1
|
Morgado LAL, Rodrigues LMZ, Silva DCF, da Silva BD, Irigoyen MCC, Takano APC. NF-κB-Specific Suppression in Cardiomyocytes Unveils Aging-Associated Responses in Cardiac Tissue. Biomedicines 2025; 13:224. [PMID: 39857807 PMCID: PMC11762954 DOI: 10.3390/biomedicines13010224] [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: 12/20/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Aging is associated with structural and functional changes in the heart, including hypertrophy, fibrosis, and impaired contractility. Cellular mechanisms such as senescence, telomere shortening, and DNA damage contribute to these processes. Nuclear factor kappa B (NF-κB) has been implicated in mediating cellular responses in aging tissues, and increased NF-κB expression has been observed in the hearts of aging rodents. Therefore, NF-κB is suspected to play an important regulatory role in the cellular and molecular processes occurring in the heart during aging. This study investigates the in vivo role of NF-κB in aging-related cardiac alterations, focusing on senescence and associated cellular events. Methods: Young and old wild-type (WT) and transgenic male mice with cardiomyocyte-specific NF-κB suppression (3M) were used to assess cardiac function, morphology, senescence markers, lipofuscin deposition, DNA damage, and apoptosis. Results: Kaplan-Meier analysis revealed reduced survival in 3M mice compared to WT. Echocardiography showed evidence of eccentric hypertrophy, and both diastolic and systolic dysfunction in 3M mice. Both aged WT and 3M mice exhibited cardiac hypertrophy, with more pronounced hypertrophic changes in cardiomyocytes from 3M mice. Additionally, cardiac fibrosis, senescence-associated β-galactosidase activity, p21 protein expression, and DNA damage (marked by phosphorylated H2A.X) were elevated in aged WT and both young and aged 3M mice. Conclusions: The suppression of NF-κB in cardiomyocytes leads to pronounced cardiac remodeling, dysfunction, and cellular damage associated with the aging process. These findings suggest that NF-κB plays a critical regulatory role in cardiac aging, influencing both cellular senescence and molecular damage pathways. This has important implications for the development of therapeutic strategies aimed at mitigating age-related cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | - Bruno Durante da Silva
- Unidade de Hipertensao, Instituto do Coracao, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-000, Brazil
| | - Maria Claudia Costa Irigoyen
- Unidade de Hipertensao, Instituto do Coracao, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-000, Brazil
| | - Ana Paula Cremasco Takano
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| |
Collapse
|
2
|
Zhang S, Wang J, Wen J, Xin Q, Wang J, Ju Z, Luan Y. MSC-derived exosomes attenuates pulmonary hypertension via inhibiting pulmonary vascular remodeling. Exp Cell Res 2024; 442:114256. [PMID: 39299482 DOI: 10.1016/j.yexcr.2024.114256] [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: 07/18/2024] [Revised: 09/05/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a serious cardiopulmonary disease with significant morbidity and mortality. Vascular obstruction leads to a continuous increase in pulmonary vascular resistance, vascular remodeling, and right ventricular hypertrophy and failure, which are the main pathological features of PH. Currently, the treatments for PH are very limited, so new methods are urgently needed. Msenchymal stem cells-derived exosomes have been shown to have significant therapeutic effects in PH, however, the mechanism still very blurry. Here, we investigated the possible mechanism by which umbilical cord mesenchymal stem cell-derived exosomes (hUC-MSC-EXO) inhibited monocrotaline (MCT)-induced pulmonary vascular remodeling in a rat model of PH by regulating the NF-κB/BMP signaling pathway. Our data revealed that hUC-MSC-EXO could significantly attenuate MCT-induced PH and right ventricular hypertrophy. Moreover, the protein expression level of BMPR2, BMP-4, BMP-9 and ID1 was significantly increased, but NF-κB p65, p-NF-κB-p65 and BMP antagonists Gremlin-1 was increased in vitro and vivo. Collectively, this study revealed that the mechanism of hUC-MSC-EXO attenuates pulmonary hypertension may be related to inhibition of NF-κB signaling to further activation of BMP signaling. The present study provided a promising therapeutic strategy for PH vascular remodeling.
Collapse
Affiliation(s)
- Shanshan Zhang
- Department of Emergency, The Second Hospital of Shandong University, PR China
| | - Junfu Wang
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, PR China
| | - Jiang Wen
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China
| | - Qian Xin
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China
| | - Jue Wang
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China
| | - Zhiye Ju
- Department of Ultrasound, Shandong Provincial Public Health Clinical Center, No. 46, Lishan Road, Jinan, 250000, PR China.
| | - Yun Luan
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China.
| |
Collapse
|
3
|
Wang L, Tang T, Tian X, Peng C, Wu S. Animal models of pulmonary arterial hypertension associated with atrial septal defect. Sci Rep 2024; 14:18287. [PMID: 39112674 PMCID: PMC11306221 DOI: 10.1038/s41598-024-69002-5] [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: 01/05/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a well-known complication of congenital heart disease (CHD). The lack of a satisfactory animal model for PAH associated with CHD (PAH-CHD) has limited progress in understanding the pathogenesis of PAH and the development of therapeutic agents. The development of a rat model for PAH associated with atrial septal defect (ASD) was achieved through atrial septal puncture and thermal ablation. Two and 4 weeks after modeling, hematoxylin and eosin staining showed that the vascular thickness, vascular thickness index, vascular area, and vascular area index in pulmonary arteries with an outer diameter of 50-300 μm in the PAH-ASD 2 and 4 weeks group were higher than those in the sham group (all P < 0.05). Alpha-smooth muscle actin (ɑ-SMA) staining showed that the medial thickness, medial thickness index, medial area, and medial area index in pulmonary arteries with an outer diameter of 50-300 µm at 2 and 4 weeks after modeling were significantly higher than those in the sham group (all P < 0.05). Additionally, mean pulmonary arterial pressure (mPAP) and pulmonary vascular resistance (PVR) in the PAH-ASD 2 and 4 weeks groups were significantly higher than those in the sham group (both P < 0.05). Elastin van Gieson staining showed that the vascular obstruction score in the PAH-ASD 2 and 4 weeks group was significantly higher than that in the sham group (both P < 0.05). The PAH-ASD rats were successfully generated. These findings suggest that our model would be useful for further research into the pathogenesis, prevention, and treatment of PAH-ASD.
Collapse
Affiliation(s)
- Li Wang
- Department of Pediatrics, Guizhou Children's Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
| | - Ting Tang
- Department of Pediatrics, Guizhou Children's Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
| | - Xiaochun Tian
- Department of Pediatrics, Guizhou Children's Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
| | - Chang Peng
- Department of Pediatrics, Guizhou Children's Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China.
| | - Shuqi Wu
- Department of Pediatrics, Guizhou Children's Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
| |
Collapse
|
4
|
Burman P, Jaiswal R, Devi K, Moharana B. Aurintricarboxylic acid protects isoproterenol induced left ventricular hypertrophy by modulating TWEAK signaling. Cardiovasc Pathol 2022; 61:107468. [PMID: 35977688 DOI: 10.1016/j.carpath.2022.107468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cardiac hypertrophy is regarded as a compensation mechanism to overcome the increased workload. Aurintricarboxylic acid (ATA) is a derivative of quinomethanes and a selective inhibitor of TWEAK/Fn14 pathway. In this study, we investigated the effect of ATA on isoproterenol (ISO)-induced pathological cardiac hypertrophy. METHODS Cardiac hypertrophy in H9C2 cells was induced using ISO 20 μM dissolved in PBS. H9C2 cells were treated with ATA (5 µM, 10 µM, 20 µM) followed by ISO stimulation for 24 h. Male SD rats were injected ISO (5 mg/kg/day, s.c) for 21 days and followed by treatment with ATA (10 mg/kg, i.p.) for 14 days. Cardiac functions were assessed. After sacrifice, hearts were subjected to histopathological and western blot analysis. RESULTS In in-vitro results, upon ATA treatment, ICC results showed significant decrease in TWEAK and ANP expression. In in-vivo results, echocardiography showed significant restoration of cardiac function in ATA treated rats. Histopathological analysis showed a significant decrease in left ventricular wall thickness, cardiomyocytes width and reduced fibrosis in ATA treated rats. Western blotting showed decreased expression of the cardiac hypertrophy maker ANP, inflammatory markers including TWEAK and apoptotic markers after ATA treatment. CONCLUSION These findings suggested that the TWEAK/Fn14 pathway could be a potential target for therapeutic exploration in ISO induced cardiac hypertrophy. ATA, as an inhibitor of this pathway, exerted significant cardioprotective effect against ISO-induced cardiac hypertrophy in rats.
Collapse
Affiliation(s)
- Prabha Burman
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Rahul Jaiswal
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Kusum Devi
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, UP, 201002, India
| | - Baisakhi Moharana
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, UP, 201002, India.
| |
Collapse
|
5
|
Becker CU, Sartório CL, Campos-Carraro C, Siqueira R, Colombo R, Zimmer A, Belló-Klein A. Exercise training decreases oxidative stress in skeletal muscle of rats with pulmonary arterial hypertension. Arch Physiol Biochem 2022; 128:1330-1338. [PMID: 32449880 DOI: 10.1080/13813455.2020.1769679] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The effects of exercise training on oxidative stress in gastrocnemius of rats with pulmonary hypertension were studied. Four groups were established: sedentary control (SC), sedentary monocrotaline (SM), trained control (TC), trained monocrotaline (TM). Exercise was applied for 4 weeks, 5 days/week, 50-60 min/session, at 60% of VO2 max. Right ventricular (RV) pressures were measured, heart and gastrocnemius were removed for morphometric/biochemical analysis. Lipid peroxidation (LPO), H2O2, GSH/GSSG, and activity/expression of antioxidant enzymes were evaluated. Increased RV hypertrophy, systolic and end-diastolic pressures (RVEDP) were observed in SM animals, and the RVEDP was decreased in TM vs. SM. H2O2, SOD-1, and LPO were higher in the SM group than in SC. In TM, H2O2 was further increased when compared to SM, with a rise in antioxidant defences and a decrease in LPO. GSH/GSSG was higher only in the TC group. Exercise induced an efficient antioxidant adaptation, preventing oxidative damage to lipids.
Collapse
Affiliation(s)
- C U Becker
- Cardiovascular Physiology Laboratory, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - C L Sartório
- Department of Physiological Sciences, Federal University of Espírito Santo (UFES), Vitória, Brazil
| | - C Campos-Carraro
- Cardiovascular Physiology Laboratory, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - R Siqueira
- Cardiovascular Physiology Laboratory, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - R Colombo
- Pharmacology and Physiology Laboratory, University of Caxias do Sul, Caxias do Sul, Brazil
| | - A Zimmer
- Cardiovascular Physiology Laboratory, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - A Belló-Klein
- Cardiovascular Physiology Laboratory, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
6
|
Liu Y, Xu Y, Yao Y, Cao Y, Chen G, Cai Y, Chen W, Chen X, Qiu Z. I-κB kinase-ε deficiency improves doxorubicin-induced dilated cardiomyopathy by inhibiting the NF-κB pathway. Front Physiol 2022; 13:934899. [PMID: 35991177 PMCID: PMC9386238 DOI: 10.3389/fphys.2022.934899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/04/2022] [Indexed: 11/23/2022] Open
Abstract
Dilated cardiomyopathy (DCM) can lead to heart expansion and severe heart failure, but its specific pathogenesis is still elusive. In many cardiovascular diseases, I-κB kinase-ε (IKKε) has been recognized as a pro-inflammatory molecule. In this study, wild-type mice (WT, n = 14) and IKKε knockout mice (IKKε-KO, n = 14) were intraperitoneally injected with a cumulative dose of 25 mg/kg with Dox or Saline five times in 30 days. Finally, the experimental mice were divided into WT + Saline group、WT + DOX group、IKKε-KO + Saline group and IKKε-KO + Dox group. Echocardiography was performed to assess cardiac structure and function. Moreover, the mechanism was validated by immunohistochemistry and western blotting. Our results demonstrated that compared to WT + Dox mice, IKKε-KO + Dox mice exhibited attenuation of dilated cardiomyopathy-related morphological changes and alleviation of heart failure. Additionally, compared to the WT mice after Dox-injected, the expression of fibrosis and proinflammatory were decreased in IKKε-KO mice, and the expression of cardiac gap junction proteins was much higher in IKKε-KO mice. Further testing found that pyroptosis and apoptosis in the myocardium were also ameliorated in IKKε-KO mice compared to WT mice after Dox was injected. Mechanistically, our results showed that deficiency of IKKε might inhibit the phosphorylation of IκBα, p65, RelB, and p100 in mouse heart tissues after Dox stimulation. In summary, our research suggests that IKKε might play an essential role in the development of Dox-induced dilated cardiomyopathy and may be a potential target for the treatment of dilated cardiomyopathy in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xin Chen
- *Correspondence: Xin Chen, ; Zhibing Qiu,
| | | |
Collapse
|
7
|
Thakur D, Taliaferro O, Atkinson M, Stoffel R, Guleria RS, Gupta S. Inhibition of nuclear factor κB in the lungs protect bleomycin-induced lung fibrosis in mice. Mol Biol Rep 2022; 49:3481-3490. [PMID: 35083615 PMCID: PMC9174314 DOI: 10.1007/s11033-022-07185-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/20/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Pulmonary fibrosis is a debilitating condition with limited therapeutic avenues. The pathogenicity of pulmonary fibrosis constitutes involvement of cellular proliferation, activation, and transformational changes of fibroblast to myofibroblasts. It is a progressive lung disease and is primarily characterized by aberrant accumulation of extracellular matrix proteins in the lungs with poor prognosis. The inflammatory response in the pathogenesis of lung fibrosis is suggested because of release of several cytokines; however, the underlying mechanism remains undefined. A genetic model is the appropriate way to delineate the underlying mechanism of pulmonary fibrosis. METHODS AND RESULTS In this report, we have used cc-10 promoter based IκBα mutant mice (IKBM, an inhibitor of NF-κB) which were challenged with bleomycin (BLM). Compared to wild-type (WT) mice, the IKBM mice showed significant reduction in several fibrotic, vascular, and inflammatory genes. Moreover, we have identified a new set of dysregulated microRNAs (miRNAs) by miRNA array analysis in BLM-induced WT mice. Among these miRNAs, let-7a-5p and miR-503-5p were further analyzed. Our data showed that these two miRNAs were upregulated in WT-BLM and were reduced in IKBM-BLM mice. Bioinformatic analyses showed that let-7a-5p and miR-503-5p target for endothelin1 and bone morphogenic receptor 1A (BMPR1A), respectively, and were downregulated in WT-BLM mice indicating a link in pulmonary fibrosis. CONCLUSION We concluded that inhibition of NF-κB and modulation of let-7a-5p and miR-503-5p contribute a pivotal role in pulmonary fibrosis and may be considered as possible therapeutic target for the clinical management of lung fibrosis.
Collapse
Affiliation(s)
- Devaang Thakur
- Department of Biology, Baylor University, 101 Bagby Avenue, Waco, TX, 76706, US
| | - Olivia Taliaferro
- Department of Biology, Baylor University, 101 Bagby Avenue, Waco, TX, 76706, US
| | - Madeleine Atkinson
- Department of Biology, Baylor University, 101 Bagby Avenue, Waco, TX, 76706, US
| | - Ryan Stoffel
- Animal Facility, Baylor University, Baylor University, 101 Bagby Avenue, Waco, TX, 76706, US
| | - Rakeshwar S Guleria
- Biomarkers and Genetics Core, VISN 17 Center of Excellence On Returning War Veterans, 4800 Memorial Drive, Waco, TX, 76711, US.,Institute of Biomedical Studies, Baylor University, Waco, TX, 76798, US
| | - Sudhiranjan Gupta
- Biomarkers and Genetics Core, VISN 17 Center of Excellence On Returning War Veterans, 4800 Memorial Drive, Waco, TX, 76711, US. .,Department of Biology, Baylor University, 101 Bagby Avenue, Waco, TX, 76706, US. .,Animal Facility, Baylor University, Baylor University, 101 Bagby Avenue, Waco, TX, 76706, US.
| |
Collapse
|
8
|
Li YL, Li YQ, Zeng FQ, Lin XY, Li XT, Ren XQ, Yang DL. Sildenafil improves right ventricular remodelling in monocrotaline-induced rats by decreasing myocardial apoptosis and activating peroxisome proliferator-activated receptors. J Pharm Pharmacol 2021; 73:145-151. [PMID: 33793805 DOI: 10.1093/jpp/rgaa017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/05/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVES To assess the effect of sildenafil on monocrotaline-induced right ventricular (RV) remodeling and investigate the possible mechanism. METHODS Rats were subcutaneously injected with monocrotaline to establish an RV remodeling model and then administered sildenafil (25 mg/kg) from days 1 to 28. After 28 days of administration, the RV systolic pressure and the RV hypertrophy index (RVHI) were measured. The morphology of the right ventricle was observed by H&E staining. The ultrastructure of the right ventricle was observed using a transmission electron microscope. The myocardial apoptosis of the right ventricle was evaluated by TUNEL staining. The protein expression of apoptosis-related proteins and PPARs were examined by western blotting. KEY FINDINGS The results indicated that sildenafil decreased the RV systolic pressure and RVHI, and improved the microstructure and ultrastructure of the right ventricle in monocrotaline-induced rats. In addition, sildenafil suppressed myocardial apoptosis and promoted the protein expression of PPARs of the right ventricle in monocrotaline-induced rats. CONCLUSION Sildenafil inhibits RV remodeling in monocrotaline-induced rats, which might be partially mediated by reducing myocardial apoptosis and activating PPARs.
Collapse
Affiliation(s)
- Ye-Li Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yi-Qi Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| | - Fan-Qun Zeng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Xiao-Ying Lin
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Xiao-Tong Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Xing-Qiao Ren
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Dan-Li Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
9
|
Huertas A, Tu L, Humbert M, Guignabert C. Chronic inflammation within the vascular wall in pulmonary arterial hypertension: more than a spectator. Cardiovasc Res 2020; 116:885-893. [PMID: 31813986 DOI: 10.1093/cvr/cvz308] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/08/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
This review seeks to provide an update of preclinical findings and available clinical data on the chronic persistent inflammation and its direct role on the pulmonary arterial hypertension (PAH) progression. We reviewed the different mechanisms by which the inflammatory and immune pathways contribute to the structural and functional changes occurring in the three vascular compartments: the tunica intima, tunica media, and tunica adventitia. We also discussed how these inflammatory mediator changes may serve as a biomarker of the PAH progression and summarize unanswered questions and opportunities for future studies in this area.
Collapse
Affiliation(s)
- Alice Huertas
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133, Avenue de la Résistance; 92350 Le Plessis-Robinson, France.,Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France.,Service de Pneumologie, AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Ly Tu
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133, Avenue de la Résistance; 92350 Le Plessis-Robinson, France.,Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
| | - Marc Humbert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133, Avenue de la Résistance; 92350 Le Plessis-Robinson, France.,Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France.,Service de Pneumologie, AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133, Avenue de la Résistance; 92350 Le Plessis-Robinson, France.,Faculté de Médecine, Université Paris-Sud and Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
| |
Collapse
|
10
|
Krishnan S, Stearman RS, Zeng L, Fisher A, Mickler EA, Rodriguez BH, Simpson ER, Cook T, Slaven JE, Ivan M, Geraci MW, Lahm T, Tepper RS. Transcriptomic modifications in developmental cardiopulmonary adaptations to chronic hypoxia using a murine model of simulated high-altitude exposure. Am J Physiol Lung Cell Mol Physiol 2020; 319:L456-L470. [PMID: 32639867 DOI: 10.1152/ajplung.00487.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mechanisms driving adaptive developmental responses to chronic high-altitude (HA) exposure are incompletely known. We developed a novel rat model mimicking the human condition of cardiopulmonary adaptation to HA starting at conception and spanning the in utero and postnatal timeframe. We assessed lung growth and cardiopulmonary structure and function and performed transcriptome analyses to identify mechanisms facilitating developmental adaptations to chronic hypoxia. To generate the model, breeding pairs of Sprague-Dawley rats were exposed to hypobaric hypoxia (equivalent to 9,000 ft elevation). Mating, pregnancy, and delivery occurred in hypoxic conditions. Six weeks postpartum, structural and functional data were collected in the offspring. RNA-Seq was performed on right ventricle (RV) and lung tissue. Age-matched breeding pairs and offspring under room air (RA) conditions served as controls. Hypoxic rats exhibited significantly lower body weights and higher hematocrit levels, alveolar volumes, pulmonary diffusion capacities, RV mass, and RV systolic pressure, as well as increased pulmonary artery remodeling. RNA-Seq analyses revealed multiple differentially expressed genes in lungs and RVs from hypoxic rats. Although there was considerable similarity between hypoxic lungs and RVs compared with RA controls, several upstream regulators unique to lung or RV were identified. We noted a pattern of immune downregulation and regulation patterns of immune and hormonal mediators similar to the genome from patients with pulmonary arterial hypertension. In summary, we developed a novel murine model of chronic hypoxia exposure that demonstrates functional and structural phenotypes similar to human adaptation. We identified transcriptomic alterations that suggest potential mechanisms for adaptation to chronic HA.
Collapse
Affiliation(s)
- Sheila Krishnan
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Robert S Stearman
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lily Zeng
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Amanda Fisher
- Department of Anesthesiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Elizabeth A Mickler
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Brooke H Rodriguez
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Edward R Simpson
- Department of BioHealth Informatics, School of Informatics and Computing, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana.,Center for Computational Biology and Bioinformatics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Todd Cook
- Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana
| | - James E Slaven
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Medicine, Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mircea Ivan
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mark W Geraci
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Tim Lahm
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Robert S Tepper
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
11
|
Koudstaal T, Boomars KA, Kool M. Pulmonary Arterial Hypertension and Chronic Thromboembolic Pulmonary Hypertension: An Immunological Perspective. J Clin Med 2020; 9:E561. [PMID: 32092864 PMCID: PMC7074374 DOI: 10.3390/jcm9020561] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/10/2020] [Accepted: 02/16/2020] [Indexed: 12/24/2022] Open
Abstract
Pulmonary hypertension (PH) is a debilitating progressive disease characterized by increased pulmonary arterial pressures, leading to right ventricular (RV) failure, heart failure and, eventually, death. Based on the underlying conditions, PH patients can be subdivided into the following five groups: (1) pulmonary arterial hypertension (PAH), (2) PH due to left heart disease, (3) PH due to lung disease, (4) chronic thromboembolic PH (CTEPH), and (5) PH with unclear and/or multifactorial mechanisms. Currently, even with PAH-specific drug treatment, prognosis for PAH and CTEPH patients remains poor, with mean five-year survival rates of 57%-59% and 53%-69% for PAH and inoperable CTEPH, respectively. Therefore, more insight into the pathogenesis of PAH and CTEPH is highly needed, so that new therapeutic strategies can be developed. Recent studies have shown increased presence and activation of innate and adaptive immune cells in both PAH and CTEPH patients. Moreover, extensive biomarker research revealed that many inflammatory and immune markers correlate with the hemodynamics and/or prognosis of PAH and CTEPH patients. Increased evidence of the pathological role of immune cells in innate and adaptive immunity has led to many promising pre-clinical interventional studies which, in turn, are leading to innovative clinical trials which are currently being performed. A combination of immunomodulatory therapies might be required besides current treatment based on vasodilatation alone, to establish an effective treatment and prevention of progression for this disease. In this review, we describe the recent progress on our understanding of the involvement of the individual cell types of the immune system in PH. We summarize the accumulating body of evidence for inflammation and immunity in the pathogenesis of PH, as well as the use of inflammatory biomarkers and immunomodulatory therapy in PAH and CTEPH.
Collapse
Affiliation(s)
- Thomas Koudstaal
- Department of Pulmonary Medicine, Erasmus MC, Doctor Molenwaterplein 40, 3015 GD Rotterdam, The Netherlands; (K.A.B.); (M.K.)
| | | | | |
Collapse
|
12
|
Involvement of fatty acid synthase in right ventricle dysfunction in pulmonary hypertension. Exp Cell Res 2019; 383:111569. [DOI: 10.1016/j.yexcr.2019.111569] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 08/09/2019] [Accepted: 08/20/2019] [Indexed: 10/26/2022]
|
13
|
Abstract
Pulmonary hypertension (PH) and its severe subtype pulmonary arterial hypertension (PAH) encompass a set of multifactorial diseases defined by sustained elevation of pulmonary arterial pressure and pulmonary vascular resistance leading to right ventricular failure and subsequent death. Pulmonary hypertension is characterized by vascular remodeling in association with smooth muscle cell proliferation of the arterioles, medial thickening, and plexiform lesion formation. Despite our recent advances in understanding its pathogenesis and related therapeutic discoveries, PH still remains a progressive disease without a cure. Nevertheless, development of drugs that specifically target molecular pathways involved in disease pathogenesis has led to improvement in life quality and clinical outcomes in patients with PAH. There are presently more than 12 Food and Drug Administration-approved vasodilator drugs in the United States for the treatment of PAH; however, mortality with contemporary therapies remains high. More recently, there have been exuberant efforts to develop new pharmacologic therapies that target the fundamental origins of PH and thus could represent disease-modifying opportunities. This review aims to summarize recent developments on key signaling pathways and molecular targets that drive PH disease progression, with emphasis on new therapeutic options under development.
Collapse
Affiliation(s)
- Chen-Shan Chen Woodcock
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen Y. Chan
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Ren X, Johns RA, Gao WD. EXPRESS: Right Heart in Pulmonary Hypertension: From Adaptation to Failure. Pulm Circ 2019; 9:2045894019845611. [PMID: 30942134 PMCID: PMC6681271 DOI: 10.1177/2045894019845611] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/27/2019] [Indexed: 01/24/2023] Open
Abstract
Right ventricular (RV) failure (RVF) has garnered significant attention in recent years because of its negative impact on clinical outcomes in patients with pulmonary hypertension (PH). PH triggers a series of events, including activation of several signaling pathways that regulate cell growth, metabolism, extracellular matrix remodeling, and energy production. These processes render the RV adaptive to PH. However, RVF develops when PH persists, accompanied by RV ischemia, alterations in substrate and mitochondrial energy metabolism, increased free oxygen radicals, increased cell loss, downregulation of adrenergic receptors, increased inflammation and fibrosis, and pathologic microRNAs. Diastolic dysfunction is also an integral part of RVF. Emerging non-invasive technologies such as molecular or metallic imaging, cardiac MRI, and ultrafast Doppler coronary flow mapping will be valuable tools to monitor RVF, especially the transition to RVF. Most PH therapies cannot treat RVF once it has occurred. A variety of therapies are available to treat acute and chronic RVF, but they are mainly supportive, and no effective therapy directly targets the failing RV. Therapies that target cell growth, cellular metabolism, oxidative stress, and myocyte regeneration are being tested preclinically. Future research should include establishing novel RVF models based on existing models, increasing use of human samples, creating human stem cell-based in vitro models, and characterizing alterations in cardiac excitation–contraction coupling during transition from adaptive RV to RVF. More successful strategies to manage RVF will likely be developed as we learn more about the transition from adaptive remodeling to maladaptive RVF in the future.
Collapse
Affiliation(s)
- Xianfeng Ren
- Department of Anesthesiology,
China-Japan
Friendship Hospital, Beijing, China
| | - Roger A. Johns
- Department of Anesthesiology and
Critical Care Medicine,
Johns
Hopkins University School of Medicine,
Baltimore, MD, USA
| | - Wei Dong Gao
- Department of Anesthesiology and
Critical Care Medicine,
Johns
Hopkins University School of Medicine,
Baltimore, MD, USA
| |
Collapse
|
15
|
Pyruvate dehydrogenase activation precedes the down-regulation of fatty acid oxidation in monocrotaline-induced myocardial toxicity in mice. Heart Vessels 2018; 34:545-555. [DOI: 10.1007/s00380-018-1293-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/26/2018] [Indexed: 12/27/2022]
|
16
|
Yoshida K, Abe K, Ishikawa M, Saku K, Shinoda-Sakamoto M, Ishikawa T, Watanabe T, Oka M, Sunagawa K, Tsutsui H. Inhibition of TLR9-NF-κB-mediated sterile inflammation improves pressure overload-induced right ventricular dysfunction in rats. Cardiovasc Res 2018; 115:658-668. [DOI: 10.1093/cvr/cvy209] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 06/23/2018] [Accepted: 09/14/2018] [Indexed: 12/16/2022] Open
Abstract
Abstract
Aims
Recent accumulating evidence suggests that sterile inflammation plays a crucial role in the progression of various cardiovascular diseases. However, its contribution to right ventricular (RV) dysfunction remains unknown. The aim of this study was to elucidate whether toll-like receptor 9 (TLR9)-NF-κB-mediated sterile inflammation plays a critical role in the pathogenesis of RV dysfunction.
Methods and results
We performed main pulmonary artery banding (PAB) in rats to induce RV pressure overload and dysfunction. On Day 14 after PAB, the pressure overload impaired RV function as indicated by increased RV end-diastolic pressure concomitant with macrophage infiltration and fibrosis, as well as maximal activation of NF-κB and TLR9. Short-term administration (days 14–16 after PAB) of a specific TLR9 inhibitor, E6446, or an NF-κB inhibitor, pyrrolidine dithiocarbamate (PDTC) significantly attenuated NF-κB activation. Furthermore, long-term administration of E6446 (treatment: days 14–28) or PDTC (prevention: days −1 to 28; treatment: days 14 to 28) improved RV dysfunction associated with mitigated macrophage infiltration and fibrosis in right ventricle and decreased serum brain natriuretic peptide levels.
Conclusion
Inhibition of TLR9-NF-κB pathway-mediated sterile inflammation improved PAB-induced RV dysfunction in rats. This pathway plays a major role in the progression of pressure overload-induced RV dysfunction and is potentially a novel therapeutic target for the disorder.
Collapse
Affiliation(s)
- Keimei Yoshida
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kohtaro Abe
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mariko Ishikawa
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Anesthesiology and Critical Care Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Keita Saku
- Department of Therapeutic Regulation of Cardiovascular Homeostasis, Center for Disruptive Cardiovascular Medicine, Kyushu University, Fukuoka, Japan
| | - Masako Shinoda-Sakamoto
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tomohito Ishikawa
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takanori Watanabe
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masahiko Oka
- Department of Pharmacology and Medicine and Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Kenji Sunagawa
- Department of Therapeutic Regulation of Cardiovascular Homeostasis, Center for Disruptive Cardiovascular Medicine, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
17
|
Dewachter L, Dewachter C. Inflammation in Right Ventricular Failure: Does It Matter? Front Physiol 2018; 9:1056. [PMID: 30177883 PMCID: PMC6109764 DOI: 10.3389/fphys.2018.01056] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/16/2018] [Indexed: 01/22/2023] Open
Abstract
Right ventricular (RV) failure is a common consequence of acute and chronic RV overload of pressure, such as after pulmonary embolism and pulmonary hypertension. It has been recently realized that symptomatology and survival of patients with pulmonary hypertension are essentially determined by RV function adaptation to increased afterload. Therefore, improvement of RV function and reversal of RV failure are treatment goals. Currently, the pathophysiology and the pathobiology underlying RV failure remain largely unknown. A better understanding of the pathophysiological processes involved in RV failure is needed, as there is no proven treatment for this disease at the moment. The present review aims to summarize the current understanding of the pathogenesis of RV failure, focusing on inflammation. We attempt to formally emphasize the importance of inflammation and associated representative inflammatory molecules and cells in the primum movens and development of RV failure in humans and in experimental models. We present inflammatory biomarkers and immune mediators involved in RV failure. We focus on inflammatory mediators and cells which seem to correlate with the deterioration of RV function and also explain how all these inflammatory mediators and cells might impact RV function adaptation to increased afterload. Finally, we also discuss the evidence on potential beneficial effects of targeted anti-inflammatory agents in the setting of acute and chronic RV failure.
Collapse
Affiliation(s)
- Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Céline Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Cardiology, Erasmus Academic Hospital, Brussels, Belgium
| |
Collapse
|
18
|
Shi R, Zhu D, Wei Z, Fu N, Wang C, Liu L, Zhang H, Liang Y, Xing J, Wang X, Wang Y. Baicalein attenuates monocrotaline-induced pulmonary arterial hypertension by inhibiting endothelial-to-mesenchymal transition. Life Sci 2018; 207:442-450. [PMID: 29969608 DOI: 10.1016/j.lfs.2018.06.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/23/2018] [Accepted: 06/28/2018] [Indexed: 12/20/2022]
Abstract
AIMS Endothelial-to-mesenchymal transition (EndoMT) was shown to lead to endothelial cell (EC) dysfunction in pulmonary arterial hypertension (PAH). Baicalein was reported to inhibit epithelial-to-mesenchymal transition (EMT), a biological process that has many regulatory pathways in common with EndoMT. Whether it can attenuate PAH by inhibiting EndoMT remains obscure. MAIN METHODS PAH was induced by a single subcutaneous injection of MCT (60 mg/kg) in male Sprague Dawley rats. Two weeks after MCT administration, the rats in the treatment groups received baicalein orally (50 or 100 mg/kg/day) for an additional 2 weeks. Hemodynamic changes and right ventricular hypertrophy (RVH) were evaluated on day 28. Cardiopulmonary interstitial fibrosis was detected using Masson's trichrome, Picrosirius-red, and immunohistochemical staining. The reactivity of pulmonary arteries (PAs) was examined ex vivo. The protein expresson of EndoMT molecules, bone morphogenetic protein receptor 2 (BMPR2), and nuclear factor-κB (NF-κB) was examined to explore the mechanism of protective action of baicalein. KEY FINDINGS Baicalein (50 and 100 mg/kg) significantly alleviated MCT-induced PAH and cardiopulmonary interstitial fibrosis. Furthermore, baicalein treatment enhanced PA responsiveness to acetylcholine (ACh) in PAH rats. The upregulation of EndoMT molecules (N-cadherin, vimentin, Snail, and Slug) strongly suggest that EndoMT participates in MCT-induced PAH, which was reversed by baicalein (50 and 100 mg/kg) treatment. Moreover, baicalein partially reversed MCT-induced reductions in BMPR2 and NF-κB activation in the PAs. SIGNIFICANCE Baicalein attenuated MCT-induced PAH in rats by inhibiting EndoMT partially via the NF-κB-BMPR2 pathway. Thus, baicalein might be considered as a promising treatment option for PAH.
Collapse
Affiliation(s)
- Ruizan Shi
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China.
| | - Diying Zhu
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China
| | - Zehui Wei
- Department of Pharmacology, Peace Hospital Affiliated to Changzhi Medical College, Changzhi 046000, China
| | - Naijie Fu
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China
| | - Chang Wang
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China
| | - Linhong Liu
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China
| | - Huifeng Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China
| | - Yueqin Liang
- Medical Functional Experimental Center, Shanxi Medical University, Taiyuan 030001, China
| | - Jianfeng Xing
- Medical Functional Experimental Center, Shanxi Medical University, Taiyuan 030001, China
| | - Xuening Wang
- Department of Cardiovascular Surgery, Shanxi Academy of Medical Sciences, Shanxi Dayi Hospital, Taiyuan 030032, China
| | - Yan Wang
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
19
|
Scott TE, Kemp-Harper BK, Hobbs AJ. Inflammasomes: a novel therapeutic target in pulmonary hypertension? Br J Pharmacol 2018; 176:1880-1896. [PMID: 29847700 DOI: 10.1111/bph.14375] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/26/2018] [Accepted: 05/18/2018] [Indexed: 01/01/2023] Open
Abstract
Pulmonary hypertension (PH) is a rare, progressive pulmonary vasculopathy characterized by increased mean pulmonary arterial pressure, pulmonary vascular remodelling and right ventricular failure. Current treatments are not curative, and new therapeutic strategies are urgently required. Clinical and preclinical evidence has established that inflammation plays a key role in PH pathogenesis, and recently, inflammasomes have been suggested to be central to this process. Inflammasomes are important regulators of inflammation, releasing the pro-inflammatory cytokines IL-1β and IL-18 in response to exogenous pathogen- and endogenous damage-associated molecular patterns. These cytokines are elevated in PH patients, but whether this is a consequence of inflammasome activation remains to be determined. This review will briefly summarize current PH therapies and their pitfalls, introduce inflammasomes and the mechanisms by which they promote inflammation and, finally, highlight the preclinical and clinical evidence for the potential involvement of inflammasomes in PH pathobiology and how they may be targeted therapeutically. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Tara Elizabeth Scott
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK.,Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Barbara K Kemp-Harper
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| |
Collapse
|
20
|
AT1 receptor blockage impairs NF-κB activation mediated by thyroid hormone in cardiomyocytes. Pflugers Arch 2017; 470:549-558. [DOI: 10.1007/s00424-017-2088-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/06/2017] [Accepted: 11/15/2017] [Indexed: 10/18/2022]
|
21
|
Li Y, Li Y, Shi F, Wang L, Li L, Yang D. Osthole attenuates right ventricular remodeling via decreased myocardial apoptosis and inflammation in monocrotaline-induced rats. Eur J Pharmacol 2017; 818:525-533. [PMID: 29146527 DOI: 10.1016/j.ejphar.2017.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/08/2017] [Accepted: 11/03/2017] [Indexed: 11/29/2022]
Abstract
Osthole (Ost) is a coumarin that exhibits wide pharmacological effects in the cardiovascular system. However, whether Ost can inhibit apoptosis and inflammation in right ventricle (RV) cardiomyocytes and prevent RV remodeling is not clear. This study was designed to investigate the effect of Ost on RV remodeling and the underlying mechanism. By applying a monocrotaline (MCT)-induced rat model, the effect of Ost on RV remodeling was investigated. Rats were given a single dose of MCT (50mg/kg) subcutaneously (s.c.) to establish the RV remodeling model, followed by treatment with 10 or 20mg/kg Ost via daily gavage for 28 days. The RV pressure was measured, and a histological analysis was performed. The results suggested that Ost remarkably decreased RV pressure and improved myocardial hypertrophy and mitochondrial swelling, vacuolization, and sarcoplasmic reticulum enlargement when compared with the model group. To further investigate the roles of apoptosis and inflammation in the effects of Ost on MCT-induced RV remodeling, apoptosis-related factors and inflammatory-associated factors were examined by western blot. Ost was found to inhibit myocardial apoptosis and inflammation in the RV. Overall, the present results indicate that Ost suppresses the RV remodeling process induced by MCT in rats, which may be at least partially mediated through the reduction of myocardial apoptosis and inflammation.
Collapse
Affiliation(s)
- Yeli Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Yiqi Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai, Guangdong 519041, China
| | - Fuguo Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Lina Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Lisheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Danli Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, China.
| |
Collapse
|
22
|
Abstract
Background Cardiac fibrosis occurs because of disruption of the extracellular matrix network leading to myocardial dysfunction. Angiotensin II has been implicated in the development of cardiac fibrosis. Recently, microRNAs have been identified as an attractive target for therapeutic intervention in cardiac pathologies; however, the underlying mechanism of microRNAs in cardiac fibrosis remains unclear. MicroRNA‐130a (miR‐130a) has been shown to participate in angiogenesis and cardiac arrhythmia; however, its role in cardiac fibrosis is unknown. Methods and Results In this study, we found that miR‐130a was significantly upregulated in angiotensin II‐infused mice. The in vivo inhibition of miR‐130a by locked nucleic acid– based anti‐miR‐130a in mice significantly reduced angiotensin II‐induced cardiac fibrosis. Upregulation of miR‐130a was confirmed in failing human hearts. Overexpressing miR‐130a in cardiac fibroblasts promoted profibrotic gene expression and myofibroblasts differentiation, and the inhibition of miR‐130a reversed the processes. Using the constitutive and dominant negative constructs of peroxisome proliferator‐activated receptor γ 3‐′untranslated region (UTR), data revealed that the protective mechanism was associated with restoration of peroxisome proliferator‐activated receptor γ level leading to the inhibition of angiotensin II‐induced cardiac fibrosis. Conclusions Our findings provide evidence that miR‐130a plays a critical role in cardiac fibrosis by directly targeting peroxisome proliferator‐activated receptor γ. We conclude that inhibition of miR‐130a would be a promising strategy for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Li Li
- Department of Medical Physiology, Texas A & M Health Science Center, Central Texas Veterans Health Care System, Temple, TX.,Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Kelsey R Bounds
- Division of Nephrology and Hypertension, Department of Internal Medicine, Baylor Scott White Health, Temple, TX
| | - Piyali Chatterjee
- Division of Nephrology and Hypertension, Department of Internal Medicine, Baylor Scott White Health, Temple, TX
| | - Sudhiranjan Gupta
- Department of Medical Physiology, Texas A & M Health Science Center, Central Texas Veterans Health Care System, Temple, TX
| |
Collapse
|
23
|
Ribeiro EL, Fragoso IT, Gomes FODS, Oliveira AC, Silva AKSE, Silva PME, Ciambarella BT, Ramos IPR, Peixoto CA. Diethylcarbamazine: A potential treatment drug for pulmonary hypertension? Toxicol Appl Pharmacol 2017; 333:92-99. [PMID: 28851623 DOI: 10.1016/j.taap.2017.08.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/09/2017] [Accepted: 08/25/2017] [Indexed: 01/16/2023]
Abstract
The present study demonstrated the potential effects of diethylcarbamazine (DEC) on monocrotaline (MCT)-induced pulmonary hypertension. MCT solution (600mg/kg) was administered once per week, and 50mg/kg body weight of DEC for 28days. Three C57Bl/6 male mice groups (n=10) were studied: Control; MCT28, and MCT28/DEC. Echocardiography analysis was performed and lung tissues were collected for light microscopy (hematoxylin-eosin and Masson's trichrome staining), immunohistochemistry (αSMA, FADD, caspase 8, caspase 3, BAX, BCL2, cytochrome C and caspase 9) western blot (FADD, caspase 8, caspase 3, BAX, BCL2, cytochrome C and caspase 9) and qRt-PCR (COL-1α and αSMA). Echocardiography analysis demonstrated an increase in the pulmonary arterial blood flow gradient and velocity in the systole and RV area in the MCT28 group, while treatment with DEC resulted in a significant reduction in these parameters. Deposition of collagen fibers and αSMA staining around the pulmonary arteries was evident in the MCT28 group, while treatment with DEC reduced both. Western blot analysis revealed a decrease in BMPR2 in the MCT28 group, in contrast DEC treatment resulted in a significant increase in the level of BMPR2. DEC also significantly reduced the level of VEGF compared to the MCT28 group. Apoptosis extrinsic and intrinsic pathway markers were reduced in the MCT28 group. After treatment with DEC these levels returned to baseline. The results of this study indicate that DEC attenuates PH in an experimental monocrotaline-induced model by inhibiting a series of markers involved in cell proliferation/death.
Collapse
Affiliation(s)
- Edlene Lima Ribeiro
- Laboratory of Ultrastructure, Aggeu Magalhães Research Center - CPqAM, Pernambuco, Brazil; Federal University of Pernambuco, Brazil
| | - Ingrid Tavares Fragoso
- Laboratory of Ultrastructure, Aggeu Magalhães Research Center - CPqAM, Pernambuco, Brazil; Federal University of Pernambuco, Brazil
| | | | - Amanda Costa Oliveira
- Laboratory of Ultrastructure, Aggeu Magalhães Research Center - CPqAM, Pernambuco, Brazil; Federal University of Pernambuco, Brazil
| | - Amanda Karoline Soares E Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Research Center - CPqAM, Pernambuco, Brazil; Federal University of Pernambuco, Brazil
| | | | | | - Isalira Peroba Rezende Ramos
- National Center Structural Biology and Bio-imaging, Carlos Chagas Filho Biophysics Institute, Department of Radiology, University Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
24
|
Li L, Kim IK, Chiasson V, Chatterjee P, Gupta S. NF-κB mediated miR-130a modulation in lung microvascular cell remodeling: Implication in pulmonary hypertension. Exp Cell Res 2017; 359:235-242. [DOI: 10.1016/j.yexcr.2017.07.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/03/2017] [Accepted: 07/19/2017] [Indexed: 12/15/2022]
|
25
|
Gong Y, Yang Y, Wu Q, Gao G, Liu Y, Xiong Y, Huang C, Wu S. Activation of LXRα improves cardiac remodeling induced by pulmonary artery hypertension in rats. Sci Rep 2017; 7:6169. [PMID: 28733583 PMCID: PMC5522383 DOI: 10.1038/s41598-017-04640-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/17/2017] [Indexed: 12/25/2022] Open
Abstract
Inflammatory factors regulated by NF-κB play a significant role in PAH and myocardial hypertrophy. LXR activation may inhibit myocardial hypertrophy via suppressing inflammatory pathways; it is unknown whether LXR is also involved in PAH-induced myocardial hypertrophy or remodeling. To further explore the protective effect of LXR in PAH-induced cardiac hypertrophy and remodeling, a PAH model was developed, and T0901317, an agonist of LXR, was used to examine the effect of LXR activation. PAH rats demonstrated obvious cardiac hypertrophy and remodeling in the right ventricle, but significant improvement of cardiac hypertrophy and remodeling was observed in PAH rats treated with T0901317. Through RT-PCR, Western blot and ELISA examination, NF-κB, IL-6, TNF-α, and iNOS were found to be significantly reduced in PAH rats treated with T0901317 compared to PAH rats treated with DMSO. Apoptosis was also significantly reduced in PAH rats treated with T0901317. Thus, LXR activation may inhibit PAH-induced cardiac hypertrophy and remodeling by inhibiting NF-κB-mediated inflammatory pathways.
Collapse
Affiliation(s)
- Yibo Gong
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qin Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ge Gao
- Faculty of Laboratory Medicine, Xiangya Medical College, Central South University, Changsha, China
| | - Yin Liu
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yaoyao Xiong
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Can Huang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Sijie Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
26
|
Zhang Z, Zhang L, Sun C, Kong F, Wang J, Xin Q, Jiang W, Li K, Chen O, Luan Y. Baicalin attenuates monocrotaline-induced pulmonary hypertension through bone morphogenetic protein signaling pathway. Oncotarget 2017; 8:63430-63441. [PMID: 28969002 PMCID: PMC5609934 DOI: 10.18632/oncotarget.18825] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 06/01/2017] [Indexed: 11/25/2022] Open
Abstract
Baicalin, a flavonoid compound extracted from roots of Scutellaria baicalensis Georgi (huang qin), it has been shown to effectively attenuates pulmonary hypertension (PH), however, the potential mechanism remains unexplored. In this study, we investigated the potential mechanism of baicalin on monocrotaline (MCT)-induced PH in rats. The results showed that baicalin attenuated lung damage in PH rat model through inhibiting the pulmonary arterial smooth muscle cell proliferation and induction of cells apoptosis. Furthermore, we demonstrated that baicalin inhibition the expression of nuclear factor-κB (NF-κB) p65 and bone morphogenetic protein (BMP) antagonists gremlin-1, but increased the expression of inhibitor of NF-κB (I-κBα), BMPR2, BMP-4, BMP-9 and Smad1/5/8. Additionally, baicalin suppression endothelial-to-mesenchymal transition in PH lung tissue. Collectively, we confirmed that baicalin via inhibition of NF-κB signaling to further activation of BMP signaling to have a therapeutic effect on PH and providing a promising therapeutic strategy for PH.
Collapse
Affiliation(s)
- Zhaohua Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, China
| | - Luan Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, China
| | - Chao Sun
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Feng Kong
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Jue Wang
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Qian Xin
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Wen Jiang
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Kaili Li
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Ou Chen
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Yun Luan
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
27
|
Yu CJ, Liang C, Li YX, Hu QQ, Zheng WW, Niu N, Yang X, Wang ZR, Yu XD, Zhang BL, Song BL, Zhang ZR. ZNF307 (Zinc Finger Protein 307) Acts as a Negative Regulator of Pressure Overload–Induced Cardiac Hypertrophy. Hypertension 2017; 69:615-624. [DOI: 10.1161/hypertensionaha.116.08500] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/05/2016] [Accepted: 01/23/2017] [Indexed: 12/13/2022]
Abstract
Pathological cardiac hypertrophy is a key risk factor for heart failure. We found that the protein expression levels of the ZNF307 (zinc finger protein 307) were significantly increased in heart samples from both human patients with dilated cardiomyopathy and mice subjected to aortic banding. Therefore, we aimed to elucidate the role of ZNF307 in the development of cardiac hypertrophy and to explore the signal transduction events that mediate the effect of ZNF307 on cardiac hypertrophy, using cardiac-specific ZNF307 transgenic (ZNF307-TG) mice and ZNF307 global knockout (ZNF307-KO) mice. The results showed that the deletion of ZNF307 potentiated aortic banding–induced pathological cardiac hypertrophy, fibrosis, and cardiac dysfunction; however, the aortic banding–induced cardiac hypertrophic phenotype was dramatically diminished by ZNF307 overexpression in mouse heart. Mechanistically, the antihypertrophic effects mediated by ZNF307 in response to pathological stimuli were associated with the direct inactivation of NF-κB (nuclear factor-κB) signaling and blockade of the nuclear translocation of NF-κB subunit p65. Furthermore, the overexpression of a degradation-resistant mutant of IκBα (IκBα
S32A/S36A
) reversed the exacerbation of cardiac hypertrophy, fibrosis, and dysfunction shown in aortic banding–treated ZNF307-KO mice. In conclusion, our findings demonstrate that ZNF307 ameliorates pressure overload–induced cardiac hypertrophy by inhibiting the activity of NF-κB–signaling pathway.
Collapse
Affiliation(s)
- Chang-Jiang Yu
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Chen Liang
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Yu-Xia Li
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Qing-Qing Hu
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Wei-Wan Zheng
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Na Niu
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Xu Yang
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Zi-Rui Wang
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Xiao-Di Yu
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Bao-Long Zhang
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Bin-Lin Song
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| | - Zhi-Ren Zhang
- From the Institute of Metabolic Disease, Department of Cardiology (X.-D.Y., B.-L.Z., Z.-R.Z.), and Department of Clinical Pharmacy (C.-J.Y., C.L., Y.-X.L, Q.-Q.H., W.-W.Z., N.N., X.Y., Z.-R.W., B.-L.S., Z.-R.Z.), Harbin Medical University Cancer Hospital, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, P. R. China
| |
Collapse
|
28
|
Chaudhry A, Carthan KA, Kang BY, Calvert J, Sutliff RL, Michael Hart C. PPARγ attenuates hypoxia-induced hypertrophic transcriptional pathways in the heart. Pulm Circ 2017; 7:98-107. [PMID: 28680569 PMCID: PMC5448534 DOI: 10.1086/689749] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/15/2016] [Indexed: 02/01/2023] Open
Abstract
Chronic hypoxia-induced pulmonary hypertension (PH) is characterized by increased pressure and resistance in the pulmonary vasculature and hypertrophy of the right ventricle (RV). The transcription factors, nuclear factor activated T-cells (NFAT), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB/p65) contribute to RV hypertrophy (RVH). Because peroxisome proliferator-activated receptor gamma (PPARγ) activation attenuates hypoxia-induced PH and RVH, we hypothesized that PPARγ inhibits activation of RV hypertrophic transcriptional signaling mechanisms. C57BL/6J mice were exposed to normoxia (21% O2) or hypoxia (10% O2) for 21 days. During the final 10 days of exposure, selected mice were treated with the PPARγ ligand, pioglitazone. RV systolic pressure (RVSP) and RVH were measured, and NFATc2 and NF-kB/p65 protein levels were measured in RV and LV nuclear and cytosolic fractions. Cardiomyocyte hypertrophy was assessed with wheatgerm agglutinin staining. NFAT activation was also examined with luciferase reporter mice and analysis of protein levels of selected transcriptional targets. Chronic-hypoxia increased: (1) RVH, RVSP, and RV cardiomyocyte hypertrophy; (2) NFATc2 and NF-κB activation in RV nuclear homogenates; (3) RV and LV NFAT luciferase activity; and (4) RV protein levels of brain natriuretic peptide (BNP) and β-myosin heavy chain (β-MyHC). Treatment with pioglitazone attenuated hypoxia-induced increases in both RV and LV NFAT luciferase activity. Chronic hypoxia caused sustained RV NFATc2 and NF-κB activation. Pioglitazone attenuated PH, RVH, cardiomyocyte hypertrophy, and activation of RV hypertrophic signaling and also attenuated LV NFAT activation. PPARγ favorably modulates signaling derangements in the heart as well as in the pulmonary vascular wall.
Collapse
Affiliation(s)
- Abubakr Chaudhry
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - Kristal A Carthan
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - Bum-Yong Kang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - John Calvert
- Department of Surgery, Emory University, Atlanta, GA, USA
| | - Roy L Sutliff
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - C Michael Hart
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| |
Collapse
|
29
|
Maarman GJ, Schulz R, Sliwa K, Schermuly RT, Lecour S. Novel putative pharmacological therapies to protect the right ventricle in pulmonary hypertension: a review of current literature. Br J Pharmacol 2017; 174:497-511. [PMID: 28099680 DOI: 10.1111/bph.13721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/06/2016] [Accepted: 09/27/2016] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension (PH) is defined by elevated mean pulmonary artery pressure following the pathological remodelling of small pulmonary arteries. An increase in right ventricular (RV) afterload results in RV hypertrophy and RV failure. The pathophysiology of PH, and RV remodelling in particular, is not well understood, thus explaining, at least in part, why current PH therapies have a limited effect. Existing therapies mostly target the pulmonary circulation. Because the remodelled RV fails to support normal cardiac function, patients eventually succumb from RV failure. Developing novel therapies that directly target the function of the RV may therefore benefit patients with PH. In the past decade, several promising studies have investigated novel cardioprotective strategies in experimental models of PH. This review aims to comprehensively discuss and highlight these novel experimental approaches to confer, in the long-term, greater health benefit in patients with PH.
Collapse
Affiliation(s)
- Gerald J Maarman
- Hatter Institute for Cardiovascular Research in Africa (HICRA) and MRC Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Karen Sliwa
- Hatter Institute for Cardiovascular Research in Africa (HICRA) and MRC Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ralph Theo Schermuly
- Universities of Giessen and Marburg Lung Centre, Member of the German Lung Centre (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa (HICRA) and MRC Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
30
|
Dimethyl Fumarate ameliorates pulmonary arterial hypertension and lung fibrosis by targeting multiple pathways. Sci Rep 2017; 7:41605. [PMID: 28150703 PMCID: PMC5288696 DOI: 10.1038/srep41605] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 12/21/2016] [Indexed: 12/28/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a fatal condition for which there is no cure. Dimethyl Fumarate (DMF) is an FDA approved anti-oxidative and anti-inflammatory agent with a favorable safety record. The goal of this study was to assess the effectiveness of DMF as a therapy for PAH using patient-derived cells and murine models. We show that DMF treatment is effective in reversing hemodynamic changes, reducing inflammation, oxidative damage, and fibrosis in the experimental models of PAH and lung fibrosis. Our findings indicate that effects of DMF are facilitated by inhibiting pro-inflammatory NFκB, STAT3 and cJUN signaling, as well as βTRCP-dependent degradation of the pro-fibrogenic mediators Sp1, TAZ and β-catenin. These results provide a novel insight into the mechanism of its action. Collectively, preclinical results demonstrate beneficial effects of DMF on key molecular pathways contributing to PAH, and support its testing in PAH treatment in patients.
Collapse
|
31
|
Li L, Guleria RS, Thakur S, Zhang CL, Pan J, Baker KM, Gupta S. Thymosin β4 Prevents Angiotensin II-Induced Cardiomyocyte Growth by Regulating Wnt/WISP Signaling. J Cell Physiol 2016; 231:1737-44. [PMID: 26627308 DOI: 10.1002/jcp.25275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 12/01/2015] [Indexed: 11/08/2022]
Abstract
Thymosin beta-4 (Tβ4) is a ubiquitous protein with many properties relating to cell proliferation and differentiation that promotes wound healing and modulates inflammatory mediators. However, the role of Tβ4 in cardiomyocyte hypertrophy is currently unknown. The purpose of this study was to determine the cardio-protective effect of Tβ4 in angiotensin II (Ang II)-induced cardiomyocyte growth. Neonatal rat ventricular cardiomyocytes (NRVM) were pretreated with Tβ4 followed by Ang II stimulation. Cell size, hypertrophy marker gene expression and Wnt signaling components, β-catenin, and Wnt-induced secreted protein-1 (WISP-1) were evaluated by quantitative real-time PCR, Western blotting and fluorescent microscopy. Pre-treatment of Tβ4 resulted in reduction of cell size, hypertrophy marker genes and Wnt-associated gene expression, and protein levels; induced by Ang II in cardiomyocyte. WISP-1 was overexpressed in NRVM and, the effect of Tβ4 in Ang II-induced cardiomyocyte growth was evaluated. WISP-1 overexpression promoted cardiomyocytes growth and was reversed by pretreatment with Tβ4. This is the first report which demonstrates that Tβ4 targets Wnt/WISP-1 to protect Ang II-induced cardiomyocyte growth. J. Cell. Physiol. 231: 1737-1744, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Li Li
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas.,Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Rakeshwar S Guleria
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas
| | - Suresh Thakur
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas
| | - Cheng-Lin Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Jing Pan
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas
| | - Kenneth M Baker
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas
| | - Sudhiranjan Gupta
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, Texas.,Baylor Scott and White Health, Temple, Texas.,Central Texas Veterans Health Care System, Temple, Texas
| |
Collapse
|
32
|
Yang L, Yu D, Mo R, Zhang J, Hua H, Hu L, Feng Y, Wang S, Zhang WY, Yin N, Mo XM. The Succinate Receptor GPR91 Is Involved in Pressure Overload-Induced Ventricular Hypertrophy. PLoS One 2016; 11:e0147597. [PMID: 26824665 PMCID: PMC4732750 DOI: 10.1371/journal.pone.0147597] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 01/06/2016] [Indexed: 01/03/2023] Open
Abstract
Background Pulmonary arterial hypertension is characterized by increased pressure overload that leads to right ventricular hypertrophy (RVH). GPR91 is a formerly orphan G-protein-coupled receptor (GPCR) that has been characterized as a receptor for succinate; however, its role in RVH remains unknown. Methods and Results We investigated the role of succinate-GPR91 signaling in a pulmonary arterial banding (PAB) model of RVH induced by pressure overload in SD rats. GPR91 was shown to be located in cardiomyocytes. In the sham and PAB rats, succinate treatment further aggravated RVH, up-regulated RVH-associated genes and increased p-Akt/t-Akt levels in vivo. In vitro, succinate treatment up-regulated the levels of the hypertrophic gene marker anp and p-Akt/t-Akt in cardiomyocytes. All these effects were inhibited by the PI3K antagonist wortmannin both in vivo and in vitro. Finally, we noted that the GPR91-PI3K/Akt axis was also up-regulated compared to that in human RVH. Conclusions Our findings indicate that succinate-GPR91 signaling may be involved in RVH via PI3K/Akt signaling in vivo and in vitro. Therefore, GPR91 may be a novel therapeutic target for treating pressure overload-induced RVH.
Collapse
MESH Headings
- Androstadienes/pharmacology
- Animals
- Atrial Natriuretic Factor/genetics
- Atrial Natriuretic Factor/metabolism
- Gene Expression Regulation
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Heart Ventricles/physiopathology
- Humans
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/physiopathology
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Phosphorylation
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/surgery
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
- Stroke Volume
- Succinic Acid/metabolism
- Succinic Acid/pharmacology
- Wortmannin
Collapse
Affiliation(s)
- Lei Yang
- Department of Gastroenterology, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| | - Di Yu
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| | - Ran Mo
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, the affiliated hospital of Nanjing University Medical School, Nanjing, China
| | - Jiru Zhang
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, Wuxi No.4 People’s Hospital, Nanjing, China
| | - Hu Hua
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| | - Liang Hu
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| | - Yu Feng
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| | - Song Wang
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| | - Wei-yan Zhang
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| | - Ning Yin
- Department of Anesthesiology, Zhongda Hospital, Southeast University, Nanjing, China
- * E-mail: (XMM); (NY)
| | - Xu-Ming Mo
- Department of Cardiothoracic Surgery, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, China
- * E-mail: (XMM); (NY)
| |
Collapse
|
33
|
Kaur G, Singh N, Lingeshwar P, Siddiqui HH, Hanif K. Poly (ADP-ribose) polymerase-1: an emerging target in right ventricle dysfunction associated with pulmonary hypertension. Pulm Pharmacol Ther 2014; 30:66-79. [PMID: 25481773 DOI: 10.1016/j.pupt.2014.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 10/20/2014] [Accepted: 11/25/2014] [Indexed: 12/19/2022]
Abstract
Recently, inhibition of poly (ADP-ribose) polymerase-1 (PARP1) was shown to be protective in experimental pulmonary hypertension (PH) and prevented right ventricular hypertrophy (RVH) associated with it. However, molecular mechanism behind cardioprotection by PARP1 inhibition in PH still needs detailed exploration. Therefore, effect of inhibition of PARP1 on the right ventricle (RV) dysfunction was studied in monocrotaline (MCT) induced PH model. Following a single dose administration of MCT (60 mg/kg, s.c.), male Sprague-Dawley rats were treated with PARP1 inhibitor 1,5-Isoquinolinediol (ISO, 3 mg/kg, i.p.) for 35 days for preventive study and from day 21-35 for curative study. RV pressure (RVP) and RVH were measured after 35 days. Histophathological studies, PARP1 activity, mRNA and protein expression were studied in isolated RV. Oxidative and nitosative stress, inflammation and Matrix metalloproteinases (MMPs)/Tissue inhibitor of metalloproteinase 2 (TIMP2) were also assessed. Mitochondrial dysfunction was studied by mitochondrial membrane permeability and estimation of Nicotinamide adenine dinucleotide (NAD) and Adenosine triphosphate (ATP). Apoptosis in RV was assessed by Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), caspase 3 activity and cleaved PARP1 expression. PARP1 inhibition significantly reversed the increase in RVP and RVH in both preventive and curative treatment in the MCT-injected rats. ISO lowered oxidative and nitrosative stress and inflammation and restored the balance of MMPs/TIMP2 expression. PARP1 inhibition prevented mitochondrial dysfunction and the release of cell death factors from mitochondria. ISO also decreased apoptosis by decreasing number of TUNEL positive cells, caspase 3 activity and PARP1 cleavage in RV. Thus, PARP1 inhibition ameliorated PH induced RV hypertrophy and may emerge as a new therapeutic target for PH.
Collapse
Affiliation(s)
- Gurpreet Kaur
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Faculty of Pharmacy, Integral University, Lucknow 226001, India
| | - Neetu Singh
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Poorella Lingeshwar
- National Institute of Pharmaceutical Education and Research, Rae Bareli 229010, India
| | | | - Kashif Hanif
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; National Institute of Pharmaceutical Education and Research, Rae Bareli 229010, India; Academy of Scientific and Innovative Research, New Delhi, India.
| |
Collapse
|
34
|
Wei C, Kim IK, Li L, Wu L, Gupta S. Thymosin Beta 4 protects mice from monocrotaline-induced pulmonary hypertension and right ventricular hypertrophy. PLoS One 2014; 9:e110598. [PMID: 25412097 PMCID: PMC4239012 DOI: 10.1371/journal.pone.0110598] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/19/2014] [Indexed: 01/25/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive vascular disease of pulmonary arteries that impedes ejection of blood by the right ventricle. As a result there is an increase in pulmonary vascular resistance and pulmonary arterial pressure causing right ventricular hypertrophy (RVH) and RV failure. The pathology of PAH involves vascular cell remodeling including pulmonary arterial endothelial cell (PAEC) dysfunction and pulmonary arterial smooth muscle cell (PASMC) proliferation. Current therapies are limited to reverse the vascular remodeling. Investigating a key molecule is required for development of new therapeutic intervention. Thymosin beta-4 (Tβ4) is a ubiquitous G-actin sequestering protein with diverse biological function and promotes wound healing and modulates inflammatory responses. However, it remains unknown whether Tβ4 has any protective role in PH. The purpose of this study is to evaluate the whether Tβ4 can be used as a vascular-protective agent. In monocrotaline (MCT)-induced PH mouse model, we showed that mice treated with Tβ4 significantly attenuated the systolic pressure and RVH, compared to the MCT treated mice. Our data revealed for the first time that Tβ4 selectively targets Notch3-Col 3A-CTGF gene axis in preventing MCT-induced PH and RVH. Our study may provide pre-clinical evidence for Tβ4 and may consider as vasculo-protective agent for the treatment of PH induced RVH.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Collagen Type III/genetics
- Collagen Type III/metabolism
- Connective Tissue Growth Factor/genetics
- Connective Tissue Growth Factor/metabolism
- Disease Models, Animal
- Endothelial Cells/drug effects
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/prevention & control
- Hypertrophy, Right Ventricular/chemically induced
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/prevention & control
- Injections, Intraperitoneal
- Lung/drug effects
- Lung/metabolism
- Lung/pathology
- Male
- Mice
- Monocrotaline/toxicity
- Receptor, Notch3
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction/drug effects
- Thymosin/administration & dosage
- Thymosin/pharmacology
Collapse
Affiliation(s)
- Chuanyu Wei
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center and Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | - Il-Kwon Kim
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center and Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | - Li Li
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center and Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Liling Wu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Sudhiranjan Gupta
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center and Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
- * E-mail:
| |
Collapse
|
35
|
Thomas CM, Yong QC, Rosa RM, Seqqat R, Gopal S, Casarini DE, Jones WK, Gupta S, Baker KM, Kumar R. Cardiac-specific suppression of NF-κB signaling prevents diabetic cardiomyopathy via inhibition of the renin-angiotensin system. Am J Physiol Heart Circ Physiol 2014; 307:H1036-45. [PMID: 25085967 DOI: 10.1152/ajpheart.00340.2014] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activation of NF-κB signaling in the heart may be protective or deleterious depending on the pathological context. In diabetes, the role of NF-κB in cardiac dysfunction has been investigated using pharmacological approaches that have a limitation of being nonspecific. Furthermore, the specific cellular pathways by which NF-κB modulates heart function in diabetes have not been identified. To address these questions, we used a transgenic mouse line expressing mutated IκB-α in the heart (3M mice), which prevented activation of canonical NF-κB signaling. Diabetes was developed by streptozotocin injections in wild-type (WT) and 3M mice. Diabetic WT mice developed systolic and diastolic cardiac dysfunction by the 12th week, as measured by echocardiography. In contrast, cardiac function was preserved in 3M mice up to 24 wk of diabetes. Diabetes induced an elevation in cardiac oxidative stress in diabetic WT mice but not 3M mice compared with nondiabetic control mice. In diabetic WT mice, an increase in the phospholamban/sarco(endo)plasmic reticulum Ca(2+)-ATPase 2 ratio and decrease in ryanodine receptor expression were observed, whereas diabetic 3M mice showed an opposite effect on these parameters of Ca(2+) handling. Significantly, renin-angiotensin system activity was suppressed in diabetic 3M mice compared with an increase in WT animals. In conclusion, these results demonstrate that inhibition of NF-κB signaling in the heart prevents diabetes-induced cardiac dysfunction through preserved Ca(2+) handling and inhibition of the cardiac renin-angiotensin system.
Collapse
Affiliation(s)
- Candice M Thomas
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Qian Chen Yong
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Rodolfo M Rosa
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil; and
| | - Rachid Seqqat
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Shanthi Gopal
- Central Texas Veterans Health Care System, Temple, Texas
| | - Dulce E Casarini
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil; and
| | - W Keith Jones
- Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, Illinois
| | - Sudhiranjan Gupta
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Kenneth M Baker
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Rajesh Kumar
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas;
| |
Collapse
|
36
|
Li L, Wei C, Kim IK, Janssen-Heininger Y, Gupta S. Inhibition of Nuclear Factor-κB in the Lungs Prevents Monocrotaline-Induced Pulmonary Hypertension in Mice. Hypertension 2014; 63:1260-9. [DOI: 10.1161/hypertensionaha.114.03220] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Li Li
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, TX (L.L., C.W., I.-K.K., S.G.); Internal Medicine, Scott & White, Temple, TX (L.L., C.W., I.-K.K., S.G.); Central Texas Veterans Health Care System, Temple, TX (L.L., C.W., I.-K.K., S.G.); and Department of Pathology, University of Vermont, Burlington, VT (Y.J.-H.)
| | - Chuanyu Wei
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, TX (L.L., C.W., I.-K.K., S.G.); Internal Medicine, Scott & White, Temple, TX (L.L., C.W., I.-K.K., S.G.); Central Texas Veterans Health Care System, Temple, TX (L.L., C.W., I.-K.K., S.G.); and Department of Pathology, University of Vermont, Burlington, VT (Y.J.-H.)
| | - Il-Kwon Kim
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, TX (L.L., C.W., I.-K.K., S.G.); Internal Medicine, Scott & White, Temple, TX (L.L., C.W., I.-K.K., S.G.); Central Texas Veterans Health Care System, Temple, TX (L.L., C.W., I.-K.K., S.G.); and Department of Pathology, University of Vermont, Burlington, VT (Y.J.-H.)
| | - Yvonne Janssen-Heininger
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, TX (L.L., C.W., I.-K.K., S.G.); Internal Medicine, Scott & White, Temple, TX (L.L., C.W., I.-K.K., S.G.); Central Texas Veterans Health Care System, Temple, TX (L.L., C.W., I.-K.K., S.G.); and Department of Pathology, University of Vermont, Burlington, VT (Y.J.-H.)
| | - Sudhiranjan Gupta
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Temple, TX (L.L., C.W., I.-K.K., S.G.); Internal Medicine, Scott & White, Temple, TX (L.L., C.W., I.-K.K., S.G.); Central Texas Veterans Health Care System, Temple, TX (L.L., C.W., I.-K.K., S.G.); and Department of Pathology, University of Vermont, Burlington, VT (Y.J.-H.)
| |
Collapse
|
37
|
Novoyatleva T, Sajjad A, Engel FB. TWEAK-Fn14 Cytokine-Receptor Axis: A New Player of Myocardial Remodeling and Cardiac Failure. Front Immunol 2014; 5:50. [PMID: 24611063 PMCID: PMC3920183 DOI: 10.3389/fimmu.2014.00050] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/28/2014] [Indexed: 01/01/2023] Open
Abstract
Tumor necrosis factor (TNF) has been firmly established as a pathogenic factor in heart failure, a significant socio-economic burden. In this review, we will explore the role of other members of the TNF/TNF receptor superfamily (TNFSF/TNFRSF) in cardiovascular diseases (CVDs) focusing on TWEAK and its receptor Fn14, new players in myocardial remodeling and heart failure. The TWEAK/Fn14 pathway controls a variety of cellular activities such as proliferation, differentiation, and apoptosis and has diverse biological functions in pathological mechanisms like inflammation and fibrosis that are associated with CVDs. Furthermore, it has recently been shown that the TWEAK/Fn14 axis is a positive regulator of cardiac hypertrophy and that deletion of Fn14 receptor protects from right heart fibrosis and dysfunction. We discuss the potential use of the TWEAK/Fn14 axis as biomarker for CVDs as well as therapeutic target for future treatment of human heart failure based on supporting data from animal models and in vitro studies. Collectively, existing data strongly suggest the TWEAK/Fn14 axis as a potential new therapeutic target for achieving cardiac protection in patients with CVDs.
Collapse
Affiliation(s)
- Tatyana Novoyatleva
- Department of Cardiac Development and Remodelling, Max-Planck-Institute for Heart and Lung Research , Bad Nauheim , Germany
| | - Amna Sajjad
- Department of Cardiac Development and Remodelling, Max-Planck-Institute for Heart and Lung Research , Bad Nauheim , Germany ; Government College University Faisalabad , Faisalabad , Pakistan
| | - Felix B Engel
- Department of Nephropathology, Experimental Renal and Cardiovascular Research, Institute of Pathology, University of Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|
38
|
Wei C, Henderson H, Spradley C, Li L, Kim IK, Kumar S, Hong N, Arroliga AC, Gupta S. Circulating miRNAs as potential marker for pulmonary hypertension. PLoS One 2013; 8:e64396. [PMID: 23717609 PMCID: PMC3662705 DOI: 10.1371/journal.pone.0064396] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 04/12/2013] [Indexed: 01/20/2023] Open
Abstract
MircoRNAs (miRNAs) are small non-coding RNAs that govern the gene expression and, play significant role in the pathogenesis of heart failure. The detection of miRNAs in circulation of pulmonary hypertensive (PH) human subjects remains elusive. In the current study, we determined the pattern of miRNAs of mild-to-severe human PH subjects and, compared them with the control subjects by miRNA array. Blood was obtained using fluoroscopic and waveform guided catheterization from the distal (pulmonary artery) port of the catheter. A total 40 human subjects were included in the study and, the degree of PH was determined by mean pulmonary arterial pressure. Among several miRNAs in the array, we validated 14 miRNAs and, the data were consistent with the array profile. We identified several novel downregulated miRNAs (miR-451, miR-1246) and upregulated miRNAs (miR-23b, miR-130a and miR-191) in the circulation of PH subjects. Our study showed novel set of miRNAs which are dysregulated in PH and, are directly proportional to the degree of PH. These miRNAs may be considered as potential biomarker for early detection of PH.
Collapse
Affiliation(s)
- Chuanyu Wei
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | - Heather Henderson
- Scott & White Memorial Hospital, Temple, Texas, United States of America
| | | | - Li Li
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | - Il-Kwon Kim
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | - Sandeep Kumar
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | - Nayeon Hong
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
| | | | - Sudhiranjan Gupta
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M Health Science Center, Scott & White, Central Texas Veterans Health Care System, Temple, Texas, United States of America
- * E-mail:
| |
Collapse
|
39
|
Colombo R, Siqueira R, Becker CU, Fernandes TG, Pires KM, Valença SS, Souza-Rabbo MP, Araujo AS, Belló-Klein A. Effects of exercise on monocrotaline-induced changes in right heart function and pulmonary artery remodeling in rats. Can J Physiol Pharmacol 2013; 91:38-44. [DOI: 10.1139/cjpp-2012-0261] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pulmonary arterial hypertension (PAH) induced by monocrotaline (MCT) is an experimental protocol of right heart failure. We analyzed the role of exercise training on the right ventricle structure and function, pulmonary artery remodeling, and GSK-3β expression. Rats were divided among the following groups: sedentary control (SC), sedentary monocrotaline (SM), trained control (TC), and trained monocrotaline (TM). Rats underwent exercise training for a period of 5 weeks, with 3 weeks post-MCT injection. Rats in the SM and TM groups presented with an increase in right ventricle hypertrophy indexes and lung congestion. The right ventricular end diastolic pressure (RVEDP), right ventricular systolic pressure (RVSP), and its minimum and maximal pressure derivates were increased in the SM and TM groups. The right ventricle interstitial volume pulmonary artery thickness and p-GSK-3β/GSK-3β were increased in the MCT groups as compared with the control groups. The TM group had a reduction in interstitial volume, p-GSK-3β/GSK-3β ratio, pulmonary artery thickness, RVEDP, and an increase in intramyocardial vessels volume as compared with the SM group. The overall results have shown that the exercise protocol used promoted positive changes in right ventricle and pulmonary artery remodeling. These observations also suggest that structural remodeling may be influenced by signaling proteins, such as GSK-3β.
Collapse
Affiliation(s)
- Rafael Colombo
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| | - Rafaela Siqueira
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| | - Cristiano Urbano Becker
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| | - Tânia Gatelli Fernandes
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| | - Karla Maria Pires
- Laboratório de Inflamação, Estresse Oxidativo e Câncer, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Samuel Santos Valença
- Laboratório de Inflamação, Estresse Oxidativo e Câncer, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | | | - Alex Sander Araujo
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| | - Adriane Belló-Klein
- Laboratório de Fisiologia Cardiovascular e Espécies Ativas de Oxigênio, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, CEP 90050-170, Porto Alegre, Brasil
| |
Collapse
|