1β caused a synergistic activation of the NF-κB pathway and a marked upregulation of proinflammatory factors under NF-κB control, including monocyte chemoattractant protein-1. The AT1, but not AT2, antagonist abolished the synergistic effect on NF-κB activation and proinflammatory genes caused by coincubation of Ang II and IL-1β. CONCLUSIONS These data indicates that Ang II, via AT1/NF-κB pathway activation, cooperates with IL-β to increase the inflammatory response in mesangial cells.
Collapse
Affiliation(s)
- Matilde Alique
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz Universidad Autónoma Madrid, Spain
| | - Elsa Sánchez-López
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz Universidad Autónoma Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz Universidad Autónoma Madrid, Spain
| | - Jesús Egido
- Renal Research Laboratory, IIS-Fundación Jiménez Díaz Universidad Autónoma Madrid, Spain
| | - Alberto Ortiz
- Dialysis Unit, IIS-Fundación Jiménez Díaz Universidad Autónoma Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz Universidad Autónoma Madrid, Spain
| |
Collapse
|
28
|
Nagasawa T, Matsushima-Nishiwaki R, Yasuda E, Matsuura J, Toyoda H, Kaneoka Y, Kumada T, Kozawa O. Heat shock protein 20 (HSPB6) regulates TNF-α-induced intracellular signaling pathway in human hepatocellular carcinoma cells. Arch Biochem Biophys 2014; 565:1-8. [PMID: 25447820 DOI: 10.1016/j.abb.2014.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/02/2014] [Accepted: 10/20/2014] [Indexed: 12/19/2022]
Abstract
We previously demonstrated that the expression of HSP20, a small heat shock protein, is inversely correlated with the progression of HCC. Inflammation is associated with HCC, and numerous cytokines, including TNF-α, act as key mediators in the progression of HCC. In the present study, we investigated whether HSP20 is implicated in the TNF-α-stimulated intracellular signaling in HCC using human HCC-derived HuH7 cells in the presence of TNF-α. In HSP20-overexpressing HCC cells, the cell growth was retarded compared with that in the control cells under long-term exposure of TNF-α. Because NF-κB pathway is the main intracellular signaling system activated by TNF-α, we investigated the effects of HSP20-overexpression of this pathway. The protein levels of IKK-α, but not IKK-β, in the HSP20-overexpressing cells were decreased. Short-term exposure to TNF-α-induced phosphorylation and degradation of IκB, and the phosphorylation and transactivational activity of NF-κB were suppressed in the HSP20-overexpressing HCC cells. Furthermore, the increase in IKK-α levels was accompanied by a decrease in the HSP20 levels in human HCC tissues. These findings strongly suggest that HSP20 might decrease the IKK-α protein level and that it down-regulates the TNF-α-stimulated intracellular signaling in HCC, thus resulting in the suppression of HCC progression.
Collapse
Affiliation(s)
- Tomoaki Nagasawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | | | - Eisuke Yasuda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Gifu 503-8502, Japan; Department of Radiological Technology, Suzuka University of Medical Science, Suzuka 513-8670, Japan
| | - Junya Matsuura
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Gifu 503-8502, Japan
| | - Yuji Kaneoka
- Department of Surgery, Ogaki Municipal Hospital, Ogaki, Gifu 503-8502, Japan
| | - Takashi Kumada
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Gifu 503-8502, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan.
| |
Collapse
|
29
|
Class A scavenger receptor deficiency augments angiotensin II-induced vascular remodeling. Biochem Pharmacol 2014; 90:254-64. [DOI: 10.1016/j.bcp.2014.05.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/17/2014] [Accepted: 05/19/2014] [Indexed: 11/23/2022]
|
30
|
Borrajo A, Rodriguez-Perez AI, Diaz-Ruiz C, Guerra MJ, Labandeira-Garcia JL. Microglial TNF-α mediates enhancement of dopaminergic degeneration by brain angiotensin. Glia 2014; 62:145-57. [PMID: 24272709 DOI: 10.1002/glia.22595] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/14/2013] [Accepted: 10/16/2013] [Indexed: 11/11/2022]
Abstract
In vitro and in vivo models of Parkinson's disease were used to investigate whether TNF-α plays a major role in the enhancement of the microglial response and dopaminergic degeneration induced by brain angiotensin hyperactivity. Treatment of primary mesencephalic cultures with low doses of the neurotoxin MPP(+) induced a significant loss of dopaminergic neurons, which was enhanced by cotreatment with angiotensin II and inhibited by TNF-α inhibitors. Treatment of primary cultures with angiotensin induced a marked increase in levels of TNF-α, which was inhibited by treatment with angiotensin type-1-receptor antagonists, NADPH-oxidase inhibitors and NFK-β inhibitors. However, TNF-α levels were not significantly affected by treatment with angiotensin in the absence of microglia. The microglial origin of the angiotensin-induced increase in TNF-α levels was confirmed using dopaminergic (MES 23.5) and microglial (N9) cell lines. Inhibition of the microglial Rho-kinase activity also blocked the AII-induced increase in TNF-α levels. Treatment of the dopaminergic cell line with TNF-α revealed that NFK-β activation mediates the deleterious effect of microglial TNF-α on dopaminergic neurons. Treatment of mice with MPTP also induced significant increases in striatal and nigral TNF-α levels, which were inhibited by angiotensin type-1-receptor antagonists or NFK-β inhibitors. The present results show that microglial TNF-α plays a major role in angiotensin-induced dopaminergic cell death and that the microglial release of TNF-α is mediated by activation of angiotensin type-1 receptors, NADPH-oxidase, Rho-kinase and NFK-β.
Collapse
Affiliation(s)
- Ana Borrajo
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, Santiago de Compostela, Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | | | | | | | | |
Collapse
|
31
|
Integrin-linked kinase plays a key role in the regulation of angiotensin II-induced renal inflammation. Clin Sci (Lond) 2014; 127:19-31. [PMID: 24383472 DOI: 10.1042/cs20130412] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
ILK (integrin-linked kinase) is an intracellular serine/threonine kinase involved in cell-matrix interactions. ILK dysregulation has been described in chronic renal disease and modulates podocyte function and fibrosis, whereas data about its role in inflammation are scarce. AngII (angiotensin II) is a pro-inflammatory cytokine that promotes renal inflammation. AngII blockers are renoprotective and down-regulate ILK in experimental kidney disease, but the involvement of ILK in the actions of AngII in the kidney has not been addressed. Therefore we have investigated whether ILK signalling modulates the kidney response to systemic AngII infusion in wild-type and ILK-conditional knockout mice. In wild-type mice, AngII induced an inflammatory response, characterized by infiltration of monocytes/macrophages and lymphocytes, and up-regulation of pro-inflammatory factors (chemokines, adhesion molecules and cytokines). AngII activated several intracellular signalling mechanisms, such as the NF-κB (nuclear factor κB) transcription factor, Akt and production of ROS (reactive oxygen species). All these responses were prevented in AngII-infused ILK-deficient mice. In vitro studies characterized further the mechanisms regulating the inflammatory response modulated by ILK. In cultured tubular epithelial cells ILK blockade, by siRNA, inhibited AngII-induced NF-κB subunit p65 phosphorylation and its nuclear translocation. Moreover, ILK gene silencing prevented NF-κB-related pro-inflammatory gene up-regulation. The results of the present study demonstrate that ILK plays a key role in the regulation of renal inflammation by modulating the canonical NF-κB pathway, and suggest a potential therapeutic target for inflammatory renal diseases.
Collapse
|
32
|
Jasińska-Stroschein M, Orszulak-Michalak D. The current approach into signaling pathways in pulmonary arterial hypertension and their implication in novel therapeutic strategies. Pharmacol Rep 2014; 66:552-64. [PMID: 24948054 DOI: 10.1016/j.pharep.2014.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 04/02/2014] [Accepted: 04/07/2014] [Indexed: 02/01/2023]
Abstract
Many mediators and signaling pathways, with their downstream effectors, have been implicated in the pathogenesis of pulmonary hypertension. Currently approved drugs, representing an option of specific therapy, target NO, prostacyclin or ET-1 pathways and provide a significant improvement in the symptomatic status of patients and a slower rate of clinical deterioration. However, despite such improvements in the treatment, PAH remains a chronic disease without a cure, the mortality associated with PAH remains high and effective therapeutic regimens are still required. Knowledge about the role of the pathways involved in PAH and their interactions provides a better understanding of the pathogenesis of the disease and may highlight directions for novel therapeutic strategies for PAH. This paper reviews some novel, promising PAH-associated signaling pathways, such as RAAS, RhoA/ROCK, PDGF, PPAR, and TGF, focusing also on their possible interactions with well-established ones such as NO, ET-1 and prostacyclin pathways.
Collapse
|
33
|
Osako MK, Nakagami H, Shimamura M, Koriyama H, Nakagami F, Shimizu H, Miyake T, Yoshizumi M, Rakugi H, Morishita R. Cross-talk of receptor activator of nuclear factor-κB ligand signaling with renin-angiotensin system in vascular calcification. Arterioscler Thromb Vasc Biol 2013; 33:1287-96. [PMID: 23580147 DOI: 10.1161/atvbaha.112.301099] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular calcification is accelerated by hypertension and also contributes to hypertension; however, it is an enigma why hypertension and vascular calcification are a vicious spiral. The present study elucidates the cross-talk between renin-angiotensin II system and receptor activator of nuclear factor-κB ligand (RANKL) system in vascular calcification. APPROACH AND RESULTS Angiotensin (Ang) II (10(-7) mol/L) significantly increased calcium deposition as assessed by Alizarin Red staining, associated with a significant increase in the expression of RANKL, RANK, and bone-related genes, such as cbfa1 and msx2, in human aortic vascular smooth muscle cells. Infusion of Ang II (100 ng/kg per minute) in ovariectomized ApoE(-/-) mice under high-fat diet significantly increased the expression of RANKL system and calcification in vivo, whereas administration of Ang II receptor blocker (olmesartan, 3 mg/kg per day) decreased the calcification and bone markers' expression. In addition, male OPG(-/-) mice showed a significant increase in vascular calcification followed by Ang II infusion as compared with wild type. Conversely, RANKL significantly increased Ang II type 1 receptor and angiotensin II-converting enzyme expression in vascular smooth muscle cells via extracellular signal-regulated protein kinase phosphorylation. CONCLUSIONS The present study demonstrated that Ang II significantly induced vascular calcification in vitro and in vivo through RANKL activation. In addition, RANKL activated renin-angiotensin II system, especially angiotensin II-converting enzyme and Ang II type 1 receptor. Cross-talk between renin-angiotensin II system and RANKL system might work as a vicious cycle to promote vascular calcification in atherosclerosis. Further studies to inhibit renin-angiotensin II system and RANKL may provide new therapeutic options to prevent and regress vascular calcification.
Collapse
Affiliation(s)
- Mariana Kiomy Osako
- Division of Vascular Medicine and Epigenetics, United Graduate School of Child Development, Osaka University, Suita, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Morinelli TA, Lee MH, Kendall RT, Luttrell LM, Walker LP, Ullian ME. Angiotensin II activates NF-κB through AT1A receptor recruitment of β-arrestin in cultured rat vascular smooth muscle cells. Am J Physiol Cell Physiol 2013; 304:C1176-86. [PMID: 23576578 DOI: 10.1152/ajpcell.00235.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of the angiotensin type 1A receptor (AT1AR) in rat aorta vascular smooth muscle cells (RASMC) results in increased synthesis of the proinflammatory enzyme cyclooxygenase-2 (COX-2). We previously showed that nuclear localization of internalized AT1AR results in activation of transcription of the gene for COX-2, i.e., prostaglandin-endoperoxide synthase-2. Others have suggested that ANG II stimulation of COX-2 protein synthesis is mediated by NF-κB. The purpose of the present study was to examine the interrelationship between AT1AR activation, β-arrestin recruitment, and NF-κB activation in the ability of ANG II to increase COX-2 protein synthesis in RASMC. In the present study we utilized RASMC, inhibitors of the NF-κB pathway, β-arrestin knockdown, radioligand binding, immunoblotting, and immunofluorescence to characterize the roles of AT1AR internalization, NF-κB activation, and β-arrestin in ANG II-induced COX-2 synthesis. Ro-106-9920 or parthenolide, agents that inhibit the initial steps of NF-κB activation, blocked ANG II-induced p65 NF-κB nuclear localization, COX-2 protein expression, β-arrestin recruitment, and AT1AR internalization without inhibiting ANG II-induced p42/44 ERK activation. Curcumin, an inhibitor of NF-κB-induced transcription, blocked ANG II-induced COX-2 protein expression without altering AT1AR internalization, ANG II-induced p65 NF-κB nuclear localization, or p42/44 ERK activation. Small interfering RNA-induced knockdown of β-arrestin-1 and -2 inhibited ANG II-induced p65 NF-κB nuclear localization. In vascular smooth muscle cells, internalization of the activated AT1AR mediated by β-arrestins activates the NF-κB pathway, producing nuclear localization of the transcription factor and initiation of COX-2 protein synthesis, thereby linking internalization of the receptor with the NF-κB pathway.
Collapse
Affiliation(s)
- Thomas A Morinelli
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Moles A, Sanchez AM, Banks PS, Murphy LB, Luli S, Borthwick L, Fisher A, O’Reilly S, van Laar JM, White SA, Perkins ND, Burt AD, Mann DA, Oakley F. Inhibition of RelA-Ser536 phosphorylation by a competing peptide reduces mouse liver fibrosis without blocking the innate immune response. Hepatology 2013; 57:817-28. [PMID: 22996371 PMCID: PMC3807604 DOI: 10.1002/hep.26068] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 05/24/2012] [Accepted: 08/29/2012] [Indexed: 12/15/2022]
Abstract
UNLABELLED Phosphorylation of the RelA subunit at serine 536 (RelA-P-Ser536) is important for hepatic myofibroblast survival and is mechanistically implicated in liver fibrosis. Here, we show that a cell-permeable competing peptide (P6) functions as a specific targeted inhibitor of RelA-P-Ser536 in vivo and exerts an antifibrogenic effect in two progressive liver disease models, but does not impair hepatic inflammation or innate immune responses after lipopolysaccharide challenge. Using kinase assays and western blotting, we confirm that P6 is a substrate for the inhibitory kappa B kinases (IKKs), IKKα and IKKβ, and, in human hepatic myofibroblasts, P6 prevents RelA-P-Ser536, but does not affect IKK activation of IκBα. We demonstrate that RelA-P-Ser536 is a feature of human lung and skin fibroblasts, but not lung epithelial cells, in vitro and is present in sclerotic skin and diseased lungs of patients suffering from idiopathic pulmonary fibrosis. CONCLUSION RelA-P-Ser536 may be a core fibrogenic regulator of fibroblast phenotype.
Collapse
Affiliation(s)
- Anna Moles
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Ana M Sanchez
- Institute for Cell and Molecular Biosciences, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Paul S Banks
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Lindsay B Murphy
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Saimir Luli
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Lee Borthwick
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Andrew Fisher
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Steven O’Reilly
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Jacob M van Laar
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Steven A White
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Neil D Perkins
- Institute for Cell and Molecular Biosciences, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Alastair D Burt
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Derek A Mann
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom
| | - Fiona Oakley
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, United Kingdom,Address reprint requests to: Fiona Oakley, Ph.D., Fibrosis Research Group, Institute of cellular Medicine, Newcastle University, Room M4.158, 4th Floor Leech Building, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom. E-mail: ; fax: +44 191 222 5455
| |
Collapse
|
36
|
Kim JM, Heo HS, Ha YM, Ye BH, Lee EK, Choi YJ, Yu BP, Chung HY. Mechanism of Ang II involvement in activation of NF-κB through phosphorylation of p65 during aging. AGE (DORDRECHT, NETHERLANDS) 2012; 34:11-25. [PMID: 21318332 PMCID: PMC3260361 DOI: 10.1007/s11357-011-9207-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 01/03/2011] [Indexed: 05/30/2023]
Abstract
Angiotensin II (Ang II), a major effector of the renin-angiotensin system, is now recognized as a pro-inflammatory mediator. This Ang II signaling, which causes transcription of pro-inflammatory genes, is regulated through nuclear factor-κB (NF-κB). At present, the molecular mechanisms underlying the effect of aging on Ang II signaling and NF-κB activation are not fully understood. The purpose of this study was to document altered molecular events involved in age-related changes in Ang II signaling and NF-κB activation. Experimentations were carried out using kidney tissues from Fischer 344 rats at 6, 12, 18, and 24 months of age, and the rat endothelial cell line, YPEN-1 for the detailed molecular work. Results show that increases in Ang II and Ang II type 1 receptor during aging were accompanied by the generation of reactive species. Increased Ang II activated NF-κB by phosphorylating IκBα and p65. Increased phosphorylation of p65 at Ser 536 was mediated by the enhanced phosphorylation of IκB kinase αβ, while phosphorylation site Ser 276 of p65 was mediated by upregulated mitogen-activated and stress-activated protein kinase-1. These altered molecular events in aged animals were partly verified by experiments using YPEN-1 cells. Collectively, our findings provide molecular insights into the pro-inflammatory actions of Ang II, actions that influence the phosphorylation of p65-mediated NF-κB activation during aging. Our study demonstrates the age-related pleiotropic nature of the physiologically important Ang II can change into a deleterious culprit that contributes to an increased incidence of many chronic diseases such as atherosclerosis, diabetes, and dementia.
Collapse
Affiliation(s)
- Ji Min Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Hyoung-Sam Heo
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Young Mi Ha
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Byeong Hyeok Ye
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Eun Kyeong Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Yeon Ja Choi
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| | - Byung Pal Yu
- Department of Physiology, University of Texas Health Science Center, San Antonio, TX 78229-3900 USA
| | - Hae Young Chung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
- College of Pharmacy, Pusan National University, San 30, Jangjun-dong, Gumjung-gu, Busan, 609-735 South Korea
| |
Collapse
|
37
|
Sugiyama T, Takahashi K, Tokoro S, Gotou T, Neri P, Mori H. Inhibitory effect of 10-hydroxy-trans-2-decenoic acid on LPS-induced IL-6 production via reducing IκB-ζ expression. Innate Immun 2011; 18:429-37. [PMID: 21948282 DOI: 10.1177/1753425911416022] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The effect of 10-hydroxy-trans-2-decenoic acid (10H2DA), a major fatty acid component of royal jelly, was investigated on LPS-induced cytokine production in murine macrophage cell line, RAW264 cells. 10H2DA inhibited LPS-induced IL-6 production dose-dependently, but did not inhibit TNF-α production. 10H2DA inhibited LPS-induced NF-κB activation in a dose-dependent fashion. In addition, NF-κB activation induced by over-expression of either MyD88 or Toll/IL-1 receptor domain-containing adaptor inducing IFN-β (TRIF) was also inhibited by 10H2DA. Degradation of IκB-α and phosphorylation of IκB kinase-α were not inhibited by 10H2DA. On the other hand, reduction of LPS-induced IκB-ζ expression was discovered. Production of lipocalin-2 and granulocyte colony-stimulating factor (G-CSF), which is dependent on IκB-ζ, was also inhibited by 10H2DA, whereas that of IκB-ζ-independent cytokines/chemokines, such as IFN-β, murine monocyte chemotactic protein-1 (JE), macrophage inflammatory protein (MIP)-1α and MIP-2, was not. Together, 10H2DA specifically inhibited LPS-induced IκB-ζ expression, followed by inhibition of IκB-ζ-dependent gene production. These results suggest that 10H2DA is one of the components of royal jelly to show anti-inflammatory effects and could be a therapeutic drug candidate for inflammatory and autoimmune diseases associated with IκB-ζ and IL-6 production.
Collapse
Affiliation(s)
- Tsuyoshi Sugiyama
- Department of Biopharmaceutical Sciences, Laboratory of Microbiology, Gifu Pharmaceutical University, Gifu, Japan.
| | | | | | | | | | | |
Collapse
|
38
|
Astrocytic P2Y(1) receptor is involved in the regulation of cytokine/chemokine transcription and cerebral damage in a rat model of cerebral ischemia. J Cereb Blood Flow Metab 2011; 31:1930-41. [PMID: 21487414 PMCID: PMC3185880 DOI: 10.1038/jcbfm.2011.49] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
After brain ischemia, significant amounts of adenosine 5'-triphosphate are released or leaked from damaged cells, thus activating purinergic receptors in the central nervous system. A number of P2X/P2Y receptors have been implicated in ischemic conditions, but to date the P2Y(1) receptor (P2Y(1)R) has not been implicated in cerebral ischemia. In this study, we found that the astrocytic P2Y(1)R, via phosphorylated-RelA (p-RelA), has a negative effect during cerebral ischemia/reperfusion. Intracerebroventricular administration of the P2Y(1)R agonist, MRS 2365, led to an increase in cerebral infarct volume 72 hours after transient middle cerebral artery occlusion (tMCAO). Administration of the P2Y(1)R antagonist, MRS 2179, significantly decreased infarct volume and led to recovered motor coordination. The effects of MRS 2179 occurred within 24 hours of tMCAO, and also markedly reduced the expression of p-RelA and interleukin-6, tumor necrosis factor-α, monocyte chemotactic protein-1/chemokine (C-C motif) ligand 2 (CCL2), and interferon-inducible protein-10/chemokine (C-X-C motif) ligand 10 (CXCL10) mRNA. P2Y(1)R and p-RelA were colocalized in glial fibrillary acidic protein-positive astrocytes, and an increase in infarct volume after MRS 2365 treatment was inhibited by the nuclear factor (NF)-κB inhibitor ammonium pyrrolidine dithiocarbamate. These results provide evidence that the P2Y(1)R expressed in cortical astrocytes may help regulate the cytokine/chemokine response after tMCAO/reperfusion through a p-RelA-mediated NF-κB pathway.
Collapse
|
39
|
Hou T, Tieu BC, Ray S, Recinos Iii A, Cui R, Tilton RG, Brasier AR. Roles of IL-6-gp130 Signaling in Vascular Inflammation. Curr Cardiol Rev 2011; 4:179-92. [PMID: 19936194 PMCID: PMC2780819 DOI: 10.2174/157340308785160570] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Revised: 05/13/2008] [Accepted: 05/13/2008] [Indexed: 02/07/2023] Open
Abstract
Interleukin-6 (IL-6) is a well-established, independent indicator of multiple distinct types of cardiovascular disease and all-cause mortality. In this review, we present current understanding of the multiple roles that IL-6 and its signaling pathways through glycoprotein 130 (gp130) play in cardiovascular homeostasis. IL-6 is highly inducible in vascular tissues through the actions of the angiotensin II (Ang II) peptide, where it acts in a paracrine manner to signal through two distinct mechanisms, the first being a classic membrane receptor initiated pathway and the second, a trans-signaling pathway, being able to induce responses even in tissues lacking the IL-6 receptor. Recent advances and new concepts in how its intracellular signaling pathways operate via the Janus kinase (JAK)-Signal Transducer and Activator of Transcription (STAT) are described. IL-6 has diverse actions in multiple cell types of cardiovascular importance, including endothelial cells, monocytes, platelets, hepatocytes and adipocytes. We discuss central roles of IL-6 in endothelial dysfunction, cellular inflammation by affecting monocyte activation/differentiation, cellular cytoprotective functions from reactive oxygen species (ROS) stress, modulation of pro-coagulant state, myocardial growth control, and its implications in metabolic control and insulin resistance. These multiple actions indicate that IL-6 is not merely a passive biomarker, but actively modulates adaptive and pathological responses to cardiovascular stress.
Collapse
Affiliation(s)
- Tieying Hou
- Departments of Biochemistry and Molecular Biology, Internal Medicine, and the Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX-77555-1060, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Zhao Y, Banerjee S, Dey N, LeJeune WS, Sarkar PS, Brobey R, Rosenblatt KP, Tilton RG, Choudhary S. Klotho depletion contributes to increased inflammation in kidney of the db/db mouse model of diabetes via RelA (serine)536 phosphorylation. Diabetes 2011; 60:1907-16. [PMID: 21593200 PMCID: PMC3121423 DOI: 10.2337/db10-1262] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Klotho is an antiaging hormone present in the kidney that extends the lifespan, regulates kidney function, and modulates cellular responses to oxidative stress. We investigated whether Klotho levels and signaling modulate inflammation in diabetic kidneys. RESEARCH DESIGN AND METHODS Renal Klotho expression was determined by quantitative real-time PCR and immunoblot analysis. Primary mouse tubular epithelial cells were treated with methylglyoxalated albumin, and Klotho expression and inflammatory cytokines were measured. Nuclear factor (NF)-κB activation was assessed by treating human embryonic kidney (HEK) 293 and HK-2 cells with tumor necrosis factor (TNF)-α in the presence or absence of Klotho, followed by immunoblot analysis to evaluate inhibitor of κB (IκB)α degradation, IκB kinase (IKK) and p38 activation, RelA nuclear translocation, and phosphorylation. A chromatin immunoprecipitation assay was performed to analyze the effects of Klotho signaling on interleukin-8 and monocyte chemoattractant protein-1 promoter recruitment of RelA and RelA serine (Ser)(536). RESULTS Renal Klotho mRNA and protein were significantly decreased in db/db mice, and a similar decline was observed in the primary cultures of mouse tubule epithelial cells treated with methylglyoxal-modified albumin. The exogenous addition of soluble Klotho or overexpression of membranous Klotho in tissue culture suppressed NF-κB activation and subsequent production of inflammatory cytokines in response to TNF-α stimulation. Klotho specifically inhibited RelA Ser(536) phosphorylation as well as promoter DNA binding of this phosphorylated form of RelA without affecting IKK-mediated IκBα degradation, total RelA nuclear translocation, and total RelA DNA binding. CONCLUSIONS These findings suggest that Klotho serves as an anti-inflammatory modulator, negatively regulating the production of NF-κB-linked inflammatory proteins via a mechanism that involves phosphorylation of Ser(536) in the transactivation domain of RelA.
Collapse
Affiliation(s)
- Yanhua Zhao
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Srijita Banerjee
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Nilay Dey
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Wanda S. LeJeune
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Partha S. Sarkar
- Department of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Reynolds Brobey
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas
| | - Kevin P. Rosenblatt
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas
| | - Ronald G. Tilton
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Sanjeev Choudhary
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas
- Corresponding author: Sanjeev Choudhary,
| |
Collapse
|
41
|
Zhao Y, Widen SG, Jamaluddin M, Tian B, Wood TG, Edeh CB, Brasier AR. Quantification of activated NF-kappaB/RelA complexes using ssDNA aptamer affinity-stable isotope dilution-selected reaction monitoring-mass spectrometry. Mol Cell Proteomics 2011; 10:M111.008771. [PMID: 21502374 PMCID: PMC3108844 DOI: 10.1074/mcp.m111.008771] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 04/08/2011] [Indexed: 12/12/2022] Open
Abstract
Nuclear Factor-κB (NF-κB) is a family of inducible transcription factors regulated by stimulus-induced protein interactions. In the cytoplasm, the NF-κB member RelA transactivator is inactivated by binding inhibitory IκBs, whereas in its activated state, the serine-phosphorylated protein binds the p300 histone acetyltransferase. Here we describe the isolation of a ssDNA aptamer (termed P028F4) that binds to the activated (IκBα-dissociated) form of RelA with a K(D) of 6.4 × 10(-10), and its application in an enrichment-mass spectrometric quantification assay. ssDNA P028F4 competes with cognate duplex high affinity NF-κB binding sites for RelA binding in vitro, binds activated RelA in eukaryotic nuclei and reduces TNFα-stimulated endogenous NF-κB dependent gene expression. Incorporation of P028F4 as an affinity isolation step enriches for serine 536 phosphorylated and p300 coactivator complexed RelA, simultaneously depleting IκBα·RelA complexes. A stable isotope dilution (SID)-selected reaction monitoring (SRM)- mass spectrometry (MS) assay for RelA was developed that produced a linear response over 1,000 fold dilution range of input protein and had a 200 amol lower limit of quantification. This multiplex SID-SRM-MS RelA assay was used to quantify activated endogenous RelA in cytokine-stimulated eukaryotic cells isolated by single-step P028F4 enrichment. The aptamer-SID-SRM-MS assay quantified the fraction of activated RelA in subcellular extracts, detecting the presence of a cytoplasmic RelA reservoir unresponsive to TNFα stimulation. We conclude that aptamer-SID-SRM-MS is a versatile tool for quantification of activated NF-κB/RelA and its associated complexes in response to pathway activation.
Collapse
Affiliation(s)
- Yingxin Zhao
- From the ‡Sealy Center for Molecular Medicine
- §Department of Internal Medicine
| | | | | | | | - Thomas G. Wood
- From the ‡Sealy Center for Molecular Medicine
- the ¶¶Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555
| | | | - Allan R. Brasier
- From the ‡Sealy Center for Molecular Medicine
- §Department of Internal Medicine
- ¶Institute for Translational Sciences, and
| |
Collapse
|
42
|
Alonso F, Krattinger N, Mazzolai L, Simon A, Waeber G, Meda P, Haefliger JA. An angiotensin II- and NF-kappaB-dependent mechanism increases connexin 43 in murine arteries targeted by renin-dependent hypertension. Cardiovasc Res 2010; 87:166-76. [PMID: 20110337 PMCID: PMC2883896 DOI: 10.1093/cvr/cvq031] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 01/20/2010] [Accepted: 01/25/2010] [Indexed: 01/17/2023] Open
Abstract
AIMS Connexins (Cxs) play a role in the contractility of the aorta wall. We investigated how connexins of the endothelial cells (ECs; Cx37, Cx40) and smooth muscle cells (SMCs; Cx43, Cx45) of the aorta change during renin-dependent and -independent hypertension. METHODS AND RESULTS We subjected both wild-type (WT) mice and mice lacking Cx40 (Cx40(-/-)), to either a two-kidney, one-clip procedure or to N-nitro-l-arginine-methyl-ester treatment, which induce renin-dependent and -independent hypertension, respectively. All hypertensive mice featured a thickened aortic wall, increased levels of Cx37 and Cx45 in SMC, and of Cx40 in EC (except in Cx40(-/-) mice). Cx43 was up-regulated, with no effect on its S368 phosphorylation, only in the SMCs of renin-dependent models of hypertension. Blockade of the renin-angiotensin system of Cx40(-/-) mice normalized blood pressure and prevented both aortic thickening and Cx alterations. Ex vivo exposure of WT aortas, carotids, and mesenteric arteries to physiologically relevant levels of angiotensin II (AngII) increased the levels of Cx43, but not of other Cx. In the aortic SMC line of A7r5 cells, AngII activated kinase-dependent pathways and induced binding of the nuclear factor-kappa B (NF-kappaB) to the Cx43 gene promoter, increasing Cx43 expression. CONCLUSION In both large and small arteries, hypertension differently regulates Cx expression in SMC and EC layers. Cx43 is selectively increased in renin-dependent hypertension via an AngII activation of the extracellular signal-regulated kinase and NF-kappaB pathways.
Collapse
MESH Headings
- Angiotensin II/metabolism
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Angiotensin-Converting Enzyme Inhibitors/pharmacology
- Animals
- Antihypertensive Agents/pharmacology
- Aorta/drug effects
- Aorta/metabolism
- Aorta/physiopathology
- Binding Sites
- Blood Pressure
- Carotid Arteries/metabolism
- Carotid Arteries/physiopathology
- Cell Line
- Connexin 43/genetics
- Connexin 43/metabolism
- Connexins/deficiency
- Connexins/genetics
- Disease Models, Animal
- Endothelial Cells/metabolism
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Genes, Reporter
- Hypertension, Renovascular/drug therapy
- Hypertension, Renovascular/etiology
- Hypertension, Renovascular/metabolism
- Hypertension, Renovascular/physiopathology
- Mesenteric Arteries/metabolism
- Mesenteric Arteries/physiopathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Smooth Muscle/metabolism
- NF-kappa B/metabolism
- NG-Nitroarginine Methyl Ester
- Nephrectomy
- Phosphorylation
- Promoter Regions, Genetic
- Rats
- Renin/blood
- Time Factors
- Transfection
- Up-Regulation
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Florian Alonso
- Service of Internal Medicine, Laboratory of Experimental Medicine 19-135S, University Hospital, CHUV-1011 Lausanne, Switzerland
| | - Nathalie Krattinger
- Service of Internal Medicine, Laboratory of Experimental Medicine 19-135S, University Hospital, CHUV-1011 Lausanne, Switzerland
| | - Lucia Mazzolai
- Service of Internal Medicine, Laboratory of Experimental Medicine 19-135S, University Hospital, CHUV-1011 Lausanne, Switzerland
| | - Alexander Simon
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Gérard Waeber
- Service of Internal Medicine, Laboratory of Experimental Medicine 19-135S, University Hospital, CHUV-1011 Lausanne, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, School of Medicine, CMU, 1211 Genève 4, Switzerland
| | - Jacques-Antoine Haefliger
- Service of Internal Medicine, Laboratory of Experimental Medicine 19-135S, University Hospital, CHUV-1011 Lausanne, Switzerland
| |
Collapse
|
43
|
NF-kappaB signaling mediates vascular smooth muscle endothelin type B receptor expression in resistance arteries. Eur J Pharmacol 2010; 637:148-54. [DOI: 10.1016/j.ejphar.2010.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 03/09/2010] [Accepted: 04/04/2010] [Indexed: 12/15/2022]
|
44
|
Ye Y, Pringle LM, Lau AW, Riquelme DN, Wang H, Jiang T, Lev D, Welman A, Blobel GA, Oliveira AM, Chou MM. TRE17/USP6 oncogene translocated in aneurysmal bone cyst induces matrix metalloproteinase production via activation of NF-kappaB. Oncogene 2010; 29:3619-29. [PMID: 20418905 PMCID: PMC2892027 DOI: 10.1038/onc.2010.116] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Aneurysmal bone cyst (ABC) is an aggressive, pediatric bone tumor characterized by extensive destruction of the surrounding bone. Though first described over 60 years ago, its molecular etiology remains poorly understood. Recent work revealed that ABCs harbor translocation of TRE17/USP6, leading to its transcriptional upregulation. TRE17 encodes a ubiquitin-specific protease (USP), and a TBC domain that mediates binding to the Arf6 GTPase. However, the mechanisms by which TRE17 overexpression contributes to tumor pathogenesis, and the role of its USP and TBC domains are unknown. ABCs are characterized by osteolysis, inflammatory recruitment, and extensive vascularization, processes in which matrix proteases play a prominent role. This led us to explore whether TRE17 regulates the production of matrix metalloproteinases (MMPs). In the current study, we demonstrate that TRE17 is sufficient to induce expression of MMP-9 and MMP-10, in a manner requiring its USP activity, but not its ability to bind Arf6. TRE17 induces transcription of MMP-9 through activation of NFκB, mediated in part by the GTPase RhoA and its effector kinase, ROCK. Furthermore, xenograft studies demonstrate that TRE17 induces formation of tumors that reproduce multiple features of ABC, including a high degree of vascularization, with an essential role for the USP domain. In sum, these studies reveal that TRE17 is sufficient to initiate tumorigenesis, identify MMPs as novel TRE17 effectors that likely contribute to ABC pathogenesis, and define the underlying signaling mechanism of their induction.
Collapse
Affiliation(s)
- Y Ye
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Brasier AR. The nuclear factor-kappaB-interleukin-6 signalling pathway mediating vascular inflammation. Cardiovasc Res 2010; 86:211-8. [PMID: 20202975 DOI: 10.1093/cvr/cvq076] [Citation(s) in RCA: 430] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vascular inflammation is a common pathophysiological response to diverse cardiovascular disease processes, including atherosclerosis, myocardial infarction, congestive heart failure, and aortic aneurysms/dissection. Inflammation is an ordered process initiated by vascular injury that produces enhanced leucocyte adherence, chemotaxis, and finally activation in situ. This process is coordinated by local secretion of adhesion molecules, chemotactic factors, and cytokines whose expression is the result of vascular injury-induced signal transduction networks. A wide variety of mediators of the vascular injury response have been identified; these factors include vasoactive peptides (angiotensin II, Ang II), CD40 ligands, oxidized cholesterol, and advanced glycation end-products. Downstream, the nuclear factor-kappaB (NF-kappaB) transcription factor performs an important signal integration step, responding to mediators of vascular injury in a stimulus-dependent and cell type-specific manner. The ultimate consequence of NF-kappaB signalling is the activation of inflammatory genes including adhesion molecules and chemotaxins. However, clinically, the hallmark of vascular NF-kappaB activation is the production of interleukin-6 (IL-6), whose local role in vascular inflammation is relatively unknown. The recent elucidation for the role of the IL-6 signalling pathway in Ang II-induced vascular inflammation as one that controls monocyte activation as well as its diverse signalling mechanism will be reviewed. These new discoveries further our understanding for the important role of the NF-kappaB-IL-6 signalling pathway in the process of vascular inflammation.
Collapse
Affiliation(s)
- Allan R Brasier
- Division of Endocrinology, Department of Internal Medicine, Sealy Center for Molecular Medicine and Institute for Translational Sciences, University of Texas Medical Branch, MRB 8.122, 301 University Blvd, Galveston, TX 77555-1060, USA.
| |
Collapse
|
46
|
Tan X, He W, Liu Y. Combination therapy with paricalcitol and trandolapril reduces renal fibrosis in obstructive nephropathy. Kidney Int 2009; 76:1248-1257. [PMID: 19759524 PMCID: PMC5527548 DOI: 10.1038/ki.2009.346] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Growing evidence suggests that active vitamin D slows the progression of chronic kidney diseases. Here we compared the individual renal protective efficacy of paricalcitol and trandolapril (an angiotensin-converting enzyme inhibitor) in obstructive nephropathy, and examined any potential additive effects of their combination on attenuating renal fibrosis and inflammation. Mice underwent unilateral ureteral obstruction and were treated individually with paricalcitol or trandolapril or their combination. Compared to vehicle-treated controls, monotherapy with paricalcitol or trandolapril inhibited the expression and accumulation of fibronectin and type I and type III collagen, suppressed alpha-smooth muscle actin, vimentin, and Snail1 expression, and reduced total collagen content in the obstructed kidney. Combination therapy led to a more profound inhibition of all parameters. Monotherapy also suppressed renal RANTES (regulated on activation, normal T cell expressed and secreted) and tumor necrosis factor (TNF)-alpha expression and inhibited renal infiltration of T cells and macrophages, whereas the combination had additive effects. Renin expression was induced in the fibrotic kidney and was augmented by trandolapril. Paricalcitol blocked renin induction in the absence or presence of trandolapril. Our study indicates that paricalcitol has renal protective effects, comparable to that of trandolapril, in reducing interstitial fibrosis and inflammation. Combination therapy had additive efficacy in retarding renal scar formation during obstructive nephropathy.
Collapse
Affiliation(s)
- Xiaoyue Tan
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Weichun He
- Department of Medicine, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Youhua Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
47
|
Takeda K, Ichiki T, Narabayashi E, Inanaga K, Miyazaki R, Hashimoto T, Matsuura H, Ikeda J, Miyata T, Sunagawa K. Inhibition of Prolyl Hydroxylase Domain-Containing Protein Suppressed Lipopolysaccharide-Induced TNF-α Expression. Arterioscler Thromb Vasc Biol 2009; 29:2132-7. [DOI: 10.1161/atvbaha.109.196071] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Kotaro Takeda
- From the Departments of Advanced Therapeutics for Cardiovascular Diseases (K.T., T.I., K.S.) and Cardiovascular Medicine (E.N., K.I., R.M., T.H., H.M., J.I., K.S.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and the Center for Translational and Advanced Research (T.M.), Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Toshihiro Ichiki
- From the Departments of Advanced Therapeutics for Cardiovascular Diseases (K.T., T.I., K.S.) and Cardiovascular Medicine (E.N., K.I., R.M., T.H., H.M., J.I., K.S.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and the Center for Translational and Advanced Research (T.M.), Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Eriko Narabayashi
- From the Departments of Advanced Therapeutics for Cardiovascular Diseases (K.T., T.I., K.S.) and Cardiovascular Medicine (E.N., K.I., R.M., T.H., H.M., J.I., K.S.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and the Center for Translational and Advanced Research (T.M.), Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Keita Inanaga
- From the Departments of Advanced Therapeutics for Cardiovascular Diseases (K.T., T.I., K.S.) and Cardiovascular Medicine (E.N., K.I., R.M., T.H., H.M., J.I., K.S.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and the Center for Translational and Advanced Research (T.M.), Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Ryohei Miyazaki
- From the Departments of Advanced Therapeutics for Cardiovascular Diseases (K.T., T.I., K.S.) and Cardiovascular Medicine (E.N., K.I., R.M., T.H., H.M., J.I., K.S.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and the Center for Translational and Advanced Research (T.M.), Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Toru Hashimoto
- From the Departments of Advanced Therapeutics for Cardiovascular Diseases (K.T., T.I., K.S.) and Cardiovascular Medicine (E.N., K.I., R.M., T.H., H.M., J.I., K.S.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and the Center for Translational and Advanced Research (T.M.), Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hirohide Matsuura
- From the Departments of Advanced Therapeutics for Cardiovascular Diseases (K.T., T.I., K.S.) and Cardiovascular Medicine (E.N., K.I., R.M., T.H., H.M., J.I., K.S.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and the Center for Translational and Advanced Research (T.M.), Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Jiro Ikeda
- From the Departments of Advanced Therapeutics for Cardiovascular Diseases (K.T., T.I., K.S.) and Cardiovascular Medicine (E.N., K.I., R.M., T.H., H.M., J.I., K.S.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and the Center for Translational and Advanced Research (T.M.), Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Toshio Miyata
- From the Departments of Advanced Therapeutics for Cardiovascular Diseases (K.T., T.I., K.S.) and Cardiovascular Medicine (E.N., K.I., R.M., T.H., H.M., J.I., K.S.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and the Center for Translational and Advanced Research (T.M.), Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kenji Sunagawa
- From the Departments of Advanced Therapeutics for Cardiovascular Diseases (K.T., T.I., K.S.) and Cardiovascular Medicine (E.N., K.I., R.M., T.H., H.M., J.I., K.S.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan; and the Center for Translational and Advanced Research (T.M.), Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
48
|
Tieu BC, Lee C, Sun H, Lejeune W, Recinos A, Ju X, Spratt H, Guo DC, Milewicz D, Tilton RG, Brasier AR. An adventitial IL-6/MCP1 amplification loop accelerates macrophage-mediated vascular inflammation leading to aortic dissection in mice. J Clin Invest 2009; 119:3637-51. [PMID: 19920349 DOI: 10.1172/jci38308] [Citation(s) in RCA: 368] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Accepted: 09/23/2009] [Indexed: 12/14/2022] Open
Abstract
Vascular inflammation contributes to cardiovascular diseases such as aortic aneurysm and dissection. However, the precise inflammatory pathways involved have not been clearly defined. We have shown here that subcutaneous infusion of Ang II, a vasopressor known to promote vascular inflammation, into older C57BL/6J mice induced aortic production of the proinflammatory cytokine IL-6 and the monocyte chemoattractant MCP-1. Production of these factors occurred predominantly in the tunica adventitia, along with macrophage recruitment, adventitial expansion, and development of thoracic and suprarenal aortic dissections. In contrast, a reduced incidence of dissections was observed after Ang II infusion into mice lacking either IL-6 or the MCP-1 receptor CCR2. Further analysis revealed that Ang II induced CCR2+CD14hiCD11bhiF4/80- macrophage accumulation selectively in aortic dissections and not in aortas from Il6-/- mice. Adoptive transfer of Ccr2+/+ monocytes into Ccr2-/- mice resulted in selective monocyte uptake into the ascending and suprarenal aorta in regions of enhanced ROS stress, with restoration of IL-6 secretion and increased incidence of dissection. In vitro, coculture of monocytes and aortic adventitial fibroblasts produced MCP-1- and IL-6-enriched conditioned medium that promoted differentiation of monocytes into macrophages, induced CD14 and CD11b upregulation, and induced MCP-1 and MMP-9 expression. These results suggest that leukocyte-fibroblast interactions in the aortic adventitia potentiate IL-6 production, inducing local monocyte recruitment and activation, thereby promoting MCP-1 secretion, vascular inflammation, ECM remodeling, and aortic destabilization.
Collapse
Affiliation(s)
- Brian C Tieu
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Respiratory syncytial virus infection induces a reactive oxygen species-MSK1-phospho-Ser-276 RelA pathway required for cytokine expression. J Virol 2009; 83:10605-15. [PMID: 19706715 DOI: 10.1128/jvi.01090-09] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a human pathogen that induces airway inflammation, at least in part, by modulating gene expression programs in airway epithelial cells. The presence of RSV replication is detected by the intracellular retinoic acid-inducible gene I (RIG-I) RNA helicase that forms a productive signaling complex with the mitochondrion-anchored MAVS protein, resulting in nuclear translocation of the NF-kappaB transcription factor. Although nuclear translocation is a prerequisite for activation of the innate inflammatory response, recent studies show that separate pathways governing RelA activation are also required for target gene expression. In this study, we examine the mechanism of RelA phosphorylation and its requirement for RSV-induced gene expression. RSV infection produced a time-dependent RelA phosphorylation on serine (Ser) residues Ser-276 and Ser-536 in parallel with enhanced reactive oxygen species (ROS) stress. Inhibition of RSV-induced ROS inhibited formation of phospho-Ser-276 RelA without affecting phospho-Ser-536 RelA formation. RSV potently induced activation of cytoplasmic mitogen- and stress-related kinase 1 (MSK1) in an ROS-dependent manner. Inhibition of MSK1 using H89 and small interfering RNA knockdown both reduced RSV-induced phospho-Ser-276 RelA formation and expression of a subset of NF-kappaB-dependent genes. Direct examination of the role of phospho-Ser-276 in target gene expression by expression of a RelA Ser-276-to-Ala site mutation in RelA(-/-) mouse embryonic fibroblasts showed that the mutation was unable to mediate RSV-induced NF-kappaB-dependent gene expression. We conclude that RSV induces RelA activation in the innate inflammatory response via a pathway separate from that controlling RelA cytoplasmic release, mediated by ROS signaling to cytoplasmic MSK1 activation and RelA Ser-276 phosphorylation.
Collapse
|
50
|
Olivares-Reyes JA, Arellano-Plancarte A, Castillo-Hernandez JR. Angiotensin II and the development of insulin resistance: implications for diabetes. Mol Cell Endocrinol 2009; 302:128-39. [PMID: 19150387 DOI: 10.1016/j.mce.2008.12.011] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 12/12/2008] [Accepted: 12/13/2008] [Indexed: 12/31/2022]
Abstract
Angiotensin II (Ang II), the major effector hormone of the renin-angiotensin system (RAS), has an important role in the regulation of vascular and renal homeostasis. Clinical and pharmacological studies have recently shown that Ang II is a critical promoter of insulin resistance and diabetes mellitus type 2. Ang II exerts its actions on insulin-sensitive tissues such as liver, muscle and adipose tissue where it has effects on the insulin receptor (IR), insulin receptor substrate (IRS) proteins and the downstream effectors PI3K, Akt and GLUT4. The molecular mechanisms involved have not been completely identified, but the role of serine/threonine phosphorylation of the IR and IRS-1 proteins in desensitization of insulin action has been well established. The purpose of this review is to highlight recent advances in the understanding of Ang II actions which lead to the development of insulin resistance and its implications for diabetes.
Collapse
Affiliation(s)
- J Alberto Olivares-Reyes
- Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico, DF, Mexico.
| | | | | |
Collapse
|