1
|
Allam A, Ali AA, Abdel Baky NA, Balah A. Omeprazole induces profibrotic gene expression in rat kidney: implication of TGF-β/Smad signaling pathway. Drug Chem Toxicol 2024; 47:748-755. [PMID: 37982208 DOI: 10.1080/01480545.2023.2282377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/11/2023] [Accepted: 10/31/2023] [Indexed: 11/21/2023]
Abstract
Proton pump inhibitors (PPIs) are one of the most commonly prescribed medications. However, PPI usage is linked to a higher risk of both acute and chronic renal damage by mechanisms not entirely known. The present study demonstrates that omeprazole (10 mg/kg body weight, i.p.) causes TGF-β/Smad signaling activation and subsequent expression of the profibrotic genes CTGF and TIMP-1 in rat kidney. Increased production of CTGF and TIMP-1 accompany activation of the TGF-β/Smad signaling cascade. However, simultaneous treatment of omeprazole and the TGF-β inhibitor, disitertide (P144) (1 mg/kg body weight i.p.) suppresses the TGF-β/Smad signaling pathway and subsequent production of CTGF and TIMP-1. Additionally, TGF-β level in rat kidney was highly reduced in animals treated with the ROS (reactive oxygen species) scavenger, N-acetyl cysteine (NAC) (100 mg/kg body weight i.p.) before omeprazole administration. Furthermore, the reduction in SOD activity brought by omeprazole was returned to the normal level in those animals. However, MDA level increased by omeprazole was highly reduced in the presence of NAC. Collectively, the current findings demonstrate that omeprazole has the ability to promote the expression of the profibrotic genes CTGF and TIMP-1 in a ROS and TGF-β dependent manner. The present study suggests the co-use of ROS scavenger to improve the therapeutic use of the PPI omeprazole.
Collapse
Affiliation(s)
- Albatoul Allam
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Azza A Ali
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Naira A Abdel Baky
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Amany Balah
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
2
|
Mounieb F, Abdel-Sattar SA, Balah A, Akool ES. P2 X 7 receptor is a critical regulator of extracellular ATP-induced profibrotic genes expression in rat kidney: implication of transforming growth factor-β/Smad signaling pathway. Purinergic Signal 2024; 20:421-430. [PMID: 37934321 PMCID: PMC11303607 DOI: 10.1007/s11302-023-09977-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023] Open
Abstract
This study was designed to investigate the potential of extracellular adenosine 5'-triphosphate (ATP) via the P2 X 7 receptor to activate the renal fibrotic processes in rats. The present study demonstrates that administration of ATP rapidly activated transforming growth factor-β (TGF-β) to induce phosphorylation of Smad-2/3. Renal connective tissue growth factor (CTGF) and tissue inhibitor of metalloproteinase-1 (TIMP-1) mRNA and protein expressions were also increased following ATP administration. A decrease in TGF-β amount in serum as well as renal Smad-2/3 phosphorylation was noticed in animals pre-treated with the specific antagonist of P2 X 7 receptor, A 438,079. In addition, a significant reduction in mRNA and protein expression of CTGF and TIMP-1were also observed in the kidneys of those animals. Collectively, the current findings demonstrate that ATP has the ability to augment TGF-β-mediated Smad-2/3 phosphorylation and enhance the expression of the pro-fibrotic genes, CTGF and TIMP-1, an effect that is largely mediated via P2 X 7 receptor.
Collapse
Affiliation(s)
- Fatma Mounieb
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, 11751 El Nasr St, Nasr City, Cairo, Egypt
| | - Somaia A Abdel-Sattar
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, 11751 El Nasr St, Nasr City, Cairo, Egypt
| | - Amany Balah
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, 11751 El Nasr St, Nasr City, Cairo, Egypt.
| | - El-Sayed Akool
- Pharmacology and Toxicology Department, Faculty of Pharmacy (boys), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
3
|
Li A, Wu S, Li Q, Wang Q, Chen Y. Elucidating the Molecular Pathways and Therapeutic Interventions of Gaseous Mediators in the Context of Fibrosis. Antioxidants (Basel) 2024; 13:515. [PMID: 38790620 PMCID: PMC11117599 DOI: 10.3390/antiox13050515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/13/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Fibrosis, a pathological alteration of the repair response, involves continuous organ damage, scar formation, and eventual functional failure in various chronic inflammatory disorders. Unfortunately, clinical practice offers limited treatment strategies, leading to high mortality rates in chronic diseases. As part of investigations into gaseous mediators, or gasotransmitters, including nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), numerous studies have confirmed their beneficial roles in attenuating fibrosis. Their therapeutic mechanisms, which involve inhibiting oxidative stress, inflammation, apoptosis, and proliferation, have been increasingly elucidated. Additionally, novel gasotransmitters like hydrogen (H2) and sulfur dioxide (SO2) have emerged as promising options for fibrosis treatment. In this review, we primarily demonstrate and summarize the protective and therapeutic effects of gaseous mediators in the process of fibrosis, with a focus on elucidating the underlying molecular mechanisms involved in combating fibrosis.
Collapse
Affiliation(s)
- Aohan Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian 116622, China; (A.L.); (S.W.); (Q.L.)
| | - Siyuan Wu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian 116622, China; (A.L.); (S.W.); (Q.L.)
| | - Qian Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian 116622, China; (A.L.); (S.W.); (Q.L.)
| | - Qianqian Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian 116622, China; (A.L.); (S.W.); (Q.L.)
- Engineering Technology Research Center for The Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian 116622, China
| | - Yingqing Chen
- Chronic Disease Research Center, Medical College, Dalian University, Dalian 116622, China; (A.L.); (S.W.); (Q.L.)
- Engineering Technology Research Center for The Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian 116622, China
| |
Collapse
|
4
|
West TM, Howsmon DP, Massidda MW, Vo HN, Janobas AA, Baker AB, Sacks MS. The effects of strain history on aortic valve interstitial cell activation in a 3D hydrogel environment. APL Bioeng 2023; 7:026101. [PMID: 37035541 PMCID: PMC10076067 DOI: 10.1063/5.0138030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
Aortic valves (AVs) undergo unique stretch histories that include high rates and magnitudes. While major differences in deformation patterns have been observed between normal and congenitally defective bicuspid aortic valves (BAVs), the relation to underlying mechanisms of rapid disease onset in BAV patients remains unknown. To evaluate how the variations in stretch history affect AV interstitial cell (AVIC) activation, high-throughput methods were developed to impart varied cyclical biaxial stretch histories into 3D poly(ethylene) glycol hydrogels seeded with AVICs for 48 h. Specifically, a physiologically mimicking stretch history was compared to two stretch histories with varied peak stretch and stretch rate. Post-conditioned AVICs were imaged for nuclear shape, alpha smooth muscle actin (αSMA) and vimentin (VMN) polymerization, and small mothers against decapentaplegic homologs 2 and 3 (SMAD 2/3) nuclear activity. The results indicated that bulk gel deformations were accurately transduced to the AVICs. Lower peak stretches lead to increased αSMA polymerization. In contrast, VMN polymerization was a function of stretch rate, with SMAD 2/3 nuclear localization and nuclear shape also trending toward stretch rate dependency. Lower than physiological levels of stretch rate led to higher SMAD 2/3 activity, higher VMN polymerization around the nucleus, and lower nuclear elongation. αSMA polymerization did not correlate with VMN polymerization, SMAD 2/3 activity, nor nuclear shape. These results suggest that a negative feedback loop may form between SMAD 2/3, VMN, and nuclear shape to maintain AVIC homeostatic nuclear deformations, which is dependent on stretch rate. These novel results suggest that AVIC mechanobiological responses are sensitive to stretch history and provide insight into the mechanisms of AV disease.
Collapse
Affiliation(s)
- Toni M. West
- James T. Willerson Center for Cardiovascular Modelling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, Austin, Texas 78711, USA
| | - Daniel P. Howsmon
- James T. Willerson Center for Cardiovascular Modelling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, Austin, Texas 78711, USA
| | - Miles W. Massidda
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78711, USA
| | | | | | - Aaron B. Baker
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78711, USA
| | - Michael S. Sacks
- James T. Willerson Center for Cardiovascular Modelling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, Austin, Texas 78711, USA
| |
Collapse
|
5
|
Rodkin S, Nwosu C, Sannikov A, Tyurin A, Chulkov VS, Raevskaya M, Ermakov A, Kirichenko E, Gasanov M. The Role of Gasotransmitter-Dependent Signaling Mechanisms in Apoptotic Cell Death in Cardiovascular, Rheumatic, Kidney, and Neurodegenerative Diseases and Mental Disorders. Int J Mol Sci 2023; 24:ijms24076014. [PMID: 37046987 PMCID: PMC10094524 DOI: 10.3390/ijms24076014] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 04/14/2023] Open
Abstract
Cardiovascular, rheumatic, kidney, and neurodegenerative diseases and mental disorders are a common cause of deterioration in the quality of life up to severe disability and death worldwide. Many pathological conditions, including this group of diseases, are based on increased cell death through apoptosis. It is known that this process is associated with signaling pathways controlled by a group of gaseous signaling molecules called gasotransmitters. They are unique messengers that can control the process of apoptosis at different stages of its implementation. However, their role in the regulation of apoptotic signaling in these pathological conditions is often controversial and not completely clear. This review analyzes the role of nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H2S), and sulfur dioxide (SO2) in apoptotic cell death in cardiovascular, rheumatic, kidney, and neurodegenerative diseases. The signaling processes involved in apoptosis in schizophrenia, bipolar, depressive, and anxiety disorders are also considered. The role of gasotransmitters in apoptosis in these diseases is largely determined by cell specificity and concentration. NO has the greatest dualism; scales are more prone to apoptosis. At the same time, CO, H2S, and SO2 are more involved in cytoprotective processes.
Collapse
Affiliation(s)
- Stanislav Rodkin
- Faculty of Bioengineering and Veterinary Medicine, Department of Bioengineering, Don State Technical University, Rostov-on-Don 344000, Russia
| | - Chizaram Nwosu
- Faculty of Bioengineering and Veterinary Medicine, Department of Bioengineering, Don State Technical University, Rostov-on-Don 344000, Russia
| | - Alexander Sannikov
- Department of Psychiatry, Rostov State Medical University, Rostov-on-Don 344022, Russia
| | - Anton Tyurin
- Internal Medicine Department, Bashkir State Medical University, Ufa 450008, Russia
| | | | - Margarita Raevskaya
- Faculty of Bioengineering and Veterinary Medicine, Department of Bioengineering, Don State Technical University, Rostov-on-Don 344000, Russia
| | - Alexey Ermakov
- Faculty of Bioengineering and Veterinary Medicine, Department of Bioengineering, Don State Technical University, Rostov-on-Don 344000, Russia
| | - Evgeniya Kirichenko
- Faculty of Bioengineering and Veterinary Medicine, Department of Bioengineering, Don State Technical University, Rostov-on-Don 344000, Russia
| | - Mitkhat Gasanov
- Department of Internal Diseases #1, Rostov State Medical University, Rostov-on-Don 344022, Russia
| |
Collapse
|
6
|
LaRue MM, Parker S, Puccini J, Cammer M, Kimmelman AC, Bar-Sagi D. Metabolic reprogramming of tumor-associated macrophages by collagen turnover promotes fibrosis in pancreatic cancer. Proc Natl Acad Sci U S A 2022; 119:e2119168119. [PMID: 35412885 PMCID: PMC9169723 DOI: 10.1073/pnas.2119168119] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/25/2022] [Indexed: 12/27/2022] Open
Abstract
A hallmark of pancreatic tumors is their highly desmoplastic stroma composed of fibroblasts, immune cells, and a dense network of collagen fibers. Tumor-associated macrophages are one of the most abundant immune cell populations in the pancreatic tumor stroma. Their protumorigenic function has been attributed predominantly to their capacity to promote immune evasion and metastasis. Tumor-assoc iated macrophages are also well known for their role in the remodeling of the stroma via collagen production and degradation, with the latter being mediated by mannose receptor (MRC1)-dependent endocytosis of collagen. Here we show that MRC1-mediated collagen internalization and subsequent lysosomal degradation by macrophages harboring a tumor-associated phenotype are accompanied by the accumulation of collagen-derived intracellular free amino acids and increased arginine biosynthesis. The resulting increase in intracellular arginine levels leads to the up-regulation of inducible nitric oxide synthase and the production of reactive nitrogen species. Furthermore, reactive nitrogen species derived from internalized and degraded collagen promotes a profibrotic phenotype in pancreatic stellate cells resulting in enhanced intratumoral collagen deposition. Overall, our findings identify a role for extracellular matrix remodeling in the functional modulation of tumor-associated macrophages via metabolic rewiring.
Collapse
Affiliation(s)
- Madeleine M. LaRue
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016
| | - Seth Parker
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016
| | - Joseph Puccini
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016
| | - Michael Cammer
- Microscopy Core, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY 10016
| | - Alec C. Kimmelman
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016
- Department of Radiation Oncology, New York University Medical Center, New York, NY 10016
| | - Dafna Bar-Sagi
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
7
|
Matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases in kidney disease. Adv Clin Chem 2021; 105:141-212. [PMID: 34809827 DOI: 10.1016/bs.acc.2021.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Matrix metalloproteinases (MMPs) are a group of zinc and calcium endopeptidases which cleave extracellular matrix (ECM) proteins. They are also involved in the degradation of cell surface components and regulate multiple cellular processes, cell to cell interactions, cell proliferation, and cell signaling pathways. MMPs function in close interaction with the endogenous tissue inhibitors of matrix metalloproteinases (TIMPs), both of which regulate cell turnover, modulate various growth factors, and participate in the progression of tissue fibrosis and apoptosis. The multiple roles of MMPs and TIMPs are continuously elucidated in kidney development and repair, as well as in a number of kidney diseases. This chapter focuses on the current findings of the significance of MMPs and TIMPs in a wide range of kidney diseases, whether they result from kidney tissue changes, hemodynamic alterations, tubular epithelial cell apoptosis, inflammation, or fibrosis. In addition, the potential use of these endopeptidases as biomarkers of renal dysfunction and as targets for therapeutic interventions to attenuate kidney disease are also explored in this review.
Collapse
|
8
|
Gasotransmitters: Potential Therapeutic Molecules of Fibrotic Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3206982. [PMID: 34594474 PMCID: PMC8478550 DOI: 10.1155/2021/3206982] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/31/2021] [Indexed: 02/06/2023]
Abstract
Fibrosis is defined as the pathological progress of excessive extracellular matrix (ECM), such as collagen, fibronectin, and elastin deposition, as the regenerative capacity of cells cannot satisfy the dynamic repair of chronic damage. The well-known features of tissue fibrosis are characterized as the presence of excessive activated and proliferated fibroblasts and the differentiation of fibroblasts into myofibroblasts, and epithelial cells undergo the epithelial-mesenchymal transition (EMT) to expand the number of fibroblasts and myofibroblasts thereby driving fibrogenesis. In terms of mechanism, during the process of fibrosis, the activations of the TGF-β signaling pathway, oxidative stress, cellular senescence, and inflammatory response play crucial roles in the activation and proliferation of fibroblasts to generate ECM. The deaths due to severe fibrosis account for almost half of the total deaths from various diseases, and few treatment strategies are available for the prevention of fibrosis as yet. Recently, numerous studies demonstrated that three well-defined bioactive gasotransmitters, including nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), generally exhibited anti-inflammatory, antioxidative, antiapoptotic, and antiproliferative properties. Besides these effects, a number of studies have reported that low-dose exogenous and endogenous gasotransmitters can delay and interfere with the occurrence and development of fibrotic diseases, including myocardial fibrosis, idiopathic pulmonary fibrosis, liver fibrosis, renal fibrosis, diabetic diaphragm fibrosis, and peritoneal fibrosis. Furthermore, in animal and clinical experiments, the inhalation of low-dose exogenous gas and intraperitoneal injection of gaseous donors, such as SNAP, CINOD, CORM, SAC, and NaHS, showed a significant therapeutic effect on the inhibition of fibrosis through modulating the TGF-β signaling pathway, attenuating oxidative stress and inflammatory response, and delaying the cellular senescence, while promoting the process of autophagy. In this review, we first demonstrate and summarize the therapeutic effects of gasotransmitters on diverse fibrotic diseases and highlight their molecular mechanisms in the process and development of fibrosis.
Collapse
|
9
|
Yang H, Zhou T, Sorenson CM, Sheibani N, Liu B. Myeloid-Derived TSP1 (Thrombospondin-1) Contributes to Abdominal Aortic Aneurysm Through Suppressing Tissue Inhibitor of Metalloproteinases-1. Arterioscler Thromb Vasc Biol 2020; 40:e350-e366. [PMID: 33028100 PMCID: PMC7686278 DOI: 10.1161/atvbaha.120.314913] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm is characterized by the progressive loss of aortic integrity and accumulation of inflammatory cells primarily macrophages. We previously reported that global deletion of matricellular protein TSP1 (thrombospondin-1) protects mice from aneurysm formation. The objective of the current study is to investigate the cellular and molecular mechanisms underlying TSP1's action in aneurysm. Approach and Results: Using RNA fluorescent in situ hybridization, we identified macrophages being the major source of TSP1 in human and mouse aneurysmal tissues, accounting for over 70% of cells that actively expressed Thbs1 mRNA. Lack of TSP1 in macrophages decreased solution-based gelatinase activities by elevating TIMP1 (tissue inhibitor of metalloproteinases-1) without affecting the major MMPs (matrix metalloproteinases). Knocking down Timp1 restored the ability of Thbs1-/- macrophages to invade matrix. Finally, we generated Thbs1flox/flox mice and crossed them with Lyz2-cre mice. In the CaCl2-induced model of abdominal aortic aneurysm, lacking TSP1 in myeloid cells was sufficient to protect mice from aneurysm by reducing macrophage accumulation and preserving aortic integrity. CONCLUSIONS TSP1 contributes to aneurysm pathogenesis, at least in part, by suppressing TIMP1 expression, which subsequently enables inflammatory macrophages to infiltrate vascular tissues.
Collapse
MESH Headings
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Cells, Cultured
- Dilatation, Pathologic
- Disease Models, Animal
- Down-Regulation
- Humans
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Matrix Metalloproteinases/metabolism
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Signal Transduction
- Thrombospondin 1/deficiency
- Thrombospondin 1/genetics
- Thrombospondin 1/metabolism
- Tissue Inhibitor of Metalloproteinase-1/genetics
- Tissue Inhibitor of Metalloproteinase-1/metabolism
Collapse
Affiliation(s)
- Huan Yang
- Department of Surgery,School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705
| | - Ting Zhou
- Department of Surgery,School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705
| | - Christine M. Sorenson
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53705
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53705
| | - Bo Liu
- Department of Surgery,School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705
- Department of Cellular and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
10
|
Vitamin E inhibits cyclosporin A-induced CTGF and TIMP-1 expression by repressing ROS-mediated activation of TGF-β/Smad signaling pathway in rat liver. Int Immunopharmacol 2018; 65:493-502. [DOI: 10.1016/j.intimp.2018.09.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 08/31/2018] [Accepted: 09/22/2018] [Indexed: 01/11/2023]
|
11
|
Grünwald B, Harant V, Schaten S, Frühschütz M, Spallek R, Höchst B, Stutzer K, Berchtold S, Erkan M, Prokopchuk O, Martignoni M, Esposito I, Heikenwalder M, Gupta A, Siveke J, Saftig P, Knolle P, Wohlleber D, Krüger A. Pancreatic Premalignant Lesions Secrete Tissue Inhibitor of Metalloproteinases-1, Which Activates Hepatic Stellate Cells Via CD63 Signaling to Create a Premetastatic Niche in the Liver. Gastroenterology 2016; 151:1011-1024.e7. [PMID: 27506299 DOI: 10.1053/j.gastro.2016.07.043] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 07/04/2016] [Accepted: 07/25/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDAC) metastasizes to liver at early stages, making this disease highly lethal. Tissue inhibitor of metalloproteinases-1 (TIMP1) creates a metastasis-susceptible environment in the liver. We investigated the role of TIMP1 and its receptor CD63 in metastasis of early-stage pancreatic tumors using mice and human cell lines and tissue samples. METHODS We obtained liver and plasma samples from patients in Germany with chronic pancreatitis, pancreatic intra-epithelial neoplasia, or PDAC, as well as hepatic stellate cells (HSCs). We performed studies with Ptf1a+/Cre;Kras+/LSL-G12D;Trp53loxP/loxP (CPK) mice, Pdx-1+/Cre;Kras+/LSL-G12D;Trp53+/LSL-R172H (KPC) mice, and their respective healthy littermates as control, and Cd63-/- mice with their wild-type littermates. KPC mice were bred with Timp1-/- mice to produce KPCxTimp1-/- mice. TIMP1 was overexpressed and CD63 was knocked down in mice using adenoviral vectors AdTIMP1 or AdshCD63, respectively. Hepatic susceptibility to metastases was determined after intravenous inoculation of syngeneic 9801L pancreas carcinoma cells. Pancreata and liver tissues were collected and analyzed by histology, immunohistochemical, immunoblot, enzyme-linked immunosorbent assay, and quantitative polymerase chain reaction analyses. We analyzed the effects of TIMP1 overexpression or knockdown and CD63 knockdown in transduced human primary HSCs and HSC cell lines. RESULTS Chronic pancreatitis, pancreatic intra-epithelial neoplasia, and PDAC tissues from patients expressed higher levels of TIMP1 protein than normal pancreas. The premalignant pancreatic lesions that developed in KPC and CPK mice expressed TIMP1 and secreted it into the circulation. In vitro and in vivo, TIMP1 activated human or mouse HSCs, which required interaction between TIMP1 and CD63 and signaling via phosphatidylinositol 3-kinase, but not TIMP1 protease inhibitor activity. This signaling pathway induced expression of endogenous TIMP1. TIMP1 knockdown in HSCs reduced their activation. Cultured TIMP1-activated human and mouse HSCs began to express stromal-derived factor-1, which induced neutrophil migration, a marker of the premetastatic niche. Mice with pancreatic intra-epithelial neoplasia-derived systemic increases in TIMP1 developed more liver metastases after injections of pancreatic cancer cells than mice without increased levels of TIMP1. This increase in formation of liver metastases from injected pancreatic cancer cells was not observed in TIMP1 or CD63 knockout mice. CONCLUSIONS Expression of TIMP1 is increased in chronic pancreatitis, pancreatic intra-epithelial neoplasia, and PDAC tissues from patients. TIMP1 signaling via CD63 leads to activation of HSCs, which create an environment in the liver that increases its susceptibility to pancreatic tumor cells. Strategies to block TIMP1 signaling via CD63 might be developed to prevent PDAC metastasis to the liver.
Collapse
Affiliation(s)
- Barbara Grünwald
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany
| | - Veronika Harant
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany
| | - Susanne Schaten
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany
| | - Monika Frühschütz
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany
| | - Ria Spallek
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany
| | - Bastian Höchst
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany
| | - Katharina Stutzer
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany
| | - Sonja Berchtold
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany
| | - Mert Erkan
- Chirurgische Klinik Technische Universität München, München, Germany
| | - Olga Prokopchuk
- Chirurgische Klinik Technische Universität München, München, Germany
| | - Marc Martignoni
- Chirurgische Klinik Technische Universität München, München, Germany
| | - Irene Esposito
- Institut für Pathologie, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | | | - Aayush Gupta
- II. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Jens Siveke
- II. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Percy Knolle
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany
| | - Dirk Wohlleber
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany
| | - Achim Krüger
- Institut für Molekulare Immunologie und Experimentelle Onkologie, Technische Universität München, München, Germany.
| |
Collapse
|
12
|
Imeri F, Fallegger D, Zivkovic A, Schwalm S, Enzmann G, Blankenbach K, Meyer zu Heringdorf D, Homann T, Kleuser B, Pfeilschifter J, Engelhardt B, Stark H, Huwiler A. Novel oxazolo-oxazole derivatives of FTY720 reduce endothelial cell permeability, immune cell chemotaxis and symptoms of experimental autoimmune encephalomyelitis in mice. Neuropharmacology 2014; 85:314-27. [PMID: 24863045 DOI: 10.1016/j.neuropharm.2014.05.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 04/15/2014] [Accepted: 05/13/2014] [Indexed: 12/29/2022]
Abstract
The immunomodulatory FTY720 (fingolimod) is presently approved for the treatment of relapsing-remitting multiple sclerosis. It is a prodrug that acts by modulating sphingosine 1-phosphate (S1P) receptor signaling. In this study, we have developed and characterized two novel oxazolo-oxazole derivatives of FTY720, ST-968 and the oxy analog ST-1071, which require no preceding activating phosphorylation, and proved to be active in intact cells and triggered S1P1 and S1P3, but not S1P2, receptor internalization as a result of receptor activation. Functionally, ST-968 and ST-1071 acted similar to FTY720 to abrogate S1P-triggered chemotaxis of mouse splenocytes, mouse T cells and human U937 cells, and reduced TNFa- and LPS-stimulated endothelial cell permeability. The compounds also reduced TNFα-induced ICAM-1 and VCAM-1 mRNA expression, but restored TNFα-mediated downregulation of PECAM-1 mRNA expression. In an in vivo setting, the application of ST-968 or ST-1071 to mice resulted in a reduction of blood lymphocytes and significantly reduced the clinical symptoms of experimental autoimmune encephalomyelitis (EAE) in C57BL/6 mice comparable to FTY720 either by prophylactic or therapeutic treatment. In parallel to the reduced clinical symptoms, infiltration of immune cells in the brain was strongly reduced, and in isolated tissues of brain and spinal cord, the mRNA and protein expressions of ICAM-1 and VCAM-1, as well as of matrix metalloproteinase-9 were reduced by all compounds, whereas PECAM-1 and tissue inhibitor of metalloproteinase TIMP-1 were upregulated. In summary, the data suggest that these novel butterfly derivatives of FTY720 could have considerable implication for future therapies of multiple sclerosis and other autoimmune diseases.
Collapse
Affiliation(s)
- Faik Imeri
- Institute of Pharmacology, University of Bern, Friedbühlstrasse 49, CH-3010 Bern, Switzerland
| | - Daniel Fallegger
- Institute of Pharmacology, University of Bern, Friedbühlstrasse 49, CH-3010 Bern, Switzerland
| | - Aleksandra Zivkovic
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, Germany; Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, D-40225 Düsseldorf, Germany
| | - Stephanie Schwalm
- Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Gaby Enzmann
- Theodor-Kocher Institute, University of Bern, Freiestrasse 1, CH-3012 Bern, Switzerland
| | - Kira Blankenbach
- Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Dagmar Meyer zu Heringdorf
- Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Thomas Homann
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert Allee 114-116, D-14558 Nuthetal, Germany
| | - Burkhard Kleuser
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert Allee 114-116, D-14558 Nuthetal, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Britta Engelhardt
- Theodor-Kocher Institute, University of Bern, Freiestrasse 1, CH-3012 Bern, Switzerland
| | - Holger Stark
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, Germany; Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, D-40225 Düsseldorf, Germany
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Friedbühlstrasse 49, CH-3010 Bern, Switzerland; Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany.
| |
Collapse
|
13
|
Uchida C, Haas TL. Endothelial cell TIMP-1 is upregulated by shear stress via Sp-1 and the TGFβ1 signaling pathways. Biochem Cell Biol 2013; 92:77-83. [PMID: 24471921 DOI: 10.1139/bcb-2013-0086] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Laminar shear stress promotes vascular integrity by inhibiting proteolysis of the extracellular matrix (ECM) surrounding the microvasculature. We hypothesized that the matrix metalloproteinase inhibitor TIMP-1 would be upregulated in endothelial cells exposed to shear stress. Microvascular endothelial cells isolated from rat or mouse skeletal muscles were exposed to laminar shear stress for 2, 4, or 24 h. A biphasic increase in TIMP-1 protein was observed at 2 and 24 h of shear stress exposure. Sp-1 siRNA prevented the increase in TIMP-1 after 2, but not 24, hours of shear exposure. TGFβ production and Smad2/3 phosphorylation are increased by shear stress. Inhibition of TGFβ signaling, either by use of the TGFβ receptor 1 inhibitor SB-431542 or with Smad 2/3 siRNA, abrogated the shear stress-induced increase in TIMP-1 mRNA after 24 h of shear stress exposure. These results suggest that both acute and chronic elevated laminar shear stress act to maintain vessel integrity through increasing TIMP-1 production, but that the TGFβ signaling pathway is essential to maintain TIMP-1 expression during chronic shear stress.
Collapse
Affiliation(s)
- Cassandra Uchida
- Angiogenesis Research Group, Faculty of Health, York University, 4700 Keele St., Toronto, ON M3J 1P3, Canada
| | | |
Collapse
|
14
|
Nitric oxide synthase-3 promotes embryonic development of atrioventricular valves. PLoS One 2013; 8:e77611. [PMID: 24204893 PMCID: PMC3812218 DOI: 10.1371/journal.pone.0077611] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 09/11/2013] [Indexed: 11/19/2022] Open
Abstract
Nitric oxide synthase-3 (NOS3) has recently been shown to promote endothelial-to-mesenchymal transition (EndMT) in the developing atrioventricular (AV) canal. The present study was aimed to investigate the role of NOS3 in embryonic development of AV valves. We hypothesized that NOS3 promotes embryonic development of AV valves via EndMT. To test this hypothesis, morphological and functional analysis of AV valves were performed in wild-type (WT) and NOS3−/− mice at postnatal day 0. Our data show that the overall size and length of mitral and tricuspid valves were decreased in NOS3−/− compared with WT mice. Echocardiographic assessment showed significant regurgitation of mitral and tricuspid valves during systole in NOS3−/− mice. These phenotypes were all rescued by cardiac specific NOS3 overexpression. To assess EndMT, immunostaining of Snail1 was performed in the embryonic heart. Both total mesenchymal and Snail1+ cells in the AV cushion were decreased in NOS3−/− compared with WT mice at E10.5 and E12.5, which was completely restored by cardiac specific NOS3 overexpression. In cultured embryonic hearts, NOS3 promoted transforming growth factor (TGFβ), bone morphogenetic protein (BMP2) and Snail1expression through cGMP. Furthermore, mesenchymal cell formation and migration from cultured AV cushion explants were decreased in the NOS3−/− compared with WT mice. We conclude that NOS3 promotes AV valve formation during embryonic heart development and deficiency in NOS3 results in AV valve insufficiency.
Collapse
|
15
|
Abstract
Underlying the dynamic regulation of tropoelastin expression and elastin formation in development and disease are transcriptional and post-transcriptional mechanisms that have been the focus of much research. Of particular importance is the cytokine-governed elastin regulatory axis in which the pro-elastogenic activities of transforming growth factor β-1 (TGFβ1) and insulin-like growth factor-I (IGF-I) are opposed by anti-elastogenic activities of basic fibroblast growth factor (bFGF/FGF-2), heparin-binding epidermal growth factor-like growth factor (HB-EGF), EGF, PDGF-BB, TGFα, tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β and noncanonical TGFβ1 signaling. A key mechanistic feature of the regulatory axis is that cytokines influence elastin formation through effects on the cell cycle involving control of cyclin-cyclin dependent kinase complexes and activation of the Ras/MEK/ERK signaling pathway. In this article we provide an overview of the major cytokines/growth factors that modulate elastogenesis and describe the underlying molecular mechanisms for their action on elastin production.
Collapse
Affiliation(s)
- Erin P Sproul
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | |
Collapse
|
16
|
Wang B, Li W, Chen Y, Wang Y, Sun C, Chen Y, Lu H, Fan J, Li D. Coexpression of Smad7 and UPA attenuates carbon tetrachloride-induced rat liver fibrosis. Med Sci Monit 2013; 18:BR394-401. [PMID: 23018346 PMCID: PMC3560566 DOI: 10.12659/msm.883479] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background There is a great need for developing novel therapies to treat liver fibrosis. Previous studies showed that both Smad7 and uPA were inhibitors of liver fibrosis. Therefore, we explored the therapeutic effects of combinational gene therapy with Smad7 and uPA on CCl4-induced liver fibrosis. Material/Methods Smad7 and uPA genes were cloned into an adenovirus vector. To observe the therapeutic effects of coexpression of Smad7 and uPA genes, the recombinant adenovirus were delivered into CCL4-induced fibrosis models. Fibrillar collagen, hydroxyproline, α-SMA, TGF-β1, MMP-13, TIMP-1, HGF and PCNA were detected to evaluate the fibrosis and to explore the mechanisms underlying the treatment with Smad7 and uPA. Results The results showed that single Smad7 or uPA adenovirus reduced CCL4 induced liver fibrosis significantly; while combination of Smad7 and uPA had more significant therapeutic effect on CCl4 induced liver fibrosis. Then the markers underlying the therapeutic effect of combination of Smad7 and uPA were also explored. Over-expression of Smad7 and uPA inhibited the expression of α-SMA and TGF-β1 significantly. Combinational gene therapy also enhanced extracellular matrix degradation by increasing the expression of MMP-13, inhibiting TIMP-1 expression, and promoted hepatocyte proliferation, while single Smad7 or uPA only induced part of these changes. Conclusions These results suggest that combinational gene therapy with Smad7 and uPA inhibited CCl4-induced rat liver fibrosis by simultaneously targeting multiple pathogenic pathways.
Collapse
Affiliation(s)
- Baocan Wang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Ueberham U, Hilbrich I, Ueberham E, Rohn S, Glöckner P, Dietrich K, Brückner MK, Arendt T. Transcriptional control of cell cycle-dependent kinase 4 by Smad proteins--implications for Alzheimer's disease. Neurobiol Aging 2012; 33:2827-40. [PMID: 22418736 DOI: 10.1016/j.neurobiolaging.2012.01.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 01/23/2012] [Accepted: 01/25/2012] [Indexed: 01/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by deregulation of neuronal cell cycle and differentiation control eventually resulting in cell death. During brain development, neuronal differentiation is regulated by Smad proteins, which are elements of the canonical transforming growth factor β (TGF-β) signaling pathway, linking receptor activation to gene expression. In the normal adult brain, Smad proteins are constitutively phosphorylated and predominantly localized in neuronal nuclei. Under neurodegenerative conditions such as AD, the subcellular localization of their phosphorylated forms is heavily disturbed, raising the question of whether a nuclear Smad deficiency in neurons might contribute to a loss of neuronal differentiation control and subsequent cell cycle re-entry. Here, we show by luciferase reporter assays, electromobility shift, and RNA interference (RNAi) technique a direct binding of Smad proteins to the CDK4 promoter inducing transcriptional inhibition of cell cycle-dependent kinase 4 (Cdk4). Mimicking the neuronal deficiency of Smad proteins observed in AD in cell culture by RNAi results in elevation of Cdk4 and retardation of neurite outgrowth. The results identify Smad proteins as direct transcriptional regulators of Cdk4 and add further evidence to a Smad-dependent deregulation of Cdk4 in AD, giving rise to neuronal dedifferentiation and cell death.
Collapse
Affiliation(s)
- Uwe Ueberham
- Department for Molecular and Cellular Mechanisms of Neurodegeneration, Universität Leipzig, Paul Flechsig Institute of Brain Research, Leipzig, D-04109, Jahnallee 59, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Baghy K, Iozzo RV, Kovalszky I. Decorin-TGFβ axis in hepatic fibrosis and cirrhosis. J Histochem Cytochem 2012; 60:262-8. [PMID: 22260996 DOI: 10.1369/0022155412438104] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatic fibrosis and cirrhosis are worldwide health care problems, especially in regions with a high rate of hepatitis infection. As these diseases affect a major part of the human population, the search for antifibrotic therapies has a high priority in medical research. Transforming growth factor β1 (TGF-β1) is one of the most powerful profibrotic cytokines. Thus, blocking TGF-β1 activity by natural inhibitors represents a valid and logical strategy to combat hepatic fibrosis. One of the natural inhibitors of TGF-β1 is decorin, a small leucine-rich proteoglycan that binds with high affinity to this cytokine and prevents its interaction with pro-fibrotic receptors. Recent evidence has shown that decorin has a protective role in liver fibrogenesis insofar as its genetic ablation in mice leads to enhanced matrix deposition, impaired matrix degradation, and "activation" of hepatic stellate cells, the main producers of fibrotic tissue. Moreover, TGF-β1 exerts a stronger effect when functional decorin is absent. These data provide robust genetic evidence for a direct role of endogenous decorin in preventing and retarding hepatic fibrosis. Thus, boosting the endogenous production of decorin or systemic delivery of recombinant decorin could represent an additional therapeutic modality against hepatic fibrosis.
Collapse
Affiliation(s)
- Kornélia Baghy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | | |
Collapse
|
19
|
Huang D, Wang Y, Wang L, Zhang F, Deng S, Wang R, Zhang Y, Huang K. Poly(ADP-ribose) polymerase 1 is indispensable for transforming growth factor-β Induced Smad3 activation in vascular smooth muscle cell. PLoS One 2011; 6:e27123. [PMID: 22073128 PMCID: PMC3205050 DOI: 10.1371/journal.pone.0027123] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 10/11/2011] [Indexed: 12/31/2022] Open
Abstract
Background Transforming growth factor type-β (TGF-β)/Smad pathway plays an essential role in vascular fibrosis. Reactive oxygen species (ROS) generation also mediates TGF-β signaling-induced vascular fibrosis, suggesting that some sort of interaction exists between Smad and redox pathways. However, the underlying molecular mechanism is largely unknown. This study aims to investigate the influence of poly(ADP-ribose) polymerase 1 (PARP1), a downstream effector of ROS, on TGF-β signaling transduction through Smad3 pathway in rat vascular smooth muscle cells (VSMCs). Methods and Results TGF-β1 treatment promoted PARP1 activation through induction of ROS generation in rat VSMCs. TGF-β1-induced phosphorylation and nuclear accumulation of Smad3 was prevented by treatment of cells with PARP inhibitor, 3-aminobenzamide (3AB) or N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-2-(N,N-dimethylamino)acetami (PJ34), or PARP1 siRNA. TGF-β1 treatment promoted poly(ADP-ribosy)lation of Smad3 via activation of PARP1 in the nucleus. Poly(ADP-ribosy)lation enhanced Smad-Smad binding element (SBE) complex formation in nuclear extracts and increased DNA binding activity of Smad3. Pretreatment with 3AB, PJ34, or PARP1 siRNA prevented TGF-β1-induced Smad3 transactivation and expression of Smad3 target genes, including collagen Iα1, collagen IIIα1 and tissue inhibitor of metalloproteinase 1, in rat VSMCs. Conclusions PARP1 is indispensable for TGF-β1 induced Smad3 activation in rat VSMCs. Targeting PARP1 may be a promising therapeutic approach against vascular diseases induced by dysregulation of TGF-β/Smad3 pathway.
Collapse
MESH Headings
- Animals
- Benzamides/pharmacology
- Blotting, Southwestern
- Blotting, Western
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cells, Cultured
- Electrophoretic Mobility Shift Assay
- Enzyme Inhibitors/pharmacology
- Enzyme-Linked Immunosorbent Assay
- Fluorescent Antibody Technique
- Immunoenzyme Techniques
- Immunoprecipitation
- Luciferases/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Phenanthrenes/pharmacology
- Phosphorylation/drug effects
- Poly(ADP-ribose) Polymerase Inhibitors
- Poly(ADP-ribose) Polymerases/genetics
- Poly(ADP-ribose) Polymerases/metabolism
- Promoter Regions, Genetic
- Protein Binding
- RNA, Messenger/genetics
- RNA, Small Interfering/pharmacology
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Real-Time Polymerase Chain Reaction
- Signal Transduction/drug effects
- Smad3 Protein/genetics
- Smad3 Protein/metabolism
- Trans-Activators
- Transcription, Genetic
- Transfection
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Dan Huang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Wang
- Central Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengxiao Zhang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Deng
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Wang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
- * E-mail: (KH); (YZ)
| | - Kai Huang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
- * E-mail: (KH); (YZ)
| |
Collapse
|
20
|
Alvira CM, Guignabert C, Kim YM, Chen C, Wang L, Duong TT, Yeung RSM, Li DY, Rabinovitch M. Inhibition of transforming growth factor β worsens elastin degradation in a murine model of Kawasaki disease. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1210-20. [PMID: 21356372 DOI: 10.1016/j.ajpath.2010.11.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Revised: 11/03/2010] [Accepted: 11/09/2010] [Indexed: 12/13/2022]
Abstract
Kawasaki disease (KD) is an acute inflammatory illness marked by coronary arteritis. However, the factors increasing susceptibility to coronary artery lesions are unknown. Because transforming growth factor (TGF) β increases elastin synthesis and suppresses proteolysis, we hypothesized that, in contrast to the benefit observed in aneurysms forming in those with Marfan syndrome, inhibition of TGF-β would worsen inflammatory-induced coronary artery lesions. By using a murine model of KD in which injection of Lactobacillus casei wall extract (LCWE) induces coronary arteritis, we show that LCWE increased TGF-β signaling in the coronary smooth muscle cells beginning at 2 days and continuing through 14 days, the point of peak coronary inflammation. By 42 days, LCWE caused fragmentation of the internal and external elastic lamina. Blocking TGF-β by administration of a neutralizing antibody accentuated the LCWE-mediated fragmentation of elastin and induced an overall loss of medial elastin without increasing the inflammatory response. We attributed these increased pathological characteristics to a reduction in the proteolytic inhibitor, plasminogen activator inhibitor-1, and an associated threefold increase in matrix metalloproteinase 9 activity compared with LCWE alone. Therefore, our data demonstrate that in the coronary arteritis associated with KD, TGF-β suppresses elastin degradation by inhibiting plasmin-mediated matrix metalloproteinase 9 activation. Thus, strategies to block TGF-β, used in those with Marfan syndrome, are unlikely to be beneficial and could be detrimental.
Collapse
Affiliation(s)
- Cristina M Alvira
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305-5162, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Schulman IH, Hare JM. Regulation of cardiovascular cellular processes by S-nitrosylation. Biochim Biophys Acta Gen Subj 2011; 1820:752-62. [PMID: 21536106 DOI: 10.1016/j.bbagen.2011.04.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Accepted: 04/07/2011] [Indexed: 12/27/2022]
Abstract
BACKGROUND Nitric oxide (NO), a highly versatile signaling molecule, exerts a broad range of regulatory influences in the cardiovascular system that extends from vasodilation to myocardial contractility, angiogenesis, inflammation, and energy metabolism. Considerable attention has been paid to deciphering the mechanisms for such diversity in signaling. S-nitrosylation of cysteine thiols is a major signaling pathway through which NO exerts its actions. An emerging concept of NO pathophysiology is that the interplay between NO and reactive oxygen species (ROS), the nitroso/redox balance, is an important regulator of cardiovascular homeostasis. SCOPE OF REVIEW ROS react with NO, limit its bioavailability, and compete with NO for binding to the same thiol in effector molecules. The interplay between NO and ROS appears to be tightly regulated and spatially confined based on the co-localization of specific NO synthase (NOS) isoforms and oxidative enzymes in unique subcellular compartments. NOS isoforms are also in close contact with denitrosylases, leading to crucial regulation of S-nitrosylation. MAJOR CONCLUSIONS Nitroso/redox balance is an emerging regulatory pathway for multiple cells and tissues, including the cardiovascular system. Studies using relevant knockout models, isoform specific NOS inhibitors, and both in vitro and in vivo methods have provided novel insights into NO- and ROS-based signaling interactions responsible for numerous cardiovascular disorders. GENERAL SIGNIFICANCE An integrated view of the role of nitroso/redox balance in cardiovascular pathophysiology has significant therapeutic implications. This is highlighted by human studies where pharmacologic manipulation of oxidative and nitrosative pathways exerted salutary effects in patients with advanced heart failure. This article is part of a Special Issue entitled Regulation of Cellular Processes by S-nitrosylation.
Collapse
Affiliation(s)
- Ivonne Hernandez Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | | |
Collapse
|
22
|
Medina C, Santos-Martinez MJ, Santana A, Paz-Cabrera MC, Johnston MJ, Mourelle M, Salas A, Guarner F. Transforming growth factor-beta type 1 receptor (ALK5) and Smad proteins mediate TIMP-1 and collagen synthesis in experimental intestinal fibrosis. J Pathol 2011; 224:461-72. [PMID: 21465486 DOI: 10.1002/path.2870] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 12/15/2010] [Accepted: 01/31/2011] [Indexed: 12/16/2022]
Abstract
Transforming growth factor β (TGF-β) is known to play a key role in intestinal fibrosis; however, the underlying mechanisms are not well understood. TGF-β signal transduction is through TGF-β receptors, including the TGF-β type 1 receptor. Most cell types contain a TGF-β type 1 receptor form known as activin receptor-like kinase 5 (ALK5), which propagates the signal to the nucleus through the phosphorylation of Smad2 and Smad3 proteins. Therefore, we assessed the effect of the disruption of TGF-β/ALK5/Smad signalling by an ALK5 inhibitor (SD-208) in two experimental animal models of intestinal fibrosis: anaerobic bacteria- and trinitrobenzensulphonic acid-induced colitis. In addition, isolated myofibroblasts were pretreated with SD-208 and exposed to recombinant TGF-β1. Finally, myofibroblasts were transfected with ALK5, Smad2, and Smad3-specific siRNA. Up-regulation of ALK5 and TIMP-1, phosphorylation of Smad2 and Smad3 proteins, and increased intestinal wall collagen deposition were found in both experimental animal models. These effects were decreased by SD-208. TGF-β1 treatment also induced phosphorylation of Smad2 and Smad3 and up-regulation of ALK5 protein, TIMP-1, and α2 type 1 collagen gene expression in isolated myofibroblasts. Again these effects were inhibited by SD-208. Also, ALK5, Smad2, and Smad3 siRNA abolished the induction of TIMP-1 and α2 type 1 collagen. Our findings provide evidence that the TGF-β/ALK5/Smad pathway participates in the pathogenesis of experimental intestinal fibrosis. These data show promise for the development of an effective therapeutic intervention in this condition.
Collapse
Affiliation(s)
- Carlos Medina
- Department of Pharmacology, Trinity College Dublin, Dublin 2, Ireland.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Baghy K, Dezső K, László V, Fullár A, Péterfia B, Paku S, Nagy P, Schaff Z, Iozzo RV, Kovalszky I. Ablation of the decorin gene enhances experimental hepatic fibrosis and impairs hepatic healing in mice. J Transl Med 2011; 91:439-51. [PMID: 20956977 PMCID: PMC5074558 DOI: 10.1038/labinvest.2010.172] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Accumulation of connective tissue is a typical feature of chronic liver diseases. Decorin, a small leucine-rich proteoglycan, regulates collagen fibrillogenesis during development, and by directly blocking the bioactivity of transforming growth factor-β1 (TGFβ1), it exerts a protective effect against fibrosis. However, no in vivo investigations on the role of decorin in liver have been performed before. In this study we used decorin-null (Dcn-/-) mice to establish the role of decorin in experimental liver fibrosis and repair. Not only the extent of experimentally induced liver fibrosis was more severe in Dcn-/- animals, but also the healing process was significantly delayed vis-à-vis wild-type mice. Collagen I, III, and IV mRNA levels in Dcn-/- livers were higher than those of wild-type livers only in the first 2 months, but no difference was observed after 4 months of fibrosis induction, suggesting that the elevation of these proteins reflects a specific impairment of their degradation. Gelatinase assays confirmed this hypothesis as we found decreased MMP-2 and MMP-9 activity and higher expression of TIMP-1 and PAI-1 mRNA in Dcn-/- livers. In contrast, at the end of the recovery phase increased production rather than impaired degradation was found to be responsible for the excessive connective tissue deposition in livers of Dcn-/- mice. Higher expression of TGFβ1-inducible early responsive gene in decorin-null livers indicated enhanced bioactivity of TGFβ1 known to upregulate TIMP-1 and PAI-1 as well. Moreover, two main axes of TGFβ1-evoked signaling pathways were affected by decorin deficiency, namely the Erk1/2 and Smad3 were activated in Dcn-/- samples, whereas no significant difference in phospho-Smad2 was observed between mice with different genotypes. Collectively, our results indicate that the lack of decorin favors the development of hepatic fibrosis and attenuates its subsequent healing process at least in part by affecting the bioactivity of TGFβ1.
Collapse
Affiliation(s)
- Kornélia Baghy
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Katalin Dezső
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Viktória László
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Alexandra Fullár
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Bálint Péterfia
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Sándor Paku
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Péter Nagy
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zsuzsa Schaff
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ilona Kovalszky
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary,Corresponding author. 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Ulloi ut 26., Budapest, Hungary 1085., Tel.: +36-1-459-1500, Ext. 54449, Fax.: +36-1-317-1074, (I. Kovalszky)
| |
Collapse
|
24
|
Rodrigues Díez R, Rodrigues-Díez R, Lavoz C, Rayego-Mateos S, Civantos E, Rodríguez-Vita J, Mezzano S, Ortiz A, Egido J, Ruiz-Ortega M. Statins inhibit angiotensin II/Smad pathway and related vascular fibrosis, by a TGF-β-independent process. PLoS One 2010; 5:e14145. [PMID: 21152444 PMCID: PMC2994748 DOI: 10.1371/journal.pone.0014145] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Accepted: 10/29/2010] [Indexed: 12/16/2022] Open
Abstract
We have recently described that in an experimental model of atherosclerosis and in vascular smooth muscle cells (VSMCs) statins increased the activation of the Smad pathway by transforming growth factor-β (TGF-β), leading to an increase in TGF-β-dependent matrix accumulation and plaque stabilization. Angiotensin II (AngII) activates the Smad pathway and contributes to vascular fibrosis, although the in vivo contribution of TGF-β has not been completely elucidated. Our aim was to further investigate the mechanisms involved in AngII-induced Smad activation in the vasculature, and to clarify the beneficial effects of statins on AngII-induced vascular fibrosis. Infusion of AngII into rats for 3 days activates the Smad pathway and increases fibrotic-related factors, independently of TGF-β, in rat aorta. Treatment with atorvastatin or simvastatin inhibited AngII-induced Smad activation and related-fibrosis. In cultured rat VSMCs, direct AngII/Smad pathway activation was mediated by p38 MAPK and ROCK activation. Preincubation of VSMCs with statins inhibited AngII-induced Smad activation at all time points studied (from 20 minutes to 24 hours). All these data show that statins inhibited several AngII-activated intracellular signaling systems, including p38-MAPK and ROCK, which regulates the AngII/Smad pathway and related profibrotic factors and matrix proteins, independently of TGF-β responses. The inhibitory effect of statins on the AngII/Smad pathway could explain, at least in part, their beneficial effects on hypertension-induced vascular damage.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Atorvastatin
- Blotting, Western
- Cells, Cultured
- Fibrosis/metabolism
- Heptanoic Acids/pharmacology
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Phosphorylation/drug effects
- Pyrroles/pharmacology
- Rats
- Rats, Wistar
- Signal Transduction/drug effects
- Simvastatin/pharmacology
- Smad Proteins/metabolism
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/pharmacology
- Vasoconstrictor Agents/pharmacology
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Raúl Rodrigues Díez
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma de Madrid, Madrid, Spain
| | - Raquel Rodrigues-Díez
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carolina Lavoz
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma de Madrid, Madrid, Spain
| | - Esther Civantos
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Rodríguez-Vita
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral, Valdivia, Chile
| | - Alberto Ortiz
- Dialysis Unit, Fundación Jiménez Díaz, Madrid, Spain
| | - Jesús Egido
- Renal Research Laboratory, Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|
25
|
Differential modulation of the cytokine-induced MMP-9/TIMP-1 protease-antiprotease system by the mTOR inhibitor rapamycin. Biochem Pharmacol 2010; 81:134-43. [PMID: 20854798 DOI: 10.1016/j.bcp.2010.09.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 09/09/2010] [Accepted: 09/10/2010] [Indexed: 01/07/2023]
Abstract
The mTOR-inhibitor rapamycin is a potent drug used in many immunosuppressive and antiinflammatory therapeutic regimes. In renal transplantation despite its beneficial roles rapamycin in some cases can promote renal fibrosis in the kidney but the underlying mechanisms are unknown. In this study, we tested for possible modulatory effects of rapamycin on the cytokine-triggered matrix metalloproteinase 9 (MMP-9)/tissue inhibitor of metalloproteinase (TIMP)-1 protease-antiprotease system which is critically involved in renal inflammation and fibrosis. Treatment of rat mesangial cells (MC) with rapamycin dose-dependently reduced the interleukin 1β (IL-1β)-triggered increase in gelatinolytic levels as demonstrated by zymography. The reduction in the extracellular MMP-9 content by rapamycin coincided with an attenuation in cytokine-induced steady-state MMP-9 mRNA levels. Conversely, rapamycin caused a dose-dependent increase in cytokine-evoked TIMP-1 expression in a Smad binding element (SBE)-dependent manner. Surprisingly, the attenuation of MMP-9 mRNA levels by rapamycin is accompanied by a potentiation of IL-1β-induced MMP-9 promoter activity in which the stimulatory effects by rapamycin are mainly attributed to a proximal AP-1 binding site. Furthermore, the rapamycin-dependent potentiation of MMP-9 expression is accompanied by an amplification of cytokine-triggered activities of nuclear factor κB (NF-κB) and activator protein 1 (AP-1) transcription factors. Importantly, rapamycin-triggered increase in MMP-9 promoter activity is fully impaired when we used a MMP-9 reporter construct which is under the additional control of the 3' untranslated region (3'-UTR) of MMP-9. Collectively, these data imply that rapamycin inhibits the cytokine-induced MMP-9 mainly through posttranscriptional events and thereby exerts profibrotic activities.
Collapse
|
26
|
Kim HJ, Kim MY, Hwang JS, Kim HJ, Lee JH, Chang KC, Kim JH, Han CW, Kim JH, Seo HG. PPARdelta inhibits IL-1beta-stimulated proliferation and migration of vascular smooth muscle cells via up-regulation of IL-1Ra. Cell Mol Life Sci 2010; 67:2119-30. [PMID: 20221783 PMCID: PMC11115654 DOI: 10.1007/s00018-010-0328-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 02/07/2010] [Accepted: 02/19/2010] [Indexed: 11/26/2022]
Abstract
Activation of peroxisome proliferator-activated receptor (PPAR) delta by GW501516, a specific PPARdelta ligand, significantly inhibited interleukin (IL)-1beta-induced proliferation and migration of vascular smooth muscle cells (VSMCs). This effect of GW501516 was dependent on transforming growth factor-beta, and was mediated through the up-regulation of IL-1 receptor antagonist. The inhibitory effect of GW501516 on VSMC proliferation was associated with cell cycle arrest at the G1 to S phase transition, which was accompanied by the induction of p21 and p53 along with decreased cyclin-dependent kinase 4 expression. Inhibition of cell migration by GW501516 was associated with the down-regulation of matrix metalloproteinase (MMP)-2 and MMP-9 in IL-1beta-treated VSMCs. Inhibition of extracellular signal-regulated kinase significantly reduced the GW501516-mediated inhibition of IL-1beta-stimulated VSMC proliferation. These results suggest that PPARdelta plays an important role in the pathophysiology of diseases associated with the proliferation and migration of VSMCs.
Collapse
Affiliation(s)
- H. J. Kim
- Department of Pharmacology, Gyeongsang Institute of Health Science, Gyeongsang National University School of Medicine, 92 Chilam-Dong, Jinju, 660-751 Korea
| | - M. Y. Kim
- Department of Pharmacology, Gyeongsang Institute of Health Science, Gyeongsang National University School of Medicine, 92 Chilam-Dong, Jinju, 660-751 Korea
| | - J. S. Hwang
- Department of Pharmacology, Gyeongsang Institute of Health Science, Gyeongsang National University School of Medicine, 92 Chilam-Dong, Jinju, 660-751 Korea
| | - H. J. Kim
- Department of Pharmacology, Gyeongsang Institute of Health Science, Gyeongsang National University School of Medicine, 92 Chilam-Dong, Jinju, 660-751 Korea
| | - J. H. Lee
- Department of Pharmacology, Gyeongsang Institute of Health Science, Gyeongsang National University School of Medicine, 92 Chilam-Dong, Jinju, 660-751 Korea
| | - K. C. Chang
- Department of Pharmacology, Gyeongsang Institute of Health Science, Gyeongsang National University School of Medicine, 92 Chilam-Dong, Jinju, 660-751 Korea
| | - J. -H. Kim
- Department of Animal Biotechnology, Kon-Kuk University, 1 Hwayang-Dong, Gwangjin-Gu, Seoul, 143-701 Korea
| | - C. W. Han
- Department of Oriental Internal Medicine, College of Oriental Medicine, Kyung-Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul, 130-701 Korea
| | - J.-H. Kim
- Department of Biomedical Science, CHA Stem Cell Institute, College of Life Science, CHA University, Pochon-si, Gyeonggi-do, 487-010 Korea
| | - H. G. Seo
- Department of Pharmacology, Gyeongsang Institute of Health Science, Gyeongsang National University School of Medicine, 92 Chilam-Dong, Jinju, 660-751 Korea
| |
Collapse
|
27
|
Malik M, Shukla A, Amin P, Niedelman W, Lee J, Jividen K, Phang JM, Ding J, Suh KS, Curmi PMG, Yuspa SH. S-nitrosylation regulates nuclear translocation of chloride intracellular channel protein CLIC4. J Biol Chem 2010; 285:23818-28. [PMID: 20504765 DOI: 10.1074/jbc.m109.091611] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nuclear translocation of chloride intracellular channel protein CLIC4 is essential for its role in Ca(2+)-induced differentiation, stress-induced apoptosis, and modulating TGF-beta signaling in mouse epidermal keratinocytes. However, post-translational modifications on CLIC4 that govern nuclear translocation and thus these activities remain to be elucidated. The structure of CLIC4 is dependent on the redox environment, in vitro, and translocation may depend on reactive oxygen and nitrogen species in the cell. Here we show that NO directly induces nuclear translocation of CLIC4 that is independent of the NO-cGMP pathway. Indeed, CLIC4 is directly modified by NO through S-nitrosylation of a cysteine residue, as measured by the biotin switch assay. NO enhances association of CLIC4 with the nuclear import proteins importin alpha and Ran. This is likely a result of the conformational change induced by S-nitrosylated CLIC4 that leads to unfolding of the protein, as exhibited by CD spectra analysis and trypsinolysis of the modified protein. Cysteine mutants of CLIC4 exhibit altered nitrosylation, nuclear residence, and stability, compared with the wild type protein likely as a consequence of altered tertiary structure. Moreover, tumor necrosis factor alpha-induced nuclear translocation of CLIC4 is dependent on nitric-oxide synthase activity. Inhibition of nitric-oxide synthase activity inhibits tumor necrosis factor alpha-induced nitrosylation and association with importin alpha and Ran and ablates CLIC4 nuclear translocation. These results suggest that S-nitrosylation governs CLIC4 structure, its association with protein partners, and thus its intracellular distribution.
Collapse
Affiliation(s)
- Mariam Malik
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Schiller M, Dennler S, Anderegg U, Kokot A, Simon JC, Luger TA, Mauviel A, Böhm M. Increased cAMP levels modulate transforming growth factor-beta/Smad-induced expression of extracellular matrix components and other key fibroblast effector functions. J Biol Chem 2009; 285:409-21. [PMID: 19858184 DOI: 10.1074/jbc.m109.038620] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
cAMP is a key messenger of many hormones and neuropeptides, some of which modulate the composition of extracellular matrix. Treatment of human dermal fibroblasts with dibutyryl cyclic AMP and forskolin antagonized the inductive effects of transforming growth factor-beta (TGF-beta) on the expression of collagen, connective tissue growth factor, tissue inhibitor of matrix metalloproteinase-1, and plasminogen activator inhibitor type I, four prototypical TGF-beta-responsive genes. Increased intracellular cAMP prevented TGF-beta-induced Smad-specific gene transactivation, although TGF-beta-mediated Smad phosphorylation and nuclear translocation remained unaffected. However, increased cAMP levels abolished TGF-beta-induced interaction of Smad3 with its transcriptional co-activator cAMP-response element-binding protein (CREB)-binding protein (CBP)/p300. Overexpression of the transcriptional co-activator CBP/p300 rescued Smad-specific gene transcription in the presence of cAMP suggesting that sequestration of limited amounts of CBP/p300 by the activated cAMP/CREB pathway is the molecular basis of this inhibitory effect. These findings were extended by two functional assays. Increased intracellular cAMP levels suppressed the inductive activity of TGF-beta to contract mechanically unloaded collagen lattices and resulted in an attenuation of fibroblast migration of mechanically induced cell layer wounds. Of note, cAMP and TGF-beta synergistically induced hyaluronan synthase 2 (HAS2) expression and hyaluronan secretion, presumably via putative CREB-binding sites adjacent to Smad-binding sites within the HAS2 promoter. Our findings identify the cAMP pathway as a potent but differential and promoter-specific regulator of TGF-beta-mediated effects involved in extracellular matrix homeostasis.
Collapse
Affiliation(s)
- Meinhard Schiller
- Department of Dermatology, Ludwig Boltzmann Institute for Cell Biology and Immunobiology of the Skin, 48149 Münster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Osman B, Doller A, Akool ES, Holdener M, Hintermann E, Pfeilschifter J, Eberhardt W. Rapamycin induces the TGFbeta1/Smad signaling cascade in renal mesangial cells upstream of mTOR. Cell Signal 2009; 21:1806-17. [PMID: 19666112 DOI: 10.1016/j.cellsig.2009.07.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 07/13/2009] [Accepted: 07/29/2009] [Indexed: 11/26/2022]
Abstract
The mTOR kinase inhibitor rapamycin (sirolimus) is a drug with potent immunosuppressive and antiproliferative properties. We found that rapamycin induces the TGFbeta/Smad signaling cascade in rat mesangial cells (MC) as depicted by the nuclear translocation of phospho-Smads 2, -3 and Smad-4, respectively. Concomitantly, rapamycin increases the nuclear DNA binding of receptor (R)- and co-Smad proteins to a cognate Smad-binding element (SBE) which in turn causes an increase in profibrotic gene expression as exemplified by the connective tissue growth factor (CTGF) and plasminogen activator inhibitor 1 (PAI-1). Using small interfering (si)RNA we demonstrate that Smad 2/3 activation by rapamycin depends on its endogenous receptor FK binding protein 12 (FKBP12). Mechanistically, Smad induction by rapamycin is initiated by an increase in active TGFbeta(1) as shown by ELISA and by the inhibitory effects of a neutralizing TGFbeta antibody. Using an activin receptor-like kinase (ALK)-5 inhibitor and by siRNA against the TGFbeta type II receptor (TGFbeta-RII) we furthermore demonstrate a functional involvement of both types of TGFbeta receptors. However, rapamycin did not compete with TGFbeta for TGFbeta-receptor binding as found in radioligand-binding assay. Besides SB203580, a specific inhibitor of the p38 MAPK, the reactive oxygen species (ROS) scavenger N-acetyl-cysteine (NAC) and a cell-permeable superoxide dismutase (SOD) mimetic strongly abrogated the stimulatory effects of rapamycin on Smad 2 and 3 phosphorylation. Furthermore, the rapid increase in dichlorofluorescein (DCF) formation implies that rapamycin mainly acts through ROS. In conclusion, activation of the profibrotic TGFbeta/Smad signaling cascade accompanies the immunosuppressive and antiproliferative actions of rapamycin.
Collapse
Affiliation(s)
- Bashier Osman
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Colasanti M, Mazzone V, Mancinelli L, Leone S, Venturini G. Involvement of nitric oxide in the head regeneration of Hydra vulgaris. Nitric Oxide 2009; 21:164-70. [PMID: 19635580 DOI: 10.1016/j.niox.2009.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2009] [Revised: 07/16/2009] [Accepted: 07/20/2009] [Indexed: 10/20/2022]
Abstract
Recent data have shown that a functional NO-cGMP signalling system plays an important role during development and seems to be operative early during the differentiation of embryonic stem cells. The intriguing possibility exists that this role can be evolutionarily conserved between vertebrates and invertebrates. In this paper, we have analyzed the effect of NO-cGMP pathway on the regeneration process in Hydra vulgaris, the most primitive invertebrate possessing a nervous system. Our results indicate that NO production increased during Hydra regeneration. The NOS inhibitor L-NAME reduced the regenerative process and the same effect was obtained by treatment with either the specific guanylate cyclase inhibitor ODQ or the protein kinase G (PKG) inhibitor KT-5823. In contrast, the regeneration process was increased by treating decapitated Hydra with the NO donor NOC-18. Furthermore, we found that cell proliferation was also increased by treating decapitated Hydra with the NO donor NOC-18 and reduced by treatment with the NOS inhibitor L-NAME. Our results strongly suggest that the NO-cGMP-PKG pathway is involved in the control of the proliferative-differentiative patterns of developing and regenerating structures in cnidarians as well as bilaterians.
Collapse
Affiliation(s)
- Marco Colasanti
- Department of Biology, University of Rome "ROMA TRE", Viale Marconi 446, 00146 Rome, Italy.
| | | | | | | | | |
Collapse
|
31
|
Dreieicher E, Beck KF, Lazaroski S, Boosen M, Tsalastra-Greul W, Beck M, Fleming I, Schaefer L, Pfeilschifter J. Nitric oxide inhibits glomerular TGF-beta signaling via SMOC-1. J Am Soc Nephrol 2009; 20:1963-74. [PMID: 19578009 DOI: 10.1681/asn.2008060653] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cytokines and nitric oxide (NO) stimulate rat mesangial cells to synthesize and secrete inflammatory mediators. To understand better the signaling pathways that contribute to this response, we exposed rat mesangial cells to the prototypic inflammatory cytokine IL-1beta and analyzed the changes in the pattern of gene expression. IL-1beta downregulated the gene encoding the matricellular glycoprotein secreted modular calcium-binding protein 1 (SMOC-1) in mesangial cells. Inflammatory cytokines attenuated SMOC-1 mRNA and protein expression through endogenous production of NO, which activated the soluble guanylyl cyclase. Silencing SMOC-1 expression with small interfering RNA decreased the formation of TGF-beta, reduced SMAD binding to DNA, and decreased mRNA expression of genes regulated by TGF-beta. In a rat model of anti-Thy-1 glomerulonephritis, glomerular SMOC-1 mRNA and protein decreased and inducible NO synthase expression increased simultaneously. Treatment of nephritic rats with the inducible NO synthase-specific inhibitor l-N(6)-(1-iminoethyl)-lysine prevented SMOC-1 downregulation. In summary, these data suggest that NO attenuates SMOC-1 expression in acute glomerular inflammation, thereby limiting TGF-beta-mediated profibrotic signaling.
Collapse
Affiliation(s)
- Ellen Dreieicher
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Gerhardinger C, Dagher Z, Sebastiani P, Park YS, Lorenzi M. The transforming growth factor-beta pathway is a common target of drugs that prevent experimental diabetic retinopathy. Diabetes 2009; 58:1659-67. [PMID: 19401417 PMCID: PMC2699853 DOI: 10.2337/db08-1008] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Accepted: 04/02/2009] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Prevention of diabetic retinopathy would benefit from availability of drugs that preempt the effects of hyperglycemia on retinal vessels. We aimed to identify candidate drug targets by investigating the molecular effects of drugs that prevent retinal capillary demise in the diabetic rat. RESEARCH DESIGN AND METHODS We examined the gene expression profile of retinal vessels isolated from rats with 6 months of streptozotocin-induced diabetes and compared it with that of control rats. We then tested whether the aldose reductase inhibitor sorbinil and aspirin, which have different mechanisms of action, prevented common molecular abnormalities induced by diabetes. The Affymetrix GeneChip Rat Genome 230 2.0 array was complemented by real-time RT-PCR, immunoblotting, and immunohistochemistry. RESULTS The retinal vessels of diabetic rats showed differential expression of 20 genes of the transforming growth factor (TGF)-beta pathway, in addition to genes involved in oxidative stress, inflammation, vascular remodeling, and apoptosis. The complete loop of TGF-beta signaling, including Smad2 phosphorylation, was enhanced in the retinal vessels, but not in the neural retina. Sorbinil normalized the expression of 71% of the genes related to oxidative stress and 62% of those related to inflammation. Aspirin had minimal or no effect on these two categories. The two drugs were instead concordant in reducing the upregulation of genes of the TGF-beta pathway (55% for sorbinil and 40% for aspirin) and apoptosis (74 and 42%, respectively). CONCLUSIONS Oxidative and inflammatory stress is the distinct signature that the polyol pathway leaves on retinal vessels. TGF-beta and apoptosis are, however, the ultimate targets to prevent the capillary demise in diabetic retinopathy.
Collapse
Affiliation(s)
- Chiara Gerhardinger
- Schepens Eye Research Institute and Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| | | | | | | | | |
Collapse
|
33
|
Kim HJ, Kim MY, Jin H, Kim HJ, Kang SS, Kim HJ, Lee JH, Chang KC, Hwang JY, Yabe-Nishimura C, Kim JH, Seo HG. Peroxisome proliferator-activated receptor {delta} regulates extracellular matrix and apoptosis of vascular smooth muscle cells through the activation of transforming growth factor-{beta}1/Smad3. Circ Res 2009; 105:16-24. [PMID: 19461048 DOI: 10.1161/circresaha.108.189159] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Homeostasis of the extracellular matrix and apoptosis of vascular smooth muscle cells (VSMCs) are key components in the regulation of the stability of atherosclerotic plaques. Here, we demonstrate that peroxisome proliferator-activated receptor (PPAR)delta regulates extracellular matrix synthesis and degradation through transforming growth factor-beta1 and its effector, Smad3. Activation of PPARdelta strongly amplified the expression of types I and III collagen, fibronectin, elastin, and TIMP-3 (tissue inhibitor of metalloproteinases 3), but not of TIMP-1, matrix metalloproteinase-2 or -9. The effect of PPARdelta on the expression of type III collagen was dually regulated by the direct binding of PPARdelta and Smad3 to a direct repeat-1 site and a Smad-binding element, respectively, in the type III collagen gene promoter. The activation of PPARdelta attenuated apoptotic cell death in VSMCs induced by oxidized low-density lipoprotein, and similar antiapoptotic effects were observed on treatment of cells with exogenous type I and/or III collagen. Administration of a PPARdelta ligand GW501516 to mice also suppressed elastase-induced cell death of aortic VSMCs. These results suggest that PPARdelta-induced upregulation of extracellular matrix proteins exerts an antiapoptotic effect, thereby maintaining the stability of atherosclerotic plaques. Specific ligands of PPARdelta may aid in the therapeutic intervention of atherosclerosis by improving plaque stability and patient prognosis.
Collapse
Affiliation(s)
- Hyo Jung Kim
- Department of Pharmacology, Gyeongsang Institute of Health Science, Gyeongsang National University School of Medicine, Chilam-Dong, Jinju, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Qureshi HY, Ricci G, Zafarullah M. Smad signaling pathway is a pivotal component of tissue inhibitor of metalloproteinases-3 regulation by transforming growth factor beta in human chondrocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:1605-12. [DOI: 10.1016/j.bbamcr.2008.04.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 03/20/2008] [Accepted: 04/07/2008] [Indexed: 11/26/2022]
|
35
|
Akool ES, Doller A, Babelova A, Tsalastra W, Moreth K, Schaefer L, Pfeilschifter J, Eberhardt W. Molecular Mechanisms of TGFβ Receptor-Triggered Signaling Cascades Rapidly Induced by the Calcineurin Inhibitors Cyclosporin A and FK506. THE JOURNAL OF IMMUNOLOGY 2008; 181:2831-45. [DOI: 10.4049/jimmunol.181.4.2831] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
36
|
PGE2 induces angiogenesis via MT1-MMP-mediated activation of the TGFbeta/Alk5 signaling pathway. Blood 2008; 112:1120-8. [PMID: 18541723 DOI: 10.1182/blood-2007-09-112268] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The development of a new vascular network is essential for the onset and progression of many pathophysiologic processes. Cyclooxygenase-2 displays a proangiogenic activity in in vitro and in vivo models, mediated principally through its metabolite prostaglandin E(2) (PGE(2)). Here, we provide evidence for a novel signaling route through which PGE(2) activates the Alk5-Smad3 pathway in endothelial cells. PGE(2) induces Alk5-dependent Smad3 nuclear translocation and DNA binding, and the activation of this pathway involves the release of active TGFbeta from its latent form through a process mediated by the metalloproteinase MT1-MMP, whose membrane clustering is promoted by PGE(2). MT1-MMP-dependent transforming growth factor beta (TGFbeta) signaling through Alk5 is also required for PGE(2)-induced endothelial cord formation in vitro, and Alk5 kinase activity is required for PGE(2)-induced neovascularization in vivo. These findings identify a novel signaling pathway linking PGE(2) and TGFbeta, 2 effectors involved in tumor growth and angiogenesis, and reveal potential targets for the treatment of angiogenesis-related disorders.
Collapse
|
37
|
Sánchez-López E, Rodriguez-Vita J, Cartier C, Rupérez M, Esteban V, Carvajal G, Rodrígues-Díez R, Plaza JJ, Egido J, Ruiz-Ortega M. Inhibitory effect of interleukin-1beta on angiotensin II-induced connective tissue growth factor and type IV collagen production in cultured mesangial cells. Am J Physiol Renal Physiol 2007; 294:F149-60. [PMID: 17989112 DOI: 10.1152/ajprenal.00129.2007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Connective tissue growth factor (CTGF) is overexpressed in kidney diseases associated with extracellular matrix accumulation. Angiotensin II (ANG II) participates in renal fibrosis by the upregulation of growth factors, including CTGF, and extracellular matrix proteins, such as type IV collagen. During renal injury, ANG II and the macrophage-produced cytokine interleukin-1beta (IL-1beta) may be present simultaneously in the glomerular environment. However, there are no studies about the interaction between ANG II and IL-1beta in renal fibrosis. For this reason, in cultured mesangial cells (MC), we investigated whether IL-1beta could regulate ANG II-mediated collagen accumulation and the mechanisms underlying this process. In MC, CTGF is a downstream mediator of type IV collagen production induced by ANG II. IL-1beta did not increase the production of CTGF and type IV collagen but significantly inhibited ANG II-induced CTGF and type IV collagen overexpression. Moreover, IL-1beta also inhibited type IV collagen upregulation caused by exogenous recombinant CTGF. Matrix metalloproteinase-9 (MMP-9) is the main enzyme involved in type IV collagen degradation. In MC, coincubation of IL-1beta and ANG II caused a synergistic increase in MMP-9 gene expression and activity, associated with type IV collagen inhibition. The described IL-1beta effects were dependent on activation of ERK/MAPK but independent p38-MAPK, JNK, phosphatidylinositol 3-kinase/Akt, and Rho-associated kinase pathways. In summary, these data indicate that IL-1beta inhibited ANG II-mediated type IV collagen production, via CTGF downregulation, and increased type IV collagen degradation, through MMP-9 upregulation. Our in vitro data show that the proinflammatory cytokine IL-1beta abrogates ANG II-induced CTGF production, describing antagonistic activities of proinflammatory cytokines on ANG II actions.
Collapse
Affiliation(s)
- Elsa Sánchez-López
- Cellular Biology in Renal Diseases Laboratory, Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Uemura K, Li M, Tsutsumi T, Yamazaki T, Kawada T, Kamiya A, Inagaki M, Sunagawa K, Sugimachi M. Efferent vagal nerve stimulation induces tissue inhibitor of metalloproteinase-1 in myocardial ischemia-reperfusion injury in rabbit. Am J Physiol Heart Circ Physiol 2007; 293:H2254-61. [PMID: 17693545 DOI: 10.1152/ajpheart.00490.2007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vagal nerve stimulation has been suggested to ameliorate left ventricular (LV) remodeling in heart failure. However, it is not known whether and to what degree vagal nerve stimulation affects matrix metalloproteinase (MMP) and tissue inhibitor of MMP (TIMP) in myocardium, which are known to play crucial roles in LV remodeling. We therefore investigated the effects of electrical stimulation of efferent vagal nerve on myocardial expression and activation of MMPs and TIMPs in a rabbit model of myocardial ischemia-reperfusion (I/R) injury. Anesthetized rabbits were subjected to 60 min of left coronary artery occlusion and 180 min of reperfusion with (I/R-VS, n = 8) or without vagal nerve stimulation (I/R, n = 7). Rabbits not subjected to coronary occlusion with (VS, n = 7) or without vagal stimulation (sham, n = 7) were used as controls. Total MMP-9 protein increased significantly after left coronary artery occlusion in I/R-VS and I/R to a similar degree compared with VS and sham values. Endogenous active MMP-9 protein level was significantly lower in I/R-VS compared with I/R. TIMP-1 mRNA expression was significantly increased in I/R-VS compared with the I/R, VS, and sham groups. TIMP-1 protein was significantly increased in I/R-VS and VS compared with the I/R and sham groups. Cardiac microdialysis technique demonstrated that topical perfusion of acetylcholine increased dialysate TIMP-1 protein level, which was suppressed by coperfusion of atropine. Immunohistochemistry demonstrated a strong expression of TIMP-1 protein in cardiomyocytes around the dialysis probe used to perfuse acetylcholine. In conclusion, in a rabbit model of myocardial I/R injury, vagal nerve stimulation induced TIMP-1 expression in cardiomyocytes and reduced active MMP-9.
Collapse
Affiliation(s)
- Kazunori Uemura
- Dept. of Cardiovascular Dynamics, National Cardiovascular Center Research Inst., 5-7-1 Fujishirodai, Suita 565-8565, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Xin C, Ren S, Eberhardt W, Pfeilschifter J, Huwiler A. Sphingosylphosphorylcholine acts in an anti-inflammatory manner in renal mesangial cells by reducing interleukin-1β-induced prostaglandin E2 formation. J Lipid Res 2007; 48:1985-96. [PMID: 17592175 DOI: 10.1194/jlr.m700077-jlr200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Sphingosylphosphorylcholine (SPC) is a bioactive lipid that binds to G protein-coupled-receptors and activates various signaling cascades. Here, we show that in renal mesangial cells, SPC not only activates various protein kinase cascades but also activates Smad proteins, which are classical members of the transforming growth factor-beta (TGFbeta) signaling pathway. Consequently, SPC is able to mimic TGFbeta-mediated cell responses, such as an anti-inflammatory and a profibrotic response. Interleukin-1beta-stimulated prostaglandin E(2) formation is dose-dependently suppressed by SPC, which is paralleled by reduced secretory phospholipase A(2) (sPLA(2)) protein expression and activity. This effect is due to a reduction of sPLA(2) mRNA expression caused by inhibited sPLA(2) promoter activity. Furthermore, SPC upregulates the profibrotic connective tissue growth factor (CTGF) protein and mRNA expression. Blocking TGFbeta signaling by a TGFbeta receptor kinase inhibitor causes an inhibition of SPC-stimulated Smad activation and reverses both the negative effect of SPC on sPLA(2) expression and the positive effect on CTGF expression. In summary, our data show that SPC, by mimicking TGFbeta, leads to a suppression of proinflammatory mediator production and stimulates a profibrotic cell response that is often the end point of an anti-inflammatory reaction. Thus, targeting SPC receptors may represent a novel therapeutic strategy to cope with inflammatory diseases.
Collapse
Affiliation(s)
- Cuiyan Xin
- Institute of Pharmacology, University of Bern, CH-3010 Bern, Switzerland
| | | | | | | | | |
Collapse
|
40
|
Abstract
Extracellular matrix (ECM) remodeling with successive tissue fibrosis is a key feature of chronic cardiovascular diseases, including atherosclerosis and restenosis. The atherogenic changes underlying these pathologies result from chronification of an acute repair response towards injurious and inflammatory stimuli. Thereby functional tissue is replaced by excessive ECM deposition. In the kidney, impaired remodeling is a major cause of perivascular, interstitial, and glomerular fibrosis but also a common complication of chronic hypertension. Experimental evidence points to the matrix metalloproteases (MMPs) and their intrinsic inhibitors, the tissue inhibitors of MMPs as key mediators of atherogenic and fibrotic pathologies. Mechanistically, a deregulation in ECM turnover tightly correlates with an increased production and release of proinflammatory and profibrotic factors including interleukin-1beta, transforming growth factor beta, angiotensin II, and reactive oxygen species. Unlike these factors the pleiotropic messenger molecule nitric oxide (NO) by acting as the major physiological vasodilator has emerged as one of the most atheroprotective factors. However, under inflammatory conditions NO does acquire proatherogenic and profibrotic properties thereby exacerbating tissue fibrosis. In this review, the mechanisms underlying both opposing properties of NO on perivascular ECM remodeling will exemplarily be discussed for renal fibrosis with a particular focus on the MMPs and intrinsic protease inhibitors.
Collapse
Affiliation(s)
- W Eberhardt
- Pharmazentrum frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Theodor-Stern-Kai 7 Frankfurt am Main, Germany.
| | | |
Collapse
|
41
|
Dooley A, Gao B, Shi-Wen X, Abraham DJ, Black CM, Jacobs M, Bruckdorfer KR. Effect of nitric oxide and peroxynitrite on type I collagen synthesis in normal and scleroderma dermal fibroblasts. Free Radic Biol Med 2007; 43:253-64. [PMID: 17603934 DOI: 10.1016/j.freeradbiomed.2007.04.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Revised: 03/30/2007] [Accepted: 04/11/2007] [Indexed: 11/26/2022]
Abstract
Nitric oxide ((.-)NO) is an important physiological signaling molecule and potent vasodilator. Recently, we have shown abnormal (.-)NO metabolism in the plasma of patients with systemic sclerosis (SSc), a disease that features excessive collagen overproduction as well as vascular dysfunction. The current study investigates the effects of (.-)NO and peroxynitrite (ONOO(-)) on secretion of type I collagen by SSc dermal fibroblasts, compared with those from normal dermal fibroblasts (CON) and a dermal fibroblast cell line (AG). Dermal fibroblasts were incubated with (.-)NO donors (SNP, DETA-NONOate) with or without the antioxidant ascorbic acid, or ONOO(-) for 24-72 h. In CON and AG fibroblasts, type I collagen was dose dependently decreased by SNP or DETA-NONOate. However, (.-)NO had no effect in SSc fibroblasts. Furthermore, the inhibition of collagen synthesis by (.-)NO was reversed by ascorbic acid and was not affected by 1H-[1,2,4]oxadiazole[4,3-a]quinoxalin-1-one, an inhibitor of soluble guanyl cyclase, or 8-bromoguanosine cyclic 3',5'-monophosphate, a cGMP agonist. SNP also showed a significant up-regulation of matrix metalloproteinase-1 (MMP-1) protein and activity levels, an essential collagenase involved in collagen degradation, in the AG fibroblasts. Additionally, (.-)NO-treated fibroblasts had lower prolyl hydroxylase activity, an enzyme important in the post-translational processing of collagen, while there was no effect on total protein levels. There were no significant effects on type I collagen levels when dermal fibroblasts were treated with ONOO(-). Taken together, ()NO inhibits collagen secretion in normal dermal fibroblasts but regulation is lost in SSc fibroblasts, while ONOO(-) itself is ineffective. (.-)NO inhibition of collagen was by cGMP-independent regulatory mechanisms and in part may be due to up-regulation of MMP-1 and/or inhibition of prolyl hydroxylase. These differences may contribute to the observed pathology of SSc.
Collapse
Affiliation(s)
- Audrey Dooley
- Department of Biochemistry and Molecular Biology, Royal Free and University College Medical School, London NW3 2PF, UK
| | | | | | | | | | | | | |
Collapse
|
42
|
Chuang TD, Guh JY, Chiou SJ, Chen HC, Huang JS, Yang YL, Chuang LY. Phosphoinositide 3-kinase is required for high glucose-induced hypertrophy and p21WAF1 expression in LLC-PK1 cells. Kidney Int 2007; 71:867-74. [PMID: 17332736 DOI: 10.1038/sj.ki.5002155] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Transforming growth factor-beta (TGF-beta), Smads, and the cyclin-dependent kinase (cdk) inhibitor p21(WAF1) are important in the pathogenesis of diabetic tubular hypertrophy. Phosphoinositide 3 kinase (PI3K)/Akt kinase activity is increased in diabetic glomerular hypertrophy. Thus, we studied the role of PI3K in high glucose (30 mM)-induced p21(WAF1), Smad2/3, and cell cycle-dependent hypertrophy in LLC-PK1 cells. We found that high glucose time-dependently (1-48 h) increased PI3K/Akt kinase activity. LY294002 (a PI3K inhibitor) attenuated high glucose-induced cell cycle-dependent (G(0)/G(1) phase) hypertrophy at 72 h while attenuating high glucose-induced p21(WAF1) gene transcription and protein expression at 36-48 h. LY294002 also attenuated high glucose-induced binding of p21(WAF1) to the cyclin E/cdk2 complex, whereas attenuating high glucose-induced TGF-beta bioactivity, Smad2/3 phosphorylation, and Smad2/3 DNA-binding activity at 36-48 h. We concluded that PI3K is required for high glucose-induced cell cycle-dependent hypertrophy, p21(WAF1) transcription and expression, p21(WAF1) binding to the cyclin E/cdk2 complex, TGF-beta bioactivity, and Smad2/3 activity in LLC-PK1 cells.
Collapse
Affiliation(s)
- T-D Chuang
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
43
|
Li SZ, McDill BW, Kovach PA, Ding L, Go WY, Ho SN, Chen F. Calcineurin-NFATc signaling pathway regulates AQP2 expression in response to calcium signals and osmotic stress. Am J Physiol Cell Physiol 2006; 292:C1606-16. [PMID: 17166937 DOI: 10.1152/ajpcell.00588.2005] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The aquaporin (AQP)2 channel mediates the reabsorption of water in renal collecting ducts in response to arginine vasopressin (AVP) and hypertonicity. Here we show that AQP2 expression is induced not only by the tonicity-responsive enhancer binding protein (TonEBP)/nuclear factor of activated T cells (NFAT)5-mediated hypertonic stress response but also by the calcium-dependent calcineurin-NFATc pathway. The induction of AQP2 expression by the calcineurin-NFATc pathway can occur in the absence of TonEBP/NFAT5. Mutational and chromatin immunoprecipitation analyses revealed the existence of functional NFAT binding sites within the proximal AQP2 promoter responsible for regulation of AQP2 by NFATc proteins and TonEBP/NFAT5. Contrary to the notion that TonEBP/NFAT5 is the only Rel/NFAT family member regulated by tonicity, we found that hypertonicity promotes the nuclear translocation of NFATc proteins for the subsequent induction of AQP2 expression. Calcineurin activity was also found to be involved in the induction of TonEBP/NFAT5 expression by hypertonicity, thus further defining the signaling mechanisms that underlie the TonEBP/NFAT5 osmotic stress response pathway. The coordinate regulation of AQP2 expression by both osmotic stress and calcium signaling appears to provide a means to integrate diverse extracellular signals into optimal cellular responses.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Aquaporin 2/genetics
- Aquaporin 2/metabolism
- Calcineurin/metabolism
- Calcium Signaling
- Cell Line
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Ionomycin/pharmacology
- Ionophores/pharmacology
- Kidney Tubules, Collecting/cytology
- Kidney Tubules, Collecting/drug effects
- Kidney Tubules, Collecting/metabolism
- Kidney Tubules, Collecting/physiopathology
- Mice
- Mice, Knockout
- Mutation
- NFATC Transcription Factors/genetics
- NFATC Transcription Factors/metabolism
- Osmosis
- Promoter Regions, Genetic
- Saline Solution, Hypertonic/pharmacology
- Signal Transduction/drug effects
- Stress, Physiological/metabolism
- Stress, Physiological/physiopathology
- Tetradecanoylphorbol Acetate/pharmacology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
- Transfection
- Up-Regulation
- Water-Electrolyte Balance
Collapse
Affiliation(s)
- Song-Zhe Li
- Department of Internal Medicine/Renal Division, Campus Box 8126, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Induction of TIMP-1 and HSP47 synthesis in primary keloid fibroblasts by exogenous nitric oxide. J Dermatol Sci 2006; 45:37-44. [PMID: 17145168 DOI: 10.1016/j.jdermsci.2006.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2006] [Revised: 10/03/2006] [Accepted: 10/04/2006] [Indexed: 01/27/2023]
Abstract
BACKGROUND The excessive accumulation of extracellular matrix is a hallmark of many fibrotic diseases, including the hypertrophic scar and keloid. Recent reports from this research team had shown that exogenous nitric oxide (NO) participates in the keloid formation; however, its role on the synthesis of fibrotic factor (TGF-beta1, TIMP-1 and HSP47) in the keloid fibroblasts (KF) remained unclear. OBJECTIVE In this study, to better define the potential effect of exogenous NO on the expression of fibrotic factors in KF, the enhancing effect of exogenous NO, released from a NO donor, on the synthesis of fibrotic factors in KF was investigated. METHODS The seven primary KF cultures were set up to measure the effect of exogenous NO on enhancing the expression of fibrotic factor. RESULTS Elevation of cellular cGMP levels was observed to be induced by NO or blocked by the hydrolysis activity of phosphodiesterase (PDE) by the PDE inhibitor. The elevated levels of cellular cGMP were noted to enhance the expression of TIMP-1 and HSP47 in KF. Exogenous NO was found to significantly accelerate the production of TIMP-1 and HSP47 in the seven primary KFs with a corresponding increase in the production of TGF-beta1. CONCLUSION The results have led to a conclusion, that is: the excess collagen formations in the keloid lesion may be attributed to the NO/cGMP signal pathway by initiating a rapid increase in the expression of TGF-beta1, TIMP-1 and HSP47 in the KF cells.
Collapse
|
45
|
Exogenous nitric oxide stimulated collagen type I expression and TGF-beta1 production in keloid fibroblasts by a cGMP-dependent manner. Nitric Oxide 2006; 16:258-65. [PMID: 17052928 DOI: 10.1016/j.niox.2006.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Revised: 09/04/2006] [Accepted: 09/05/2006] [Indexed: 10/24/2022]
Abstract
Keloids arise from the aberrant wound healing process and nitric oxide (NO) plays an important role in the inflammation stage of wound healing. In order to better define the potential effect of NO/cGMP signal pathway in the keloid pathogenesis, the enhancing effect of exogenous NO (released from NO donor) on collagen expression in the keloid fibroblast (KF) as well as on the induction of collagen type I protein and TGF-beta1 expression in the KF were studied in this investigation. The DETA NONOate, an NO donor, was added to the KF, as the exogenous NO, to release NO in the culture medium. The expression of collagens was then determined by assaying the total soluble collagens and collagen type I in the KF. The cellular concentration of cGMP was measured by EIA in the KF. Exogenous NO was found to enhance the expression of collagens and elevate the cellular levels of cGMP. Moreover, to evaluate the effect of the elevated cellular cGMP levels on the expression of collagen and TGF-beta1, both cGMP and TGF-beta1 were measured by ELISA. The inhibitors for phosphodiesterase (PDE), such as IBMX (3-isobutyl-1-methylxanthine), Vinpocetine, EHNA, Milrinone and Zapriast, which have been reported to reduce the ability of PDE and subsequently produce an increase of cellular cGMP, induce the production of autocrine TGF-beta1 as well as the synthesis of collagen in the KF. In this investigation, the inhibition of the PDE enzyme activity was observed to enhance the effect on the collagen synthesis, and was induced by exogenous NO. Taken together, these results have suggested that the NO/cGMP pathway could positively influence the progression of keloid formation, via the TGF-beta1 expression in the KF.
Collapse
|
46
|
Hubert B, Troncy E, Gauvin D, Taha R, Pang D, Beauchamp G, Radomski A, Radomski MW, Blaise GA. Increased Alveolar and Plasma Gelatinases Activity during Postpump Syndrome: Inhibition by Inhaled Nitric Oxide. J Cardiovasc Pharmacol 2006; 48:71-8. [PMID: 17031259 DOI: 10.1097/01.fjc.0000242054.66031.5c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Postpump syndrome is associated with systemic inflammation. Matrix metalloproteinases (MMP)-2 and -9 contribute to proinflammatory and platelet-activator reactions. Nitric oxide (NO) is involved in the regulation of MMPs. The objectives of our study were to investigate the intensity of inflammation induced by 3 different surgical procedures, the effects of inflammation on the activity of MMPs, and the regulation of inflammation by inhaled NO (20 ppm). Inhaled NO was initiated immediately after tracheal intubation and maintained for the total duration of the experiments. Thirty pigs were equally randomized into 6 groups [sham; sham + NO; cardiopulmonary bypass; bypass + NO; bypass + lipopolysaccharide (1 microg/kg for 50 min); bypass + lipopolysaccharide + NO] and animals were subjected to anesthesia and mechanical ventilation up to 24 h. The levels of MMP-2 and MMP-9 in plasma and bronchoalveolar lavage were measured using zymography. Bypass resulted in a time-dependent rise in MMP activity, an effect potentiated by lipopolysaccharide. Inhaled NO attenuated the effects of bypass + lipopolysaccharide. These results confirm that MMP-2 and MMP-9 are associated with the inflammatory process causing the postpump syndrome. Preemptive and continuous administration of inhaled NO helps to prevent increased MMP-2 and MMP-9 activity.
Collapse
Affiliation(s)
- Bernard Hubert
- Department of Anesthesia, Centre Hospitalier de l'Université de Montréal, Montréal, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Nath AK, Madri JA. The roles of nitric oxide in murine cardiovascular development. Dev Biol 2006; 292:25-33. [PMID: 16442519 DOI: 10.1016/j.ydbio.2005.12.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2005] [Revised: 11/22/2005] [Accepted: 12/16/2005] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) participates in a diverse array of biological functions in mammalian organ systems. Depending on the biochemical environment, the production of NO may result in cytoprotection or cytotoxicity. The paradoxical actions of NO arise from the complexities generated by the redox milieu, NO concentration/bioavailability, and tissue/cell context, which ultimately result in the wide range of regulatory roles observed. Additionally, in physiological versus pathological states, NO often displays diametrically opposing affects in several organ systems. Here, we will discuss the roles of NO during reproduction, organ system development, in particular, the cardiovascular system, and its potential implications in diabetes-induced fetal defects.
Collapse
Affiliation(s)
- Anjali K Nath
- Yale University, Department of Molecular, Cellular and Developmental Biology, New Haven, CT 06520, USA
| | | |
Collapse
|