1
|
Cirilo MADS, Ribeiro FPB, Lima NKDS, Silva JK, Gomes JADS, Albuquerque JSS, Siqueira LCDS, Santos VBDS, Carvalho JMD, Tenorio FDCAM, Vieira LD. Paricalcitol prevents renal tubular injury induced by ischemia-reperfusion: Role of oxidative stress, inflammation and AT 1R. Mol Cell Endocrinol 2024; 594:112349. [PMID: 39233041 DOI: 10.1016/j.mce.2024.112349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
The vitamin D receptor (VDR) is associated with antioxidative and anti-inflammatory effects and modulation of the renin-angiotensin-aldosterone system. This study evaluated whether VDR agonist paricalcitol protects renal ischemia-reperfusion (IR) induced tubular injury in rats by evaluating: 1) ATP-dependent tubular Na+ transport; 2) renal redox signaling; 3) renal content of proinflammatory cytokines TNF-α and IL-6; and 4) renal content of renin and angiotensin II receptor type 1 (AT1R). Paricalcitol prevented IR-induced tubular injury, evidenced by the prevention of histopathological changes and renal fibrosis with preservation of the activity of ATP-dependent Na+ transporters in the renal cortex. Paricalcitol decreased renal oxidative stress by reducing NADPH oxidase activity and increasing catalase. Paricalcitol also decreased the renal content of TNF-α, IL-6, and AT1R. The NADPH oxidase inhibitor apocynin did not present additive protection to paricalcitol-induced effects. The protective effects of paricalcitol on tubular injury induced by renal IR may dependent on the modulation of redox and proinflammatory signaling and renal angiotensin II/AT1R signaling.
Collapse
Affiliation(s)
| | | | | | - Jeoadã Karollyne Silva
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil.
| | - José Anderson da Silva Gomes
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil; Department of Histology and Embriology, Federal University of Pernambuco, Recife, Brazil.
| | | | | | | | | | | | - Leucio Duarte Vieira
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife, Brazil.
| |
Collapse
|
2
|
Wu H, Sun Q, Yuan S, Wang J, Li F, Gao H, Chen X, Yang R, Xu J. AT1 Receptors: Their Actions from Hypertension to Cognitive Impairment. Cardiovasc Toxicol 2022; 22:311-325. [PMID: 35211833 PMCID: PMC8868040 DOI: 10.1007/s12012-022-09730-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022]
Abstract
Hypertension is one of the most prevalent cardiovascular disorders worldwide, affecting 1.13 billion people, or 14% of the global population. Hypertension is the single biggest risk factor for cerebrovascular dysfunction. According to the American Heart Association, high blood pressure (BP), especially in middle-aged individuals (~ 40 to 60 years old), is associated with an increased risk of dementia, later in life. Alzheimer’s disease and cerebrovascular disease are the two leading causes of dementia, accounting for around 80% of the total cases and usually combining mixed pathologies from both. Little is known regarding how hypertension affects cognitive function, so the impact of its treatment on cognitive impairment has been difficult to assess. The brain renin-angiotensin system (RAS) is essential for BP regulation and overactivity of this system has been established to precede the development and maintenance of hypertension. Angiotensin II (Ang-II), the main peptide within this system, induces vasoconstriction and impairs neuro-vascular coupling by acting on brain Ang-II type 1 receptors (AT1R). In this review, we systemically analyzed the association between RAS and biological mechanisms of cognitive impairment, from the perspective of AT1R located in the central nervous system. Additionally, the possible contribution of brain AT1R to global cognition decline in COVID-19 cases will be discussed as well.
Collapse
Affiliation(s)
- Hanxue Wu
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Qi Sun
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shenglan Yuan
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Jiawei Wang
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Fanni Li
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hongli Gao
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Rui Yang
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Jiaxi Xu
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China.
| |
Collapse
|
3
|
Kovács MG, Kovács ZZA, Varga Z, Szűcs G, Freiwan M, Farkas K, Kővári B, Cserni G, Kriston A, Kovács F, Horváth P, Földesi I, Csont T, Kahán Z, Sárközy M. Investigation of the Antihypertrophic and Antifibrotic Effects of Losartan in a Rat Model of Radiation-Induced Heart Disease. Int J Mol Sci 2021; 22:12963. [PMID: 34884782 PMCID: PMC8657420 DOI: 10.3390/ijms222312963] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/27/2022] Open
Abstract
Radiation-induced heart disease (RIHD) is a potential late side-effect of thoracic radiotherapy resulting in left ventricular hypertrophy (LVH) and fibrosis due to a complex pathomechanism leading to heart failure. Angiotensin-II receptor blockers (ARBs), including losartan, are frequently used to control heart failure of various etiologies. Preclinical evidence is lacking on the anti-remodeling effects of ARBs in RIHD, while the results of clinical studies are controversial. We aimed at investigating the effects of losartan in a rat model of RIHD. Male Sprague-Dawley rats were studied in three groups: (1) control, (2) radiotherapy (RT) only, (3) RT treated with losartan (per os 10 mg/kg/day), and were followed for 1, 3, or 15 weeks. At 15 weeks post-irradiation, losartan alleviated the echocardiographic and histological signs of LVH and fibrosis and reduced the overexpression of chymase, connective tissue growth factor, and transforming growth factor-beta in the myocardium measured by qPCR; likewise, the level of the SMAD2/3 protein determined by Western blot decreased. In both RT groups, the pro-survival phospho-AKT/AKT and the phospho-ERK1,2/ERK1,2 ratios were increased at week 15. The antiremodeling effects of losartan seem to be associated with the repression of chymase and several elements of the TGF-β/SMAD signaling pathway in our RIHD model.
Collapse
Affiliation(s)
- Mónika Gabriella Kovács
- Interdisciplinary Center of Excellence and MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (M.G.K.); (Z.Z.A.K.); (G.S.); (M.F.)
| | - Zsuzsanna Z. A. Kovács
- Interdisciplinary Center of Excellence and MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (M.G.K.); (Z.Z.A.K.); (G.S.); (M.F.)
| | - Zoltán Varga
- Department of Oncotherapy, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (Z.V.); (Z.K.)
| | - Gergő Szűcs
- Interdisciplinary Center of Excellence and MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (M.G.K.); (Z.Z.A.K.); (G.S.); (M.F.)
| | - Marah Freiwan
- Interdisciplinary Center of Excellence and MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (M.G.K.); (Z.Z.A.K.); (G.S.); (M.F.)
| | - Katalin Farkas
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (K.F.); (I.F.)
| | - Bence Kővári
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (B.K.); (G.C.)
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (B.K.); (G.C.)
| | - András Kriston
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary; (A.K.); (F.K.); (P.H.)
- Single-Cell Technologies Ltd., H-6726 Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FIN-00014 Helsinki, Finland
| | - Ferenc Kovács
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary; (A.K.); (F.K.); (P.H.)
- Single-Cell Technologies Ltd., H-6726 Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FIN-00014 Helsinki, Finland
| | - Péter Horváth
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary; (A.K.); (F.K.); (P.H.)
- Single-Cell Technologies Ltd., H-6726 Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FIN-00014 Helsinki, Finland
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (K.F.); (I.F.)
| | - Tamás Csont
- Interdisciplinary Center of Excellence and MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (M.G.K.); (Z.Z.A.K.); (G.S.); (M.F.)
| | - Zsuzsanna Kahán
- Department of Oncotherapy, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (Z.V.); (Z.K.)
| | - Márta Sárközy
- Interdisciplinary Center of Excellence and MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (M.G.K.); (Z.Z.A.K.); (G.S.); (M.F.)
| |
Collapse
|
4
|
Chen X, Liu F, Xue Q, Weng X, Xu F. Metastatic pancreatic cancer: Mechanisms and detection (Review). Oncol Rep 2021; 46:231. [PMID: 34498718 PMCID: PMC8444192 DOI: 10.3892/or.2021.8182] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer (PC) is a lethal malignancy. Its prevalence rate remains low but continues to grow each year. Among all stages of PC, metastatic PC is defined as late-stage (stage IV) PC and has an even higher fatality rate. Patients with PC do not have any specific clinical manifestations. Most cases are inoperable at the time-point of diagnosis. Prognosis is also poor even with curative-intent surgery. Complications during surgery, postoperative pancreatic fistula and recurrence with metastatic foci make the management of metastatic PC difficult. While extensive efforts were made to improve survival outcomes, further elucidation of the molecular mechanisms of metastasis poses a formidable challenge. The present review provided an overview of the mechanisms of metastatic PC, summarizing currently known signaling pathways (e.g. epithelial-mesenchymal transition, NF-κB and KRAS), imaging that may be utilized for early detection and biomarkers (e.g. carbohydrate antigen 19-9, prostate cancer-associated transcript-1, F-box/LRR-repeat protein 7 and tumor stroma), giving insight into promising therapeutic targets.
Collapse
Affiliation(s)
- Xiangling Chen
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Fangfang Liu
- Department of Art, Art College, Southwest Minzu University, Chengdu, Sichuan 610041, P.R. China
| | - Qingping Xue
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xiechuan Weng
- Department of Neuroscience, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Fan Xu
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
5
|
Bruijn LE, van den Akker BEWM, van Rhijn CM, Hamming JF, Lindeman JHN. Extreme Diversity of the Human Vascular Mesenchymal Cell Landscape. J Am Heart Assoc 2020; 9:e017094. [PMID: 33190596 PMCID: PMC7763765 DOI: 10.1161/jaha.120.017094] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
Background Human mesenchymal cells are culprit factors in vascular (patho)physiology and are hallmarked by phenotypic and functional heterogeneity. At present, they are subdivided by classic umbrella terms, such as "fibroblasts," "myofibroblasts," "smooth muscle cells," "fibrocytes," "mesangial cells," and "pericytes." However, a discriminative marker-based subclassification has to date not been established. Methods and Results As a first effort toward a classification scheme, a systematic literature search was performed to identify the most commonly used phenotypical and functional protein markers for characterizing and classifying vascular mesenchymal cell subpopulation(s). We next applied immunohistochemistry and immunofluorescence to inventory the expression pattern of identified markers on human aorta specimens representing early, intermediate, and end stages of human atherosclerotic disease. Included markers comprise markers for mesenchymal lineage (vimentin, FSP-1 [fibroblast-specific protein-1]/S100A4, cluster of differentiation (CD) 90/thymocyte differentiation antigen 1, and FAP [fibroblast activation protein]), contractile/non-contractile phenotype (α-smooth muscle actin, smooth muscle myosin heavy chain, and nonmuscle myosin heavy chain), and auxiliary contractile markers (h1-Calponin, h-Caldesmon, Desmin, SM22α [smooth muscle protein 22α], non-muscle myosin heavy chain, smooth muscle myosin heavy chain, Smoothelin-B, α-Tropomyosin, and Telokin) or adhesion proteins (Paxillin and Vinculin). Vimentin classified as the most inclusive lineage marker. Subset markers did not separate along classic lines of smooth muscle cell, myofibroblast, or fibroblast, but showed clear temporal and spatial diversity. Strong indications were found for presence of stem cells/Endothelial-to-Mesenchymal cell Transition and fibrocytes in specific aspects of the human atherosclerotic process. Conclusions This systematic evaluation shows a highly diverse and dynamic landscape for the human vascular mesenchymal cell population that is not captured by the classic nomenclature. Our observations stress the need for a consensus multiparameter subclass designation along the lines of the cluster of differentiation classification for leucocytes.
Collapse
Affiliation(s)
- Laura E. Bruijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | | | - Connie M. van Rhijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jaap F. Hamming
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jan H. N. Lindeman
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
6
|
Gibb AA, Lazaropoulos MP, Elrod JW. Myofibroblasts and Fibrosis: Mitochondrial and Metabolic Control of Cellular Differentiation. Circ Res 2020; 127:427-447. [PMID: 32673537 DOI: 10.1161/circresaha.120.316958] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiac fibrosis is mediated by the activation of resident cardiac fibroblasts, which differentiate into myofibroblasts in response to injury or stress. Although myofibroblast formation is a physiological response to acute injury, such as myocardial infarction, myofibroblast persistence, as occurs in heart failure, contributes to maladaptive remodeling and progressive functional decline. Although traditional pathways of activation, such as TGFβ (transforming growth factor β) and AngII (angiotensin II), have been well characterized, less understood are the alterations in mitochondrial function and cellular metabolism that are necessary to initiate and sustain myofibroblast formation and function. In this review, we highlight recent reports detailing the mitochondrial and metabolic mechanisms that contribute to myofibroblast differentiation, persistence, and function with the hope of identifying novel therapeutic targets to treat, and potentially reverse, tissue organ fibrosis.
Collapse
Affiliation(s)
- Andrew A Gibb
- From the Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Michael P Lazaropoulos
- From the Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - John W Elrod
- From the Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| |
Collapse
|
7
|
Wang A, Cao S, Aboelkassem Y, Valdez-Jasso D. Quantification of uncertainty in a new network model of pulmonary arterial adventitial fibroblast pro-fibrotic signalling. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2020; 378:20190338. [PMID: 32448066 PMCID: PMC7287331 DOI: 10.1098/rsta.2019.0338] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/16/2020] [Indexed: 05/21/2023]
Abstract
Here, we present a novel network model of the pulmonary arterial adventitial fibroblast (PAAF) that represents seven signalling pathways, confirmed to be important in pulmonary arterial fibrosis, as 92 reactions and 64 state variables. Without optimizing parameters, the model correctly predicted 80% of 39 results of input-output and inhibition experiments reported in 20 independent papers not used to formulate the original network. Parameter uncertainty quantification (UQ) showed that this measure of model accuracy is robust to changes in input weights and half-maximal activation levels (EC50), but is more affected by uncertainty in the Hill coefficient (n), which governs the biochemical cooperativity or steepness of the sigmoidal activation function of each state variable. Epistemic uncertainty in model structure, due to the reliance of some network components and interactions on experiments using non-PAAF cell types, suggested that this source of uncertainty had a smaller impact on model accuracy than the alternative of reducing the network to only those interactions reported in PAAFs. UQ highlighted model parameters that can be optimized to improve prediction accuracy and network modules where there is the greatest need for new experiments. This article is part of the theme issue 'Uncertainty quantification in cardiac and cardiovascular modelling and simulation'.
Collapse
Affiliation(s)
| | | | | | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92092, USA
| |
Collapse
|
8
|
Ogola BO, Zimmerman MA, Sure VN, Gentry KM, Duong JL, Clark GL, Miller KS, Katakam PVG, Lindsey SH. G Protein-Coupled Estrogen Receptor Protects From Angiotensin II-Induced Increases in Pulse Pressure and Oxidative Stress. Front Endocrinol (Lausanne) 2019; 10:586. [PMID: 31507536 PMCID: PMC6718465 DOI: 10.3389/fendo.2019.00586] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022] Open
Abstract
Our previous work showed that the G protein-coupled estrogen receptor (GPER) is protective in the vasculature and kidneys during angiotensin (Ang) II-dependent hypertension by inhibiting oxidative stress. The goal of the current study was to assess the impact of GPER deletion on sex differences in Ang II-induced hypertension and oxidative stress. Male and female wildtype and GPER knockout mice were implanted with radiotelemetry probes for measurement of baseline blood pressure before infusion of Ang II (700 ng/kg/min) for 2 weeks. Mean arterial pressure was increased to the same extent in all groups, but female wildtype mice were protected from Ang II-induced increases in pulse pressure, aortic wall thickness, and Nox4 mRNA. In vitro studies using vascular smooth muscle cells found that pre-treatment with the GPER agonist G-1 inhibited Ang II-induced ROS and NADP/NADPH. Ang II increased while G-1 decreased Nox4 mRNA and protein. The effects of Ang II were blocked by losartan and Nox4 siRNA, while the effects of G-1 were inhibited by adenylyl cyclase inhibition and mimicked by phosphodiesterase inhibition. We conclude that during conditions of elevated Ang II, GPER via the cAMP pathway suppresses Nox4 transcription to limit ROS production and prevent arterial stiffening. Taken together with our previous work, this study provides insight into how acute estrogen signaling via GPER provides cardiovascular protection during Ang II hypertension and potentially other diseases characterized by increased oxidative stress.
Collapse
Affiliation(s)
- Benard O. Ogola
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| | | | - Venkata N. Sure
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| | - Kaylee M. Gentry
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| | - Jennifer L. Duong
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| | - Gabrielle L. Clark
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, United States
| | - Kristin S. Miller
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, United States
| | | | - Sarah H. Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
- *Correspondence: Sarah H. Lindsey
| |
Collapse
|
9
|
Chen G, Pan SF, Cui XL, Liu LH. Puerarin attenuates angiotensin II-induced cardiac fibroblast proliferation via the promotion of catalase activity and the inhibition of hydrogen peroxide-dependent Rac-1 activation. Chin J Nat Med 2018; 16:41-52. [PMID: 29425589 DOI: 10.1016/s1875-5364(18)30028-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Indexed: 12/09/2022]
Abstract
The aims of the present study were to evaluate the effects of puerarin on angiotensin II-induced cardiac fibroblast proliferation and to explore the molecular mechanisms of action. Considering the role of H2O2 in nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation, we hypothesized that modulating catalase activity would be a potential target in regulating the redox-sensitive pathways. Our results showed that the activation of Rac1 was dependent on the levels of intracellular H2O2. Puerarin blocked the phosphorylation of extracellular regulated protein kinases (ERK)1/2, abolished activator protein (AP)-1 binding activity, and eventually attenuated cardiac fibroblast proliferation through the inhibition of H2O2-dependent Rac1 activation. Further studies revealed that angiotensin II treatment resulted in decreased catalase protein expression and enzyme activity, which was disrupted by puerarin via the upregulation of catalase protein expression at the transcriptional level and the prolonged protein degradation. These findings indicated that the anti-proliferation mechanism of puerarin was mainly through blocking angiontensin II-triggered downregulation of catalase expression and H2O2-dependent Rac1 activation.
Collapse
Affiliation(s)
- Gang Chen
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Shi-Fen Pan
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiang-Li Cui
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Li-Hong Liu
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
10
|
Peña C, Hernández-Fonseca JP, Pedreañez A, Viera N, Mosquera J. Renal oxidative stress and renal CD8(+) T-cell infiltration in mercuric chloride-induced nephropathy in rats: role of angiotensin II. J Immunotoxicol 2015; 13:324-34. [PMID: 26536500 DOI: 10.3109/1547691x.2015.1089960] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Mercuric chloride (HgCl2) induces kidney damage, in part, through oxidative stress. A role for angiotensin II (Ang II) in pro-inflammatory events in a model of acute HgCl2-induced nephropathy was reported. Ang II is a potent oxidative stress inducer; however, its role in oxidative/anti-oxidative events in HgCl2-induced nephropathy remains unknown. The aim of this study was to determine the role of Ang II in the oxidative stress and renal infiltration of CD8(+) T-cells after an acute HgCl2 intoxication. Three groups of Sprague Dawley rats were treated with a single subcutaneous dose of 2.5 mg/kg HgCl2: for 3 days prior to and for 4 days after that injection, rats in one group received Losartan (30 mg/kg), in another group Enalapril (30 mg/kg) or normal saline in the last group. Two other groups of drug-treated rats received saline in place of HgCl2. A final group of rats received saline in place of HgCl2 and the test drugs. All treatments were via gastric gavage. At 96 h after the vehicle/HgCl2 injection, blood and kidney samples were harvested. Renal sections were homogenized for measures of malondialdehyde (MDA), reduced glutathione (GSH) and catalase activity. Frozen sections were studied for the presence of superoxide anion ([Formula: see text]) and CD8(+) T-cells. HgCl2-treated rats had increased interstitial and tubular expression of [Formula: see text], high levels of MDA, normal catalase activity and GSH content, increased levels of interstitial CD8(+) T-cells and an increased percentage of necrotic tubules. Anti-Ang II treatments diminished the HgCl2-induced increases in interstitial [Formula: see text], CD8(+) T-cells and tubular damage and increased catalase and GSH expression above that due to HgCl2 alone; the HgCl2-induced high MDA levels were unaffected by the drugs. These data provide new information regarding the potential role of Ang II in the oxidative stress and renal CD8(+) T-cell infiltration that occur during HgCl2 nephropathy.
Collapse
Affiliation(s)
- Caterina Peña
- a Department of Genetics , School of Bioanalysis, Faculty of Medicine, Universidad of Zulia , Maracaibo , Venezuela
| | - Juan P Hernández-Fonseca
- b Dr. Américo Negrette Institute for Clinical Investigation, Faculty of Medicine, Universidad of Zulia , Maracaibo , Venezuela
| | - Adriana Pedreañez
- c Department of Immunology , School of Bioanalysis, Faculty of Medicine, Universidad of Zulia , Maracaibo , Venezuela and
| | - Ninoska Viera
- d Institute for Investigation, Faculty of Odontology, Universidad of Zulia , Maracaibo , Venezuela
| | - Jesús Mosquera
- b Dr. Américo Negrette Institute for Clinical Investigation, Faculty of Medicine, Universidad of Zulia , Maracaibo , Venezuela
| |
Collapse
|
11
|
Liu CF, Zhang J, Shen K, Gao PJ, Wang HY, Jin X, Meng C, Fang NY. Adventitial gene transfer of catalase attenuates angiotensin II-induced vascular remodeling. Mol Med Rep 2014; 11:2608-14. [PMID: 25503998 PMCID: PMC4337488 DOI: 10.3892/mmr.2014.3069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 11/19/2014] [Indexed: 02/07/2023] Open
Abstract
Vascular adventitia and adventitia-derived reactive oxygen species (ROS) contribute to vascular remodeling following vascular injury. A previous ex vivo study in adventitial fibroblasts showed that catalase, one of most important anti-oxide enzymes, was downregulated by angiotensin II (AngII). The aim of the present study was to investigate whether adventitial gene transfer of catalase affects AngII-induced vascular remodeling in vivo. Adenoviruses co-expressing catalase and enhanced green fluorescent protein (eGFP) or expressing eGFP only were applied to the adventitial surface of common carotid arteries of Sprague-Dawley rats. Alzet minipumps administering AngII (0.75 mg/kg/day) were then implanted subcutaneously for 14 days. Systolic blood pressure and biological parameters of vascular remodeling were measured in each group. Adventitial fibroblasts were cultured and p38 mitogen-activated protein kinase (MAPK) phosphorylation was measured using western blot analysis. The results showed that adventitial gene transfer of catalase had no effect on AngII-induced systolic blood pressure elevation. However, catalase adenovirus transfection significantly inhibited AngII-induced media hypertrophy compared with that of the control virus (P<0.05). In addition, catalase transfection significantly attenuated AngII-induced ROS generation, macrophage infiltration, collagen deposition and adventitial α-smooth muscle actin expression. Furthermore, catalase transfection significantly inhibited the AngII-induced increase in p38MAPK phosphorylation. In conclusion, the results of the present study demonstrated that adventitial gene transfer of catalase significantly attenuated AngII-induced vascular remodeling in rats via inhibition of adventitial p38MAPK phosphorylation.
Collapse
Affiliation(s)
- Cun-Fei Liu
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| | - Jia Zhang
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| | - Kai Shen
- Division of Cardiology, Zhoushan People's Hospital, Zhoushan, Zhejiang 316000, P.R. China
| | - Ping-Jin Gao
- Shanghai Key Laboratory of Vascular Biology at Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Hai-Ya Wang
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| | - Xin Jin
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| | - Chao Meng
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| | - Ning-Yuan Fang
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| |
Collapse
|
12
|
Yang T, Li X, Zhu W, Chen C, Sun Z, Tan Z, Kang J. Alteration of antioxidant enzymes and associated genes induced by grape seed extracts in the primary muscle cells of goats in vitro. PLoS One 2014; 9:e107670. [PMID: 25238394 PMCID: PMC4169554 DOI: 10.1371/journal.pone.0107670] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 08/22/2014] [Indexed: 11/20/2022] Open
Abstract
This study was conducted to investigate how the activity and expression of certain paramount antioxidant enzymes respond to grape seed extract (GSE) addition in primary muscle cells of goats. Gluteal primary muscle cells (PMCs) isolated from a 3-week old goat were cultivated as an unstressed cell model, or they were exposed to 100 µM H2O2 to establish a H2O2-stimulated cell model. The activities of catalase (CAT), superoxide dismutases (SOD) and glutathione peroxidases (GPx) in combination with other relevant antioxidant indexes [i.e., reduced glutathione (GSH) and total antioxidant capacity (TAOC)] in response to GSE addition were tested in the unstressed and H2O2-stimulated cell models, and the relative mRNA levels of the CAT, GuZu-SOD, and GPx-1 genes were measured by qPCR. In unstressed PMCs, GSE addition at the dose of 10 µg/ml strikingly attenuated the expression levels of CAT and CuZn-SOD as well as the corresponding enzyme activities. By contrast, in cells pretreated with 100 µM H2O2, the expression and activity levels of these two antioxidant enzymes were enhanced by GSE addition at 10 µg/ml. GSE addition promoted GPx activity in both unstressed and stressed PMCs, while the expression of the GPx 1 gene displayed partial divergence with GPx activity, which was mitigated by GSE addition at 10 µg/ml in unstressed PMCs. GSH remained comparatively stable except for GSE addition to H2O2-stimulated PMCs at 60 µg/ml, in which a dramatic depletion of GSH occurred. Moreover, GSE addition enhanced TAOC in unstressed (but not H2O2-stimulated) PMCs. GSE addition exerted a bidirectional modulating effect on the mRNA levels and activities of CAT and SOD in unstressed and stressed PMCs at a moderate dose, and it only exhibited a unidirectional effect on the promotion of GPx activity, reflecting its potential to improve antioxidant protection in ruminants.
Collapse
Affiliation(s)
- Tan Yang
- Key Laboratory for Bio-Feed and Molecular Nutrition, Southwest University, Chongqing, P. R. China
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P.R. China
| | - Xiaomin Li
- Key Laboratory for Bio-Feed and Molecular Nutrition, Southwest University, Chongqing, P. R. China
| | - Wang Zhu
- Key Laboratory for Bio-Feed and Molecular Nutrition, Southwest University, Chongqing, P. R. China
| | - Cheng Chen
- Key Laboratory for Bio-Feed and Molecular Nutrition, Southwest University, Chongqing, P. R. China
| | - Zhihong Sun
- Key Laboratory for Bio-Feed and Molecular Nutrition, Southwest University, Chongqing, P. R. China
- * E-mail: (ZS); (ZT)
| | - Zhiliang Tan
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P.R. China
- * E-mail: (ZS); (ZT)
| | - Jinghe Kang
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, P.R. China
| |
Collapse
|
13
|
Wang Z, Ren Z, Hu Z, Hu X, Zhang H, Wu H, Zhang M. Angiotensin-II induces phosphorylation of ERK1/2 and promotes aortic adventitial fibroblasts differentiating into myofibroblasts during aortic dissection formation. J Mol Histol 2013; 45:401-12. [DOI: 10.1007/s10735-013-9558-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/26/2013] [Indexed: 10/25/2022]
|
14
|
Rajamohan SB, Raghuraman G, Prabhakar NR, Kumar GK. NADPH oxidase-derived H(2)O(2) contributes to angiotensin II-induced aldosterone synthesis in human and rat adrenal cortical cells. Antioxid Redox Signal 2012; 17:445-59. [PMID: 22214405 PMCID: PMC3365360 DOI: 10.1089/ars.2011.4176] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The Renin-Angiotensin-Aldosterone-System plays a pivotal role in hypertension. Angiotensin II (Ang II) is a major regulator of aldosterone synthesis and secretion, and it is known to facilitate reactive oxygen species (ROS) generation in many cell types. AIMS Here, we assessed the role of ROS signaling in Ang II-induced aldosterone synthesis by focusing on the regulation of aldosterone synthase (CYP11B2), a cytochrome P450 oxidase that catalyzes the final step in aldosterone biosynthetic pathway. RESULTS Ang II increased CYP11B2 activity, mRNA and protein with a concomitant elevation of 6-Carboxy- 2',7'-dichlorodihydrofluorescein diacetate fluorescence, malondialdehyde and protein carbonyl levels (indices of ROS), NADPH oxidase (Nox) activity, and H(2)O(2) levels in human and rat adrenal cortical cells. The expression of nuclear receptor related 1 protein, a transcription factor known to regulate CYP11B2 expression, was also augmented by Ang II. These Ang II-evoked effects were either abolished or attenuated by pretreatment of cells with either Ang II type I receptor (AT(1)R) antagonist, or antioxidants or Nox inhibitor or siRNA silencing of Nox1, 2 and 4, or inhibitors of phospholipase C and protein kinase C. Exogenous H(2)O(2) mimicked the facilitatory effects of Ang II on CYP11B2 activity, mRNA, and protein expression, and these changes were significantly reduced by PEG-catalase. INNOVATION ROS, particularly H(2)O(2), is identified as a key regulator of aldosterone production. CONCLUSION Our results suggest that Ang II facilitates CYP11B2 activity and the ensuing aldosterone production via activation of AT(1)R-Nox-H(2)O(2) signaling pathway.
Collapse
Affiliation(s)
- Senthilkumar B Rajamohan
- Department of Medicine, Institute for Integrative Physiology, University of Chicago, Chicago, IL 60637-1470, USA
| | | | | | | |
Collapse
|
15
|
Li Y, Yan CH, Han YL. CREG mediated adventitial fibroblast phenotype modulation: A possible therapeutic target for proliferative vascular disease. Med Hypotheses 2012; 79:95-7. [DOI: 10.1016/j.mehy.2012.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 04/03/2012] [Indexed: 11/16/2022]
|