1
|
Ayik S, Gunata M, Ozhan O, Yildiz A, Vardi N, Sonmez E, Ermis N, Ates N, Kilic E, Noma SAA, Ulu A, Inan ST, Acet HA, Parlakpinar H. Alamandin and especially melatonin attenuate pulmonary arterial hypertension induced by monocrotalin. Fundam Clin Pharmacol 2024; 38:1143-1154. [PMID: 39128482 DOI: 10.1111/fcp.13033] [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: 06/15/2023] [Revised: 02/22/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Despite the available treatments, pulmonary arterial hypertension (PAH) prognosis is poor. OBJECTIVES We aimed to investigate the effects of the alamandine (ALA), melatonin (MEL), and ALA + MEL in PAH. METHODS The rats were randomly divided into Control (n = 10), monocrotaline (MCT) (n = 12), ALA (n = 12), MEL (n = 12), and ALA + MEL (n = 12) groups. PAH was induced by MCT. The ALA, MEL, and ALA + MEL groups received 50 μg/kg/day ALA, 10 mg/kg/day MEL, and ALA + MEL, respectively, for 35 days. Echocardiographic and hemodynamic measurements and tissue analyses (morphometric, histopathological, ELISA, and western blot) were performed. RESULTS Monotherapies, especially MEL, reduced the right ventricular (RV) systolic pressure. Only MEL increased the pulmonary artery acceleration time. MCT increased the RV/left ventricle (LV) + interventricular septum (IVS) ratio. While ALA and ALA + MEL slightly decreased the RV/(LV + IVS), MEL significantly restored it. MCT increased the tunica intima-media (TIM) thickness, PCNA and α-SMA of pulmonary arterioles, histopathological score (HS) (inflammatory infiltration etc.) of the lung, and RV. All treatments reduced the TIM thickness (especially MEL), PCNA, and α-SMA. All treatments significantly decreased the HS of the lung; however, MEL and ALA + MEL produced greater benefits. All treatments attenuated the HS of RV. MCT caused a significant increase in lung lysyl oxidase (LOX) activity. All treatments restored the LOX; however, MEL and ALA + MEL provided greater improvement. While lung Nrf-2 was increased in MCT-treated rats, MEL reduced it. CONCLUSION ALA, MEL, and ALA + MEL attenuate PAH and protect RV via antiproliferative, anti-remodeling, antihypertrophic, anti-inflammatory, and free radical scavenging (only MEL) capabilities. Overall, MEL produced the best outcomes.
Collapse
Affiliation(s)
- Seyhan Ayik
- Department of Medical Pharmacology, Inonu University, Malatya, Turkey
| | - Mehmet Gunata
- Department of Medical Pharmacology, Inonu University, Malatya, Turkey
| | - Onural Ozhan
- Department of Medical Pharmacology, Inonu University, Malatya, Turkey
| | - Azibe Yildiz
- Department of Histology and Embryology, Inonu University, Malatya, Turkey
| | - Nigar Vardi
- Department of Histology and Embryology, Inonu University, Malatya, Turkey
| | - Emre Sonmez
- Department of Cardiology, Inonu University, Malatya, Turkey
| | - Necip Ermis
- Department of Cardiology, Inonu University, Malatya, Turkey
| | - Nilay Ates
- Department of Medical Pharmacology, Istanbul Medipol University, Istanbul, Turkey
| | - Ertugrul Kilic
- Department of Physiology, Istanbul Medipol University, Istanbul, Turkey
| | - Samir Abbas Ali Noma
- Biochemistry and Biomaterials Research Laboratory, Department of Chemistry, Inonu University, Malatya, Turkey
| | - Ahmet Ulu
- Biochemistry and Biomaterials Research Laboratory, Department of Chemistry, Inonu University, Malatya, Turkey
| | | | - Haci Ahmet Acet
- Department of Medical Pharmacology, Inonu University, Malatya, Turkey
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Inonu University, Malatya, Turkey
| |
Collapse
|
2
|
Lisboa CD, Maciel de Souza JL, Gaspar CJ, Turck P, Ortiz VD, Teixeira Proença IC, Fernandes TRG, Fernandes E, Tasca S, Carraro CC, Belló-Klein A, Sander da Rosa Araujo A, Luz de Castro A. Melatonin effects on oxidative stress and on TLR4/NF-kβ inflammatory pathway in the right ventricle of rats with pulmonary arterial hypertension. Mol Cell Endocrinol 2024; 592:112330. [PMID: 39002930 DOI: 10.1016/j.mce.2024.112330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/19/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Pulmonary arterial hypertension (PAH) is characterised by an increase in mean pulmonary arterial pressure and a compromised the right ventricle (RV), together with progression to heart failure and premature death. Studies have evaluated the role of melatonin as a promising therapeutic strategy for PAH. The objective of this study was to evaluate melatonin's effects on oxidative stress and on the TLR4/NF-kβ inflammatory pathway in the RV of rats with PAH. Male Wistar rats were divided into the following groups: control, monocrotaline (MCT), and monocrotaline plus melatonin groups. These two last groups received one intraperitoneal injection of MCT (60 mg/kg) on the first day of experimental protocol. The monocrotaline plus melatonin group received 10 mg/kg/day of melatonin by gavage for 21 days. Echocardiographic analysis was performed, and the RV was collected for morphometric analysis oxidative stress and molecular evaluations. The main findings of the present study were that melatonin administration attenuated the reduction in RV function that was induced by monocrotaline, as assessed by TAPSE. In addition, melatonin prevented RV diastolic area reduction caused by PAH. Furthermore, animals treated with melatonin did not show an increase in ROS levels or in NF-kβ expression. In addition, the monocrotaline plus melatonin group showed a reduction in TLR4 expression when compared with control and monocrotaline groups. To our knowledge, this is the first study demonstrating a positive effect of melatonin on the TLR4/NF-kβ pathway in the RV of rats with PAH. In this sense, this study makes it possible to think of melatonin as a possible ally in mitigating RV alterations caused by PAH.
Collapse
Affiliation(s)
- Cristiane Dias Lisboa
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - José Luciano Maciel de Souza
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Custódio José Gaspar
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Patrick Turck
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Vanessa Duarte Ortiz
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Isabel Cristina Teixeira Proença
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Tânia Regina G Fernandes
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Elissa Fernandes
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Silvio Tasca
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Cristina Campos Carraro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Adriane Belló-Klein
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Alex Sander da Rosa Araujo
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil
| | - Alexandre Luz de Castro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Science (ICBS), Federal University of Rio Grande do Sul (UFRGS), Ramiro Barcelos Street, 2600, Santa Cecília, CEP: 90035-003, Porto Alegre, RS, Brazil.
| |
Collapse
|
3
|
Jacobs S, Payne C, Shaboodien S, Kgatla T, Pretorius A, Jumaar C, Maarman G, Sanni O. Pulmonary hypertension and the potential of 'drug' repurposing: A case for African medicinal plants. Afr J Thorac Crit Care Med 2024; 30:e1352. [PMID: 39171151 PMCID: PMC11334905 DOI: 10.7196/ajtccm.2024.v30i2.1352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 03/25/2024] [Indexed: 08/23/2024] Open
Abstract
Abstract Pulmonary hypertension (PH) is a haemodynamic disorder in which elevated blood pressure in the pulmonary circulation is caused by abnormal vascular tone. Despite advances in treatment, PH mortality remains high, and drug repurposing has been proposed as a mitigating approach. This article reviews the studies that have investigated drug repurposing as a viable option for PH. We provide an overview of PH and highlight pharmaceutical drugs with repurposing potential, based on limited evidence of their mechanisms of action. Moreover, studies have demonstrated the benefits of medicinal plants in PH, most of which are of Indian or Asian origin. Africa is a rich source of many medicinal plants that have been scientifically proven to counteract myriad pathologies. When perusing these studies, one will notice that some African medicinal plants can counteract the molecular pathways (e.g. proliferation, vasoconstriction, inflammation, oxidative stress and mitochondrial dysfunction) that are also involved in the pathogenesis of PH. We review the actions of these plants with actions applicable to PH and highlight that they could be repurposed as adjunct PH therapies. However, these plants have either never been tested in PH, or there is little evidence of their actions against PH. We therefore encourage caution, as more research is needed to study these plants further in experimental models of PH while acknowledging that the outcomes of such proof of-concept studies may not always yield promising findings. Regardless, this article aims to stimulate future research that could make timely contributions to the field. Study synopsis What the study adds. Pulmonary hypertension (PH) remains a fatal disease, and 80% of the patients live in developing countries where resources are scarce and specialised therapies are often unavailable. Drug repurposing is a viable option to try to improve treatment outcomes.Implications of the findings. We propose that another form of 'drug' repurposing is the use of medicinal plants, many of which have demonstrated benefits against pathological processes that are also key in PH, e.g. apoptosis, tumour-like growth of cells, proliferation, oxidative stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- S Jacobs
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - C Payne
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - S Shaboodien
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - T Kgatla
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - A Pretorius
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - C Jumaar
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - G Maarman
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - O Sanni
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
4
|
Gu P, Wu Y, Lu W. New Perspectives on the Role and Therapeutic Potential of Melatonin in Cardiovascular Diseases. Am J Cardiovasc Drugs 2024; 24:171-195. [PMID: 38436867 DOI: 10.1007/s40256-024-00631-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2024] [Indexed: 03/05/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death and disability worldwide. It is essential to develop novel interventions to prevent/delay CVDs by targeting their fundamental cellular and molecular processes. Melatonin is a small indole molecule acting both as a hormone of the pineal gland and as a local regulator molecule in various tissues. It has multiple features that may contribute to its cardiovascular protection. Moreover, melatonin enters all cells and subcellular compartments and crosses morphophysiological barriers. Additionally, this indoleamine also serves as a safe exogenous therapeutic agent. Increasing evidence has demonstrated the beneficial effects of melatonin in preventing and improving cardiovascular risk factors. Exogenous administration of melatonin, as a result of its antioxidant and anti-inflammatory properties, has been reported to decrease blood pressure, protect against atherosclerosis, attenuate molecular and cellular damage resulting from cardiac ischemia/reperfusion, and improve the prognosis of myocardial infarction and heart failure. This review aims to summarize the beneficial effects of melatonin against these conditions, the possible protective mechanisms of melatonin, and its potential clinical applicability in CVDs.
Collapse
Affiliation(s)
- Pengchen Gu
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiang Su Prov., China
| | - Yuxin Wu
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiang Su Prov., China
| | - Weiwei Lu
- Department of Physiology and Neurobiology, Suzhou Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, Jiang Su Prov., China.
| |
Collapse
|
5
|
Hosseinzadeh A, Pourhanifeh MH, Amiri S, Sheibani M, Irilouzadian R, Reiter RJ, Mehrzadi S. Therapeutic potential of melatonin in targeting molecular pathways of organ fibrosis. Pharmacol Rep 2024; 76:25-50. [PMID: 37995089 DOI: 10.1007/s43440-023-00554-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/24/2023]
Abstract
Fibrosis, the excessive deposition of fibrous connective tissue in an organ in response to injury, is a pathological condition affecting many individuals worldwide. Fibrosis causes the failure of tissue function and is largely irreversible as the disease progresses. Pharmacologic treatment options for organ fibrosis are limited, but studies suggest that antioxidants, particularly melatonin, can aid in preventing and controlling fibrotic damage to the organs. Melatonin, an indole nocturnally released from the pineal gland, is commonly used to regulate circadian and seasonal biological rhythms and is indicated for treating sleep disorders. While it is often effective in treating sleep disorders, melatonin's anti-inflammatory and antioxidant properties also make it a promising molecule for treating other disorders such as organ fibrosis. Melatonin ameliorates the necrotic and apoptotic changes that lead to fibrosis in various organs including the heart, liver, lung, and kidney. Moreover, melatonin reduces the infiltration of inflammatory cells during fibrosis development. This article outlines the protective effects of melatonin against fibrosis, including its safety and potential therapeutic effects. The goal of this article is to provide a summary of data accumulated to date and to encourage further experimentation with melatonin and increase its use as an anti-fibrotic agent in clinical settings.
Collapse
Affiliation(s)
- Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Shiva Amiri
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Sheibani
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rana Irilouzadian
- Clinical Research Development Unit of Shohada-e Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Jacobs S, Payne C, Shaboodien S, Kgatla T, Pretorius A, Jumaar C, Sanni O, Butrous G, Maarman G. Gut microbiota crosstalk mechanisms are key in pulmonary hypertension: The involvement of melatonin is instrumental too. Pulm Circ 2023; 13:e12277. [PMID: 37583483 PMCID: PMC10423855 DOI: 10.1002/pul2.12277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023] Open
Abstract
The microbiota refers to a plethora of microorganisms with a gene pool of approximately three million, which inhabits the human gastrointestinal tract or gut. The latter, not only promotes the transport of nutrients, ions, and fluids from the lumen to the internal environment but is linked with the development of diseases including coronary artery disease, heart failure, and lung diseases. The exact mechanism of how the microbiota achieves crosstalk between itself and distant organs/tissues is not clear, but factors released to other organs may play a role, like inflammatory and genetic factors, and now we highlight melatonin as a novel mediator of the gut-lung crosstalk. Melatonin is present in high concentrations in the gut and the lung and has recently been linked to the pathogenesis of pulmonary hypertension (PH). In this comprehensive review of the literature, we suggest that melatonin is an important link between the gut microbiota and the development of PH (where suppressed melatonin-crosstalk between the gut and lungs could promote the development of PH). More studies are needed to investigate the link between the gut microbiota, melatonin and PH. Studies could also investigate whether microbiota genes play a role in the epigenetic aspects of PH. This is relevant because, for example, dysbiosis (caused by epigenetic factors) could reduce melatonin signaling between the gut and lungs, reduce subcellular melatonin concentrations in the gut/lungs, or reduce melatonin serum levels secondary to epigenetic factors. This area of research is largely unexplored and further studies are warranted.
Collapse
Affiliation(s)
- Steve Jacobs
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Carmen Payne
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Sara Shaboodien
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Thato Kgatla
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Amy Pretorius
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Chrisstoffel Jumaar
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Olakunle Sanni
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Ghazwan Butrous
- School of Pharmacy, Imperial College of LondonUniversity of KentCanterburyUK
| | - Gerald Maarman
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| |
Collapse
|
7
|
Li D, Pan JH, Huang XF, Liao YQ, Ling YJ, Luo JY. Effect of melatonin on oxidative stress indicators in animal models of fibrosis: A systematic review and meta-analysis. Free Radic Biol Med 2023; 195:158-177. [PMID: 36586451 DOI: 10.1016/j.freeradbiomed.2022.12.094] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/20/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Imbalance of oxidative stress has been detected in a range of fibrotic diseases. Melatonin as an indoleamine hormone plays an important role in regulating the circadian rhythm of human, while in recent years, its antioxidant effect has also attracted increasing attention. This study aimed to perform a systematic review and meta-analysis to comprehensively evaluate the antioxidant effect of melatonin in animal models of fibrosis. METHODS The PubMed, Cochrane Library, EMBASE, Web of Science, China National Knowledge Infrastructure (CNKI), Wanfang database, China Science and Technology Journal Database (VIP), and SinoMed databases were searched from inception to March 1st, 2022 to retrieve eligible studies that evaluated the effect of melatonin supplementation on the levels of malondialdehyde (MDA), lipid peroxidation (LPO), nitric oxide (NO), superoxide dismutase (SOD), glutathione (GSH), glutathione peroxidase (GPx), and catalase (CAT) in animal models of fibrosis. RESULTS A total of 64 studies were included in this meta-analysis. The results showed that melatonin supplementation significantly reduced the levels of oxidative indicators including MDA (P < 0.00001), LPO (P < 0.00001) and NO (P < 0.0001), and elevated the levels of antioxidant indicators including GSH (P < 0.00001), GPx (P < 0.00001) and SOD (P < 0.00001) in fibrotic diseases. CONCLUSIONS Our research findings showed that melatonin supplementation could significantly reduce the levels of oxidative indicators including MDA, LPO and NO and elevate the levels of antioxidant indicators including GSH, GPx and SOD so as to correct oxidative stress in animal models of fibrosis. However, no significant changes were observed in CAT level. More clinical studies are needed to further confirm the beneficial role of melatonin in fibrotic diseases.
Collapse
Affiliation(s)
- Dan Li
- Department of Dermatology, The Second Affiliated Hospital of Guilin Medical University, 212 Ren-Min Road, Guilin, Guangxi, 541199, China
| | - Jun-Hua Pan
- Department of Dermatology, The Second Affiliated Hospital of Guilin Medical University, 212 Ren-Min Road, Guilin, Guangxi, 541199, China
| | - Xiao-Fang Huang
- Department of Dermatology, The Second Affiliated Hospital of Guilin Medical University, 212 Ren-Min Road, Guilin, Guangxi, 541199, China
| | - Yu-Qing Liao
- Department of Dermatology, The Second Affiliated Hospital of Guilin Medical University, 212 Ren-Min Road, Guilin, Guangxi, 541199, China
| | - Yong-Jin Ling
- Department of Dermatology, The Second Affiliated Hospital of Guilin Medical University, 212 Ren-Min Road, Guilin, Guangxi, 541199, China
| | - Jing-Ying Luo
- Department of Dermatology, The Second Affiliated Hospital of Guilin Medical University, 212 Ren-Min Road, Guilin, Guangxi, 541199, China.
| |
Collapse
|
8
|
Daiber A, Frenis K, Kuntic M, Li H, Wolf E, Kilgallen AB, Lecour S, Van Laake LW, Schulz R, Hahad O, Münzel T. Redox Regulatory Changes of Circadian Rhythm by the Environmental Risk Factors Traffic Noise and Air Pollution. Antioxid Redox Signal 2022; 37:679-703. [PMID: 35088601 PMCID: PMC9618394 DOI: 10.1089/ars.2021.0272] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/31/2021] [Indexed: 12/13/2022]
Abstract
Significance: Risk factors in the environment such as air pollution and traffic noise contribute to the development of chronic noncommunicable diseases. Recent Advances: Epidemiological data suggest that air pollution and traffic noise are associated with a higher risk for cardiovascular, metabolic, and mental disease, including hypertension, heart failure, myocardial infarction, diabetes, arrhythmia, stroke, neurodegeneration, depression, and anxiety disorders, mainly by activation of stress hormone signaling, inflammation, and oxidative stress. Critical Issues: We here provide an in-depth review on the impact of the environmental risk factors air pollution and traffic noise exposure (components of the external exposome) on cardiovascular health, with special emphasis on the role of environmentally triggered oxidative stress and dysregulation of the circadian clock. Also, a general introduction on the contribution of circadian rhythms to cardiovascular health and disease as well as a detailed mechanistic discussion of redox regulatory pathways of the circadian clock system is provided. Future Directions: Finally, we discuss the potential of preventive strategies or "chrono" therapy for cardioprotection. Antioxid. Redox Signal. 37, 679-703.
Collapse
Affiliation(s)
- Andreas Daiber
- Molecular Cardiology, Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Katie Frenis
- Molecular Cardiology, Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Marin Kuntic
- Molecular Cardiology, Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Eva Wolf
- Structural Chronobiology, Institute of Molecular Physiology, Johannes Gutenberg University, Mainz, Germany
- Institute of Molecular Biology, Mainz, Germany
| | - Aoife B. Kilgallen
- Division Heart and Lungs, Regenerative Medicine Centre, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Linda W. Van Laake
- Division Heart and Lungs, Regenerative Medicine Centre, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Omar Hahad
- Molecular Cardiology, Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Thomas Münzel
- Molecular Cardiology, Department of Cardiology 1, Medical Center of the Johannes Gutenberg University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| |
Collapse
|
9
|
Wei H, Xiao L, Yao H, Li X, Wang W, Lee Y, Li D, Wei J. Melatonin through blockade of Hif-1α signaling mediates the anti-fibrosis under hypoxia in canine Sertoli cells. Reprod Biol 2022; 22:100677. [PMID: 36152357 DOI: 10.1016/j.repbio.2022.100677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 06/28/2022] [Accepted: 07/17/2022] [Indexed: 11/28/2022]
Abstract
The hypoxic microenvironment of cryptorchidism is an important factor in the impairment and fibrosis of Sertoli cells which result in blood-testis barrier (BTB) destruction and spermatogenesis loss. Recent studies have shown that melatonin, a well-known pineal hormone exerts beneficial effects against pathological fibrosis in a various of organs. However, it is still unknown whether melatonin can regulate hypoxia-induced fibrosis of Sertoli cells. In this study we evaluate melatonin levels, and its synthesizing enzymes, AANAT and HIOMT expression patterns in canine cryptorchidism and contralateral normal testis. Results show abdominal testes presented low melatonin levels and AANAT and HIOMT expression compared with testes located in the scrotum. Moreover, we established a hypoxia-induced fibrosis model in canine Sertoli cells induced by cobalt chloride (CoCl2) and found that melatonin inhibited the EMT markers expression and ECM production as well as Hif-1α expression of Sertoli cells in a dose-dependent manner. Furthermore, use of Lificiguat (synonyms YC-1, Hif-1α inhibitor) to interfere with the Hif-1α pathway showed a similar effect with melatonin suppression of the fibrosis in Sertoli cells. The results indicate that melatonin supplementation can alleviate the fibrosis process of Sertoli cells caused by hypoxia, which is associated with regulating the inhibition of Hif-1α signaling.
Collapse
Affiliation(s)
- Huawei Wei
- Beijing Detector Dog Program Facility of China Customs, Beijing, China
| | - Longfei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Hua Yao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Xiang Li
- RuiLi Detector Dog Program Facility of China Customs, Yunnan, China
| | - Wei Wang
- China-Japan Friendship Hospital, Beijing, China
| | - Yuan Lee
- Beijing Customs of PRC, Beijing, China
| | | | - Jingwen Wei
- Beijing Detector Dog Program Facility of China Customs, Beijing, China.
| |
Collapse
|
10
|
Tobeiha M, Jafari A, Fadaei S, Mirazimi SMA, Dashti F, Amiri A, Khan H, Asemi Z, Reiter RJ, Hamblin MR, Mirzaei H. Evidence for the Benefits of Melatonin in Cardiovascular Disease. Front Cardiovasc Med 2022; 9:888319. [PMID: 35795371 PMCID: PMC9251346 DOI: 10.3389/fcvm.2022.888319] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022] Open
Abstract
The pineal gland is a neuroendocrine gland which produces melatonin, a neuroendocrine hormone with critical physiological roles in the circadian rhythm and sleep-wake cycle. Melatonin has been shown to possess anti-oxidant activity and neuroprotective properties. Numerous studies have shown that melatonin has significant functions in cardiovascular disease, and may have anti-aging properties. The ability of melatonin to decrease primary hypertension needs to be more extensively evaluated. Melatonin has shown significant benefits in reducing cardiac pathology, and preventing the death of cardiac muscle in response to ischemia-reperfusion in rodent species. Moreover, melatonin may also prevent the hypertrophy of the heart muscle under some circumstances, which in turn would lessen the development of heart failure. Several currently used conventional drugs show cardiotoxicity as an adverse effect. Recent rodent studies have shown that melatonin acts as an anti-oxidant and is effective in suppressing heart damage mediated by pharmacologic drugs. Therefore, melatonin has been shown to have cardioprotective activity in multiple animal and human studies. Herein, we summarize the most established benefits of melatonin in the cardiovascular system with a focus on the molecular mechanisms of action.
Collapse
Affiliation(s)
- Mohammad Tobeiha
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ameneh Jafari
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Fadaei
- Department of Internal Medicine and Endocrinology, Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, United States
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Johannesburg, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
11
|
Pongkan W, Piamsiri C, Dechvongya S, Punyapornwitthaya V, Boonyapakorn C. Short-term melatonin supplementation decreases oxidative stress but does not affect left ventricular structure and function in myxomatous mitral valve degenerative dogs. BMC Vet Res 2022; 18:24. [PMID: 34996468 PMCID: PMC8740462 DOI: 10.1186/s12917-021-03125-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 12/20/2021] [Indexed: 11/27/2022] Open
Abstract
Background Cardiac wall stress and high oxidative stress are often found in cases of myxomatous mitral valve degenerative (MMVD) disease and can lead to myocardial injuries and cardiac dysfunction. Melatonin, an antioxidant, has been shown to exert cardioprotection in laboratory animal models. However, its effect on metabolic parameters and left ventricular (LV) adaptation in MMVD dogs has rarely been investigated. This clinical trial hypothesized that a melatonin supplement for 4 weeks would improve metabolic parameters, LV structure (diameters and wall thickness), and LV function in MMVD dogs. Blood profiles, echocardiograms, and oxidative stress levels were obtained from 18 dogs with MMVD stage B2 and C at baseline and after prescribed Melatonin (2 mg/kg) for 4 weeks. Eleven dogs with MMVD stage B2 and C, which received a placebo, were evaluated as a control group. Results In this clinical trial, the baseline plasma malondialdehyde (MDA) was no different between the treatment and placebo groups. The post-treatment plasma MDA levels (4.50 ± 0.63 mg/mL) in the treatment group was significantly decreased after 4 weeks of melatonin supplementation compared to pre-treatment levels (7.51 ± 1.11 mg/mL) (P = 0.038). However, blood profiles and LV structure and function investigated using echocardiography were found not to different between pre-and post-treatment in each group. No adverse effects were observed following melatonin supplementation. Conclusions This clinical trial demonstrated that a melatonin supplement for 4 weeks can attenuate oxidative stress levels in MMVD dogs, especially in MMVD stage C, but does not result in LV structural changes or LV function in MMVD dogs of either stage B2 or stage C.
Collapse
Affiliation(s)
- Wanpitak Pongkan
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand.,Integrative Research Center for Veterinary Circulatory Sciences, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand.,Veterinary Cardiopulmonary Clinic, Small Animal Hospital, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand
| | - Chanon Piamsiri
- Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand
| | - Sirada Dechvongya
- Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand
| | - Verasak Punyapornwitthaya
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand
| | - Chavalit Boonyapakorn
- Integrative Research Center for Veterinary Circulatory Sciences, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand. .,Veterinary Cardiopulmonary Clinic, Small Animal Hospital, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand. .,Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand.
| |
Collapse
|
12
|
Bryant AJ, Ebrahimi E, Nguyen A, Wolff CA, Gumz ML, Liu AC, Esser KA. A wrinkle in time: circadian biology in pulmonary vascular health and disease. Am J Physiol Lung Cell Mol Physiol 2022; 322:L84-L101. [PMID: 34850650 PMCID: PMC8759967 DOI: 10.1152/ajplung.00037.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
An often overlooked element of pulmonary vascular disease is time. Cellular responses to time, which are regulated directly by the core circadian clock, have only recently been elucidated. Despite an extensive collection of data regarding the role of rhythmic contribution to disease pathogenesis (such as systemic hypertension, coronary artery, and renal disease), the roles of key circadian transcription factors in pulmonary hypertension remain understudied. This is despite a large degree of overlap in the pulmonary hypertension and circadian rhythm fields, not only including shared signaling pathways, but also cell-specific effects of the core clock that are known to result in both protective and adverse lung vessel changes. Therefore, the goal of this review is to summarize the current dialogue regarding common pathways in circadian biology, with a specific emphasis on its implications in the progression of pulmonary hypertension. In this work, we emphasize specific proteins involved in the regulation of the core molecular clock while noting the circadian cell-specific changes relevant to vascular remodeling. Finally, we apply this knowledge to the optimization of medical therapy, with a focus on sleep hygiene and the role of chronopharmacology in patients with this disease. In dissecting the unique relationship between time and cellular biology, we aim to provide valuable insight into the practical implications of considering time as a therapeutic variable. Armed with this information, physicians will be positioned to more efficiently use the full four dimensions of patient care, resulting in improved morbidity and mortality of pulmonary hypertension patients.
Collapse
Affiliation(s)
- Andrew J. Bryant
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Elnaz Ebrahimi
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Amy Nguyen
- 1Department of Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Christopher A. Wolff
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Michelle L. Gumz
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Andrew C. Liu
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| | - Karyn A. Esser
- 2Department of Physiology, University of Florida College of Medicine, Gainesville, Florida
| |
Collapse
|
13
|
Mannino G, Pernici C, Serio G, Gentile C, Bertea CM. Melatonin and Phytomelatonin: Chemistry, Biosynthesis, Metabolism, Distribution and Bioactivity in Plants and Animals-An Overview. Int J Mol Sci 2021; 22:ijms22189996. [PMID: 34576159 PMCID: PMC8469784 DOI: 10.3390/ijms22189996] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/21/2022] Open
Abstract
Melatonin is a ubiquitous indolamine, largely investigated for its key role in the regulation of several physiological processes in both animals and plants. In the last century, it was reported that this molecule may be produced in high concentrations by several species belonging to the plant kingdom and stored in specialized tissues. In this review, the main information related to the chemistry of melatonin and its metabolism has been summarized. Furthermore, the biosynthetic pathway characteristics of animal and plant cells have been compared, and the main differences between the two systems highlighted. Additionally, in order to investigate the distribution of this indolamine in the plant kingdom, distribution cluster analysis was performed using a database composed by 47 previously published articles reporting the content of melatonin in different plant families, species and tissues. Finally, the potential pharmacological and biostimulant benefits derived from the administration of exogenous melatonin on animals or plants via the intake of dietary supplements or the application of biostimulant formulation have been largely discussed.
Collapse
Affiliation(s)
- Giuseppe Mannino
- Department of Life Sciences and Systems Biology, Plant Physiology Unit, University of Turin, Via Quarello 15/A, 10135 Turin, Italy; (G.M.); (C.P.)
| | - Carlo Pernici
- Department of Life Sciences and Systems Biology, Plant Physiology Unit, University of Turin, Via Quarello 15/A, 10135 Turin, Italy; (G.M.); (C.P.)
| | - Graziella Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy;
| | - Carla Gentile
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy;
- Correspondence: (C.G.); (C.M.B.); Tel.: +39-091-2389-7423 (C.G.); +39-011-670-6361 (C.M.B.)
| | - Cinzia M. Bertea
- Department of Life Sciences and Systems Biology, Plant Physiology Unit, University of Turin, Via Quarello 15/A, 10135 Turin, Italy; (G.M.); (C.P.)
- Correspondence: (C.G.); (C.M.B.); Tel.: +39-091-2389-7423 (C.G.); +39-011-670-6361 (C.M.B.)
| |
Collapse
|
14
|
Diaba-Nuhoho P, Cour M, Hadebe N, Marais D, Lecour S, Blackhurst D. Chronic and moderate consumption of reduced-alcohol wine confers cardiac benefits in a rat model of pulmonary arterial hypertension. BMC Res Notes 2021; 14:324. [PMID: 34425891 PMCID: PMC8381534 DOI: 10.1186/s13104-021-05738-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/12/2021] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES In pulmonary arterial hypertension (PAH), right ventricular (RV) dysfunction develops via mechanisms involving oxidative stress. Moderate and chronic red wine (RW) consumption reduces oxidative stress and confers cardioprotection but its effect on PAH is unknown. We evaluated whether moderate and chronic consumption of reduced-alcohol RW (RARW) confers cardioprotection in a monocrotaline (MCT)-induced PAH rat model. RESULTS Rats were randomly grouped: control; MCT; RARW; MCT + RARW. Wine was diluted to mimic moderate intake for humans, and consumed from 7 days before, until 28 days after MCT-injection. Echocardiography measured pulmonary artery acceleration time (PAAT) and RV thickness. Conjugated dienes (CD), and thiobarbituric acid reactive substances (TBARS) concentrations were assessed. MCT induced RV thickness and decreased PAAT compared to controls [1.22 ± 0.09 mm vs 0.46 ± 0.02 mm and 14 ± 1 vs 23 ± 2 m/s, respectively (p < 0.001)]. Chronic RARW consumption limited MCT-induced RV hypertrophy and increased PAAT. CD and TBARS increased in MCT-treated animals compared to controls (672 ± 43 nmol/L vs 453 ± 35 nmol/L; p < 0.01 and 13 ± 2 µmol/L vs 4 ± 0.3 µmol/L; p < 0.01). RARW reduced MCT-induced CD (472 ± 27 nmol/L vs 672 ± 43 nmol/L; p < 0.01). CONCLUSION Chronic and moderate intake of RARW ameliorates MCT-induced PAH in rats, which may be partly attributable to reduction of lipid peroxidation.
Collapse
Affiliation(s)
- Patrick Diaba-Nuhoho
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University of Technology Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Martin Cour
- Cardioprotection Group, Hatter Cardiovascular Institute and Lionel Opie Preclinical Imaging Core Facility, University of Cape Town, Cape Town, South Africa
| | - Nkanyiso Hadebe
- Cardioprotection Group, Hatter Cardiovascular Institute and Lionel Opie Preclinical Imaging Core Facility, University of Cape Town, Cape Town, South Africa
- Department of Anaesthesia, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
| | - David Marais
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Hatter Cardiovascular Institute and Lionel Opie Preclinical Imaging Core Facility, University of Cape Town, Cape Town, South Africa
| | - Dee Blackhurst
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
15
|
Zhang J, Lu X, Liu M, Fan H, Zheng H, Zhang S, Rahman N, Wołczyński S, Kretowski A, Li X. Melatonin inhibits inflammasome-associated activation of endothelium and macrophages attenuating pulmonary arterial hypertension. Cardiovasc Res 2021; 116:2156-2169. [PMID: 31774487 DOI: 10.1093/cvr/cvz312] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/25/2019] [Accepted: 11/25/2019] [Indexed: 01/08/2023] Open
Abstract
AIMS Pulmonary arterial hypertension (PAH) is a pathophysiological syndrome associated with pulmonary/systemic inflammation. Melatonin relieves PAH, but the molecular mode of action remains unclear. Here, we investigated the role of melatonin in normalizing vascular homeostasis. METHODS AND RESULTS Light-time mean serum melatonin concentration was lower in patients with PAH than in normal controls [11.06 ± 3.44 (7.13-15.6) vs. 14.55 ± 1.28 (8.0-19.4) pg/mL], which was negatively correlated with increased serum levels of interleukin-1β (IL-1β) in patients with PAH. We showed that inflammasomes were activated in the PAH mice model and that melatonin attenuated IL-1β secretion. On one hand, melatonin reduced the number of macrophages in lung by inhibiting the endothelial chemokines and adhesion factors. Moreover, use of Il1r-/- mice, Caspase1/11-/- mice, and melatonin-treated mice revealed that melatonin reduced hypoxia-induced vascular endothelial leakage in the lung. On the other hand, we verified that melatonin reduced the formation of inflammasome multiprotein complexes by modulating calcium ions in macrophages using a live cell station, and melatonin decreased inositol triphosphate and increased cAMP. Furthermore, knockdown of melatonin membrane receptors blocked melatonin function, and a melatonin membrane receptors agonist inactivated inflammasomes in macrophages. CONCLUSION Melatonin attenuated inflammasome-associated vascular disorders by directly improving endothelial leakage and decreasing the formation of inflammasome multiprotein complexes in macrophages. Taken together, our data provide a theoretical basis for applying melatonin clinically, and inflammasomes may be a possible target of PAH treatment.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xiaohui Lu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Mei Liu
- Department of Pathology, Chinese PLA General Hospital, Beijing 102628, China
| | - Hanlu Fan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Han Zheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Shanshan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Nafis Rahman
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Sławomir Wołczyński
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Adam Kretowski
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Białystok, Poland
| | - Xiangdong Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.,Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
16
|
Zimmer A, Teixeira RB, Constantin RL, Campos-Carraro C, Aparicio Cordero EA, Ortiz VD, Donatti L, Gonzalez E, Bahr AC, Visioli F, Baldo G, Luz de Castro A, Araujo AS, Belló-Klein A. The progression of pulmonary arterial hypertension induced by monocrotaline is characterized by lung nitrosative and oxidative stress, and impaired pulmonary artery reactivity. Eur J Pharmacol 2021; 891:173699. [PMID: 33160936 DOI: 10.1016/j.ejphar.2020.173699] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
The time-course of pulmonary arterial hypertension in the monocrotaline (MCT) model was investigated. Male rats were divided into two groups: MCT (received a 60 mg/kg i.p. injection) and control (received saline). The MCT and control groups were further divided into three cohorts, based on the follow-up interval: 1, 2, and 3 weeks. Right ventricle (RV) catheterization was performed and RV hypertrophy (RVH) was estimated. The lungs were used for biochemical, histological, molecular, and immunohistochemical analysis, while pulmonary artery rings were used for vascular reactivity. MCT promoted lung perivascular edema, inflammatory cells exudation, greater neutrophils and lymphocytes profile, and arteriolar wall thickness, compared to CTR group. Increases in pulmonary artery pressure and in RVH were observed in the MCT 2- and 3-week groups. The first week was marked by the presence of nitrosative stress (50% moderate and 33% accentuated staining by nitrotyrosine). These alterations lead to an adaptation of NO production by NO synthase activity after 2 weeks. Oxidative stress was evident in the third week, probably by an imbalance between endothelin-1 receptors, resulting in extracellular matrix remodeling, endothelial dysfunction, and RVH. Also, it was found a reduced pulmonary arterial vasodilatory response to acetylcholine after 2 (55%) and 3 (45%) weeks in MCT groups. The relevance of this study is precisely to show that nitrosative and oxidative stress predominate in distinct time windows of the disease progression.
Collapse
Affiliation(s)
- Alexsandra Zimmer
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Rayane Brinck Teixeira
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Rosalia Lempk Constantin
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Cristina Campos-Carraro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | | | - Vanessa Duarte Ortiz
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Luiza Donatti
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Esteban Gonzalez
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| | - Alan Christhian Bahr
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Fernanda Visioli
- Faculty of Dentistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Guilherme Baldo
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| | - Alexandre Luz de Castro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Alex Sander Araujo
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| | - Adriane Belló-Klein
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
17
|
Ding Z, Wu X, Wang Y, Ji S, Zhang W, Kang J, Li J, Fei G. Melatonin prevents LPS-induced epithelial-mesenchymal transition in human alveolar epithelial cells via the GSK-3β/Nrf2 pathway. Biomed Pharmacother 2020; 132:110827. [PMID: 33065391 DOI: 10.1016/j.biopha.2020.110827] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Oxidative stress plays a critical role in pulmonary fibrosis after acute lung injury (ALI), and epithelial-mesenchymal transition (EMT) events are involved in this process. The purpose of this study was to investigate the protective effects of melatonin, a natural antioxidant, on lipopolysaccharide (LPS)-induced EMT in human alveolar epithelial cells. METHODS Human type II alveolar epithelial cell-derived A549 cells were incubated with LPS and melatonin alone or in combination for up to 24 h. The morphological changes of the treated cells were evaluated as well as indexes of oxidative stress. EMT-related proteins and the Nrf2 signaling pathway were detected by western blot analysis and immunofluorescence staining, respectively. To further investigate the underlying mechanisms, the effects of melatonin on cells transfected Nrf2 short hairpin RNA (shRNA) and the PI3K / GSK-3β signaling pathway were evaluated. RESULTS Treatment with melatonin upregulated Nrf2 expression, inhibited LPS-induced cell morphological change, reversed the expressions of EMT-related proteins, and reduced reactive oxygen species (ROS) production in A549 cells, as well as the levels of malondialdehyde (MDA) and anti-oxidative enzymes. Yet, the effects of melatonin were almost completely abolished in cells transfected Nrf2 shRNA. Furthermore, the data demonstrated that melatonin could activate the PI3K/AKT signaling pathway, resulting in phosphorylation of GSK-3β (Ser9) and upregulation of the Nrf2 protein in A549 cells, which ultimately attenuated LPS-induced EMT. CONCLUSION The present study is the first to demonstrate that melatonin can protect human alveolar epithelial cells against oxidative stress by effectively inhibiting LPS-induced EMT, which was mostly dependent on upregulation of the Nrf2 pathway via the PI3K/GSK-3β axis. Further studies are warranted to investigate the role of melatonin for the treatment of oxidative stress-associated diseases, as well as pulmonary fibrosis after ALI.
Collapse
Affiliation(s)
- Zhenxing Ding
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Xu Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Yueguo Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Shuang Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Wenying Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Jiaying Kang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Jiajia Li
- Center Lab of The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China
| | - Guanghe Fei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022 Anhui, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, China.
| |
Collapse
|
18
|
Chitimus DM, Popescu MR, Voiculescu SE, Panaitescu AM, Pavel B, Zagrean L, Zagrean AM. Melatonin's Impact on Antioxidative and Anti-Inflammatory Reprogramming in Homeostasis and Disease. Biomolecules 2020; 10:biom10091211. [PMID: 32825327 PMCID: PMC7563541 DOI: 10.3390/biom10091211] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/30/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022] Open
Abstract
There is a growing consensus that the antioxidant and anti-inflammatory properties of melatonin are of great importance in preserving the body functions and homeostasis, with great impact in the peripartum period and adult life. Melatonin promotes adaptation through allostasis and stands out as an endogenous, dietary, and therapeutic molecule with important health benefits. The anti-inflammatory and antioxidant effects of melatonin are intertwined and are exerted throughout pregnancy and later during development and aging. Melatonin supplementation during pregnancy can reduce ischemia-induced oxidative damage in the fetal brain, increase offspring survival in inflammatory states, and reduce blood pressure in the adult offspring. In adulthood, disturbances in melatonin production negatively impact the progression of cardiovascular risk factors and promote cardiovascular and neurodegenerative diseases. The most studied cardiovascular effects of melatonin are linked to hypertension and myocardial ischemia/reperfusion injury, while the most promising ones are linked to regaining control of metabolic syndrome components. In addition, there might be an emerging role for melatonin as an adjuvant in treating coronavirus disease 2019 (COVID 19). The present review summarizes and comments on important data regarding the roles exerted by melatonin in homeostasis and oxidative stress and inflammation related pathologies.
Collapse
Affiliation(s)
- Diana Maria Chitimus
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Mihaela Roxana Popescu
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, Elias University Hospital, 010164 Bucharest, Romania;
| | - Suzana Elena Voiculescu
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Anca Maria Panaitescu
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, Filantropia Clinical Hospital, 010164 Bucharest, Romania;
| | - Bogdan Pavel
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Leon Zagrean
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Ana-Maria Zagrean
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
- Correspondence:
| |
Collapse
|
19
|
|
20
|
Cai Z, Klein T, Geenen LW, Tu L, Tian S, van den Bosch AE, de Rijke YB, Reiss IKM, Boersma E, Duncker DJ, Boomars KA, Guignabert C, Merkus D. Lower Plasma Melatonin Levels Predict Worse Long-Term Survival in Pulmonary Arterial Hypertension. J Clin Med 2020; 9:jcm9051248. [PMID: 32344923 PMCID: PMC7287676 DOI: 10.3390/jcm9051248] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
Exogenous melatonin has been reported to be beneficial in the treatment of pulmonary hypertension (PH) in animal models. Multiple mechanisms are involved, with melatonin exerting anti-oxidant and anti-inflammatory effects, as well as inducing vasodilation and cardio-protection. However, endogenous levels of melatonin in treatment-naïve patients with PH and their clinical significance are still unknown. Plasma levels of endogenous melatonin were measured by liquid chromatography-tandem mass spectrometry in PH patients (n = 64, 43 pulmonary arterial hypertension (PAH) and 21 chronic thromboembolic PH (CTEPH)) and healthy controls (n = 111). Melatonin levels were higher in PH, PAH, and CTEPH patients when compared with controls (Median 118.7 (IQR 108.2–139.9), 118.9 (109.3–147.7), 118.3 (106.8–130.1) versus 108.0 (102.3–115.2) pM, respectively, p all <0.001). The mortality was 26% (11/43) in the PAH subgroup during a long-term follow-up of 42 (IQR: 32–58) months. Kaplan–Meier analysis showed that, in the PAH subgroup, patients with melatonin levels in the 1st quartile (<109.3 pM) had a worse survival than those in quartile 2–4 (Mean survival times were 46 (95% CI: 30–65) versus 68 (58–77) months, Log-rank, p = 0.026) with an increased hazard ratio of 3.5 (95% CI: 1.1–11.6, p = 0.038). Endogenous melatonin was increased in treatment-naïve patients with PH, and lower levels of melatonin were associated with worse long-term survival in patient with PAH.
Collapse
Affiliation(s)
- Zongye Cai
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Z.C.); (L.W.G.); (S.T.); (A.E.v.d.B.); (E.B.); (D.J.D.)
| | - Theo Klein
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, 3000 CB Rotterdam, The Netherlands; (T.K.); (Y.B.d.R.)
| | - Laurie W. Geenen
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Z.C.); (L.W.G.); (S.T.); (A.E.v.d.B.); (E.B.); (D.J.D.)
| | - Ly Tu
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, 92350 Paris, France; (L.T.); (C.G.)
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, 94270 Paris, France
| | - Siyu Tian
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Z.C.); (L.W.G.); (S.T.); (A.E.v.d.B.); (E.B.); (D.J.D.)
| | - Annemien E. van den Bosch
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Z.C.); (L.W.G.); (S.T.); (A.E.v.d.B.); (E.B.); (D.J.D.)
| | - Yolanda B. de Rijke
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, 3000 CB Rotterdam, The Netherlands; (T.K.); (Y.B.d.R.)
| | - Irwin K. M. Reiss
- Department of Pediatrics/Neonatology, Sophia Children’s Hospital, Erasmus MC, University Medical Center Rotterdam, 3000 CB Rotterdam, The Netherlands;
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Z.C.); (L.W.G.); (S.T.); (A.E.v.d.B.); (E.B.); (D.J.D.)
- Department of Clinical Epidemiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | - Dirk J. Duncker
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Z.C.); (L.W.G.); (S.T.); (A.E.v.d.B.); (E.B.); (D.J.D.)
| | - Karin A. Boomars
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands;
| | - Christophe Guignabert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, 92350 Paris, France; (L.T.); (C.G.)
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, 94270 Paris, France
| | - Daphne Merkus
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (Z.C.); (L.W.G.); (S.T.); (A.E.v.d.B.); (E.B.); (D.J.D.)
- Walter Brendel Center of Experimental Medicine (WBex), LMU Munich, 81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), 81377 Munich, Germany
- Correspondence: ; Tel.: +31-10-7030955
| |
Collapse
|
21
|
Making a case for metallothioneins conferring cardioprotection in pulmonary hypertension. Med Hypotheses 2020; 137:109572. [DOI: 10.1016/j.mehy.2020.109572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/30/2019] [Accepted: 01/15/2020] [Indexed: 11/23/2022]
|
22
|
Boengler K, Schlüter KD, Schermuly RT, Schulz R. Cardioprotection in right heart failure. Br J Pharmacol 2020; 177:5413-5431. [PMID: 31995639 PMCID: PMC7680005 DOI: 10.1111/bph.14992] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/04/2019] [Accepted: 01/06/2020] [Indexed: 02/06/2023] Open
Abstract
Ischaemic and pharmacological conditioning of the left ventricle is mediated by the activation of signalling cascades, which finally converge at the mitochondria and reduce ischaemia/reperfusion (I/R) injury. Whereas the molecular mechanisms of conditioning in the left ventricle are well characterized, cardioprotection of the right ventricle is principally feasible but less established. Similar to what is known for the left ventricle, a dysregulation in signalling pathways seems to play a role in I/R injury of the healthy and failing right ventricle and in the ability/inability of the right ventricle to respond to a conditioning stimulus. The maintenance of mitochondrial function seems to be crucial in both ventricles to reduce I/R injury. As far as currently known, similar molecular mechanisms mediate ischaemic and pharmacological preconditioning in the left and right ventricles. However, the two ventricles seem to respond differently towards exercise‐induced preconditioning. LINKED ARTICLES This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc
Collapse
Affiliation(s)
- Kerstin Boengler
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | | | | | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
23
|
Effects of melatonin on cardiovascular risk factors and metabolic syndrome: a comprehensive review. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:521-536. [DOI: 10.1007/s00210-020-01822-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
|
24
|
The protective effects of caffeine and melatonin on antioxidant enzymes in rat fetal lung tissues. JOURNAL OF SURGERY AND MEDICINE 2019. [DOI: 10.28982/josam.598742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
25
|
Kim JY, Park JH, Kim K, Leem J, Park KK. Melatonin Inhibits Transforming Growth Factor-β1-Induced Epithelial-Mesenchymal Transition in AML12 Hepatocytes. BIOLOGY 2019; 8:biology8040084. [PMID: 31717992 PMCID: PMC6956139 DOI: 10.3390/biology8040084] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/01/2019] [Accepted: 11/08/2019] [Indexed: 12/30/2022]
Abstract
Recent studies showed that melatonin, a well-known pineal hormone that modulates the circadian rhythm, exerts beneficial effects against liver fibrosis. However, mechanisms for its protective action against the fibrotic processes remain incompletely understood. Here, we aimed to explore the effects of the hormone on transforming growth factor-β1 (TGF-β1)-stimulated epithelial–mesenchymal transition (EMT) in AML12 hepatocytes. Pretreatment with melatonin dose-dependently reversed downregulation of an epithelial marker and upregulation of mesenchymal markers after TGF-β1 stimulation. Additionally, melatonin dose-dependently suppressed an increased phosphorylation of Smad2/3 after TGF-β1 treatment. Besides the canonical Smad signaling pathway, an increase in phosphorylation of extracellular signal-regulated kinase 1/2 and p38 was also dose-dependently attenuated by melatonin. The suppressive effect of the hormone on EMT stimulated by TGF-β1 was not affected by luzindole, an antagonist of melatonin membrane receptors, suggesting that its membrane receptors are not required for the inhibitory action of melatonin. Moreover, melatonin suppressed elevation of intracellular reactive oxygen species (ROS) levels in TGF-β1-treated cells. Finally, TGF-β1-stimulated EMT was also inhibited by the antioxidant N-acetylcysteine. Collectively, these results suggest that melatonin prevents TGF-β1-stimulated EMT through suppression of Smad and mitogen-activated protein kinase signaling cascades by deactivating ROS-dependent mechanisms in a membrane receptor-independent manner.
Collapse
Affiliation(s)
- Jung-Yeon Kim
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| | - Jae-Hyung Park
- Department of Physiology, School of Medicine, Keimyung University, Daegu 42601, Korea; (J.-H.P.); (K.K.)
| | - Kiryeong Kim
- Department of Physiology, School of Medicine, Keimyung University, Daegu 42601, Korea; (J.-H.P.); (K.K.)
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
- Correspondence:
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| |
Collapse
|
26
|
Bosco AD, Schedler FB, Colares JR, Schemitt EG, Hartmann RM, Forgiarini Junior LA, Dias AS, Marroni NP. Melatonin effects on pulmonary tissue in the experimental model of Hepatopulmonary Syndrome. ACTA ACUST UNITED AC 2019; 45:e20170164. [PMID: 31166552 PMCID: PMC6715043 DOI: 10.1590/1806-3713/e20170164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/12/2018] [Indexed: 01/25/2023]
Abstract
Objective: To evaluate the pulmonary alterations of animals with Hepatopulmonary Syndrome (HPS) submitted to Biliary Duct Ligature (BDL), as well as the antioxidant effect of Melatonin (MEL). Methods: Sixteen male Wistar rats, divided into four Sham groups: BDL group, Sham + MEL group and BDL + MEL. The pulmonary and hepatic histology, lipoperoxidation and antioxidant activity of lung tissue, alveolar-arterial O2 difference and lung / body weight ratio (%) were evaluated. Results: When comparing the groups, could be observed an increase of vasodilation and pulmonary fibrosis in the BDL group and the reduction of this in relation to the BDL + MEL group. It was also observed significant changes in the activity of catalase, ApCO2, ApO2 in the LBD group when compared to the other groups. Conclusion: The use of MEL has been shown to be effective in reducing vasodilation, fibrosis levels and oxidative stress as well as gas exchange in an experimental HPS model.
Collapse
Affiliation(s)
| | | | | | - Elisângela Gonçalves Schemitt
- . Hospital de Clínicas de Porto Alegre, Porto Alegre (RS) Brasil.,. Universidade Federal do Rio Grande do Sul, Porto Alegre (RS) Brasil
| | - Renata Minuzzo Hartmann
- . Hospital de Clínicas de Porto Alegre, Porto Alegre (RS) Brasil.,. Universidade Federal do Rio Grande do Sul, Porto Alegre (RS) Brasil
| | | | - Alexandre Simões Dias
- . Hospital de Clínicas de Porto Alegre, Porto Alegre (RS) Brasil.,. Universidade Federal do Rio Grande do Sul, Porto Alegre (RS) Brasil
| | - Norma Possa Marroni
- . Hospital de Clínicas de Porto Alegre, Porto Alegre (RS) Brasil.,. Universidade Federal do Rio Grande do Sul, Porto Alegre (RS) Brasil
| |
Collapse
|
27
|
Prasad K. AGE-RAGE Stress in the Pathophysiology of Pulmonary Hypertension and its Treatment. Int J Angiol 2019; 28:71-79. [PMID: 31384104 DOI: 10.1055/s-0039-1687818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pulmonary hypertension (PH) is a rare and fatal disease characterized by elevation of pulmonary artery pressure ≥ 25 mm Hg. There are five groups of PH: (1) pulmonary artery (PA) hypertension (PAH), (2) PH due to heart diseases, (3) PH associated with lung diseases/hypoxia, (4) PH associated with chronic obstruction of PA, and (5) PH due to unclear and/or multifactorial mechanisms. The pathophysiologic mechanisms of group 1 have been studied in detail; however, those for groups 2 to 5 are not that well known. PH pathology is characterized by smooth muscle cells (SMC) proliferation, muscularization of peripheral PA, accumulation of extracellular matrix (ECM), plexiform lesions, thromboembolism, and recanalization of thrombi. Advanced glycation end products (AGE) and its receptor (RAGE) and soluble RAGE (sRAGE) appear to be involved in the pathogenesis of PH. AGE and its interaction with RAGE induce vascular hypertrophy through proliferation of vascular SMC, accumulation of ECM, and suppression of apoptosis. Reactive oxygen species (ROS) generated by interaction of AGE and RAGE modulates SMC proliferation, attenuate apoptosis, and constricts PA. Increased stiffness in the artery due to vascular hypertrophy, and vasoconstriction due to ROS resulted in PH. The data also suggest that reduction in consumption and formation of AGE, suppression of RAGE expression, blockage of RAGE ligand binding, elevation of sRAGE levels, and antioxidants may be novel therapeutic targets for prevention, regression, and slowing of progression of PH. In conclusion, AGE-RAGE stress may be involved in the pathogenesis of PH and the therapeutic targets should be the AGE-RAGE axis.
Collapse
Affiliation(s)
- Kailash Prasad
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
28
|
Ren X, Johns RA, Gao WD. EXPRESS: Right Heart in Pulmonary Hypertension: From Adaptation to Failure. Pulm Circ 2019; 9:2045894019845611. [PMID: 30942134 PMCID: PMC6681271 DOI: 10.1177/2045894019845611] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/27/2019] [Indexed: 01/24/2023] Open
Abstract
Right ventricular (RV) failure (RVF) has garnered significant attention in recent years because of its negative impact on clinical outcomes in patients with pulmonary hypertension (PH). PH triggers a series of events, including activation of several signaling pathways that regulate cell growth, metabolism, extracellular matrix remodeling, and energy production. These processes render the RV adaptive to PH. However, RVF develops when PH persists, accompanied by RV ischemia, alterations in substrate and mitochondrial energy metabolism, increased free oxygen radicals, increased cell loss, downregulation of adrenergic receptors, increased inflammation and fibrosis, and pathologic microRNAs. Diastolic dysfunction is also an integral part of RVF. Emerging non-invasive technologies such as molecular or metallic imaging, cardiac MRI, and ultrafast Doppler coronary flow mapping will be valuable tools to monitor RVF, especially the transition to RVF. Most PH therapies cannot treat RVF once it has occurred. A variety of therapies are available to treat acute and chronic RVF, but they are mainly supportive, and no effective therapy directly targets the failing RV. Therapies that target cell growth, cellular metabolism, oxidative stress, and myocyte regeneration are being tested preclinically. Future research should include establishing novel RVF models based on existing models, increasing use of human samples, creating human stem cell-based in vitro models, and characterizing alterations in cardiac excitation–contraction coupling during transition from adaptive RV to RVF. More successful strategies to manage RVF will likely be developed as we learn more about the transition from adaptive remodeling to maladaptive RVF in the future.
Collapse
Affiliation(s)
- Xianfeng Ren
- Department of Anesthesiology,
China-Japan
Friendship Hospital, Beijing, China
| | - Roger A. Johns
- Department of Anesthesiology and
Critical Care Medicine,
Johns
Hopkins University School of Medicine,
Baltimore, MD, USA
| | - Wei Dong Gao
- Department of Anesthesiology and
Critical Care Medicine,
Johns
Hopkins University School of Medicine,
Baltimore, MD, USA
| |
Collapse
|
29
|
Pulmonary arterial hypertension and the potential roles of metallothioneins: A focused review. Life Sci 2018; 214:77-83. [PMID: 30355531 DOI: 10.1016/j.lfs.2018.10.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/10/2018] [Accepted: 10/19/2018] [Indexed: 12/17/2022]
Abstract
The pathophysiology of pulmonary arterial hypertension (PAH) is underlined by cell proliferation and vasoconstriction of pulmonary arterioles this involves multiple molecular factors or proteins, but it is not clear what the exact roles of these factors/proteins are. In addition, there may be other factors/proteins that have not been identified that contribute to PAH pathophysiology. Therefore, research has focused on investigating novel role players, in order to facilitate a better understanding of how PAH develop. Evidence suggest that mitochondrial regulators are key role players in PAH pathophysiology, but regulators that have not received sufficient attention in PAH are metallothioneins (MTs). In PAH patients, MT expression is elevated compared to healthy individuals, suggesting that MTs may be possible biomarkers. In other disease-models, MTs have been shown to regulate cell proliferation and vasoconstriction, processes that are instrumental in PAH pathophysiology. Due to the involvement of these processes in PAH pathophysiology and the ability of MTs to modulate them, this paper propose that cellular MTs may also play a role in PAH development. This paper suggests that PAH-research should perhaps begin to investigate the involvement of cellular MTs in the development of PAH.
Collapse
|
30
|
Bonomini F, Borsani E, Favero G, Rodella LF, Rezzani R. Dietary Melatonin Supplementation Could Be a Promising Preventing/Therapeutic Approach for a Variety of Liver Diseases. Nutrients 2018; 10:nu10091135. [PMID: 30134592 PMCID: PMC6164189 DOI: 10.3390/nu10091135] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/14/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023] Open
Abstract
In the therapeutic strategies, the role of diet is a well-established factor that can also have an important role in liver diseases. Melatonin, identified in animals, has many antioxidant properties and it was after discovered also in plants, named phytomelatonin. These substances have a positive effect during aging and in pathological conditions too. In particular, it is important to underline that the amount of melatonin produced by pineal gland in human decreases during lifetime and its reduction in blood could be related to pathological conditions in which mitochondria and oxidative stress play a pivotal role. Moreover, it has been indicated that melatonin/phytomelatonin containing foods may provide dietary melatonin, so their ingestion through balanced diets could be sufficient to confer health benefits. In this review, the classification of liver diseases and an overview of the most important aspects of melatonin/phytomelatonin, concerning the differences among their synthesis, their presence in foods and their role in health and diseases, are summarized. The findings suggest that melatonin/phytomelatonin supplementation with diet should be considered important in preventing different disease settings, in particular in liver. Currently, more studies are needed to strengthen the potential beneficial effects of melatonin/phytomelatonin in liver diseases and to better clarify the molecular mechanisms of action.
Collapse
Affiliation(s)
- Francesca Bonomini
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
- Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy.
| | - Elisa Borsani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
- Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy.
| | - Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | - Luigi F Rodella
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
- Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy.
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
- Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy.
| |
Collapse
|
31
|
Nduhirabandi F, Maarman GJ. Melatonin in Heart Failure: A Promising Therapeutic Strategy? Molecules 2018; 23:molecules23071819. [PMID: 30037127 PMCID: PMC6099639 DOI: 10.3390/molecules23071819] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022] Open
Abstract
Heart failure is a multifactorial clinical syndrome characterized by the inability of the heart to pump sufficient blood to the body. Despite recent advances in medical management, poor outcomes in patients with heart failure remain very high. This highlights a need for novel paradigms for effective, preventive and curative strategies. Substantial evidence supports the importance of endogenous melatonin in cardiovascular health and the benefits of melatonin supplementation in various cardiac pathologies and cardiometabolic disorders. Melatonin plays a crucial role in major pathological processes associated with heart failure including ischemic injury, oxidative stress, apoptosis, and cardiac remodeling. In this review, available evidence for the role of melatonin in heart failure is discussed. Current challenges and possible limitations of using melatonin in heart failure are also addressed. While few clinical studies have investigated the role of melatonin in the context of heart failure, current findings from experimental studies support the potential use of melatonin as preventive and adjunctive curative therapy in heart failure.
Collapse
Affiliation(s)
- Frederic Nduhirabandi
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa.
| | - Gerald J Maarman
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa (HICRA), Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa.
| |
Collapse
|
32
|
Jiki Z, Lecour S, Nduhirabandi F. Cardiovascular Benefits of Dietary Melatonin: A Myth or a Reality? Front Physiol 2018; 9:528. [PMID: 29867569 PMCID: PMC5967231 DOI: 10.3389/fphys.2018.00528] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022] Open
Abstract
The role of the diet as well as the impact of the dietary habits on human health and disease is well established. Apart from its sleep regulatory effect, the indoleamine melatonin is a well-established antioxidant molecule with multiple health benefits. Convincing evidence supports the presence of melatonin in plants and foods with the intake of such foods affecting circulating melatonin levels in humans. While numerous actions of both endogenous melatonin and melatonin supplementation are well described, little is known about the influence of the dietary melatonin intake on human health. In the present review, evidence for the cardiovascular health benefits of melatonin supplementation and dietary melatonin is discussed. Current knowledge on the biological significance as well as the underlying physiological mechanism of action of the dietary melatonin is also summarized. Whether dietary melatonin constitutes an alternative preventive treatment for cardiovascular disease is addressed.
Collapse
Affiliation(s)
- Zukiswa Jiki
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frederic Nduhirabandi
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
33
|
Xu Y, Lu X, Hu Y, Yang B, Tsui CK, Yu S, Lu L, Liang X. Melatonin attenuated retinal neovascularization and neuroglial dysfunction by inhibition of HIF-1α-VEGF pathway in oxygen-induced retinopathy mice. J Pineal Res 2018; 64:e12473. [PMID: 29411894 DOI: 10.1111/jpi.12473] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
Retinopathy of prematurity (ROP) is a retinopathy characterized by retinal neovascularization (RNV) occurring in preterm infants treated with high concentrations of oxygen and may lead to blindness in severe cases. Currently, anti-VEGF therapy is a major treatment for ROP, but it is costly and may cause serious complications. The previous study has demonstrated that melatonin exerted neuroprotective effect against retinal ganglion cell death induced by hypoxia in neonatal rats. However, whether melatonin is anti-angiogenic and neuroglial protective in the progression of ROP remains unknown. Thus, this study was to investigate the effect of melatonin on RNV and neuroglia in the retina of oxygen-induced retinopathy (OIR) mice. The results showed a reduction in retinal vascular leakage in OIR mice after melatonin treatment. Besides, the size of retinal neovascular and avascular areas, the number of preretinal neovascular cell nuclei, and the number of proliferative vascular endothelial cells within the neovascular area were significantly decreased in mice treated with melatonin. After oxygen-induced injury, the density of astrocytes was decreased, accompanied by morphologic and functional changes of astrocytes. Besides, retinal microglia were also activated. Meanwhile, the levels of inflammatory factors were elevated. However, these pathologic processes were all hindered by melatonin treatment. Furthermore, HIF-1α-VEGF pathway was activated in the retina of OIR mice, yet was suppressed in melatonin-treated OIR mice retinas. In conclusion, melatonin prevented pathologic neovascularization, protected neuroglial cells, and exerts anti-inflammation effect via inhibition of HIF-1α-VEGF pathway in OIR retinas, suggesting that melatonin could be a promising therapeutic agent for ROP.
Collapse
Affiliation(s)
- Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yaguang Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ching-Kit Tsui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lin Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
34
|
Goedecke JH, Mendham AE, Clamp L, Nono Nankam PA, Fortuin-de Smidt MC, Phiri L, Micklesfield LK, Keswell D, Woudberg NJ, Lecour S, Alhamud A, Kaba M, Lutomia FM, van Jaarsveld PJ, de Villiers A, Kahn SE, Chorell E, Hauksson J, Olsson T. An Exercise Intervention to Unravel the Mechanisms Underlying Insulin Resistance in a Cohort of Black South African Women: Protocol for a Randomized Controlled Trial and Baseline Characteristics of Participants. JMIR Res Protoc 2018; 7:e75. [PMID: 29669711 PMCID: PMC5932332 DOI: 10.2196/resprot.9098] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 11/13/2022] Open
Abstract
Background The pathogenesis of type 2 diabetes (T2D) in black African women is complex and differs from that in their white counterparts. However, earlier studies have been cross-sectional and provide little insight into the causal pathways. Exercise training is consistently used as a model to examine the mechanisms underlying insulin resistance and risk for T2D. Objective The objective of the study was to examine the mechanisms underlying the changes in insulin sensitivity and secretion in response to a 12-week exercise intervention in obese black South African (SA) women. Methods A total of 45 obese (body mass index, BMI: 30-40 kg/m2) black SA women were randomized into a control (n=22) or experimental (exercise; n=23) group. The exercise group completed 12 weeks of supervised combined aerobic and resistance training (40-60 min, 4 days/week), while the control group maintained their typical physical activity patterns, and both groups were requested not to change their dietary patterns. Before and following the 12-week intervention period, insulin sensitivity and secretion (frequently sampled intravenous glucose tolerance test) and its primary and secondary determinants were measured. Dietary intake, sleep quality and quantity, physical activity, and sedentary behaviors were measured every 4 weeks. Results The final sample included 20 exercise and 15 control participants. Baseline sociodemographics, cardiorespiratory fitness, anthropometry, cardiometabolic risk factors, physical activity, and diet did not differ between the groups (P>.05). Conclusions The study describes a research protocol for an exercise intervention to understand the mechanisms underlying insulin sensitivity and secretion in obese black SA women and aims to identify causal pathways underlying the high prevalence of insulin resistance and risk for T2D in black SA women, targeting specific areas for therapeutic intervention. Trial Registration Pan African Clinical Trial Registry PACTR201711002789113; http://www.pactr.org/ATMWeb/ appmanager/atm/atmregistry?_nfpb=true&_pageLabel=portals_app_atmregistry_portal_page_13 (Archived by WebCite at http://www.webcitation.org/6xLEFqKr0)
Collapse
Affiliation(s)
- Julia H Goedecke
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa.,Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Amy E Mendham
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa.,Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Louise Clamp
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Pamela A Nono Nankam
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Melony C Fortuin-de Smidt
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Lindokuhle Phiri
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Lisa K Micklesfield
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa.,South African Medical Research Council / University of the Witwatersrand Developmental Pathways for Health Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Gauteng, South Africa
| | - Dheshnie Keswell
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Nicholas J Woudberg
- Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Ali Alhamud
- Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Mamadou Kaba
- Division of Medical Microbiology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Faith M Lutomia
- Division of Medical Microbiology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Paul J van Jaarsveld
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa.,Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Anniza de Villiers
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, WA, United States
| | - Elin Chorell
- Department of Public Health and Clinical Medicine, Umeå University, Umea, Sweden
| | - Jon Hauksson
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Tommy Olsson
- Department of Public Health and Clinical Medicine, Umeå University, Umea, Sweden
| |
Collapse
|
35
|
Maarman GJ. Natural Antioxidants as Potential Therapy, and a Promising Role for Melatonin Against Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:161-178. [PMID: 29047086 DOI: 10.1007/978-3-319-63245-2_10] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Plasma and serum samples, and lung/heart tissue of pulmonary hypertension (PH) patients and animal models of PH display elevated oxidative stress. Moreover, the severity of PH and levels of oxidative stress increase concurrently, which suggests that oxidative stress could be utilized as a biomarker for PH progression. Accumulating evidence has well established that oxidative stress is also key role player in the development of PH. Preclinical studies have demonstrated that natural antioxidants improved PH condition, and, therefore, antioxidant therapy has been proposed as a potential therapeutic strategy against PH. These natural antioxidants include medicinal plant extracts and compounds such as resveratrol and melatonin. Recent studies suggest that melatonin provides health benefit against PH, by enhancing antioxidant capacity, increasing vasodilation, counteracting lung and cardiac fibrosis, and stunting right ventricular (RV) hypertrophy/failure. This chapter comprehensively reviews and discusses a variety of natural antioxidants and their efficacy in modulating experimental PH. This chapter also demonstrates that antioxidant therapy remains a therapeutic strategy for PH, and particularly identifies melatonin as a safe, cost-effective, and promising antioxidant therapy.
Collapse
Affiliation(s)
- Gerald J Maarman
- Hatter Institute for Cardiovascular Research in Africa (HICRA) and MRC Inter-University, Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
36
|
Bonnet S, Boucherat O. The ROS controversy in hypoxic pulmonary hypertension revisited. Eur Respir J 2018; 51:1800276. [PMID: 29519907 DOI: 10.1183/13993003.00276-2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/13/2018] [Indexed: 01/09/2023]
Affiliation(s)
- Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
- Dept of Medicine, Université Laval, Québec, QC, Canada
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
- Dept of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
37
|
Maarman GJ, Reiter RJ. Melatonin therapy for blunt trauma and strenuous exercise: A mechanism involving cytokines, NFκB, Akt, MAFBXand MURF-1. J Sports Sci 2018; 36:1897-1901. [DOI: 10.1080/02640414.2018.1424491] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Gerald J. Maarman
- Hatter Institute for Cardiovascular Disease in Africa (HICRA), Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, UT Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
38
|
dos Santos M, Favero G, Bonomini F, Stacchiotti A, Rodella LF, Veronese FV, Rezzani R. Oral supplementation of melatonin protects against lupus nephritis renal injury in a pristane-induced lupus mouse model. Life Sci 2018; 193:242-251. [DOI: 10.1016/j.lfs.2017.10.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 12/11/2022]
|
39
|
Westerhof BE, Saouti N, van der Laarse WJ, Westerhof N, Vonk Noordegraaf A. Treatment strategies for the right heart in pulmonary hypertension. Cardiovasc Res 2017; 113:1465-1473. [PMID: 28957540 PMCID: PMC5852547 DOI: 10.1093/cvr/cvx148] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/15/2017] [Accepted: 09/01/2017] [Indexed: 02/06/2023] Open
Abstract
The function of the right ventricle (RV) determines the prognosis of patients with pulmonary hypertension. While much progress has been made in the treatment of pulmonary hypertension, therapies for the RV are less well established. In this review of treatment strategies for the RV, first we focus on ways to reduce wall stress since this is the main determinant of changes to the ventricle. Secondly, we discuss treatment strategies targeting the detrimental consequences of increased RV wall stress. To reduce wall stress, afterload reduction is the essential. Additionally, preload to the ventricle can be reduced by diuretics, by atrial septostomy, and potentially by mechanical ventricular support. Secondary to ventricular wall stress, left-to-right asynchrony, altered myocardial energy metabolism, and neurohumoral activation will occur. These may be targeted by optimising RV contraction with pacing, by iron supplement, by angiogenesis and improving mitochondrial function, and by neurohumoral modulation, respectively. We conclude that several treatment strategies for the right heart are available; however, evidence is still limited and further research is needed before clinical application can be recommended.
Collapse
MESH Headings
- Adrenergic beta-Antagonists/therapeutic use
- Animals
- Antihypertensive Agents/adverse effects
- Antihypertensive Agents/therapeutic use
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/therapy
- Arterial Pressure/drug effects
- Cardiac Resynchronization Therapy
- Cardiac Resynchronization Therapy Devices
- Diuretics/therapeutic use
- Energy Metabolism/drug effects
- Heart-Assist Devices
- Humans
- Hypertension, Pulmonary/complications
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Pulmonary Artery/drug effects
- Pulmonary Artery/physiopathology
- Treatment Outcome
- Ventricular Dysfunction, Right/etiology
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/therapy
- Ventricular Function, Right/drug effects
Collapse
Affiliation(s)
- Berend E. Westerhof
- Department of Pulmonary Diseases, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Department of Medical Biology, Academic Medical Center, Amsterdam, The Netherlands
| | - Nabil Saouti
- Department of Cardio-Thoracic Surgery, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Willem J. van der Laarse
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Nico Westerhof
- Department of Pulmonary Diseases, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Anton Vonk Noordegraaf
- Department of Pulmonary Diseases, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
40
|
Majidinia M, Sadeghpour A, Mehrzadi S, Reiter RJ, Khatami N, Yousefi B. Melatonin: A pleiotropic molecule that modulates DNA damage response and repair pathways. J Pineal Res 2017; 63. [PMID: 28439991 DOI: 10.1111/jpi.12416] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/20/2017] [Indexed: 02/06/2023]
Abstract
DNA repair is responsible for maintaining the integrity of the genome. Perturbations in the DNA repair pathways have been identified in several human cancers. Thus, compounds targeting DNA damage response (DDR) hold great promise in cancer therapy. A great deal of effort, in pursuit of new anticancer drugs, has been devoted to understanding the basic mechanisms and functions of the cellular DNA repair machinery. Melatonin, a widely produced indoleamine in all organisms, is associated with a reduced risk of cancer and has multiple regulatory roles on the different aspects of the DDR and DNA repair. Herein, we have mainly discussed how defective components in different DNA repair machineries, including homologous recombination (HR), nonhomologous end-joining (NHEJ), base excision repair (BER), nucleotide excision repair (NER), and finally DNA mismatch repair (MMR), can contribute to the risk of cancer. Melatonin biosynthesis, mode of action, and antioxidant effects are reviewed along with the means by which the indoleamine regulates DDR at the transduction, mediation, and functional levels. Finally, we summarize recent studies that illustrate how melatonin can be combined with DNA-damaging agents to improve their efficacy in cancer therapy.
Collapse
Affiliation(s)
- Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Alireza Sadeghpour
- Department of Orthopedic Surgery, School of Medicine and Shohada Educational Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Mehrzadi
- Health Promotion Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Nasrin Khatami
- Institute for Stem Cell and Regenerative Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Targeting Therapy Research Group, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Melatonin Attenuates Pulmonary Hypertension in Chronically Hypoxic Rats. Int J Mol Sci 2017; 18:ijms18061125. [PMID: 28538666 PMCID: PMC5485949 DOI: 10.3390/ijms18061125] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 11/17/2022] Open
Abstract
Chronic hypoxia induces pulmonary hypertension and vascular remodeling, which are clinically relevant to patients with chronic obstructive pulmonary disease (COPD) associated with a decreased level of nitric oxide (NO). Oxidative stress and inflammation play important roles in the pathophysiological processes in COPD. We examined the hypothesis that daily administration of melatonin (10 mg/kg) mitigates the pulmonary hypertension and vascular remodeling in chronically hypoxic rats. The right ventricular systolic pressure (RVSP) and the thickness of pulmonary arteriolar wall were measured from normoxic control, vehicle- and melatonin-treated hypoxic rats exposed to 10% O2 for 14 days. Levels of markers for oxidative stress (malondialdhyde) and inflammation (tumor necrosis factor-α (TNFα), inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2)) and the expressions of total endothelial NO synthase (eNOS) and phosphorylated eNOS at serine1177 (ser1177) were determined in the lung tissue. We found that the RVSP and the thickness of the arteriolar wall were significantly increased in the vehicle-treated hypoxic animals with elevated levels of malondialdhyde and mRNA expressions of the inflammatory mediators, when compared with the normoxic control. In addition, the phosphorylated eNOS (ser1177) level was significantly decreased, despite an increased eNOS expression in the vehicle-treated hypoxic group. Melatonin treatment significantly attenuated the levels of RVSP, thickness of the arteriolar wall, oxidative and inflammatory markers in the hypoxic animals with a marked increase in the eNOS phosphorylation in the lung. These results suggest that melatonin attenuates pulmonary hypertension by antagonizing the oxidative injury and restoration of NO production.
Collapse
|
42
|
Favero G, Franceschetti L, Buffoli B, Moghadasian MH, Reiter RJ, Rodella LF, Rezzani R. Melatonin: Protection against age-related cardiac pathology. Ageing Res Rev 2017; 35:336-349. [PMID: 27884595 DOI: 10.1016/j.arr.2016.11.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/04/2016] [Accepted: 11/18/2016] [Indexed: 12/14/2022]
Abstract
Aging is a complex and progressive process that involves physiological and metabolic deterioration in every organ and system. Cardiovascular diseases are one of the most common causes of mortality and morbidity among elderly subjects worldwide. Most age-related cardiovascular disorders can be influenced by modifiable behaviours such as a healthy diet rich in fruit and vegetables, avoidance of smoking, increased physical activity and reduced stress. The role of diet in prevention of various disorders is a well-established factor, which has an even more important role in the geriatric population. Melatonin, an indoleamine with multiple actions including antioxidant properties, has been identified in a very large number of plant species, including edible plant products and medical herbs. Among products where melatonin has been identified include wine, olive oil, tomato, beer, and others. Interestingly, consumed melatonin in plant foods or melatonin supplementation may promote health benefits by virtue of its multiple properties and it may counteract pathological conditions also related to cardiovascular disorders, carcinogenesis, neurological diseases and aging. In the present review, we summarized melatonin effects against age-related cardiac alterations and abnormalities with a special focus on heart ischemia/reperfusion (IR) injury and myocardial infarction.
Collapse
Affiliation(s)
- Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Lorenzo Franceschetti
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Barbara Buffoli
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Mohammed H Moghadasian
- Department of Human Nutritional Sciences, University of Manitoba and the Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Luigi F Rodella
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| |
Collapse
|
43
|
Maarman GJ, Andrew BM, Blackhurst DM, Ojuka EO. Melatonin protects against uric acid-induced mitochondrial dysfunction, oxidative stress, and triglyceride accumulation in C2C12myotubes. J Appl Physiol (1985) 2017; 122:1003-1010. [DOI: 10.1152/japplphysiol.00873.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/02/2016] [Accepted: 12/15/2016] [Indexed: 01/08/2023] Open
Abstract
Excess uric acid has been shown to induce oxidative stress, triglyceride accumulation, and mitochondrial dysfunction in the liver and is an independent predictor of type-2 diabetes. Skeletal muscle plays a dominant role in type 2 diabetes and presents a large surface area to plasma uric acid. However, the effects of uric acid on skeletal muscle are underinvestigated. Our aim was therefore to characterize the effects of excessive uric acid on oxidative stress, triglyceride content, and mitochondrial function in skeletal muscle C2C12myotubes and assess how these are modulated by the antioxidant molecule melatonin. Differentiated C2C12myotubes were exposed to 750 µM uric acid or uric acid + 10 nM melatonin for 72 h. Compared with control, uric acid increased triglyceride content by ~237%, oxidative stress by 32%, and antioxidant capacity by 135%. Uric acid also reduced endogenous ROUTINE respiration, complex II-linked oxidative phosphorylation, and electron transfer system capacities. Melatonin counteracted the effects of uric acid without further altering antioxidant capacity. Our data demonstrate that excess uric acid has adverse effects on skeletal muscle similar to those previously reported in hepatocytes and suggest that melatonin at a low physiological concentration of 10 nM may be a possible therapy against some adverse effects of excess uric acid.NEW & NOTEWORTHY Few studies have investigated the effects of uric acid on skeletal muscle. This study shows that hyperuricemia induces mitochondrial dysfunction and triglyceride accumulation in skeletal muscle. The findings may explain why hyperuricemia is an independent predictor of diabetes.
Collapse
Affiliation(s)
- Gerald J. Maarman
- Division of Exercise Science and Sports Medicine (ESSM), Department of Human Biology, University of Cape Town, Newlands, South Africa; and
| | - Brittany M. Andrew
- Division of Exercise Science and Sports Medicine (ESSM), Department of Human Biology, University of Cape Town, Newlands, South Africa; and
| | - Dee M. Blackhurst
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Edward O. Ojuka
- Division of Exercise Science and Sports Medicine (ESSM), Department of Human Biology, University of Cape Town, Newlands, South Africa; and
| |
Collapse
|
44
|
Pandi-Perumal SR, BaHammam AS, Ojike NI, Akinseye OA, Kendzerska T, Buttoo K, Dhandapany PS, Brown GM, Cardinali DP. Melatonin and Human Cardiovascular Disease. J Cardiovasc Pharmacol Ther 2017; 22:122-132. [DOI: 10.1177/1074248416660622] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
The possible therapeutic role of melatonin in the pathophysiology of coronary artery disorder (CAD) is increasingly being recognized. In humans, exogenous melatonin has been shown to decrease nocturnal hypertension, improve systolic and diastolic blood pressure, reduce the pulsatility index in the internal carotid artery, decrease platelet aggregation, and reduce serum catecholamine levels. Low circulating levels of melatonin are reported in individuals with CAD, arterial hypertension, and congestive heart failure. This review assesses current literature on the cardiovascular effects of melatonin in humans. It can be concluded that melatonin deserves to be considered in clinical trials evaluating novel therapeutic interventions for cardiovascular disorders.
Collapse
Affiliation(s)
- Seithikurippu R. Pandi-Perumal
- Department of Medicine, College of Medicine, The University Sleep Disorders Center, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed S. BaHammam
- Department of Medicine, College of Medicine, The University Sleep Disorders Center, King Saud University, Riyadh, Saudi Arabia
| | - Nwakile I. Ojike
- Division of Health and Behavior, Department of Population Health, New York University Medical Center, Center for Healthful Behavior Change, New York, NY, USA
| | - Oluwaseun A. Akinseye
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, Queens Hospital Center, New York, NY, USA
- CUNY School of Public Health at Brooklyn College, New York, NY, USA
| | - Tetyana Kendzerska
- Institute for Clinical Evaluative Sciences, Sunnybrook Health Sciences Center, Toronto, Ontario, Canada
| | | | - Perundurai S. Dhandapany
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Medicine, Oregon Health and Science University, Portland, OR, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India
| | - Gregory M. Brown
- Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | - Daniel P. Cardinali
- BIOMED-UCA-CONICET and Department of Teaching and Research, Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| |
Collapse
|
45
|
Maarman GJ, Schulz R, Sliwa K, Schermuly RT, Lecour S. Novel putative pharmacological therapies to protect the right ventricle in pulmonary hypertension: a review of current literature. Br J Pharmacol 2017; 174:497-511. [PMID: 28099680 DOI: 10.1111/bph.13721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/06/2016] [Accepted: 09/27/2016] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension (PH) is defined by elevated mean pulmonary artery pressure following the pathological remodelling of small pulmonary arteries. An increase in right ventricular (RV) afterload results in RV hypertrophy and RV failure. The pathophysiology of PH, and RV remodelling in particular, is not well understood, thus explaining, at least in part, why current PH therapies have a limited effect. Existing therapies mostly target the pulmonary circulation. Because the remodelled RV fails to support normal cardiac function, patients eventually succumb from RV failure. Developing novel therapies that directly target the function of the RV may therefore benefit patients with PH. In the past decade, several promising studies have investigated novel cardioprotective strategies in experimental models of PH. This review aims to comprehensively discuss and highlight these novel experimental approaches to confer, in the long-term, greater health benefit in patients with PH.
Collapse
Affiliation(s)
- Gerald J Maarman
- Hatter Institute for Cardiovascular Research in Africa (HICRA) and MRC Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Karen Sliwa
- Hatter Institute for Cardiovascular Research in Africa (HICRA) and MRC Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ralph Theo Schermuly
- Universities of Giessen and Marburg Lung Centre, Member of the German Lung Centre (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa (HICRA) and MRC Inter-University Cape Heart Group, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
46
|
Cabrera-Fuentes HA, Aragones J, Bernhagen J, Boening A, Boisvert WA, Bøtker HE, Bulluck H, Cook S, Di Lisa F, Engel FB, Engelmann B, Ferrazzi F, Ferdinandy P, Fong A, Fleming I, Gnaiger E, Hernández-Reséndiz S, Kalkhoran SB, Kim MH, Lecour S, Liehn EA, Marber MS, Mayr M, Miura T, Ong SB, Peter K, Sedding D, Singh MK, Suleiman MS, Schnittler HJ, Schulz R, Shim W, Tello D, Vogel CW, Walker M, Li QOY, Yellon DM, Hausenloy DJ, Preissner KT. From basic mechanisms to clinical applications in heart protection, new players in cardiovascular diseases and cardiac theranostics: meeting report from the third international symposium on "New frontiers in cardiovascular research". Basic Res Cardiol 2016; 111:69. [PMID: 27743118 PMCID: PMC5065587 DOI: 10.1007/s00395-016-0586-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 10/02/2016] [Accepted: 10/04/2016] [Indexed: 12/12/2022]
Abstract
In this meeting report, particularly addressing the topic of protection of the cardiovascular system from ischemia/reperfusion injury, highlights are presented that relate to conditioning strategies of the heart with respect to molecular mechanisms and outcome in patients' cohorts, the influence of co-morbidities and medications, as well as the contribution of innate immune reactions in cardioprotection. Moreover, developmental or systems biology approaches bear great potential in systematically uncovering unexpected components involved in ischemia-reperfusion injury or heart regeneration. Based on the characterization of particular platelet integrins, mitochondrial redox-linked proteins, or lipid-diol compounds in cardiovascular diseases, their targeting by newly developed theranostics and technologies opens new avenues for diagnosis and therapy of myocardial infarction to improve the patients' outcome.
Collapse
Affiliation(s)
- Hector A Cabrera-Fuentes
- Institute of Biochemistry, Medical School, Justus-Liebig University, Giessen, Germany
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, NL, Mexico
| | - Julian Aragones
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa, Autonomous University of Madrid, Madrid, Spain
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Andreas Boening
- Department of Cardiovascular Surgery, Medical School, Justus-Liebig-University, Giessen, Germany
| | - William A Boisvert
- Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA
| | - Hans E Bøtker
- Department of Cardiology, Aarhus University Hospital, Skejby, Aarhus N, Denmark
| | - Heerajnarain Bulluck
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Stuart Cook
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nuremberg, Germany
| | - Bernd Engelmann
- Institut für Laboratoriumsmedizin, Ludwig-Maximilians-Universität, Munich, Germany
| | - Fulvia Ferrazzi
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nuremberg, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Alan Fong
- Department of Cardiology, Sarawak Heart Centre, Sarawak, Malaysia
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt, Germany
| | - Erich Gnaiger
- D. Swarovski Research Lab, Department of Visceral, Transplant Thoracic Surgery, Medical Univ Innsbruck, Innsbruck, Austria
| | - Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Department of Cardiovascular Medicine, National Institute of Cardiology, Ignacio Chavez, Mexico, D.F., Mexico
| | - Siavash Beikoghli Kalkhoran
- The Hatter Cardiovascular Institute, University College London, London, UK
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - Moo Hyun Kim
- Department of Cardiology, Dong-A University Hospital, Busan, Korea
| | - Sandrine Lecour
- Hatter Institute and MRC Inter-University Cape Heart Unit, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Elisa A Liehn
- Institute for Molecular Cardiovascular Research, RWTH University Hospital, Aachen, Germany
| | - Michael S Marber
- Department of Cardiology, The Rayne Institute, St Thomas' Campus, King's College London, London, UK
| | - Manuel Mayr
- The James Black Centre, King's College, University of London, London, UK
| | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Karlheinz Peter
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Daniel Sedding
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Manvendra K Singh
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - M Saadeh Suleiman
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Bristol, UK
| | - Hans J Schnittler
- Institute of Anatomy and Vascular Biology, Westfalian-Wilhelms-University, Münster, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Winston Shim
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Daniel Tello
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa, Autonomous University of Madrid, Madrid, Spain
| | - Carl-Wilhelm Vogel
- Department of Pathology, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA
| | - Malcolm Walker
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Qilong Oscar Yang Li
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa, Autonomous University of Madrid, Madrid, Spain
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore, 169857, Singapore.
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.
- The Hatter Cardiovascular Institute, University College London, London, UK.
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK.
| | - Klaus T Preissner
- Institute of Biochemistry, Medical School, Justus-Liebig University, Giessen, Germany
- Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
47
|
Wei A, Gu Z, Li J, Liu X, Wu X, Han Y, Pu J. Clinical Adverse Effects of Endothelin Receptor Antagonists: Insights From the Meta-Analysis of 4894 Patients From 24 Randomized Double-Blind Placebo-Controlled Clinical Trials. J Am Heart Assoc 2016; 5:e003896. [PMID: 27912207 PMCID: PMC5210319 DOI: 10.1161/jaha.116.003896] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 09/09/2016] [Indexed: 12/03/2022]
Abstract
BACKGROUND Evidence of the clinical safety of endothelin receptor antagonists (ERAs) is limited and derived mainly from individual trials; therefore, we conducted a meta-analysis. METHODS AND RESULTS After systematic searches of the Medline, Embase, and Cochrane Library databases and the ClinicalTrials.gov website, randomized controlled trials with patients receiving ERAs (bosentan, macitentan, or ambrisentan) in at least 1 treatment group were included. All reported adverse events of ERAs were evaluated. Summary relative risks and 95% CIs were calculated using random- or fixed-effects models according to between-study heterogeneity. In total, 24 randomized trials including 4894 patients met the inclusion criteria. Meta-analysis showed that the incidence of abnormal liver function (7.91% versus 2.84%; risk ratio [RR] 2.38, 95% CI 1.36-4.18), peripheral edema (14.36% versus 9.68%; RR 1.44, 95% CI 1.20-1.74), and anemia (6.23% versus 2.44%; RR 2.69, 95% CI 1.78-4.07) was significantly higher in the ERA group compared with placebo. In comparisons of individual ERAs with placebo, bosentan (RR 3.78, 95% CI 2.42-5.91) but not macitentan (RR 1.17, 95% CI 0.42-3.31) significantly increased the risk of abnormal liver function, whereas ambrisentan (RR 0.06, 95% CI 0.01-0.45) significantly decreased that risk. Bosentan (RR 1.47, 95% CI 1.06-2.03) and ambrisentan (RR 2.02, 95% CI 1.40-2.91) but not macitentan (RR 1.08, 95% CI 0.81-1.46) significantly increased the risk of peripheral edema. Bosentan (RR 3.09, 95% CI 1.52-6.30) and macitentan (RR 2.63, 95% CI 1.54-4.47) but not ambrisentan (RR 1.30, 95% CI 0.20-8.48) significantly increased the risk of anemia. ERAs were not found to increase other reported adverse events compared with placebo. CONCLUSIONS The present meta-analysis showed that the main adverse effects of treatment with ERAs were hepatic transaminitis (bosentan), peripheral edema (bosentan and ambrisentan), and anemia (bosentan and macitentan).
Collapse
Affiliation(s)
- Anhua Wei
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichun Gu
- Department of Pharmacy, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Juan Li
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Liu
- Department of Pharmacy, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaofan Wu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yi Han
- Geriatric ICU, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Pu
- Department of Cardiology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Melatonin is a neuroendocrine hormone synthesized primarily by the pineal gland. Numerous studies have suggested that melatonin plays an important role in various cardiovascular diseases. In this article, recent progress regarding melatonin's effects on cardiovascular diseases is reviewed. RECENT FINDINGS In the past year, studies have focused on the mechanism of protection of melatonin on cardiovascular diseases, including myocardial ischemia-reperfusion injury, myocardial hypoxia-reoxygenation injury, pulmonary hypertension, hypertension, atherosclerosis, valvular heart diseases, and other cardiovascular diseases. SUMMARY Studies have demonstrated that melatonin has significant effects on ischemia-reperfusion injury, myocardial chronic intermittent hypoxia injury, pulmonary hypertension, hypertension, valvular heart diseases, vascular diseases, and lipid metabolism. As an inexpensive and well tolerated drug, melatonin may be a new therapeutic option for cardiovascular disease.
Collapse
Affiliation(s)
- Hang Sun
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Aaron M. Gusdon
- Department of Pathology, Division of Neuropathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Neurology, Weill Cornell Medical College, New York, USA
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
49
|
Kang JW, Hong JM, Lee SM. Melatonin enhances mitophagy and mitochondrial biogenesis in rats with carbon tetrachloride-induced liver fibrosis. J Pineal Res 2016; 60:383-93. [PMID: 26882442 DOI: 10.1111/jpi.12319] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 02/09/2016] [Indexed: 12/14/2022]
Abstract
Liver fibrosis leads to liver cirrhosis and failure, and no effective treatment is currently available. Growing evidence supports a link between mitochondrial dysfunction and liver fibrogenesis and mitochondrial quality control-based therapy has emerged as a new therapeutic target. We investigated the protective mechanisms of melatonin against mitochondrial dysfunction-involved liver fibrosis, focusing on mitophagy and mitochondrial biogenesis. Rats were treated with carbon tetrachloride (CCl4) dissolved in olive oil (0.5 mL/kg, twice a week, i.p.) for 8 wk. Melatonin was administered orally at 2.5, 5, and 10 mg/kg once a day. Chronic CCl4 exposure induced collagen deposition, hepatocellular damage, and oxidative stress, and melatonin attenuated these increases. Increases in mRNA and protein expression levels of transforming growth factor β1 and α-smooth muscle actin in response to CCl4 were attenuated by melatonin. Melatonin attenuated hallmarks of mitochondrial dysfunction, such as mitochondrial swelling and glutamate dehydrogenase release. Chronic CCl4 exposure impaired mitophagy and mitochondrial biogenesis, and melatonin attenuated this impairment, as indicated by increases in mitochondrial DNA and in protein levels of PTEN-induced putative kinase 1 (PINK1); Parkin; peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α); nuclear respiratory factor 1 (NRF1); and transcription factor A, mitochondrial (TFAM). CCl4-mediated decreases in mitochondrial fission- and fusion-related proteins, such as dynamin-related protein 1 (DRP1) and mitofusin 2, were also attenuated by melatonin. Moreover, melatonin induced AMP-activated protein kinase (AMPK) phosphorylation. These results suggest that melatonin protects against liver fibrosis via upregulation of mitophagy and mitochondrial biogenesis, and may be useful as an anti-fibrotic treatment.
Collapse
Affiliation(s)
- Jung-Woo Kang
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| | - Jeong-Min Hong
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| | - Sun-Mee Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| |
Collapse
|
50
|
Hu W, Ma Z, Jiang S, Fan C, Deng C, Yan X, Di S, Lv J, Reiter RJ, Yang Y. Melatonin: the dawning of a treatment for fibrosis? J Pineal Res 2016; 60:121-31. [PMID: 26680689 DOI: 10.1111/jpi.12302] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/11/2015] [Indexed: 12/28/2022]
Abstract
Fibrosis is a common occurrence following organ injury and failure. To date, there is no effective treatment for this condition. Melatonin targets numerous molecular pathways, a consequence of its antioxidant and anti-inflammatory actions that reduce excessive fibrosis. Herein, we review the multiple protective effects of melatonin against fibrosis. There exist four major phases of the fibrogenic response including primary injury to the organ, activation of effector cells, the elaboration of extracellular matrix (ECM) and dynamic deposition. Melatonin regulates each of these phases. Additionally, melatonin reduces fibrosis levels in numerous organs. Melatonin exhibits its anti-fibrosis effects in heart, liver, lung, kidney, and other organs. In addition, adhesions which occur following surgical procedures are also inhibited by melatonin. The information reviewed here should be significant to understanding the protective role of melatonin against fibrosis, contribute to the design of further experimental studies related to melatonin and the fibrotic response and shed light on a potential treatment for fibrosis.
Collapse
Affiliation(s)
- Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi'an, China
- Department of General Surgery, Beidaihe Sanatorium, Beijing Military Area Command, Qinhuangdao, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, Xi'an, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Chao Deng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jianjun Lv
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi'an, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, TX, USA
| | - Yang Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi'an, China
- Department of General Surgery, Beidaihe Sanatorium, Beijing Military Area Command, Qinhuangdao, China
| |
Collapse
|