1
|
Efentakis P, Symeonidi L, Gianniou DD, Mikropoulou EV, Giardoglou P, Valakos D, Vatsellas G, Tsota M, Kostomitsopoulos N, Smyrnioudis I, Trougakos IP, Halabalaki M, Dedoussis GV, Andreadou I. Antihypertensive Potential of Pistacia lentiscus var. Chia: Molecular Insights and Therapeutic Implications. Nutrients 2024; 16:2152. [PMID: 38999899 PMCID: PMC11243328 DOI: 10.3390/nu16132152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
Background: Hypertension poses a significant global health burden and is associated with cardiovascular morbidity. Chios mastic gum (CMG), derived from Pistacia lentiscus var. Chia, shows potential as a phytotherapeutic agent, due to its multifaceted beneficial effects. However, its anti-hypertensive effects and vascular, circulatory, and renal-related dysfunction, have not been thoroughly investigated. Herein, we aimed to explore the antihypertensive potential of CMG, focusing on vascular and renal endothelium, in vivo. Methods: Two models of hypertension in male rats, induced by Angiotensin II and Deoxycorticosterone acetate (DOCA)-high-salt administration, were utilized. CMG was administered at 220 mg/kg daily for four weeks after hypertension onset and blood pressure was measured non-invasively. Whole blood RNA sequencing, metabolomics, real-time PCR, and Western blot analyses of kidney and aorta tissues were additionally performed. Results: CMG significantly lowered systolic, diastolic, and mean blood pressure in both models. RNA sequencing revealed that CMG modulated immunity in the Angiotensin II model and metabolism in the DOCA-HS model. CMG downregulated genes related to oxidative stress and endothelial dysfunction and upregulated endothelial markers such as Vegfa. Metabolomic analysis indicated improved endothelial homeostasis via lysophosphatidylinositol upregulation. Conclusions: CMG emerges as a potent natural antihypertensive therapy, demonstrating beneficial effects on blood pressure and renal endothelial function.
Collapse
Affiliation(s)
- Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Zografou, Greece; (P.E.); (L.S.)
| | - Lydia Symeonidi
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Zografou, Greece; (P.E.); (L.S.)
| | - Despoina D. Gianniou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (D.D.G.); (I.P.T.)
| | - Eleni V. Mikropoulou
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece; (E.V.M.); (M.H.)
| | - Panagiota Giardoglou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17671 Athens, Greece; (P.G.); (M.T.); (G.V.D.)
| | - Dimitrios Valakos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (D.V.); (N.K.)
| | - Giannis Vatsellas
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Maria Tsota
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17671 Athens, Greece; (P.G.); (M.T.); (G.V.D.)
| | - Nikolaos Kostomitsopoulos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (D.V.); (N.K.)
| | | | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (D.D.G.); (I.P.T.)
| | - Maria Halabalaki
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece; (E.V.M.); (M.H.)
| | - Georgios V. Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17671 Athens, Greece; (P.G.); (M.T.); (G.V.D.)
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Zografou, Greece; (P.E.); (L.S.)
| |
Collapse
|
2
|
Luan J, Ji X, Liu L. PPARγ in Atherosclerotic Endothelial Dysfunction: Regulatory Compounds and PTMs. Int J Mol Sci 2023; 24:14494. [PMID: 37833942 PMCID: PMC10572723 DOI: 10.3390/ijms241914494] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
The formation of atherosclerotic plaques is one of the main sources of cardiovascular disease. In addition to known risk factors such as dyslipidemia, diabetes, obesity, and hypertension, endothelial dysfunction has been shown to play a key role in the formation and progression of atherosclerosis. Peroxisome proliferator-activated receptor-gamma (PPARγ), a transcription factor belonging to the steroid superfamily, is expressed in the aorta and plays a critical role in protecting endothelial function. It thereby serves as a target for treating both diabetes and atherosclerosis. Although many studies have examined endothelial cell disorders in atherosclerosis, the role of PPARγ in endothelial dysfunction is still not well understood. In this review, we summarize the possible mechanisms of action behind PPARγ regulatory compounds and post-translational modifications (PTMs) of PPARγ in the control of endothelial function. We also explore the potential use of endothelial PPARγ-targeted agents in the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
| | | | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200082, China
| |
Collapse
|
3
|
Xu WJ, Wu Q, He WN, Wang S, Zhao YL, Huang JX, Yan XS, Jiang R. Interleukin-6 and pulmonary hypertension: from physiopathology to therapy. Front Immunol 2023; 14:1181987. [PMID: 37449201 PMCID: PMC10337993 DOI: 10.3389/fimmu.2023.1181987] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive, pulmonary vascular disease with high morbidity and mortality. Unfortunately, the pathogenesis of PH is complex and remains unclear. Existing studies have suggested that inflammatory factors are key factors in PH. Interleukin-6 (IL-6) is a multifunctional cytokine that plays a crucial role in the regulation of the immune system. Current studies reveal that IL-6 is elevated in the serum of patients with PH and it is negatively correlated with lung function in those patients. Since IL-6 is one of the most important mediators in the pathogenesis of inflammation in PH, signaling mechanisms targeting IL-6 may become therapeutic targets for this disease. In this review, we detailed the potential role of IL-6 in accelerating PH process and the specific mechanisms and signaling pathways. We also summarized the current drugs targeting these inflammatory pathways to treat PH. We hope that this study will provide a more theoretical basis for targeted treatment in patients with PH in the future.
Collapse
Affiliation(s)
- Wei-Jie Xu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiong Wu
- Department of Pulmonary and Critical Care Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Ni He
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shang Wang
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ya-Lin Zhao
- Department of Respiratory Critical Care Medicine, The First Hospital of Kunming, Kunming, China
| | - Jun-Xia Huang
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xue-Shen Yan
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Rong Jiang
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
4
|
Abdelazeem H, Tu L, Thuillet R, Ottaviani M, Boulfrad A, Beck T, Senbel A, Mani S, Castier Y, Guyard A, Tran-Dinh A, El-Benna J, Longrois D, Silverstein AM, Guignabert C, Norel X. AMPK activation by metformin protects against pulmonary hypertension in rats and relaxes isolated human pulmonary artery. Eur J Pharmacol 2023; 946:175579. [PMID: 36914083 DOI: 10.1016/j.ejphar.2023.175579] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 03/13/2023]
Abstract
Pulmonary hypertension (PH) is associated with pulmonary vasoconstriction and endothelial dysfunction leading to impaired nitric oxide (NO) and prostacyclin (PGI2) pathways. Metformin, the first line treatment for type 2 diabetes and AMP-activated protein kinase (AMPK) activator, has been recently highlighted as a potential PH treatment. AMPK activation has been reported to improve endothelial function by enhancing endothelial NO synthase (eNOS) activity and to have relaxant effects in blood vessels. In this study, we examined the effect of metformin treatment on PH as well as on NO and PGI2 pathways in monocrotaline (MCT)-injected rats with established PH. Moreover, we investigated the anti-contractile effects of AMPK activators on endothelium-denuded human pulmonary arteries (HPA) from Non-PH and Group 3 PH patients (due to lung diseases and/or hypoxia). Furthermore, we explored the interaction between treprostinil and the AMPK/eNOS pathway. Our results showed that metformin protected against PH progression in MCT rats where it reduced the mean pulmonary artery pressure, pulmonary vascular remodeling and right ventricular hypertrophy and fibrosis compared to vehicle-treated MCT rats. The protective effects on rat lungs were mediated in part by increasing eNOS activity and protein kinase G-1 expression but not through the PGI2 pathway. In addition, incubation with AMPK activators reduced the phenylephrine-induced contraction of endothelium-denuded HPA from Non-PH and PH patients. Finally, treprostinil also augmented eNOS activity in HPA smooth muscle cells. In conclusion, we found that AMPK activation can enhance the NO pathway, attenuate vasoconstriction by direct effects on smooth muscles, and reverse established MCT-induced PH in rats.
Collapse
Affiliation(s)
- Heba Abdelazeem
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Egypt
| | - Ly Tu
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France; Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Raphaël Thuillet
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France; Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Mina Ottaviani
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France; Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Achraf Boulfrad
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France
| | - Thomas Beck
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France
| | - Amira Senbel
- Arab Academy for Science, Technology & Maritime Transport, College of Pharmacy, Alexandria, Egypt
| | - Salma Mani
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France; Université de Monastir-Tunisia, Institut Supérieur de Biotechnologie de Monastir (ISBM), Tunisia
| | - Yves Castier
- Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Alice Guyard
- Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Alexy Tran-Dinh
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France; Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Jamel El-Benna
- Université Paris Cité, INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Site Xavier Bichat, Paris, F-75018, France
| | - Dan Longrois
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France; Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | | | - Christophe Guignabert
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France; Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Xavier Norel
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France.
| |
Collapse
|
5
|
Chen D, Zhang HF, Yuan TY, Sun SC, Wang RR, Wang SB, Fang LH, Lyu Y, Du GH. Puerarin-V prevents the progression of hypoxia- and monocrotaline-induced pulmonary hypertension in rodent models. Acta Pharmacol Sin 2022; 43:2325-2339. [PMID: 35190697 PMCID: PMC9433387 DOI: 10.1038/s41401-022-00865-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/09/2022] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a cardiopulmonary disease characterized by a progressive increase in pulmonary vascular resistance. One of the initial pathogenic factors of PH is pulmonary arterial remodeling under various stimuli. Current marketed drugs against PH mainly relieve symptoms without significant improvement in overall prognosis. Discovering and developing new therapeutic drugs that interfere with vascular remodeling is in urgent need. Puerarin is an isoflavone compound extracted from the root of Kudzu vine, which is widely used in the treatment of cardiovascular diseases. In the present study, we evaluated the efficacy of puerarin in the treatment of experimental PH. PH was induced in rats by a single injection of MCT (50 mg/kg, sc), and in mice by exposure to hypoxia (10% O2) for 14 days. After MCT injection the rats were administered puerarin (10, 30, 100 mg · kg-1 · d-1, i.g.) for 28 days, whereas hypoxia-treated mice were pre-administered puerarin (60 mg · kg-1 · d-1, i.g.) for 7 days. We showed that puerarin administration exerted significant protective effects in both experimental PH rodent models, evidenced by significantly reduced right ventricular systolic pressure (RVSP) and lung injury, improved pulmonary artery blood flow as well as pulmonary vasodilation and contraction function, inhibited inflammatory responses in lung tissues, improved resistance to apoptosis and abnormal proliferation in lung tissues, attenuated right ventricular injury and remodeling, and maintained normal function of the right ventricle. We revealed that MCT and hypoxia treatment significantly downregulated BMPR2/Smad signaling in the lung tissues and PPARγ/PI3K/Akt signaling in the lung tissues and right ventricles, which were restored by puerarin administration. In addition, we showed that a novel crystal type V (Puer-V) exerted better therapeutic effects than the crude form of puerarin (Puer). Furthermore, Puer-V was more efficient than bosentan (a positive control drug) in alleviating the abnormal structural changes and dysfunction of lung tissues and right ventricles. In conclusion, this study provides experimental evidence for developing Puer-V as a novel therapeutic drug to treat PH.
Collapse
Affiliation(s)
- Di Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hui-Fang Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Tian-Yi Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shu-Chan Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ran-Ran Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shou-Bao Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lian-Hua Fang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Yang Lyu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Guan-Hua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
6
|
rhEPO Upregulates the PPARγ Pathway in Long-term Cultured Primary Nerve Cells via PI3K/Akt to Delay Cell Senescence. J Mol Neurosci 2022; 72:1586-1597. [PMID: 35505269 DOI: 10.1007/s12031-022-01998-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 03/09/2022] [Indexed: 10/18/2022]
Abstract
Previous studies have confirmed that both recombinant human erythropoietin (rhEPO) and peroxisome proliferator-activated receptors γ (PPARγ) activator pioglitazone can protect senescent nerve cells, and their mechanisms involve enhancing cell antioxidant capacity and reducing cell apoptosis. However, whether the PPARγ pathway is involved in the rhEPO anti-aging process in neuronal cells is still unclear. In this study, to explore the relationship between rhEPO and the PPARγ pathway at the cellular level, primary nerve cells cultured for 22 days were used to simulate the natural aging process of nerve cells. Starting on the 11th day of culture, rhEPO, LY294002, and GW9662 were added for treatment. Immunochemical methods and SA-β-gal staining were used to observe the changes in cellular antioxidant capacity and the fraction of senescent cells. The results showed that PPARγ blockade retarded the effect of rhEPO on the cellular antioxidant capacity and altered the fraction of senescent cells. It was confirmed that PPARγ was involved in rhEPO's anti-aging process in neuronal cells. Real-time fluorescent quantitative RT-PCR, Western blotting, and immunofluorescence staining were used to observe the changes in PPARγ pathway-related factors in nerve cells after rhEPO treatment. The results showed that rhEPO significantly upregulated the expression of PPARγ coactivator-1α (PGC-1α), PPARγ, and nuclear PPARγ in cells but did not affect the level of phosphorylated PPARγ protein, confirming that rhEPO has the ability to upregulate the PPARγ pathway. PI3K/Akt and PPARγ pathway blockade experiments were used to explore the relationships among rhEPO, PI3K/Akt, and PPARγ. The results showed that after PPARγ blockade, rhEPO had no significant effect on the PI3K/Akt pathway-related factor p-Akt, while after PI3K/Akt blockade, rhEPO's effects on PPARγ-related factors (PGC-1α, PPARγ, and nuclear PPARγ) were significantly decreased. It is suggested that rhEPO delays the PI3K/Akt pathway in the process of neuronal senescence, which is located upstream of PPARγ regulation. In conclusion, this study confirmed that rhEPO can upregulate the expression of PGC-1α and PPARγ in cells and the level of PPARγ protein in the nucleus to enhance the antioxidant capacity of cells and delay the senescence of nerve cells through the PI3K/Akt pathway. These findings will provide ideas for finding new targets for neuroprotection research and will also provide a theoretical basis and experimental evidence for rhEPO anti-aging research in neural cells.
Collapse
|
7
|
Tettey A, Jiang Y, Li X, Li Y. Therapy for Pulmonary Arterial Hypertension: Glance on Nitric Oxide Pathway. Front Pharmacol 2021; 12:767002. [PMID: 34867394 PMCID: PMC8633825 DOI: 10.3389/fphar.2021.767002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/25/2021] [Indexed: 01/11/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease with a resultant increase of the mean pulmonary arterial pressure, right ventricular hypertrophy and eventual death. Research in recent years has produced various therapeutic options for its clinical management but the high mortality even under treatment remains a big challenge attributed to the complex pathophysiology. Studies from clinical and non-clinical experiments have revealed that the nitric oxide (NO) pathway is one of the key pathways underlying the pathophysiology of PAH. Many of the essential drugs used in the management of PAH act on this pathway highlighting its significant role in PAH. Meanwhile, several novel compounds targeting on NO pathway exhibits great potential to become future therapy medications. Furthermore, the NO pathway is found to interact with other crucial pathways. Understanding such interactions could be helpful in the discovery of new drug that provide better clinical outcomes.
Collapse
Affiliation(s)
- Abraham Tettey
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
| | - Yujie Jiang
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
| | - Xiaohui Li
- Department of Pharmacology, School of Pharmaceutical Science, Central South University, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| |
Collapse
|
8
|
Guerra-Ojeda S, Marchio P, Gimeno-Raga M, Arias-Mutis ÓJ, San-Miguel T, Valles S, Aldasoro M, Vila JM, Zarzoso M, Mauricio MD. PPARγ as an indicator of vascular function in an experimental model of metabolic syndrome in rabbits. Atherosclerosis 2021; 332:16-23. [PMID: 34375909 DOI: 10.1016/j.atherosclerosis.2021.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 07/09/2021] [Accepted: 08/04/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS Underlying mechanisms associated with vascular dysfunction in metabolic syndrome (MetS) remain unclear and can even vary from one vascular bed to another. METHODS In this study, MetS was induced by a high-fat, high-sucrose diet, and after 28 weeks, aorta and renal arteries were removed and used for isometric recording of tension in organ baths, protein expression by Western blot, and histological analysis to assess the presence of atherosclerosis. RESULTS MetS induced a mild hypertension, pre-diabetes, central obesity and dyslipidaemia. Our results indicated that MetS did not change the contractile response in either the aorta or renal artery. Conversely, vasodilation was affected in both arteries in a different way. The aorta from MetS showed vascular dysfunction, including lower response to acetylcholine and sodium nitroprusside, while the renal artery from MetS presented a preserved relaxation to acetylcholine and an increased sensitivity to sodium nitroprusside. We did not find vascular oxidative stress in the aorta from MetS, but we found a significant decrease in PPARγ, phospho-Akt (p-Akt) and phospho-eNOS (p-eNOS) protein expression. On the other hand, we found oxidative stress in the renal artery from MetS, and PPARγ, Akt and p-Akt were overexpressed. No evidence of atherosclerosis was found in arteries from MetS. CONCLUSIONS MetS affects vascular function differently depending on the vessel. In the aorta, it decreases both the vasodilation and the expression of the PPARγ/Akt/eNOS pathway, while in the renal artery, it increases the expression of PPARγ/Akt signalling pathway without decreasing the vasodilation.
Collapse
Affiliation(s)
- Sol Guerra-Ojeda
- Department of Physiology. School of Medicine. University of Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Patricia Marchio
- Department of Physiology. School of Medicine. University of Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Marc Gimeno-Raga
- Department of Physiology. School of Medicine. University of Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Óscar Julián Arias-Mutis
- Department of Physiology. School of Medicine. University of Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Teresa San-Miguel
- Department of Pathology. School of Medicine. University of Valencia, Valencia, Spain
| | - Soraya Valles
- Department of Physiology. School of Medicine. University of Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Martin Aldasoro
- Department of Physiology. School of Medicine. University of Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - José M Vila
- Department of Physiology. School of Medicine. University of Valencia and Institute of Health Research INCLIVA, Valencia, Spain
| | - Manuel Zarzoso
- Department of Physiotherapy. School of Physiotherapy. University of Valencia, Valencia, Spain
| | - Maria D Mauricio
- Department of Physiology. School of Medicine. University of Valencia and Institute of Health Research INCLIVA, Valencia, Spain.
| |
Collapse
|
9
|
Chen IC, Lin JY, Liu YC, Chai CY, Yeh JL, Hsu JH, Wu BN, Dai ZK. Angiotensin-Converting Enzyme 2 Activator Ameliorates Severe Pulmonary Hypertension in a Rat Model of Left Pneumonectomy Combined With VEGF Inhibition. Front Med (Lausanne) 2021; 8:619133. [PMID: 33681251 PMCID: PMC7933511 DOI: 10.3389/fmed.2021.619133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/25/2021] [Indexed: 11/28/2022] Open
Abstract
Background: Pulmonary arterial hypertension (PAH) is a life-threatening and deteriorating disease with no promising therapy available currently due to its diversity and complexity. An imbalance between vasoconstriction and vasodilation has been proposed as the mechanism of PAH. Angiotensin-converting enzyme 2 (ACE2), which catalyzes the hydrolysis of the vasoconstrictor angiotensin (Ang) II into the vasodilator Ang-(1-7), has been shown to be an important regulator of blood pressure and cardiovascular diseases. Herein we hypothesized diminazene aceturate (DIZE), an ACE2 activator, could ameliorate the development of PAH and pulmonary vascular remodeling. Methods: A murine model of PAH was established using left pneumonectomy (PNx) on day 0 followed by injection of a single dose of the VEGF receptor-2 inhibitor SU5416 (25 mg/kg) subcutaneously on day 1. All hemodynamic and biochemical measurements were done at the end of the study on day 42. Animals were divided into 4 groups (n = 6–8/group): (1) sham-operated group, (2) vehicle-treatment group (SuPNx42), (3) early treatment group (SuPNx42/DIZE1−42) with DIZE at 15 mg/kg/day, subcutaneously from day 1 to day 42, and (4) late treatment group (SuPNx42/DIZE29−42) with DIZE from days 29–42. Results: In both the early and late treatment groups, DIZE significantly attenuated the mean pulmonary artery pressure, pulmonary arteriolar remodeling, and right ventricle brain natriuretic peptide (BNP), as well as reversed the overexpression of ACE while up-regulating the expression of Ang-(1-7) when compared with the vehicle-treatment group. In addition, the early treatment group also significantly decreased plasma BNP and increased the expression of eNOS. Conclusions: ACE2 activator has therapeutic potentials for preventing and attenuating the development of PAH in an animal model of left pneumonectomy combined with VEGF inhibition. Activation of ACE2 may thus be a useful therapeutic strategy for the treatment of human PAH.
Collapse
Affiliation(s)
- I-Chen Chen
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,College of Medicine, Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jao-Yu Lin
- Department of Surgery, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ching Liu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jwu-Lai Yeh
- College of Medicine, Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jong-Hau Hsu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bin-Nan Wu
- College of Medicine, Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zen-Kong Dai
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,College of Medicine, Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
10
|
Affiliation(s)
- Haihua Qiu
- Department of Cardiovascular Medicine The Affiliated Zhuzhou Hospital Xiangya Medical College Central South University Zhuzhou Hunan China
| | - Yi He
- Department of Cardiovascular Medicine The Affiliated Zhuzhou Hospital Xiangya Medical College Central South University Zhuzhou Hunan China
| | - Fan Ouyang
- Department of Cardiovascular Medicine The Affiliated Zhuzhou Hospital Xiangya Medical College Central South University Zhuzhou Hunan China
| | - Ping Jiang
- Department of Cardiovascular Medicine The Affiliated Zhuzhou Hospital Xiangya Medical College Central South University Zhuzhou Hunan China
| | - Shuhong Guo
- Department of Cardiovascular Medicine The Affiliated Zhuzhou Hospital Xiangya Medical College Central South University Zhuzhou Hunan China
| | - Yuan Guo
- Department of Cardiovascular Medicine The Affiliated Zhuzhou Hospital Xiangya Medical College Central South University Zhuzhou Hunan China
| |
Collapse
|
11
|
Tseng V, Sutliff RL, Hart CM. Redox Biology of Peroxisome Proliferator-Activated Receptor-γ in Pulmonary Hypertension. Antioxid Redox Signal 2019; 31:874-897. [PMID: 30582337 PMCID: PMC6751396 DOI: 10.1089/ars.2018.7695] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Peroxisome proliferator-activated receptor-gamma (PPARγ) maintains pulmonary vascular health through coordination of antioxidant defense systems, inflammation, and cellular metabolism. Insufficient PPARγ contributes to pulmonary hypertension (PH) pathogenesis, whereas therapeutic restoration of PPARγ activity attenuates PH in preclinical models. Recent Advances: Numerous studies in the past decade have elucidated the complex mechanisms by which PPARγ in the pulmonary vasculature and right ventricle (RV) protects against PH. The scope of PPARγ-interconnected pathways continues to expand and includes induction of antioxidant genes, transrepression of inflammatory signaling, regulation of mitochondrial biogenesis and bioenergetic integrity, control of cell cycle and proliferation, and regulation of vascular tone through interactions with nitric oxide and endogenous vasoactive molecules. Furthermore, PPARγ interacts with an extensive regulatory network of transcription factors and microRNAs leading to broad impact on cell signaling. Critical Issues: Abundant evidence suggests that targeting PPARγ exerts diverse salutary effects in PH and represents a novel and potentially translatable therapeutic strategy. However, progress has been slowed by an incomplete understanding of how specific PPARγ pathways are critically disrupted across PH disease subtypes and lack of optimal pharmacological ligands. Future Directions: Recent studies indicate that ligand-induced post-translational modifications of the PPARγ receptor differentially induce therapeutic benefits versus adverse side effects of PPARγ receptor activation. Strategies to selectively target PPARγ activity in diseased cells of pulmonary circulation and RV, coupled with development of ligands designed to specifically regulate post-translational PPARγ modifications, may unlock the full therapeutic potential of this versatile master transcriptional and metabolic regulator in PH.
Collapse
Affiliation(s)
- Victor Tseng
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia.,Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Roy L Sutliff
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia.,Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - C Michael Hart
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia.,Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| |
Collapse
|
12
|
Chen J, Shi M, Wang N, Yi P, Sun L, Meng Q. TSH inhibits eNOS expression in HMEC-1 cells through the TSHR/PI3K/AKT signaling pathway. ANNALES D'ENDOCRINOLOGIE 2019; 80:273-279. [PMID: 31606200 DOI: 10.1016/j.ando.2019.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate the effects of thyroid-stimulating hormone (TSH) on the expression of endothelial nitric oxide synthase (eNOS) in human microvascular endothelial cells (HMEC-1) and explore the potential mechanism. MATERIALS AND METHODS Expression of thyroid-stimulating hormone receptor (TSHR) in HMEC-1 cells was determined by immunofluorescence, reverse transcription-polymerase chain reaction (RT-PCR), and Western blotting. Cell proliferation and the production of nitric oxide (NO) and superoxide anion (SA) were measured after TSH treatment. eNOS expression and AKT phosphorylation were detected by Western blotting. RESULTS TSHR was expressed in HMEC-1 cells. TSH promoted HMEC-1 cell proliferation and SA production, but inhibited NO generation by dose-dependent blocking of mRNA and protein expression of eNOS. Mechanism studies demonstrated that TSH promoted AKT phosphorylation (P<0.05), and that LY294002 inhibited the reduction of eNOS expression by TSH. Moreover, TSH activated the AKT signaling pathway through binding to TSHR on HMEC-1 cells. CONCLUSIONS TSH inhibits NO production via the TSHR/AKT signaling pathway.
Collapse
Affiliation(s)
- Jing Chen
- Department of Endocrinology 1, Affiliated Hospital of Jining Medical University, 272029 Jining, Shandong, China
| | - Minmin Shi
- Department of Endocrinology 1, Affiliated Hospital of Jining Medical University, 272029 Jining, Shandong, China
| | - Na Wang
- Department of Endocrinology 1, Affiliated Hospital of Jining Medical University, 272029 Jining, Shandong, China
| | - Pengfei Yi
- Department of Endocrinology 1, Affiliated Hospital of Jining Medical University, 272029 Jining, Shandong, China
| | - Lin Sun
- Department of Endocrinology 1, Affiliated Hospital of Jining Medical University, 272029 Jining, Shandong, China
| | - Qiang Meng
- Department of Endocrinology 1, Affiliated Hospital of Jining Medical University, 272029 Jining, Shandong, China.
| |
Collapse
|
13
|
Monocrotaline pyrrole enhanced bone morphogenetic protein 7 signaling transduced by alternative activin A receptor type 2A in pulmonary arterial smooth muscle cells. Eur J Pharmacol 2019; 863:172679. [PMID: 31542483 DOI: 10.1016/j.ejphar.2019.172679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/11/2019] [Accepted: 09/18/2019] [Indexed: 01/20/2023]
Abstract
BACKGROUND Increased expression levels of bone morphogenetic protein 7 (BMP7) are associated with poor prognosis in pulmonary hypertension patients. However, whether BMP7 signaling conspire to involve in the proliferation of pulmonary artery smooth muscle cells (PASMC) underlying monocrotaline (MCT) induced pulmonary arterial hypertension (PAH) remain unclear. METHODS AND RESULTS Western blot experiments found BMP7 was increased in pulmonary arteries isolated from MCT-PAH rat. In addition, monocrotaline pyrrole (MCTP), the putative toxic metabolite of the MCT, increases the expression of BMP7, proliferating cell nuclear antigen (PCNA) and activin A receptor type 2A, but decreases bone morphogenetic protein receptor type 2 in cultured pulmonary artery smooth muscle cells (PASMC). In PASMCs, exogenous BMP7 leads to the decreasing expression of activin A receptor type 2, increasing phosphorylation of p38MAPK and elevation of P21. However, BMP7 treatment results in the increasing expression of activin A receptor type 2A, p38MAPK, and PCNA in bone morphogenetic protein receptor type 2 knockdown PASMCs. Knockdown of activin A receptor type 2A abrogated the MCTP-induced PCNA and cell cycle progression. CONCLUSIONS MCTP treatment lead to the expression of BMP7, suppression of bone morphogenetic protein receptor type 2 but increasing expression of activin A receptor type 2A, the BMP7 mediated PASMC proliferation via preferential activation of an activin A receptor type 2A signaling axis.
Collapse
|
14
|
Telmisartan and/or chlorogenic acid attenuates fructose-induced non-alcoholic fatty liver disease in rats: Implications of cross-talk between angiotensin, the sphingosine kinase/sphingoine-1-phosphate pathway, and TLR4 receptors. Biochem Pharmacol 2019; 164:252-262. [PMID: 31004566 DOI: 10.1016/j.bcp.2019.04.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022]
Abstract
Renin-angiotensin-aldosterone system (RAS) has been implicated in non-alcoholic fatty liver disease (NAFLD); the most common cause of chronic liver diseases. There is accumulating evidence that altered TLR4 and Sphingosine kinase 1(SphK1)/sphingosine1phosphate (S1P) signaling pathways are key players in the pathogenesis of NAFLD. Cross talk of the sphingosine signaling pathway, toll-4 (TLR4) receptors, and angiotensin II was reported in various tissues. Therefore, the aim of this study was to define the contribution of these two pathways to the hepatoprotective effects of telmisartan and/or chlorogenic acid (CGA) in NAFLD. CGA is a strong antioxidant that was previously reported to inhibit angiotensin converting enzyme. Male Wistar rats were treated with either high-fructose, with or without telmisartan, CGA, telmisartan + CGA for 8 weeks. Untreated NAFL rats showed characteristics of NAFLD, as evidenced by significant increase in the body weight, insulin resistance, and serum hepatotoxicity markers (Alanine and Aspartate transaminases) and lipids as compared to the negative control group, in addition to characteristic histopathological alterations. Treatment with either telmisartan and/or CGA improved aforementioned parameters, in addition to upregulation of antioxidant enzymes (Superoxide dismutase and Glutathione peroxidase). Effect of inhibiting RAS on both sphingosine pathway and TLR4 was evident by the suppressing effect of telmisartan and/or CGA on high fructose-induced upregulation of hepatic SPK1 and S1P, in addition to concomitant up-regulation of Sphingosine-1-Phosphate receptor (S1PR)3 protein level and increased expression of S1PR1 and TLR4. As TLR4 and SPK/S1P signaling pathways play important roles in the progression of liver inflammation, the effect on sphingosine pathway and TLR4 was associated with decreased concentrations of inflammatory markers, enzyme kB kinase (IKK), nuclear factor-kB and tumor necrosis factor-α as compared to untreated NAFL group. In conclusion, the present data strongly suggests the cross-talk between angiotensin, the Sphingosine SPK/S1P Axis and TLR4 Receptors, and their role in the pathogenesis of fructose-induced NAFLD, and the protection afforded by drugs inhibiting RAS.
Collapse
|
15
|
Dai G, Li B, Xu Y, Zeng Z, Yang H. Oxymatrine prevents the development of monocrotaline-induced pulmonary hypertension via regulation of the N G, N G-dimethyl-L-arginine metabolism pathways in rats. Eur J Pharmacol 2018; 842:338-344. [PMID: 30419238 DOI: 10.1016/j.ejphar.2018.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/06/2018] [Accepted: 11/06/2018] [Indexed: 12/21/2022]
Abstract
The purpose of this study was to investigate the potential effect of oxymatrine in monocrotaline-induced pulmonary hypertension and its possible influence on the NG,NG-dimethyl-L-arginine (ADMA) metabolism pathway. Pulmonary hypertension was induced in rats by a single-dose injection of monocrotaline (60 mg/kg). Daily oral administration of oxymatrine (25, 50 and 100 mg/kg) was started on the day following the monocrotaline injection for 28 days. Oxymatrine (50 and 100 mg/kg) significantly attenuated monocrotaline-induced lung and right ventricular hypertrophy, right ventricular systolic pressure elevation, and right ventricular dysfunction. Oxymatrine also reduced the thickening of monocrotaline-induced pulmonary arterial medial wall. Meanwhile, oxymatrine normalized the level of pulmonary asymmetric ADMA and attenuated the upregulated expression of protein arginine methyltransferase 1 (PRMT1). Oxymatrine had no effect on the expression of protein arginine methyltransferase 2 (PRMT2) and NG,NG-Dimethylarginine dimethylaminohydrolase 1 (DDAH1), which were upregulated in monocrotaline-induced pulmonary arterial hypertensive rats. However, the expression of the protein NG,NG-Dimethylarginine dimethylaminohydrolase 2 (DDAH2) did not differ among all groups (all P﹥0.05). These results suggest that oxymatrine may offer protective effects on the development of pulmonary hypertension by ameliorating pulmonary remodeling and modulating the ADMA metabolism pathway.
Collapse
Affiliation(s)
- Guidong Dai
- Key Laboratory for Modernization of Qiandongnan Miao & Dong Medicine, Qiandongnan Traditional Medicine Research & Development Center, Kaili University, 3 Kaiyuan Road, Kaili, Guizhou 556011, People's Republic of China
| | - Benpeng Li
- Key Laboratory for Modernization of Qiandongnan Miao & Dong Medicine, Qiandongnan Traditional Medicine Research & Development Center, Kaili University, 3 Kaiyuan Road, Kaili, Guizhou 556011, People's Republic of China
| | - Yuping Xu
- School of Life and Health Science, Kaili University, 3 Kaiyuan Road, Kaili, Guizhou 556011, People's Republic of China
| | - Zhuliang Zeng
- School of Life and Health Science, Kaili University, 3 Kaiyuan Road, Kaili, Guizhou 556011, People's Republic of China
| | - Hongyun Yang
- School of Life and Health Science, Kaili University, 3 Kaiyuan Road, Kaili, Guizhou 556011, People's Republic of China
| |
Collapse
|
16
|
Zhu G, Zhang W, Liu Y, Wang S. miR‑371b‑5p inhibits endothelial cell apoptosis in monocrotaline‑induced pulmonary arterial hypertension via PTEN/PI3K/Akt signaling pathways. Mol Med Rep 2018; 18:5489-5501. [PMID: 30387816 PMCID: PMC6236307 DOI: 10.3892/mmr.2018.9614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 03/16/2018] [Indexed: 01/25/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a clinical hemodynamic syndrome. It is characterized by elevated PA pressure and pulmonary vascular resistance. In the present study, the role of microRNA (miRNA/miR)-371b-5p in monocrotaline-induced PAH and the underlying mechanisms were investigated. In a monocrotaline-induced PAH rat model, gene chip and reverse transcription-quantitative polymerase chain reaction were employed to measure miRNA expression levels. The results revealed that miR-371b-5p was downregulated in PAH rats compared with the control group. In addition, in vitro results demonstrated that an miR-371b-5p inhibitor reduced miR-371b-5p expression levels, increased apoptosis and reduced proliferation of pulmonary arterial endothelial cells (PAECs) in rats with monocrotaline-induced PAH. Furthermore, inhibition of miR-371b-5p induced phosphatase and tensin homolog (PTEN) protein expression and suppressed that of phosphoinositide 3-kinase (PI3K) and phosphorylated (p)-Akt in the PAECs. In addition, VO-OHpic, a PTEN inhibitor, reduced the protein expression levels of PTEN in the PAECs and inhibited the effects of anti-miR-371b-5p on cell apoptosis. In addition, LY294002, a PI3K inhibitor, reduced the PI3K and p-Akt protein expression in the PAECs and reversed the effects of miR-371b-5p overexpression on the apoptosis of PAECs in rats with monocrotaline-induced PAH. Collectively, the results of the present study indicate that, in this animal model of PAH, miR-371b-5p inhibits apoptosis of PAECs via PTEN/PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Guangfa Zhu
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Wenmei Zhang
- Department of Pulmonary and Critical Care Medicine, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Yan Liu
- Department of Infectious Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Shenghao Wang
- Department of Infectious Diseases, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| |
Collapse
|
17
|
Guignabert C, de Man F, Lombès M. ACE2 as therapy for pulmonary arterial hypertension: the good outweighs the bad. Eur Respir J 2018; 51:51/6/1800848. [PMID: 29929959 DOI: 10.1183/13993003.00848-2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Christophe Guignabert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.,Faculté de Médecine, Université Paris-Sud and Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Frances de Man
- Dept of Pulmonology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc Lombès
- Faculté de Médecine, Université Paris-Sud and Université Paris Saclay, Le Kremlin-Bicêtre, France.,INSERM UMR_S 1185, Le Kremlin-Bicêtre, France.,Service d'Endocrinologie et des Maladies de la Reproduction, Hôpitaux Universitaires Paris-Sud, CHU Bicêtre, AH-HP, Le Kremlin Bicêtre, France
| |
Collapse
|
18
|
Feng L, Liu W, Yang J, Wang Q, Wen S. Effect of Hexadecyl Azelaoyl Phosphatidylcholine on Cardiomyocyte Apoptosis in Myocardial Ischemia-Reperfusion Injury: A Hypothesis. Med Sci Monit 2018; 24:2661-2667. [PMID: 29706617 PMCID: PMC5949054 DOI: 10.12659/msm.907578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reperfusion after myocardial ischemia can induce cardiomyocyte death, known as myocardial reperfusion injury. The pathophysiology of the process of reperfusion suggests the confluence multiple pathways. Recent studies have focused on the inflammatory response, which is considered to be the main mechanism during the process of myocardial ischemia-reperfusion injury and can cause cardiomyocyte apoptosis. Peroxisome proliferator-activated receptors gamma activated by endogenous ligands and exogenous ligand can decrease the inflammatory response in cardiomyocytes. Thiazolidinediones are synthetic, high-affinity, selective ligands for peroxisome proliferator-activated receptors gamma, and can inhibit the inflammatory response, decrease myocardial infarct size, and protect cardiac function. However, thiazolidinediones, including rosiglitazone and pioglitazone, can also contribute to adverse cardiovascular events such as congestive heart failure. Therefore, there are some limitations to the use of thiazolidinediones. Most endogenous ligands were of low affinity until hexadecyl azelaoyl phosphatidylcholine was identified as a high-affinity ligand and agonist for peroxisome proliferator-activated receptors gamma. Hexadecyl azelaoyl phosphatidylcholine binds recombinant peroxisome proliferator-activated receptors with an affinity (Kd(app) ≈40 nM) which is equivalent to rosiglitazone. Therefore, hexadecyl azelaoyl phosphatidylcholine is a specific peroxisome proliferator-activated receptors gamma agonist. Given these findings, we hypothesized that the use of hexadecyl azelaoyl phosphatidylcholine can activate the peroxisome proliferator-activated receptors gamma signal pathways and prevent the inflammatory response process of myocardial ischemia-reperfusion injury, with reduced cardiomyocyte apoptosis and death.
Collapse
Affiliation(s)
- Limin Feng
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China (mainland)
| | - Wennan Liu
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Jianzhou Yang
- Department of Preventive Medicine, Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| | - Qing Wang
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Shiwu Wen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
19
|
Li X, Bilali A, Qiao R, Paerhati T, Yang Y. Association of the PPARγ/PI3K/Akt pathway with the cardioprotective effects of tacrolimus in myocardial ischemic/reperfusion injury. Mol Med Rep 2018; 17:6759-6767. [PMID: 29488613 DOI: 10.3892/mmr.2018.8649] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 11/02/2017] [Indexed: 11/06/2022] Open
Abstract
Myocardial ischemia/reperfusion injury (MIRI) induces severe arrhythmias and has a high risk of mortality. The aim of the present study was to investigate the effect of tacrolimus on arrhythmias, cardiac function, oxidative stress and myocardium apoptosis induced by MIRI, and to elucidate the underlying mechanism. The effects of MIRI and tacrolimus on arrhythmias, cardiac function parameters, myocardial oxidative stress and apoptosis were investigated in a rat model of MIRI. The phosphorylation of peroxisome proliferator‑activated receptor γ (PPARγ) and protein kinase B (Akt) was investigated via western blotting. After rats were treated with inhibitors of PPARγ/phosphoinositide 3‑kinase (PI3K)/Akt, cardiac function parameters were measured. The results demonstrated that the MIRI procedure induced arrhythmias and significant impairment of cardiac function, oxidative stress and apoptosis in cardiomyocytes (P<0.05). Tacrolimus significantly alleviated the arrhythmias and impairment of cardiac function and inhibited the oxidative stress and apoptosis in cardiomyocytes (P<0.05). The phosphorylation of PPARγ and Akt was significantly activated by tacrolimus, whereas inhibitors of PPARγ/PI3K/Akt significantly abolished the effects of tacrolimus (P<0.05). Together, these results suggest that tacrolimus may protect rats from MIRI through activation of the PPARγ/PI3K/Akt pathway.
Collapse
Affiliation(s)
- Xiufen Li
- Cardiac Care Unit, The Traditional Hospital Affiliated to Xinjiang Medical University, Ürümqi, Xinjiang 830001, P.R. China
| | - Aishan Bilali
- Cardiac Care Unit, The Traditional Hospital Affiliated to Xinjiang Medical University, Ürümqi, Xinjiang 830001, P.R. China
| | - Rui Qiao
- Cardiac Care Unit, The Traditional Hospital Affiliated to Xinjiang Medical University, Ürümqi, Xinjiang 830001, P.R. China
| | - Tuerxun Paerhati
- Cardiac Care Unit, The Traditional Hospital Affiliated to Xinjiang Medical University, Ürümqi, Xinjiang 830001, P.R. China
| | - Yan Yang
- Cardiac Care Unit, The Traditional Hospital Affiliated to Xinjiang Medical University, Ürümqi, Xinjiang 830001, P.R. China
| |
Collapse
|
20
|
Chen J, Guo J, Cui X, Dai Y, Tang Z, Qu J, Raj JU, Hu Q, Gou D. The Long Noncoding RNA LnRPT Is Regulated by PDGF-BB and Modulates the Proliferation of Pulmonary Artery Smooth Muscle Cells. Am J Respir Cell Mol Biol 2018; 58:181-193. [DOI: 10.1165/rcmb.2017-0111oc] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Jidong Chen
- Shenzhen Key Laboratory of Microbial Genetic Engineering
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences, and
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Jiao Guo
- Shenzhen Key Laboratory of Microbial Genetic Engineering
| | - Xiaolei Cui
- Shenzhen Key Laboratory of Microbial Genetic Engineering
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences, and
| | - Yan Dai
- Key Laboratory of Systems Biology, Chinese Academy of Science, Shanghai Institute for Biological Sciences, Shanghai, China
| | - Zhixiong Tang
- Shenzhen Key Laboratory of Microbial Genetic Engineering
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences, and
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - J. Usha Raj
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois; and
| | - Qinghua Hu
- Department of Pathophysiology and
- Key Laboratory of Pulmonary Diseases of Ministry of Health, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, College of Life Sciences, and
| |
Collapse
|
21
|
Jin Z, Tan Q, Sun B. Telmisartan ameliorates vascular endothelial dysfunction in coronary slow flow phenomenon (CSFP). Cell Biochem Funct 2018; 36:18-26. [PMID: 29314204 DOI: 10.1002/cbf.3313] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/22/2017] [Accepted: 11/29/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Zhe Jin
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing, China
| | - Qindong Tan
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bei Sun
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
22
|
Shen Q, Li J, Zhang C, Wang P, Mohammed A, Ni S, Tang Z. Panax notoginseng saponins reduce high-risk factors for thrombosis through peroxisome proliferator-activated receptor -γ pathway. Biomed Pharmacother 2017; 96:1163-1169. [PMID: 29174034 DOI: 10.1016/j.biopha.2017.11.106] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/01/2017] [Accepted: 11/20/2017] [Indexed: 12/09/2022] Open
Abstract
The classic Virchow theory suggests that blood stasis, hypercoagulability and endothelial dysfunction are three major factors that cause venous thrombosis (VT). It is a complicated biological process involved multi-factors. Platelet plays a central role and participates in multiple links of this process. Panax notoginseng saponins (PNS), the principal constituents derived from panax notoginseng, has been widely described for its anti-platelet activity. However, its potential mechanism against platelet aggregation has not been clarified. In this present study, we evaluated the anti-platelet effects of PNS on thrombin-induced platelet activation and its possible molecular mechanism of action, and further explored the therapeutic action of PNS on thrombin induced hypercoagulability in rat. Our results showed that PNS treatment inhibited platelet aggregation induced by thrombin, which was accompanied with over-expression of Peroxisome proliferator-activated receptor γ (PPAR-γ) protein, mRNA and upregulation of phosphatidylinositol 3 kinase (PI3K)/ protein kinase B (Akt)/ endothelial nitric oxide synthase (eNOS) pathway in platelet, and this effect could be reversed by PPAR-γ inhibitor T0070907. In vivo, PNS significantly reversed thrombin-induced hypercoagulable state in rat which was accompanied by PPAR-γ protein and mRNA upregulation in rat lung. In conclusion, these data suggested that PNS could suppress thrombin-induced platelet aggregation in vitro and effectively improve hypercoagulable state in vivo and PNS-induced activation of PPAR-γ and its downstream PI3K/Akt/eNOS pathway played the central role.
Collapse
Affiliation(s)
- Qin Shen
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Jun Li
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Caixin Zhang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Pengbo Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Anaz Mohammed
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Songshi Ni
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Zhiyuan Tang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
23
|
Li J, Xiang X, Gong X, Shi Y, Yang J, Xu Z. Cilostazol protects mice against myocardium ischemic/reperfusion injury by activating a PPARγ/JAK2/STAT3 pathway. Biomed Pharmacother 2017; 94:995-1001. [PMID: 28810537 DOI: 10.1016/j.biopha.2017.07.143] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/20/2017] [Accepted: 07/27/2017] [Indexed: 02/06/2023] Open
Abstract
Myocardial ischemia/reperfusion (MIR) injury causes severe arrhythmias and a high lethality. The present study is designed to investigate the effect of cilostazol on MIR injury and the underlying mechaism. We measured the effects of cilostazol on heart function parameters in a mouse model of MIR. Proinflammatory cytokines and apoptosis proteins in the myocardium were examined to investigate the anti-inflammatory and anti-apoptosis ability of cilostazol. The participation of PPARγ/JAK2/STAT3 pathway was investigated. Results showed that the impairment of hemodynamic parameters caused by MIR was attenuated by cilostazol. The IL-6, IL-1β and TNF-a levels were all decreased by cilostazol. Cilostazol also significantly inhibited Bax and cleaved caspase-3 levels and restored the Bcl-2 levels. PPARγ, JAK2 and STAT3 were all activated by cilostazol. Treatment of inhibitors of them abolished the protective effects of cilostazol on cardiac function, myocardial inflammation and apoptosis. In summary, cilostazol alleviated the cardiac function impairment, myocardial inflammation and apoptosis induced by MIR. The results present a novel signaling mechanism that cilostazol protects MIR injury by activating a PPARγ/JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Jiangjin Li
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, PR China.
| | - Xiaoli Xiang
- Department of Nephrology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, PR China
| | - Xiaoxuan Gong
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, PR China
| | - Yafei Shi
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, PR China
| | - Jing Yang
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, PR China
| | - Zuo Xu
- Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, PR China
| |
Collapse
|
24
|
Ahmed LA, Rizk SM, El-Maraghy SA. Pinocembrin ex vivo preconditioning improves the therapeutic efficacy of endothelial progenitor cells in monocrotaline-induced pulmonary hypertension in rats. Biochem Pharmacol 2017; 138:193-204. [PMID: 28450224 DOI: 10.1016/j.bcp.2017.04.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/21/2017] [Indexed: 11/17/2022]
Abstract
Pulmonary hypertension is still not curable and the available current therapies can only alleviate symptoms without hindering the progression of disease. The present study was directed to investigate the possible modulatory effect of pinocembrin on endothelial progenitor cells transplanted in monocrotaline-induced pulmonary hypertension in rats. Pulmonary hypertension was induced by a single subcutaneous injection of monocrotaline (60mg/kg). Endothelial progenitor cells were in vitro preconditioned with pinocembrin (25mg/L) for 30min before being i.v. injected into rats 2weeks after monocrotaline administration. Four weeks after monocrotaline administration, blood pressure, electrocardiography and right ventricular systolic pressure were recorded. Rats were sacrificed and serum was separated for determination of endothelin-1 and asymmetric dimethylarginine levels. Right ventricles and lungs were isolated for estimation of tumor necrosis factor-alpha and transforming growth factor-beta contents as well as caspase-3 activity. Moreover, protein expression of matrix metalloproteinase-9 and endothelial nitric oxide synthase in addition to myocardial connexin-43 was assessed. Finally, histological analysis of pulmonary arteries, cardiomyocyte cross-sectional area and right ventricular hypertrophy was performed and cryosections were done for estimation of cell homing. Preconditioning with pinocembrin provided a significant improvement in endothelial progenitor cells' effect towards reducing monocrotaline-induced elevation of inflammatory, fibrogenic and apoptotic markers. Furthermore, preconditioned cells induced a significant amelioration of endothelial markers and cell homing and prevented monocrotaline-induced changes in right ventricular function and histological analysis compared with native cells alone. In conclusion, pinocembrin significantly improves the therapeutic efficacy of endothelial progenitor cells in monocrotaline-induced pulmonary hypertension in rats.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Apoptosis
- Biomarkers/blood
- Biomarkers/metabolism
- Bone Marrow Cells/cytology
- Bone Marrow Cells/drug effects
- Bone Marrow Cells/immunology
- Bone Marrow Transplantation/adverse effects
- Cells, Cultured
- Cytokines/metabolism
- Disease Models, Animal
- Endothelial Progenitor Cells/cytology
- Endothelial Progenitor Cells/drug effects
- Endothelial Progenitor Cells/immunology
- Endothelial Progenitor Cells/transplantation
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Flavanones/therapeutic use
- Graft Rejection/prevention & control
- Heart Ventricles/immunology
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Heart Ventricles/physiopathology
- Hypertension, Pulmonary/immunology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/surgery
- Lung/blood supply
- Lung/immunology
- Lung/metabolism
- Lung/pathology
- Male
- Pulmonary Artery/pathology
- Random Allocation
- Rats, Wistar
Collapse
Affiliation(s)
- Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Sherine M Rizk
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Shohda A El-Maraghy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
25
|
Shan H, Zhang S, Wei X, Li X, Qi H, He Y, Liu A, Luo D, Yu X. Protection of endothelial cells against Ang II-induced impairment: Involvement of both PPARα and PPARγ via PI3K/Akt pathway. Clin Exp Hypertens 2016; 38:571-577. [PMID: 27650941 DOI: 10.3109/10641963.2016.1174248] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- HaiYan Shan
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyang Zhang
- Science Experiment Center, China Medical University, Shenyang, China
| | - Xiaojie Wei
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuelian Li
- Department of Epidemiology, College of Public Health China Medical University, Shenyang, China
| | - Huimeng Qi
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yini He
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ao Liu
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Donghui Luo
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaosong Yu
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
26
|
Li P, Zhang D, Wan M, Liu J. PPARγ affects nitric oxide in human umbilical vein endothelial cells exposed to Porphyromonas gingivalis. Arch Oral Biol 2016; 68:116-22. [DOI: 10.1016/j.archoralbio.2016.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 03/17/2016] [Accepted: 04/10/2016] [Indexed: 02/08/2023]
|
27
|
CTRP9 Ameliorates Pulmonary Arterial Hypertension Through Attenuating Inflammation and Improving Endothelial Cell Survival and Function. J Cardiovasc Pharmacol 2016; 67:394-401. [DOI: 10.1097/fjc.0000000000000364] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Michel MC, Brunner HR, Foster C, Huo Y. Angiotensin II type 1 receptor antagonists in animal models of vascular, cardiac, metabolic and renal disease. Pharmacol Ther 2016; 164:1-81. [PMID: 27130806 DOI: 10.1016/j.pharmthera.2016.03.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 02/07/2023]
Abstract
We have reviewed the effects of angiotensin II type 1 receptor antagonists (ARBs) in various animal models of hypertension, atherosclerosis, cardiac function, hypertrophy and fibrosis, glucose and lipid metabolism, and renal function and morphology. Those of azilsartan and telmisartan have been included comprehensively whereas those of other ARBs have been included systematically but without intention of completeness. ARBs as a class lower blood pressure in established hypertension and prevent hypertension development in all applicable animal models except those with a markedly suppressed renin-angiotensin system; blood pressure lowering even persists for a considerable time after discontinuation of treatment. This translates into a reduced mortality, particularly in models exhibiting marked hypertension. The retrieved data on vascular, cardiac and renal function and morphology as well as on glucose and lipid metabolism are discussed to address three main questions: 1. Can ARB effects on blood vessels, heart, kidney and metabolic function be explained by blood pressure lowering alone or are they additionally directly related to blockade of the renin-angiotensin system? 2. Are they shared by other inhibitors of the renin-angiotensin system, e.g. angiotensin converting enzyme inhibitors? 3. Are some effects specific for one or more compounds within the ARB class? Taken together these data profile ARBs as a drug class with unique properties that have beneficial effects far beyond those on blood pressure reduction and, in some cases distinct from those of angiotensin converting enzyme inhibitors. The clinical relevance of angiotensin receptor-independent effects of some ARBs remains to be determined.
Collapse
Affiliation(s)
- Martin C Michel
- Dept. Pharmacology, Johannes Gutenberg University, Mainz, Germany; Dept. Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim, Ingelheim, Germany.
| | | | - Carolyn Foster
- Retiree from Dept. of Research Networking, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Yong Huo
- Dept. Cardiology & Heart Center, Peking University First Hospital, Beijing, PR China
| |
Collapse
|
29
|
Abstract
Calorie restriction (CR) is one of the most effective nonpharmacological interventions protecting against cardiovascular disease, such as hypertension in the systemic circulation. However, whether CR could attenuate pulmonary arterial hypertension (PAH) is largely unknown. The PAH model was developed by subjecting the rats to a single subcutaneous injection of monocrotaline. CR lowered mean pulmonary arterial pressure (mPAP) and reduced vascular remodeling and right ventricular hypertrophy in PAH rats. Meanwhile, CR attenuated endothelial dysfunction as evidenced by increased relaxation in response to acetylcholine. The beneficial effects of CR were associated with restored sirtuin-1 (SIRT1) expression and endothelial nitric oxide synthase (eNOS) phosphorylation and reduced eNOS acetylation in pulmonary arteries of PAH rats. To further clarify the role of SIRT1 in the protective effects of CR, adenoviral vectors for overexpression of SIRT1 were administered intratracheally at 1 day before monocrotaline injection. Overexpression of SIRT1 exhibited similar beneficial effects on mPAP and endothelial function, and increased eNOS phosphorylation and reduced eNOS acetylation in the absence of CR. Moreover, SIRT1 overexpression attenuated the increase in mPAP in hypoxia-induced PAH animals. Overall, the present data demonstrate that CR may serve as an effective treatment of PAH, and targeting the SIRT1/eNOS pathway may improve treatment of PAH.
Collapse
|
30
|
Ti Y, Xie GL, Wang ZH, Ding WY, Zhang Y, Zhong M, Zhang W. Tribbles 3: A potential player in diabetic aortic remodelling. Diab Vasc Dis Res 2016; 13:69-80. [PMID: 26410836 DOI: 10.1177/1479164115605645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tribbles 3, whose expression is up-regulated by insulin resistance, was confirmed to be involved in diabetic cardiomyopathy in our previous study. However, it is not known whether Tribbles 3 has a role on conduit arteries such as the aorta in diabetes. Type 2 diabetic rat model was induced by high-fat diet and low-dose streptozotocin. We evaluated the characteristics of diabetic rats by serial ultrasonography and histopathologic analyses of aortic wall architecture. Diabetic rats displayed increased aortic medial thickness, excessive collagen deposition, diminished elastic fibres and reduced vascular compliance together with Tribbles 3 overexpression. To further investigate the role of Tribbles 3 in aortic remodelling, we used Tribbles 3 gene silencing in vivo 12 weeks after onset of diabetes. Silence of Tribbles 3 significantly reversed pathological aortic remodelling without blood pressure modification. In Tribbles 3-small interfering RNA group, medial thickness and perivascular fibrosis were markedly decreased; moreover, there were prominent reductions in collagen content and collagen/elastin ratio, resulting in an improved arterial compliance. Additionally, with Tribbles 3 silencing, the diminished phosphorylation of PI3K/Akt was restored, and increased activation of MKK4/JNK was decreased. Silence of Tribbles 3 is potent in mediating reversal of aortic remodelling, implicating that Tribbles 3 is proposed to be a potential therapeutic target for vascular complication in diabetes.
Collapse
Affiliation(s)
- Yun Ti
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Guo-lu Xie
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, P.R. China Department of Cardiology, Tianjin Chest Hospital, Tianjin, P.R. China
| | - Zhi-hao Wang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Wen-yuan Ding
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Ming Zhong
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, P.R. China
| | - Wei Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, P.R. China
| |
Collapse
|
31
|
Su X, Yu R, Yang X, Zhou G, Wang Y, Li L, Li D. Telmisartan attenuates peritoneal fibrosis via peroxisome proliferator-activated receptor-γactivation in rats. Clin Exp Pharmacol Physiol 2015; 42:671-9. [PMID: 25867712 DOI: 10.1111/1440-1681.12403] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/24/2015] [Accepted: 04/02/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Xuesong Su
- Department of Nephrology; Shengjing Hospital of China Medical University; Shenyang China
| | - Rui Yu
- Department of Nephrology; Shengjing Hospital of China Medical University; Shenyang China
| | - Xu Yang
- Department of Nephrology; Shengjing Hospital of China Medical University; Shenyang China
| | - Guangyu Zhou
- Department of Nephrology; Shengjing Hospital of China Medical University; Shenyang China
| | - Yanqiu Wang
- Department of Nephrology; Shengjing Hospital of China Medical University; Shenyang China
| | - Li Li
- Department of Nephrology; Shengjing Hospital of China Medical University; Shenyang China
| | - Detian Li
- Department of Nephrology; Shengjing Hospital of China Medical University; Shenyang China
| |
Collapse
|
32
|
Li X, Cheng XW, Hu L, Wu H, Hao CN, Jiang H, Zhu E, Huang Z, Inoue A, Sasaki T, Du Q, Takeshita K, Okumura K, Murohara T, Kuzuya M. Cathepsin S activity controls ischemia-induced neovascularization in mice. Int J Cardiol 2015; 183:198-208. [PMID: 25668148 DOI: 10.1016/j.ijcard.2015.01.058] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/11/2014] [Accepted: 01/25/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND Evidence from human and animal studies has demonstrated elevated levels of the cysteine protease cathepsin S (CatS) in hypoxic atherosclerotic lesions. We hypothesized that silencing of CatS gene would suppress ischemia-induced angiogenic action. METHODS AND RESULTS Left femoral artery ligation-induced ischemia in mice showed the increased expression and activity of CatS in the ischemic muscle. The CatS-deficiency (CatS(-/-)) mice showed impaired functional recovery following hindlimb ischemia and reduced levels of peroxisome proliferator-activated receptor-γ (PPAR-γ), phospho-Akt (p-Akt), p-endothelial nitric oxide synthase, p-extracellular signal-regulated kinase1/2 (Erk1/2), p-p38 mitogen-activated protein kinase, and vascular endothelial growth factor (VEGF) proteins, as well as reduced levels of matrix metalloproteinase-9 and macrophage infiltration in the ischemic muscles. In vitro, CatS silencing reduced the levels of these targeted essential molecules for angiogenesis and vasculogenesis. Together, the results indicated that the effects of CatS knockdown led to defective endothelial cell invasion, proliferation, and tube formation. This notion was reinforced by the finding that CatS inhibition led to a decreased PPAR-γ level and VEGF/Erk1/2 signaling activation in response to ischemia. CatS(-/-) resulted in decreased circulating EPC-like CD31(+)/c-Kit(+) cells, accompanied by the reduction of the cellular levels of PPAR-γ, p-Akt, and VEGF induced by ischemic stress. Transplantation of bone-marrow-derived mononuclear cells from CatS(+/+) mice restored neovascularization in CatS(-/-) mice. CONCLUSIONS CatS activity controls ischemia-induced neovascularization partially via the modulation of PPAR-γ and VEGF/Akt signaling activation.
Collapse
Affiliation(s)
- Xiang Li
- Department of Cardiology, Yanbian University Hospital, China
| | - Xian Wu Cheng
- Department of Cardiology, Yanbian University Hospital, China; Department of Geriatrics, Nagoya University Graduate School of Medicine, Japan.
| | - Lina Hu
- Department of Geriatrics, Nagoya University Graduate School of Medicine, Japan
| | - Hongxian Wu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Chang-Ning Hao
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Haiying Jiang
- Department of Geriatrics, Nagoya University Graduate School of Medicine, Japan; Department of Physiology, Yanbian University School of Medicine, China
| | - Enbo Zhu
- Department of Cardiology, Yanbian University Hospital, China
| | - Zhe Huang
- Department of Neurology, Nagoya University Graduate School of Medicine, Japan
| | - Aiko Inoue
- Department of Geriatrics, Nagoya University Graduate School of Medicine, Japan
| | - Takeshi Sasaki
- Department of Anatomy, Hamamatsu University School of Medicine, Japan
| | - Qiuna Du
- Department of Nephrology, Nagoya University Graduate School of Medicine, Japan
| | - Kyosuke Takeshita
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Kenji Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Masafumi Kuzuya
- Department of Geriatrics, Nagoya University Graduate School of Medicine, Japan
| |
Collapse
|
33
|
Li BH, Liao SQ, Yin YW, Long CY, Guo L, Cao XJ, Liu Y, Zhou Y, Gao CY, Zhang LL, Li JC. Telmisartan-induced PPARγ activity attenuates lipid accumulation in VSMCs via induction of autophagy. Mol Biol Rep 2014; 42:179-86. [DOI: 10.1007/s11033-014-3757-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 09/17/2014] [Indexed: 01/05/2023]
|
34
|
Almodovar S. The complexity of HIV persistence and pathogenesis in the lung under antiretroviral therapy: challenges beyond AIDS. Viral Immunol 2014; 27:186-99. [PMID: 24797368 DOI: 10.1089/vim.2013.0130] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Antiretroviral therapy (ART) represents a significant milestone in the battle against AIDS. However, we continue learning about HIV and confronting challenges 30 years after its discovery. HIV has cleverly tricked both the host immune system and ART. First, the many HIV subtypes and recombinant forms have different susceptibilities to antiretroviral drugs, which may represent an issue in countries where ART is just being introduced. Second, even under the suppressive pressures of ART, HIV still increases inflammatory mediators, deregulates apoptosis and proliferation, and induces oxidative stress in the host. Third, the preference of HIV for CXCR4 as a co-receptor may also have noxious outcomes, including potential malignancies. Furthermore, HIV still replicates cryptically in anatomical reservoirs, including the lung. HIV impairs bronchoalveolar T-lymphocyte and macrophage immune responses, rendering the lung susceptible to comorbidities. In addition, HIV-infected individuals are significantly more susceptible to long-term HIV-associated complications. This review focuses on chronic obstructive pulmonary disease (COPD), pulmonary arterial hypertension, and lung cancer. Almost two decades after the advent of highly active ART, we now know that HIV-infected individuals on ART live as long as the uninfected population. Fortunately, its availability is rapidly increasing in low- and middle-income countries. Nevertheless, ART is not risk-free: the developed world is facing issues with antiretroviral drug toxicity, resistance, and drug-drug interactions, while developing countries are confronting issues with immune reconstitution inflammatory syndrome. Several aspects of the complexity of HIV persistence and challenges with ART are discussed, as well as suggestions for new avenues of research.
Collapse
Affiliation(s)
- Sharilyn Almodovar
- Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus , Aurora, Colorado
| |
Collapse
|
35
|
Singh B, Sangle GV, Murugan J, Umrani R, Roy S, Kulkarni O, Semwal A, Unnikrishnan MK, Jain M. Effect of combination treatment of S-amlodipine with peroxisome proliferator-activated receptor agonists on metabolic and cardiovascular parameters in Zucker fa/fa rats. Diabetol Metab Syndr 2014; 6:45. [PMID: 24673913 PMCID: PMC3976049 DOI: 10.1186/1758-5996-6-45] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 03/24/2014] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Type 2 diabetes is a complex metabolic disorder characterized by hyperglycemia, impaired glucose tolerance and insulin resistance associated with dyslipidemia and hypertension. The available drugs are not sufficiently efficacious in reducing cardiovascular risk and restoring normal glucose metabolism associated with type 2 diabetes as a mono- or a combination therapy. The present study examined the combined effects of an antihypertensive (S-Amlodipine) and an insulin-sensitizing agent, peroxisome proliferator-activated receptor (PPAR) agonists (Pioglitazone and Ragaglitazar), on cardiovascular risk factors in aged diabetic and insulin-resistant Zucker fa/fa rats. METHODS Following combination treatment for 14 days, blood pressure (BP), serum glucose, total cholesterol and triglycerides were measured. Aortic ring study was conducted to determine the effect of combination treatments on phenylephrine-induced vasoconstriction and acetylcholine (Ach)-induced vasorelaxation. RESULTS In combination, S-Amlodipine and Pioglitazone significantly reduced blood glucose (115.1 ± 6.6 vs. 81.7 ± 4.2), BP (184.4 ± 5.0 vs. 155.1 ± 5.0), serum triglycerides (362.5 ± 47.5 vs. 211.1 ± 23.7) and glucose intolerance when compared with vehicle treated Zucker fa/fa rats. Similar results were observed with the combination of S-Amlodipine and Ragaglitazar (Triglycerides, 362.5 ± 47.5 vs. 252.34 ± 27.86; BP, 184.4 ± 5.0 vs. 159.0 ± 8.0) except for serum glucose. ACh-induced vasorelaxation in aortic rings was also superior with both of the combinations compared to individual treatment. Furthermore, there was less body weight gain and food intake with S-Amlodipine and Pioglitazone combination in Zucker fa/fa rats. S-Amlodipine itself caused significant reduction in glucose (115.1 ± 6.6 vs. 89.7 ± 2.7) and BP (184.4 ± 5.0 vs. 156.1 ± 4.0) with improvement in insulin sensitivity observed through oral glucose tolerance test. CONCLUSIONS The results suggest that a combination of PPAR agonists and S-Amlodipine has partial benefits in improving the cardiovascular risk factors such as reduction in triglyceride levels, associated with chronic type 2 diabetes, and therefore may be utilized as an approach for addressing some of these devastating metabolic syndrome complications.
Collapse
Affiliation(s)
- Bhagat Singh
- Department of Clinical Neuroscience, University of Calgary, 3330 Hospital Drive, NW, Calgary, Alberta T2N4N1, Canada
| | - Ganesh V Sangle
- Department of Physiology, University of Manitoba, Winnipeg, Canada
| | - Jeya Murugan
- Department of Pharmacology, Zydus Research Centre, Ahmedabad, India
| | - Rinku Umrani
- Department of Pharmacology, Zydus Research Centre, Ahmedabad, India
| | - Subhasis Roy
- Department of Pharmacology, Zydus Research Centre, Ahmedabad, India
| | - Onkar Kulkarni
- Department of Pharmacology, Zydus Research Centre, Ahmedabad, India
- Department of Pharmacy, BITS Pilani Hyderabad campus, Jawahar Nagar, Hyderabad, India
| | - Arvind Semwal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Karnataka, India
| | - MK Unnikrishnan
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Karnataka, India
| | - Mukul Jain
- Department of Pharmacology, Zydus Research Centre, Ahmedabad, India
| |
Collapse
|