1
|
Nevoit G, Jarusevicius G, Potyazhenko M, Mintser O, Bumblyte IA, Vainoras A. Mitochondrial Dysfunction and Atherosclerosis: The Problem and the Search for Its Solution. Biomedicines 2025; 13:963. [PMID: 40299559 PMCID: PMC12024619 DOI: 10.3390/biomedicines13040963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025] Open
Abstract
Background/Objectives: This review has been prepared to promote interest in the interdisciplinary study of mitochondrial dysfunction (MD) and atherosclerosis. This review aims to describe the state of this problem and indicate the direction for further implementation of this knowledge in clinical medicine. Methods: Extensive research of the literature was implemented to elucidate the role of the molecular mechanisms of MD in the pathogenesis of atherosclerosis. Results: A view on the pathogenesis of atherosclerosis through the prism of knowledge about MD is presented. MD is the cause and primary mechanism of the onset and progression of atherosclerosis. It is proposed that this problem be considered in the context of a continuum. Conclusions: MD and atherosclerosis are united by common molecular mechanisms of pathogenesis. Knowledge of MD should be used to argue for a healthy lifestyle as the primary way to prevent atherosclerosis. The development of new approaches to diagnosing and treating MD in atherosclerosis is an urgent task and challenge for modern science.
Collapse
Affiliation(s)
- Ganna Nevoit
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Gediminas Jarusevicius
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Maksim Potyazhenko
- Department of Internal Medicine and Emergency Medicine, Poltava State Medical University, 36011 Poltava, Ukraine
| | - Ozar Mintser
- Department of Fundamental Disciplines and Informatics, Shupyk National Healthcare University of Ukraine, 04112 Kyiv, Ukraine
| | - Inga Arune Bumblyte
- Department of Nephrology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Alfonsas Vainoras
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| |
Collapse
|
2
|
Alhasaniah AH, Alissa M, Elsaid FG, Alsugoor MH, AlQahtani MS, Alessa A, Jambi K, Albakri GS, Albaqami FMK, Bennett E. The enigmatic role of SIRT2 in the cardiovascular system: Deciphering its protective and detrimental actions to unlock new avenues for therapeutic intervention. Curr Probl Cardiol 2025; 50:102929. [PMID: 39566866 DOI: 10.1016/j.cpcardiol.2024.102929] [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: 11/12/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Cardiovascular diseases (CVDs) are leading causes of mortality throughout the world, and hence, there is a critical need to elucidate their molecular mechanisms. The Sirtuin (SIRT) family of NAD+-dependent enzymes has recently been shown to play a critical role in cardiovascular health and disease, and several SIRT isoforms, especially SIRT1 and SIRT3, have been amply investigated. However, the precise function of SIRT2 is only partially explored. Here, we review the current understanding of the involvement of SIRT2 in various cardiovascular pathologies, such as cardiac hypertrophy, ischemia-reperfusion injury, diabetic cardiomyopathy, and vascular dysfunction, with emphasis placed on the context-dependent protective or deleterious actions of SIRT2, including its wide array of catalytic activities which span beyond deacetylation. Furthermore, the review uncovers several unresolved research gaps for SIRT2 mechanisms by which SIRT2 modulates cardiac and vascular function during development and aging, thereby paving the way for the discovery of novel therapeutic targets as well as SIRT2-targeted interventions in the prevention and treatment of various cardiovascular diseases.
Collapse
Affiliation(s)
- Abdulaziz Hassan Alhasaniah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, P.O. Box 1988, Najran, Saudi Arabia
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Fahmy Gad Elsaid
- Department of Biology, College of Science, King Khalid University, PO Box 960, Asir, Abha, 61421, Saudi Arabia
| | - Mahdi H Alsugoor
- Department of Emergency Medical Services, Faculty of Health Sciences, AlQunfudah, Umm Al-Qura University, Makkah 21912, Saudi Arabia
| | - Mohammed S AlQahtani
- Department of Medical Laboratory, Prince Sultan Air Base Hospital, Al-kharj, Saudi Arabia
| | - Anwer Alessa
- Department of Medical Laboratory, Al Kharj Military Industries Corporation Hospital, Al-kharj, Saudi Arabia
| | - Khalid Jambi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Ghadah Shukri Albakri
- Department of Teching and Learning, College of Education and Human development, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Faisal Miqad K Albaqami
- Department of Biology, Faculty of Science, Islamic University of Madinah, Madinah 42351, Saudi Arabia
| | - Elizabeth Bennett
- Queen Elizabeth Hospital Birmingham (QEHB), Nuffield House, 3rd Floor Room 17/E, Mindelsohn Way, Edgbaston, Birmingham, B15 2WB, Dudley Road, Birmingham, West Midlands, B18 7QH
| |
Collapse
|
3
|
Divya KP, Kanwar N, Anuranjana PV, Kumar G, Beegum F, George KT, Kumar N, Nandakumar K, Kanwal A. SIRT6 in Regulation of Mitochondrial Damage and Associated Cardiac Dysfunctions: A Possible Therapeutic Target for CVDs. Cardiovasc Toxicol 2024; 24:598-621. [PMID: 38689163 DOI: 10.1007/s12012-024-09858-1] [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: 09/01/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Cardiovascular diseases (CVDs) can be described as a global health emergency imploring possible prevention strategies. Although the pathogenesis of CVDs has been extensively studied, the role of mitochondrial dysfunction in CVD development has yet to be investigated. Diabetic cardiomyopathy, ischemic-reperfusion injury, and heart failure are some of the CVDs resulting from mitochondrial dysfunction Recent evidence from the research states that any dysfunction of mitochondria has an impact on metabolic alteration, eventually causes the death of a healthy cell and therefore, progressively directing to the predisposition of disease. Cardiovascular research investigating the targets that both protect and treat mitochondrial damage will help reduce the risk and increase the quality of life of patients suffering from various CVDs. One such target, i.e., nuclear sirtuin SIRT6 is strongly associated with cardiac function. However, the link between mitochondrial dysfunction and SIRT6 concerning cardiovascular pathologies remains poorly understood. Although the Role of SIRT6 in skeletal muscles and cardiomyocytes through mitochondrial regulation has been well understood, its specific role in mitochondrial maintenance in cardiomyocytes is poorly determined. The review aims to explore the domain-specific function of SIRT6 in cardiomyocytes and is an effort to know how SIRT6, mitochondria, and CVDs are related.
Collapse
Affiliation(s)
- K P Divya
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Navjot Kanwar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab, Technical University, Bathinda, Punjab, 151005, India
| | - P V Anuranjana
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Gautam Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
- School of Pharmacy, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Krupa Thankam George
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Nitesh Kumar
- Department of Pharmacology, National Institute of Pharmaceutical Educations and Research, Hajipur, Bihar, 844102, India
| | - K Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India.
| | - Abhinav Kanwal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, Punjab, 151005, India.
| |
Collapse
|
4
|
Sun W, Yang T, Wang C, Li H, Lei L. Mitochondrial ROS participates in Porphyromonas gingivalis-induced pyroptosis in cementoblasts. Heliyon 2024; 10:e30814. [PMID: 38774076 PMCID: PMC11107101 DOI: 10.1016/j.heliyon.2024.e30814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 04/02/2024] [Accepted: 05/06/2024] [Indexed: 05/24/2024] Open
Abstract
This study aimed to investigate correlation between mitochondrial reactive oxygen species and Porphyromonas gingivalis in the process of cementoblast pyroptosis. Lactate dehydrogenase activity assay, enzyme-linked immunosorbent assay, western blotting and flow cytometry analysis were utilized to explore whether Porphyromonas gingivalis triggered pyroptosis in cementoblasts. Reactive oxygen species and mitochondrial reactive oxygen species were detected using flow cytometry and fluorescence staining. The effect of mitochondrial reactive oxygen species on the Porphyromonas gingivalis-induced pyroptosis of cementoblasts was assessed by Mito-Tempo, mitochondrion-targeted superoxide dismutase mimetic. Phosphorylation levels of p65 were measured by western blotting. SC75741, a nuclear factor-kappa B inhibitor, was added to block the nuclear factor-kappa B in the Porphyromonas gingivalis-infected cementoblasts. Porphyromonas gingivalis triggered pyroptosis of cementoblasts, and an elevation in reactive oxygen species generation in the mitochondria was observed. Inhibition of mitochondrial reactive oxygen species reduced pyroptosis and nuclear factor-kappa B signaling pathway mediated the pyroptotic cell death in Porphyromonas gingivalis-infected cementoblasts. Together, our findings demonstrate that mitochondrial reactive oxygen species increased by Porphyromonas gingivalis participated in the pyroptosis of cementoblasts. Targeting mitochondrial reactive oxygen species may offer therapeutic strategies for root surface remodeling or periodontal regeneration.
Collapse
Affiliation(s)
- Weiman Sun
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
| | - Tianrui Yang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
| | - Chenxu Wang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
| | - Houxuan Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
| | - Lang Lei
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
| |
Collapse
|
5
|
Solanki K, Bezsonov E, Orekhov A, Parihar SP, Vaja S, White FA, Obukhov AG, Baig MS. Effect of reactive oxygen, nitrogen, and sulfur species on signaling pathways in atherosclerosis. Vascul Pharmacol 2024; 154:107282. [PMID: 38325566 DOI: 10.1016/j.vph.2024.107282] [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: 01/28/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease in which fats, lipids, cholesterol, calcium, proliferating smooth muscle cells, and immune cells accumulate in the intima of the large arteries, forming atherosclerotic plaques. A complex interplay of various vascular and immune cells takes place during the initiation and progression of atherosclerosis. Multiple reports indicate that tight control of reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species (RSS) production is critical for maintaining vascular health. Unrestricted ROS and RNS generation may lead to activation of various inflammatory signaling pathways, facilitating atherosclerosis. Given these deleterious consequences, it is important to understand how ROS and RNS affect the signaling processes involved in atherogenesis. Conversely, RSS appears to exhibit an atheroprotective potential and can alleviate the deleterious effects of ROS and RNS. Herein, we review the literature describing the effects of ROS, RNS, and RSS on vascular smooth muscle cells, endothelial cells, and macrophages and focus on how changes in their production affect the initiation and progression of atherosclerosis. This review also discusses the contribution of ROS, RNS, and RSS in mediating various post-translational modifications, such as oxidation, nitrosylation, and sulfation, of the molecules involved in inflammatory signaling.
Collapse
Affiliation(s)
- Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Evgeny Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia; Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia; Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; The Cell Physiology and Pathology Laboratory, Turgenev State University of Orel, Orel, Russia
| | - Alexander Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Suraj P Parihar
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Medical Microbiology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Shivani Vaja
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Fletcher A White
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India.
| |
Collapse
|
6
|
Julla JB, Girard D, Diedisheim M, Saulnier PJ, Tran Vuong B, Blériot C, Carcarino E, De Keizer J, Orliaguet L, Nemazanyy I, Potier C, Khider K, Tonui DC, Ejlalmanesh T, Ballaire R, Mambu Mambueni H, Germain S, Gaborit B, Vidal-Trécan T, Riveline JP, Garchon HJ, Fenaille F, Lemoine S, Carlier A, Castelli F, Potier L, Masson D, Roussel R, Vandiedonck C, Hadjadj S, Alzaid F, Gautier JF, Venteclef N. Blood Monocyte Phenotype Is A Marker of Cardiovascular Risk in Type 2 Diabetes. Circ Res 2024; 134:189-202. [PMID: 38152893 DOI: 10.1161/circresaha.123.322757] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/13/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Diabetes is a major risk factor for atherosclerotic cardiovascular diseases with a 2-fold higher risk of cardiovascular events in people with diabetes compared with those without. Circulating monocytes are inflammatory effector cells involved in both type 2 diabetes (T2D) and atherogenesis. METHODS We investigated the relationship between circulating monocytes and cardiovascular risk progression in people with T2D, using phenotypic, transcriptomic, and metabolomic analyses. cardiovascular risk progression was estimated with coronary artery calcium score in a cohort of 672 people with T2D. RESULTS Coronary artery calcium score was positively correlated with blood monocyte count and frequency of the classical monocyte subtype. Unsupervised k-means clustering based on monocyte subtype profiles revealed 3 main endotypes of people with T2D at varying risk of cardiovascular events. These observations were confirmed in a validation cohort of 279 T2D participants. The predictive association between monocyte count and major adverse cardiovascular events was validated through an independent prospective cohort of 757 patients with T2D. Integration of monocyte transcriptome analyses and plasma metabolomes showed a disruption of mitochondrial pathways (tricarboxylic acid cycle, oxidative phosphorylation pathway) that underlined a proatherogenic phenotype. CONCLUSIONS In this study, we provide evidence that frequency and monocyte phenotypic profile are closely linked to cardiovascular risk in patients with T2D. The assessment of monocyte frequency and count is a valuable predictive marker for risk of cardiovascular events in patients with T2D. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT04353869.
Collapse
Affiliation(s)
- Jean-Baptiste Julla
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology, Endocrinology and Nutrition Department, Lariboisière Hospital, Fédération de Diabétologie, France (J.-B.J., T.V.-T., J.-P.R., J.-F.G.)
| | - Diane Girard
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Marc Diedisheim
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Clinique Saint Gatien Alliance (NCT+), Saint-Cyr-sur-Loire, France (M.D.)
| | - Pierre-Jean Saulnier
- Poitiers Université, CHU Poitiers, INSERM, Centre d'Investigation Clinique CIC1402, Poitiers, France (P.-J.S.)
| | - Bao Tran Vuong
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Camille Blériot
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Elena Carcarino
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Joe De Keizer
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France (J.D.K., S.H.)
| | - Lucie Orliaguet
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Ivan Nemazanyy
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Charline Potier
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Kennan Khider
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Dorothy Chepngenoh Tonui
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Tina Ejlalmanesh
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Raphaelle Ballaire
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Hendrick Mambu Mambueni
- Genomics platform UFR Simone Veil 1173; U, University of Versailles Paris-Saclay; Inserm UMR 1173 (H.M.M., H.-J.G.)
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France (S.G.)
| | - Bénédicte Gaborit
- C2VN, INRAE, INSERM, Aix Marseille University, Marseille, France (B.G.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, AP-HM, Marseille, France (B.G.)
| | - Tiphaine Vidal-Trécan
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology, Endocrinology and Nutrition Department, Lariboisière Hospital, Fédération de Diabétologie, France (J.-B.J., T.V.-T., J.-P.R., J.-F.G.)
| | - Jean-Pierre Riveline
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology, Endocrinology and Nutrition Department, Lariboisière Hospital, Fédération de Diabétologie, France (J.-B.J., T.V.-T., J.-P.R., J.-F.G.)
| | - Henri-Jean Garchon
- Genomics platform UFR Simone Veil 1173; U, University of Versailles Paris-Saclay; Inserm UMR 1173 (H.M.M., H.-J.G.)
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (MTS), MetaboHUB, France (F.F., F.C.)
| | - Sophie Lemoine
- Genomics core facility, Institut de Biologie de l'ENS (IBENS), Département de biologie, École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France (S.L.)
| | - Aurélie Carlier
- Diabetology and Endocrinology Department, Bichat Hospital, Fédération de Diabétologie, France (L.P., A.C., R.R.)
| | - Florence Castelli
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (MTS), MetaboHUB, France (F.F., F.C.)
| | - Louis Potier
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology and Endocrinology Department, Bichat Hospital, Fédération de Diabétologie, France (L.P., A.C., R.R.)
| | - David Masson
- INSERM, LNC UMR1231, Dijon, France (D.M.)
- University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France (D.M.)
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France (D.M.)
- Plateau Automatisé de Biochimie, Dijon University Hospital, France (D.M.)
| | - Ronan Roussel
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology and Endocrinology Department, Bichat Hospital, Fédération de Diabétologie, France (L.P., A.C., R.R.)
| | - Claire Vandiedonck
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Samy Hadjadj
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France (J.D.K., S.H.)
| | - Fawaz Alzaid
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Dasman Diabetes Institute, Kuwait (F.A.)
| | - Jean-François Gautier
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology, Endocrinology and Nutrition Department, Lariboisière Hospital, Fédération de Diabétologie, France (J.-B.J., T.V.-T., J.-P.R., J.-F.G.)
| | - Nicolas Venteclef
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| |
Collapse
|
7
|
Wang H, Zhang X, Li P, Huang F, Xiu T, Wang H, Zhang W, Zhang W, Tang B. Prediction of Early Atherosclerotic Plaques Using a Sequence-Activated Fluorescence Probe for the Simultaneous Detection of γ-Glutamyl Transpeptidase and Hypobromous Acid. Angew Chem Int Ed Engl 2024; 63:e202315861. [PMID: 37985247 DOI: 10.1002/anie.202315861] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Atherosclerosis is a lipoprotein-driven disease, and there is no effective therapy to reverse atherosclerosis or existing plaques. Therefore, it is urgently necessary to create a noninvasive and reliable approach for early atherosclerosis detection to prevent initial plaque formation. Atherosclerosis is intimately associated with inflammation, which is accompanied by an excess of reactive oxygen species (ROS), leading to cells requiring more glutathione (GSH) to resist severe oxidative stress. Therefore, the GSH-hydrolyzed protein γ-glutamyl transpeptidase (GGT) and the ROS-hypobromous acid (HBrO) are potential biomarkers for predicting atherogenesis. Hence, to avoid false-positive diagnoses caused by a single biomarker, we constructed an ingenious sequence-activated double-locked TP fluorescent probe, C-HBrO-GGT, in which two sequential triggers of GGT and HBrO are meticulously designed to ensure that the probe fluoresces in response to HBrO only after GGT hydrolyzes the probe. By utilization of C-HBrO-GGT, the voltage-gated chloride channel (CLC-1)-HBrO-catalase (CAT)-GGT signaling pathway was confirmed in cellular level. Notably, the forthcoming atherosclerotic plaques were successfully predicted before the plaques could be observed via the naked eye or classical immunofluorescent staining. Collectively, this research proposed a powerful tool to indicate the precise position of mature plaques and provide early warning of atherosclerotic plaques.
Collapse
Affiliation(s)
- Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
| | - Xiaoting Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
| | - Fang Huang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
| | - Tiancong Xiu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
| | - HongTong Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
| | - Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institutes of Biomedical Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
- People's Republic of China; Laoshan Laboratory, 168 Wenhai Middle Rd, Aoshanwei Jimo, Qingdao, 266237, Shandong, P. R. China
| |
Collapse
|
8
|
De Marchi F, Venkatesan S, Saraceno M, Mazzini L, Grossini E. Acetyl-L-carnitine and Amyotrophic Lateral Sclerosis: Current Evidence and Potential use. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:588-601. [PMID: 36998125 DOI: 10.2174/1871527322666230330083757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND The management of neurodegenerative diseases can be frustrating for clinicians, given the limited progress of conventional medicine in this context. AIM For this reason, a more comprehensive, integrative approach is urgently needed. Among various emerging focuses for intervention, the modulation of central nervous system energetics, oxidative stress, and inflammation is becoming more and more promising. METHODS In particular, electrons leakage involved in the mitochondrial energetics can generate reactive oxygen-free radical-related mitochondrial dysfunction that would contribute to the etiopathology of many disorders, such as Alzheimer's and other dementias, Parkinson's disease, multiple sclerosis, stroke, and amyotrophic lateral sclerosis (ALS). RESULTS In this context, using agents, like acetyl L-carnitine (ALCAR), provides mitochondrial support, reduces oxidative stress, and improves synaptic transmission. CONCLUSION This narrative review aims to update the existing literature on ALCAR molecular profile, tolerability, and translational clinical potential use in neurodegeneration, focusing on ALS.
Collapse
Affiliation(s)
- Fabiola De Marchi
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale 28100 Novara, Italy
| | - Sakthipriyan Venkatesan
- Laboratory of Physiology, Department of Translational Medicine, University of Piemonte Orientale 28100, Novara, Italy
| | - Massimo Saraceno
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale 28100 Novara, Italy
| | - Letizia Mazzini
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale 28100 Novara, Italy
| | - Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, University of Piemonte Orientale 28100, Novara, Italy
| |
Collapse
|
9
|
Reddy SM, Suresh V, Pitchiah S, Subramanian B. Anti-inflammatory Activities of Sulfated Polysaccharides From Ethanol Crude Extract of Spyrida Species Red Seaweed. Cureus 2023; 15:e50284. [PMID: 38205502 PMCID: PMC10776340 DOI: 10.7759/cureus.50284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 12/10/2023] [Indexed: 01/12/2024] Open
Abstract
INTRODUCTION The extracts derived from red seaweed have shown characteristics that may reduce inflammation. The abovementioned effects can potentially provide positive outcomes in managing inflammatory illnesses, including arthritis, inflammatory bowel disease, and other skin problems. AIM The polysaccharides were isolated from the Spyrida species. The water-soluble polysaccharides were extracted and fractionated from several Indian seaweeds (Red) using a simple, cost-effective approach. Anti-inflammatory effects were further evaluated and validated by FTIR and FESEM analyses. MATERIALS AND METHODS FT-IR and FESEM were used to assess the structural features of polysaccharides and the surface morphology. In addition, the red seaweed species of the genus Spyrida, which includes polysaccharides, was shown to significantly inhibit the denaturation of bovine serum albumin (BSA), further proving that the substance has anti-inflammatory qualities. RESULTS In this work, an assay to suppress protein activity was utilized to investigate the potential anti-inflammatory effects of polysaccharides derived from Spyrida. As predicted, increasing concentrations of the extract, ranging from 25 to 100 µg/ml, led to a rise in the percentage of inhibited protein denaturation. CONCLUSION A statistically significant difference was found between these findings and those obtained with aspirin, a commonly used non-steroidal anti-inflammatory medicine (NSAID). The red algae that grow in the shallow waters of the southern Indian Ocean may be used in medicine.
Collapse
Affiliation(s)
- Shweta Mary Reddy
- Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Vasugi Suresh
- Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Sivaperumal Pitchiah
- Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Balachandran Subramanian
- Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
10
|
Lee J, Lee SA, Son SH, Choi JA, Nguyen TD, Kim J, Son D, Song CH. Impaired mitophagy induces antimicrobial responses in macrophages infected with Mycobacterium tuberculosis. Cell Biosci 2023; 13:158. [PMID: 37649112 PMCID: PMC10470153 DOI: 10.1186/s13578-023-01107-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 08/18/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Mitophagy, mitochondrial selective autophagy, plays a pivotal role in the maintenance of cellular homeostasis in response to cellular stress. However, the role of mitophagy in macrophages during infection has not been elucidated. To determine whether mitophagy regulates intracellular pathogen survival, macrophages were infected with Mycobacterium tuberculosis (Mtb), an intracellular bacterium. RESULTS We showed that Mtb-infected macrophages induced mitophagy through BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3) activation. In contrast, BNIP3-deficient macrophages failed to induce mitophagy, resulting in reduced mitochondrial membrane potential in response to Mtb infection. Moreover, the accumulation of damaged mitochondria due to BNIP3 deficiency generated higher levels of mitochondrial reactive oxygen species (mROS) compared to the control, suppressing the intracellular survival of Mtb. We observed that siBNIP3 suppressed intracellular Mtb in mice lungs. CONCLUSION We found that BNIP3 plays a critical role in the regulation of mitophagy during Mtb infection. The inhibition of mitophagy suppresses Mtb growth in macrophages through increased mROS production. Therefore, BNIP3 might be a novel therapeutic target for tuberculosis treatment.
Collapse
Affiliation(s)
- Junghwan Lee
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, 34134, South Korea
| | - Seong-Ahn Lee
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Sang-Hun Son
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Ji-Ae Choi
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, 34134, South Korea
| | - Tam Doan Nguyen
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Jaewhan Kim
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Doyi Son
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Chang-Hwa Song
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea.
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea.
- Translational Immunology Institute, Chungnam National University, Daejeon, 34134, South Korea.
| |
Collapse
|
11
|
Bansal S, Burman A, Tripathi AK. Advanced glycation end products: Key mediator and therapeutic target of cardiovascular complications in diabetes. World J Diabetes 2023; 14:1146-1162. [PMID: 37664478 PMCID: PMC10473940 DOI: 10.4239/wjd.v14.i8.1146] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/21/2023] [Accepted: 05/22/2023] [Indexed: 08/11/2023] Open
Abstract
The incidence of type 2 diabetes mellitus is growing in epidemic proportions and has become one of the most critical public health concerns. Cardiovascular complications associated with diabetes are the leading cause of morbidity and mortality. The cardiovascular diseases that accompany diabetes include angina, myocardial infarction, stroke, peripheral artery disease, and congestive heart failure. Among the various risk factors generated secondary to hyperglycemic situations, advanced glycation end products (AGEs) are one of the important targets for future diagnosis and prevention of diabetes. In the last decade, AGEs have drawn a lot of attention due to their involvement in diabetic patho-physiology. AGEs can be derived exogenously and endogenously through various pathways. These are a non-homogeneous, chemically diverse group of compounds formed non-enzymatically by condensation between carbonyl groups of reducing sugars and free amino groups of protein, lipids, and nucleic acid. AGEs mediate their pathological effects at the cellular and extracellular levels by multiple pathways. At the cellular level, they activate signaling cascades via the receptor for AGEs and initiate a complex series of intracellular signaling resulting in reactive oxygen species generation, inflammation, cellular proliferation, and fibrosis that may possibly exacerbate the damaging effects on cardiac functions in diabetics. AGEs also cause covalent modifications and cross-linking of serum and extracellular matrix proteins; altering their structure, stability, and functions. Early diagnosis of diabetes may prevent its progression to complications and decrease its associated comorbidities. In the present review, we recapitulate the role of AGEs as a crucial mediator of hyperglycemia-mediated detrimental effects in diabetes-associated complications. Furthermore, this review presents an overview of future perspectives for new therapeutic interventions to ameliorate cardiovascular complications in diabetes.
Collapse
Affiliation(s)
- Savita Bansal
- Department of Biochemistry, Institute of Home Sciences, University of Delhi, New Delhi 110016, India
| | - Archana Burman
- Department of Biochemistry, Institute of Home Economics, University of Delhi, New Delhi 110016, India
| | - Asok Kumar Tripathi
- Department of Biochemistry, University College of Medical Sciences, University of Delhi, New Delhi 110095, India
| |
Collapse
|
12
|
Mitochondrial Dysfunction: The Hidden Player in the Pathogenesis of Atherosclerosis? Int J Mol Sci 2023; 24:ijms24021086. [PMID: 36674602 PMCID: PMC9861427 DOI: 10.3390/ijms24021086] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Atherosclerosis is a multifactorial inflammatory pathology that involves metabolic processes. Improvements in therapy have drastically reduced the prognosis of cardiovascular disease. Nevertheless, a significant residual risk is still relevant, and is related to unmet therapeutic targets. Endothelial dysfunction and lipid infiltration are the primary causes of atherosclerotic plaque progression. In this contest, mitochondrial dysfunction can affect arterial wall cells, in particular macrophages, smooth muscle cells, lymphocytes, and endothelial cells, causing an increase in reactive oxygen species (ROS), leading to oxidative stress, chronic inflammation, and intracellular lipid deposition. The detection and characterization of mitochondrial DNA (mtDNA) is crucial for assessing mitochondrial defects and should be considered the goal for new future therapeutic interventions. In this review, we will focus on a new idea, based on the analysis of data from many research groups, namely the link between mitochondrial impairment and endothelial dysfunction and, in particular, its effect on atherosclerosis and aging. Therefore, we discuss known and novel mitochondria-targeting therapies in the contest of atherosclerosis.
Collapse
|
13
|
Karnewar S, Pulipaka S, Katta S, Panuganti D, Neeli PK, Thennati R, Jerald MK, Kotamraju S. Mitochondria-targeted esculetin mitigates atherosclerosis in the setting of aging via the modulation of SIRT1-mediated vascular cell senescence and mitochondrial function in Apoe mice. Atherosclerosis 2022; 356:28-40. [DOI: 10.1016/j.atherosclerosis.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022]
|
14
|
Erdmann J, Kujaciński M, Wiciński M. Beneficial Effects of Ursolic Acid and Its Derivatives-Focus on Potential Biochemical Mechanisms in Cardiovascular Conditions. Nutrients 2021; 13:3900. [PMID: 34836155 PMCID: PMC8622438 DOI: 10.3390/nu13113900] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
Ursolic acid (UA) is a natural pentacyclic triterpenoid found in a number of plants such as apples, thyme, oregano, hawthorn and others. Several in vitro and in vivo studies have presented its anti-inflammatory and anti-apoptotic properties. The inhibition of NF-κB-mediated inflammatory pathways and the increased scavenging of reactive oxygen species (ROS) in numerous ways seem to be the most beneficial effects of UA. In mice and rats, administration of UA appears to slow down the development of cardiovascular diseases (CVDs), especially atherosclerosis and cardiac fibrosis. Upregulation of endothelial-type nitric oxide synthase (eNOS) and cystathionine-λ-lyase (CSE) by UA may suggest its vasorelaxant property. Inhibition of metalloproteinases activity by UA may contribute to better outcomes in aneurysms management. UA influence on lipid and glucose metabolism remains inconsistent, and additional studies are essential to verify its efficacy. Furthermore, UA derivatives appear to have a beneficial impact on the cardiovascular system. This review aims to summarize recent findings on beneficial effects of UA that may make it a promising candidate for clinical trials for the management of CVDs.
Collapse
Affiliation(s)
- Jakub Erdmann
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland; (M.K.); (M.W.)
| | | | | |
Collapse
|
15
|
Ding YG, Zhao YL, Zhang J, Zuo ZT, Zhang QZ, Wang YZ. The traditional uses, phytochemistry, and pharmacological properties of Paris L. (Liliaceae): A review. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114293. [PMID: 34102270 DOI: 10.1016/j.jep.2021.114293] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Paris L. (Liliaceae) consisted of 33 species, of which the study focused on Paris polyphylla Smith, P. polyphylla var. chinensis (Franch.) Hara, and P. polyphylla Smith var. yunnanensis (Franch.) Hand. -Mazz. Due of course to the good effects of analgesia and hemostasis, it was traditionally used to treat trauma by folk herbalists. AIM OF THIS REVIEW This study summarized the traditional uses, distributions, phytochemical components, pharmacological properties, and toxicity evaluation of the genus Paris, and reviewed the economic value of cultivate P. polyphylla. This aim was that of providing a new and comprehensive recognition of these medicinal plants for the further utilization of Paris plants. MATERIALS AND METHODS The literature about traditional and folk uses of genus Paris was obtained from Duxiu Search, and China National Knowledge Infrastructure (CNKI). The other literature about genus Paris was searched online on Web of Science, PubMed, Google Scholar, Baidu Scholar, Scifinder database, and Springer research. The Scientific Database of China Plant Species (DCP) (http://db.kib.ac.cn/Default.aspx) databases were used to check the scientific names and provide species, varieties, and distribution of genus Paris. The botany studies information of genus Paris was available online from Plant Plus of China (www.iplant.cn). All the molecular structures of chemical compounds displayed in the text were produced by ChemBioDraw Ultra 14.0. RESULTS The plants of genus Paris, containing about 33 species and 15 varieties, are mainly distributed in Southwest China (Yunnan, Sichuan, and Guizhou provinces). More than 320 chemical components have been isolated from genus Paris since 2020, including steroidal saponins, C-21 steroids, phytosterols, insect hormones, pentacyclic triterpenes, flavonoids, and other compounds. Arrays of pharmacological investigations revealed that compounds and extracts of Paris species possess a wide spectrum of pharmacological effects, such as antitumor, cytotoxic, antimicrobial, antifungal, hemostatic, and anti-inflammatory activities. The studies about toxicity evaluation suggested that Rhizome Paridis had slight liver toxicity. CONCLUSIONS The dried rhizomes of P. polyphylla, P. polyphylla var. chinensis, and P. polyphylla var. yunnanensis were used to treat wound, bleeding, and stomachache, etc. in folk medicine. Phytochemistry researches showed that different species had pretty similarities especially in terms of chemical constituents. Pharmacological studies witnessed that Rhizome Paridis has various activities. Among these activities, steroidal saponins were the main active ingredients. Furthermore, an important aspect responsible for increasing interest in genus Paris is the use of antifertility-nonhormonal contraceptives by women. Also, the development of TCM (Traditional Chinese medicine) planting industry can improve the income of ethnic minorities and promote economic development.
Collapse
Affiliation(s)
- Yu-Gang Ding
- Medicinal Plants Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, China; College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Yan-Li Zhao
- Medicinal Plants Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, China
| | - Ji Zhang
- Medicinal Plants Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, China
| | - Zhi-Tian Zuo
- Medicinal Plants Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, China
| | - Qing-Zhi Zhang
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Yuan-Zhong Wang
- Medicinal Plants Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, China.
| |
Collapse
|
16
|
Plazas Guerrero CG, Acosta Cota SDJ, Castro Sánchez FH, Vergara Jiménez MDJ, Ríos Burgueño ER, Sarmiento Sánchez JI, Picos Corrales LA, Osuna Martínez U. Evaluation of sucrose-enriched diet consumption in the development of risk factors associated to type 2 diabetes, atherosclerosis and non-alcoholic fatty liver disease in a murine model. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2021; 31:651-669. [PMID: 31668088 DOI: 10.1080/09603123.2019.1680817] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/09/2019] [Indexed: 06/10/2023]
Abstract
Overconsumption of sucrose, the main contributor of the total added sugar intake in the world, has been associated with negative metabolic effects related to non-communicable diseases. However, this relationship continues to be a controversial topic and further studies are needed. The aim of this study was to evaluate the sucrose-enriched diet consumption in the development of risk factors associated with type 2 diabetes, atherosclerosis and non-alcoholic fatty liver disease in a murine model. Sucrose-enriched diet-fed rats showed a decrease in food, lipids and protein intake as well as in serum total cholesterol levels, an increase in carbohydrates intake, glucose, insulin, triglycerides, VLDL-c and HDL-c levels and a greater degree of insulin resistance, steatosis and non-alcoholic steatohepatitis. Our results show that sucrose-enriched diet consumption during 25 weeks contribute to the development of risk factors associated with type 2 diabetes, atherosclerosis and non-alcoholic fatty liver disease in male Wistar rats.
Collapse
Affiliation(s)
| | | | | | | | - Efrén Rafael Ríos Burgueño
- Centro de Investigación y Docencia en Ciencias de la Salud, Universidad Autónoma de Sinaloa, Culiacán de Rosales, Mexico
| | | | | | - Ulises Osuna Martínez
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán de Rosales, Mexico
| |
Collapse
|
17
|
Kunz HE, Hart CR, Gries KJ, Parvizi M, Laurenti M, Dalla Man C, Moore N, Zhang X, Ryan Z, Polley EC, Jensen MD, Vella A, Lanza IR. Adipose tissue macrophage populations and inflammation are associated with systemic inflammation and insulin resistance in obesity. Am J Physiol Endocrinol Metab 2021; 321:E105-E121. [PMID: 33998291 PMCID: PMC8321823 DOI: 10.1152/ajpendo.00070.2021] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity is accompanied by numerous systemic and tissue-specific derangements, including systemic inflammation, insulin resistance, and mitochondrial abnormalities in skeletal muscle. Despite growing recognition that adipose tissue dysfunction plays a role in obesity-related disorders, the relationship between adipose tissue inflammation and other pathological features of obesity is not well-understood. We assessed macrophage populations and measured the expression of inflammatory cytokines in abdominal adipose tissue biopsies in 39 nondiabetic adults across a range of body mass indexes (BMI 20.5-45.8 kg/m2). Skeletal muscle biopsies were used to evaluate mitochondrial respiratory capacity, ATP production capacity, coupling, and reactive oxygen species production. Insulin sensitivity (SI) and β cell responsivity were determined from test meal postprandial glucose, insulin, c-peptide, and triglyceride kinetics. We examined the relationships between adipose tissue inflammatory markers, systemic inflammatory markers, SI, and skeletal muscle mitochondrial physiology. BMI was associated with increased adipose tissue and systemic inflammation, reduced SI, and reduced skeletal muscle mitochondrial oxidative capacity. Adipose-resident macrophage numbers were positively associated with circulating inflammatory markers, including tumor necrosis factor-α (TNFα) and C-reactive protein (CRP). Local adipose tissue inflammation and circulating concentrations of TNFα and CRP were negatively associated with SI, and circulating concentrations of TNFα and CRP were also negatively associated with skeletal muscle oxidative capacity. These results demonstrate that obese humans exhibit increased adipose tissue inflammation concurrently with increased systemic inflammation, reduced insulin sensitivity, and reduced muscle oxidative capacity and suggest that adipose tissue and systemic inflammation may drive obesity-associated metabolic derangements.NEW AND NOTEWORTHY Adipose inflammation is proposed to be at the nexus of the systemic inflammation and metabolic derangements associated with obesity. The present study provides evidence to support adipose inflammation as a central feature of the pathophysiology of obesity. Adipose inflammation is associated with systemic and peripheral metabolic derangements, including increased systemic inflammation, reduced insulin sensitivity, and reduced skeletal muscle mitochondrial respiration.
Collapse
Affiliation(s)
- Hawley E Kunz
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Corey R Hart
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kevin J Gries
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Mojtaba Parvizi
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Marcello Laurenti
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Chiara Dalla Man
- Biomedical Engineering and Physiology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota
| | - Natalie Moore
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Xiaoyan Zhang
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Zachary Ryan
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Eric C Polley
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Michael D Jensen
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Adrian Vella
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Ian R Lanza
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
18
|
Mitochondrial DAMPs and altered mitochondrial dynamics in OxLDL burden in atherosclerosis. Mol Cell Biochem 2021; 476:1915-1928. [PMID: 33492610 DOI: 10.1007/s11010-021-04061-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/11/2021] [Indexed: 10/22/2022]
Abstract
Atherosclerosis results in life-threatening cardiovascular pathologies, including ischemic heart disease, stroke, myocardial infarction, and peripheral arterial disease. The role of increased serum low-density lipoprotein (LDL) and resultant accumulation of oxidized-LDL (oxLDL) in atheroma formation is well established. Recent findings elucidate the significance of mitochondrial damage-associated molecular patterns (mtDAMPs) in triggering sterile inflammation in concert with oxLDL. The mtDAMPs including mitochondrial DNA (mtDNA), cytochrome C, cardiolipin, heat shock protein 60 (HSP60), mitochondrial transcription factor A (TFAM), and N-formyl peptides, are expected to possess proatherogenic roles. However, limited data are available in the literature. The mtDAMPs initiate sterile inflammation in atherosclerotic lesions via numerous signaling pathways, most of which converge to the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome. Priming the activation of the NLRP3 inflammasome, mtDAMPs promote secretion of proinflammatory cytokines, including interleukin-1β (IL-1β), implicated in atherosclerotic lesions through vascular smooth muscle and fibroblast proliferation, arterial wall thickening, and plaque formation. In this article we critically reviewed and discussed the central role of the NLRP3 inflammasome in mtDAMP-induced sterile inflammation in atherosclerosis with specific components including caspase-1, pregnane X receptor (PXR), adenosine monophosphate activated protein kinase (AMPK), protein phosphatase 2A (PP2A), thioredoxin-interacting protein (TXNIP), and downstream cytokines including IL-1β and IL-18 as potential mediators of atherosclerosis. Better understanding of the proinflammatory effects of mtDAMPs and its pathological association with oxLDL possess immense translational significance for novel therapeutic intervention.
Collapse
|
19
|
Chang R, Mamun A, Dominic A, Le NT. SARS-CoV-2 Mediated Endothelial Dysfunction: The Potential Role of Chronic Oxidative Stress. Front Physiol 2021; 11:605908. [PMID: 33519510 PMCID: PMC7844210 DOI: 10.3389/fphys.2020.605908] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/09/2020] [Indexed: 01/08/2023] Open
Abstract
Endothelial cells have emerged as key players in SARS-CoV-2 infection and COVID-19 inflammatory pathologies. Dysfunctional endothelial cells can promote chronic inflammation and disease processes like thrombosis, atherosclerosis, and lung injury. In endothelial cells, mitochondria regulate these inflammatory pathways via redox signaling, which is primarily achieved through mitochondrial reactive oxygen species (mtROS). Excess mtROS causes oxidative stress that can initiate and exacerbate senescence, a state that promotes inflammation and chronic endothelial dysfunction. Oxidative stress can also activate feedback loops that perpetuate mitochondrial dysfunction, mtROS overproduction, and inflammation. In this review, we provide an overview of phenotypes mediated by mtROS in endothelial cells - such as mitochondrial dysfunction, inflammation, and senescence - as well as how these chronic states may be initiated by SARS-CoV-2 infection of endothelial cells. We also propose that SARS-CoV-2 activates mtROS-mediated feedback loops that cause long-term changes in host redox status and endothelial function, promoting cardiovascular disease and lung injury after recovery from COVID-19. Finally, we discuss the implications of these proposed pathways on long-term vascular health and potential treatments to address these chronic conditions.
Collapse
Affiliation(s)
- Ryan Chang
- College of Arts & Sciences, Washington University in St. Louis, St. Louis, MO, United States
| | - Abrar Mamun
- Wiess School of Natural Sciences, Rice University, Houston, TX, United States
| | - Abishai Dominic
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX, United States
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
20
|
Galectin-1 is associated with the severity of coronary artery disease and adverse cardiovascular events in patients undergoing coronary angiography. Sci Rep 2020; 10:20683. [PMID: 33244142 PMCID: PMC7692553 DOI: 10.1038/s41598-020-77804-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 11/02/2020] [Indexed: 12/02/2022] Open
Abstract
Galectin-1, a β-galactoside-binding lectin mediating inflammation and neovascularization, is reported to attenuate ventricular remodeling after myocardial infarction. But its role in stable coronary artery disease (CAD) has not been fully elucidated. This study aimed to identify the relationship between the circulating galectin-1 level and the severity of CAD in patients with suspected CAD. Pre-procedure galectin-1 and high-sensitivity C-reactive protein (hs-CRP) concentrations were measured in 834 subjects who underwent scheduled coronary angiography. Subjects were grouped into tertiles of the galectin-1 levels. SYNTAX scores were calculated to evaluate the severity of CAD. All patients were followed until January 2019 or the occurrence of major adverse cardiovascular events (MACE). Patients with higher galectin-1 concentrations were older; had greater prevalence of hypertension, diabetes, chronic kidney disease, and heart failure; and were more likely to present with higher hs-CRP levels and SYNTAX scores. During the follow-up period of 1.3 ± 1.1 years, patients in the highest tertile of galectin-1 were associated with a greater risk of MACE after adjustment for age, sex, comorbidities, co-medications, serum levels of hemoglobin, creatinine, hs-CRP, ejection fraction, SYNTAX scores, and revascularization modalities (adjusted hazard ratio 10.95, 95% confidence interval 2.29–52.47, p = 0.003). Galectin-1 showed better discriminatory performance than hs-CRP, and non-inferior performance to SYNTAX scores, in predicting the incidence of MACE.
Collapse
|
21
|
Brand MD. Riding the tiger - physiological and pathological effects of superoxide and hydrogen peroxide generated in the mitochondrial matrix. Crit Rev Biochem Mol Biol 2020; 55:592-661. [PMID: 33148057 DOI: 10.1080/10409238.2020.1828258] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Elevated mitochondrial matrix superoxide and/or hydrogen peroxide concentrations drive a wide range of physiological responses and pathologies. Concentrations of superoxide and hydrogen peroxide in the mitochondrial matrix are set mainly by rates of production, the activities of superoxide dismutase-2 (SOD2) and peroxiredoxin-3 (PRDX3), and by diffusion of hydrogen peroxide to the cytosol. These considerations can be used to generate criteria for assessing whether changes in matrix superoxide or hydrogen peroxide are both necessary and sufficient to drive redox signaling and pathology: is a phenotype affected by suppressing superoxide and hydrogen peroxide production; by manipulating the levels of SOD2, PRDX3 or mitochondria-targeted catalase; and by adding mitochondria-targeted SOD/catalase mimetics or mitochondria-targeted antioxidants? Is the pathology associated with variants in SOD2 and PRDX3 genes? Filtering the large literature on mitochondrial redox signaling using these criteria highlights considerable evidence that mitochondrial superoxide and hydrogen peroxide drive physiological responses involved in cellular stress management, including apoptosis, autophagy, propagation of endoplasmic reticulum stress, cellular senescence, HIF1α signaling, and immune responses. They also affect cell proliferation, migration, differentiation, and the cell cycle. Filtering the huge literature on pathologies highlights strong experimental evidence that 30-40 pathologies may be driven by mitochondrial matrix superoxide or hydrogen peroxide. These can be grouped into overlapping and interacting categories: metabolic, cardiovascular, inflammatory, and neurological diseases; cancer; ischemia/reperfusion injury; aging and its diseases; external insults, and genetic diseases. Understanding the involvement of mitochondrial matrix superoxide and hydrogen peroxide concentrations in these diseases can facilitate the rational development of appropriate therapies.
Collapse
|
22
|
Venil CK, Dufossé L, Renuka Devi P. Bacterial Pigments: Sustainable Compounds With Market Potential for Pharma and Food Industry. FRONTIERS IN SUSTAINABLE FOOD SYSTEMS 2020. [DOI: 10.3389/fsufs.2020.00100] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
23
|
Shemiakova T, Ivanova E, Grechko AV, Gerasimova EV, Sobenin IA, Orekhov AN. Mitochondrial Dysfunction and DNA Damage in the Context of Pathogenesis of Atherosclerosis. Biomedicines 2020; 8:E166. [PMID: 32570831 PMCID: PMC7344998 DOI: 10.3390/biomedicines8060166] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/13/2020] [Accepted: 06/16/2020] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a multifactorial disease of the cardiovascular system associated with aging, inflammation, and oxidative stress. An important role in the development of atherosclerosis play elevated plasma lipoproteins. A number of external factors (smoking, diabetes, infections) can also contribute to the development of the disease. For a long time, atherosclerosis remains asymptomatic, therefore, the search for early markers of the disease is critical for the timely management and better outcomes for patients. Mitochondrial dysfunction and mitochondrial DNA (mtDNA) damage appear to connect different aspects of atherosclerosis pathogenesis. To date, multiple lines of research have demonstrated the strong association of mitochondrial dysfunction with the development of various human diseases. Therapies aimed at restoring the mitochondrial function are being actively developed, and are expected to broaden the therapeutic possibilities for several chronic human diseases. The development of such therapies depends on our understanding of the functional roles of different mtDNA variants associated with one or another disorder, and the molecular mechanisms linking mitochondrial dysfunction with a given pathological feature. These questions are, however, challenging and require future intensive research. This review summarizes the recent studies and describes the central processes of the development of atherosclerosis, and shows their relationship with mitochondrial dysfunction. One of the promising therapeutic approaches for future atherosclerosis treatments is the use of mitochondria-targeted antioxidants. Future studies should focus on characterizing the mechanisms of mitochondrial involvement in cardiovascular pathologies to better direct the search for novel therapies.
Collapse
Affiliation(s)
- Taisiia Shemiakova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia;
| | - Ekaterina Ivanova
- Department of Basic Research, Institute for Atherosclerosis Research, 121609 Moscow, Russia
| | - Andrey V. Grechko
- Federal Scientific Clinical Center for Resuscitation and Rehabilitation, 109240 Moscow, Russia;
| | - Elena V. Gerasimova
- Laboratory of Systemic Rheumatic Disorders, V.A. Nasonova Institute of Rheumatology, 115522 Moscow, Russia;
| | - Igor A. Sobenin
- Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 121552 Moscow, Russia;
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Laboratory of Infection Pathology and Molecular Microecology, Institute of Human Morphology, 117418 Moscow, Russia
| |
Collapse
|
24
|
Dias DDS, Bernardes N, Stoyell-Conti FF, dos Santos CP, de Araujo AA, Llesuy S, Irigoyen MC, De Angelis K. Impact of combined exercise training on the development of cardiometabolic and neuroimmune complications induced by fructose consumption in hypertensive rats. PLoS One 2020; 15:e0233785. [PMID: 32521542 PMCID: PMC7286703 DOI: 10.1371/journal.pone.0233785] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 05/12/2020] [Indexed: 01/19/2023] Open
Abstract
This study evaluated the impact of combined exercise training on the development of cardiovascular and neuroimmune complications induced by fructose consumption (10% in the drinking water) in hypertensive rats (SHR). After weaning, SHR were divided into 3 groups: SHR (H), SHR+fructose (HF) and SHR+fructose+combined exercise training (treadmill+ladder, 40-60% of maximum capacity) (HFTC). Metabolic, hemodynamic, autonomic, inflammatory and oxidative stress parameters were evaluated in the subgroups (n = 6 group/time) at 7, 15, 30 and 60 days of protocol. Fructose consumption (H vs. HF groups) decreased spontaneous baroreflex sensitivity and total variance of pulse interval at day 7 (7 to 60); increased IL-6 and TNFα in the heart (at day 15, 30 and 60) and NADPH oxidase activity and cardiac lipoperoxidation (LPO) (day 60); increased white adipose tissue weight, reduced insulin sensitivity and increased triglycerides (day 60); induced an additional increase in mean arterial pressure (MAP) (days 30 and 60). Combined exercise training prevented such dysfunctions and sustained increased cardiac IL-10 (day 7) and glutathione redox balance (GSH/GSSG) for the entire protocol. In conclusion, combined exercise training performed simultaneously with exacerbated fructose consumption prevented early cardiovascular autonomic dysfunction, probably trigging positive changes in inflammation and oxidative stress, resulting in a better cardiometabolic profile in rats genetically predisposed to hypertension.
Collapse
Affiliation(s)
- Danielle da Silva Dias
- Laboratory of Translational Physiology, Universidade Nove de Julho (UNINOVE), Sao Paulo, Sao Paulo, Brazil
- Department of Physiology, Federal University of Sao Paulo (UNIFESP), Sao Paulo, Sao Paulo, Brazil
| | - Nathalia Bernardes
- Laboratory of Translational Physiology, Universidade Nove de Julho (UNINOVE), Sao Paulo, Sao Paulo, Brazil
| | | | - Camila Paixão dos Santos
- Department of Physiology, Federal University of Sao Paulo (UNIFESP), Sao Paulo, Sao Paulo, Brazil
| | | | - Susana Llesuy
- Laboratory of Translational Physiology, Universidade Nove de Julho (UNINOVE), Sao Paulo, Sao Paulo, Brazil
- Instituto Universitario Hospital Italiano, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Maria Cláudia Irigoyen
- Hypertension Unit, Heart Institute (InCor), School of Medicine, University of Sao Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Kátia De Angelis
- Laboratory of Translational Physiology, Universidade Nove de Julho (UNINOVE), Sao Paulo, Sao Paulo, Brazil
- Department of Physiology, Federal University of Sao Paulo (UNIFESP), Sao Paulo, Sao Paulo, Brazil
- * E-mail:
| |
Collapse
|
25
|
Flavones' and Flavonols' Antiradical Structure-Activity Relationship-A Quantum Chemical Study. Antioxidants (Basel) 2020; 9:antiox9060461. [PMID: 32471289 PMCID: PMC7346117 DOI: 10.3390/antiox9060461] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 01/07/2023] Open
Abstract
Flavonoids are known for their antiradical capacity, and this ability is strongly structure-dependent. In this research, the activity of flavones and flavonols in a water solvent was studied with the density functional theory methods. These included examination of flavonoids’ molecular and radical structures with natural bonding orbitals analysis, spin density analysis and frontier molecular orbitals theory. Calculations of determinants were performed: specific, for the three possible mechanisms of action—hydrogen atom transfer (HAT), electron transfer–proton transfer (ETPT) and sequential proton loss electron transfer (SPLET); and the unspecific—reorganization enthalpy (RE) and hydrogen abstraction enthalpy (HAE). Intramolecular hydrogen bonding, catechol moiety activity and the probability of electron density swap between rings were all established. Hydrogen bonding seems to be much more important than the conjugation effect, because some structures tends to form more intramolecular hydrogen bonds instead of being completely planar. The very first hydrogen abstraction mechanism in a water solvent is SPLET, and the most privileged abstraction site, indicated by HAE, can be associated with the C3 hydroxyl group of flavonols and C4’ hydroxyl group of flavones. For the catechol moiety, an intramolecular reorganization to an o-benzoquinone-like structure occurs, and the ETPT is favored as the second abstraction mechanism.
Collapse
|
26
|
MitoQ and CoQ10 supplementation mildly suppresses skeletal muscle mitochondrial hydrogen peroxide levels without impacting mitochondrial function in middle-aged men. Eur J Appl Physiol 2020; 120:1657-1669. [DOI: 10.1007/s00421-020-04396-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/16/2020] [Indexed: 12/14/2022]
|
27
|
Zakirov FH, Zhang D, Grechko AV, Wu WK, Poznyak AV, Orekhov AN. Lipid-based gene delivery to macrophage mitochondria for atherosclerosis therapy. Pharmacol Res Perspect 2020; 8:e00584. [PMID: 32237116 PMCID: PMC7111069 DOI: 10.1002/prp2.584] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis with associated cardiovascular diseases remains one of the main causes of disability and death worldwide, requiring development of new solutions for prevention and treatment. Macrophages are the key effectors of a series of events involved in atherogenesis, such as inflammation, plaque formation, and changes in lipid metabolism. Some of these events were shown to be associated with mitochondrial dysfunction and excessive mitochondrial DNA (mtDNA) damage. Moreover, macrophages represent a promising target for novel therapeutic approaches that are based on the expression of various receptors and nanoparticle uptake. Lipid-based gene delivery to mitochondria is considered to be an interesting strategy for mtDNA damage correction. To date, several nanocarriers and their modifications have been developed that demonstrate high transfection efficiency and low cytotoxicity. This review discusses the possibilities of lipid-based gene delivery to macrophage mitochondria for atherosclerosis therapy.
Collapse
Affiliation(s)
- Felix H Zakirov
- I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Dongwei Zhang
- Diabetes Research Center, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Andrey V Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow, Russian Federation
| | - Wei-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Anastasia V Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Alexander N Orekhov
- Institute of Human Morphology, Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
28
|
Lemjallad L, Chabir R, Kandri Rodi Y, El Ghadraoui L, Ouazzani Chahdi F, Errachidi F. Improvement of Heliciculture by Three Medicinal Plants Belonging to the Lamiaceae Family. ScientificWorldJournal 2019; 2019:2630537. [PMID: 31885523 PMCID: PMC6925714 DOI: 10.1155/2019/2630537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/11/2019] [Indexed: 12/01/2022] Open
Abstract
Snails were fed with three medicinal plants belonging to the Lamiaceae family (rosemary, sage, and peppermint) in order to test their effects on those animals with high nutritive values. The media of raising were flour containing different percentages of the cited plants ranging from 1% to 9%. The feed had benefits on the raised snails depending on the plant and its percentage. Minerals in those aromatic plants, especially zinc and magnesium, had their effect on protein synthesis in snails fed with those plant percentages. Rosemary was the most profitable plant with the highest protein amount, the lowest mortality rate, and reduced microbial charge. Furthermore, it was a good regulator of the specific catalase activity which confirmed the role of the antioxidant activity of rosemary during raising snails.
Collapse
Affiliation(s)
- Lamiaa Lemjallad
- Department of Chemistry, Laboratory of Applied Organic Chemistry, Faculty of Sciences and Technologies, University Sidi Mohamed Ben Abdellah, B.P. 2202-route Imouzzer, Fez, Morocco
- Team of Nutrition, Agri-Food and Environment, Laboratory of Human Pathology, Biomedicine and Environment, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, P.B. 1893 km 2200 Road Sidi Harazem, Fez, Morocco
| | - Rachida Chabir
- Team of Nutrition, Agri-Food and Environment, Laboratory of Human Pathology, Biomedicine and Environment, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, P.B. 1893 km 2200 Road Sidi Harazem, Fez, Morocco
| | - Youssef Kandri Rodi
- Department of Chemistry, Laboratory of Applied Organic Chemistry, Faculty of Sciences and Technologies, University Sidi Mohamed Ben Abdellah, B.P. 2202-route Imouzzer, Fez, Morocco
| | - Lahssen El Ghadraoui
- Laboratory of Functional Ecology and Environment, Faculty of Sciences and Technologies, University Sidi Mohamed Ben Abdellah, B.P. 2202-route Imouzzer, Fez, Morocco
| | - Fouad Ouazzani Chahdi
- Department of Chemistry, Laboratory of Applied Organic Chemistry, Faculty of Sciences and Technologies, University Sidi Mohamed Ben Abdellah, B.P. 2202-route Imouzzer, Fez, Morocco
| | - Faouzi Errachidi
- Laboratory of Functional Ecology and Environment, Faculty of Sciences and Technologies, University Sidi Mohamed Ben Abdellah, B.P. 2202-route Imouzzer, Fez, Morocco
| |
Collapse
|
29
|
The effects of antibiotics and melatonin on hepato-intestinal inflammation and gut microbial dysbiosis induced by a short-term high-fat diet consumption in rats. Br J Nutr 2019; 122:841-855. [PMID: 31217044 DOI: 10.1017/s0007114519001466] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
High-fat diet (HFD) consumption leads to metabolic disorders, gastrointestinal dysfunction and intestinal dysbiosis. Antibiotics also disrupt the composition of intestinal microbiota. The aim of the present study was to investigate the impact of a short-term feeding with HFD on oxidative status, enteric microbiota, intestinal motility and the effects of antibiotics and/or melatonin treatments on diet-induced hepato-intestinal dysfunction and inflammation. Male Sprague-Dawley rats were pair-fed with either standard chow or HFD (45 % fat) and were given tap water or melatonin (4 mg/kg per d) or melatonin plus antibiotics (ABX; neomycin, ampicillin, metronidazole; each 1 g/l) in drinking water for 2 weeks. On the 14th day, colonic motility was measured and the next day intestinal transit was assessed using charcoal propagation. Trunk blood, liver and intestine samples were removed for biochemical and histopathological evaluations, and faeces were collected for microbiota analysis. A 2-week HFD feeding increased blood glucose level and perirenal fat weight, induced low-level hepatic and intestinal inflammation, delayed intestinal transit, led to deterioration of epithelial tight junctions and overgrowth of colonic bacteria. Melatonin intake in HFD-fed rats reduced ileal inflammation, colonic motility and perirenal fat accumulation. ABX abolished increases in fat accumulation and blood glucose, reduced ileal oxidative damage, suppressed HFD-induced overgrowth in colonic bacteria, and reversed HFD-induced delay in intestinal transit; however, hepatic neutrophil accumulation, hepatic injury and dysfunction were further enhanced. In conclusion, the results demonstrate that even a short-term HFD ingestion results in hepato-intestinal inflammatory state and alterations in bacterial populations, which may be worsened with antibiotic intake, but alleviated by melatonin.
Collapse
|
30
|
Sobenin IA, Zhelankin AV, Khasanova ZB, Sinyov VV, Medvedeva LV, Sagaidak MO, Makeev VJ, Kolmychkova KI, Smirnova AS, Sukhorukov VN, Postnov AY, Grechko AV, Orekhov AN. Heteroplasmic Variants of Mitochondrial DNA in Atherosclerotic Lesions of Human Aortic Intima. Biomolecules 2019; 9:biom9090455. [PMID: 31500189 PMCID: PMC6770808 DOI: 10.3390/biom9090455] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial dysfunction and oxidative stress are likely involved in atherogenesis. Since the mitochondrial genome variation can alter functional activity of cells, it is necessary to assess the presence in atherosclerotic lesions of mitochondrial DNA (mtDNA) heteroplasmic mutations known to be associated with different pathological processes and ageing. In this study, mtDNA heteroplasmy and copy number (mtCN) were evaluated in the autopsy-derived samples of aortic intima differing by the type of atherosclerotic lesions. To detect mtDNA heteroplasmic variants, next generation sequencing was used, and mtCN measurement was performed by qPCR. It was shown that mtDNA heteroplasmic mutations are characteristic for particular areas of intimal tissue; in 83 intimal samples 55 heteroplasmic variants were found; mean minor allele frequencies level accounted for 0.09, with 12% mean heteroplasmy level. The mtCN variance measured in adjacent areas of intima was high, but atherosclerotic lesions and unaffected intima did not differ significantly in mtCN values. Basing on the ratio of minor and major nucleotide mtDNA variants, we can conclude that there exists the increase in the number of heteroplasmic mtDNA variants, which corresponds to the extent of atherosclerotic morphologic phenotype.
Collapse
Affiliation(s)
- Igor A Sobenin
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 121552 Moscow, Russia.
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia.
- Research Institute of Threpsology and Healthy Longevity, Plekhanov Russian University of Economics, 115093 Moscow, Russia.
| | - Andrey V Zhelankin
- Federal Research and Clinical Center of Physical-Chemical Medicine, 119435 Moscow, Russia.
| | - Zukhra B Khasanova
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 121552 Moscow, Russia.
| | - Vasily V Sinyov
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 121552 Moscow, Russia.
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia.
| | - Lyudmila V Medvedeva
- Federal Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia.
| | - Maria O Sagaidak
- Vavilov Institute of General Genetics, 117971 Moscow, Russia.
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701 Moscow Region, Russia.
| | - Vsevolod J Makeev
- Vavilov Institute of General Genetics, 117971 Moscow, Russia.
- Moscow Institute of Physics and Technology, Dolgoprudny, 141701 Moscow Region, Russia.
- Engelhardt Institute of Molecular Biology, 119991 Moscow, Russia.
| | - Kira I Kolmychkova
- Institute for Atherosclerosis Research, Skolkovo Innovation Center, 143026 Moscow, Russia.
| | - Anna S Smirnova
- Institute for Atherosclerosis Research, Skolkovo Innovation Center, 143026 Moscow, Russia.
| | - Vasily N Sukhorukov
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 121552 Moscow, Russia.
- Research Institute of Human Morphology, 117418 Moscow, Russia.
| | - Anton Y Postnov
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 121552 Moscow, Russia.
- Research Institute of Human Morphology, 117418 Moscow, Russia.
| | - Andrey V Grechko
- Federal Scientific Clinical Center for Resuscitation and Rehabilitation, 141534 Moscow Region, Russia.
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovation Center, 143026 Moscow, Russia.
- Research Institute of Human Morphology, 117418 Moscow, Russia.
| |
Collapse
|
31
|
de Souza RGM, Gomes AC, Navarro AM, Cunha LCD, Silva MAC, Junior FB, Mota JF. Baru Almonds Increase the Activity of Glutathione Peroxidase in Overweight and Obese Women: A Randomized, Placebo-Controlled Trial. Nutrients 2019; 11:E1750. [PMID: 31366053 PMCID: PMC6723341 DOI: 10.3390/nu11081750] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/20/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Obesity-induced inflammation is frequently associated with higher oxidative stress. In vitro and experimental studies have considered baru almonds (Dipteryx alata Vog) as a legume seed with high antioxidant capacity. The aim of this study was to evaluate whether baru almonds are capable of improving the inflammatory and antioxidant status in overweight and obese women. METHODS In a parallel-arm, randomized placebo-controlled trial, 46 overweight and obese women (age: 40 ± 11 years; body mass index: 33.3 ± 4.3) were randomly assigned to receive advice to follow a normocaloric and isoenergetic diet with placebo (PLA, n = 22) or similar advice plus 20 g baru almonds (BARU, n = 24) for 8 wk. Malondialdehyde (MDA), adiponectin, tumor necrosis factor-α, interleukin-6, interleukin-10, antioxidant enzymes activities (catalase-CAT; glutathione peroxidase-GPx; superoxide dismutase-SOD), and minerals were analyzed in plasma samples. RESULTS At baseline, groups were similar regarding the body composition, oxidative, and inflammatory parameters. The BARU group increased the activity of GPx (+0.08 U/mg, 95%CI + 0.05 to +0.12 vs. -0.07, 95%CI -0.12 to -0.03, p < 0.01) and plasma copper concentration (p = 0.037) when compared to the PLA group. No differences were observed between groups in CAT and SOD activity or MDA and cytokines concentrations. CONCLUSIONS Baru almond supplementation increased the GPx activity in overweight and obese women.
Collapse
Affiliation(s)
- Rávila Graziany Machado de Souza
- Clinical and Sports Nutrition Research Laboratory (LABINCE), Faculty of Nutrition, Federal University of Goias-UFG, Goiânia, 74605-080 GO, Brazil
| | - Aline Corado Gomes
- Clinical and Sports Nutrition Research Laboratory (LABINCE), Faculty of Nutrition, Federal University of Goias-UFG, Goiânia, 74605-080 GO, Brazil
| | - Anderson Marliere Navarro
- Department of Health Sciences; Faculty of Medicine, University of São Paulo-USP, Ribeirão Preto, 14049-900 SP, Brazil
| | - Luiz Carlos da Cunha
- Nucleus of Toxic-pharmacological Studies and Research (NEPET), Federal University of Goiás-UFG, Goiânia, 74605-220 GO, Brazil
| | - Marina Alves Coelho Silva
- Nucleus of Toxic-pharmacological Studies and Research (NEPET), Federal University of Goiás-UFG, Goiânia, 74605-220 GO, Brazil
| | - Fernando Barbosa Junior
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-FCRP/USP, Ribeirão Preto, 14040-903 SP, Brazil
| | - João Felipe Mota
- Clinical and Sports Nutrition Research Laboratory (LABINCE), Faculty of Nutrition, Federal University of Goias-UFG, Goiânia, 74605-080 GO, Brazil.
| |
Collapse
|
32
|
Xia W, Li Y, Wu M, Yin J, Zhang Y, Chen H, Huang S, Jia Z, Zhang A. Inhibition of mitochondrial activity ameliorates atherosclerosis in ApoE
−/−
mice via suppressing vascular smooth cell activation and macrophage foam cell formation. J Cell Biochem 2019; 120:17767-17778. [PMID: 31131474 DOI: 10.1002/jcb.29042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 05/05/2019] [Accepted: 05/07/2019] [Indexed: 11/10/2022]
Affiliation(s)
- Weiwei Xia
- Department of Nephrology Children's Hospital of Nanjing Medical University Nanjing China
- Jiangsu Key Laboratory of Pediatrics Nanjing Medical University Nanjing China
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University Nanjing China
- Department of Clinical Laboratory Children's Hospital of Nanjing Medical University Nanjing China
| | - Yuanyuan Li
- Department of Nephrology Children's Hospital of Nanjing Medical University Nanjing China
- Jiangsu Key Laboratory of Pediatrics Nanjing Medical University Nanjing China
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University Nanjing China
| | - Mengying Wu
- Department of Nephrology Children's Hospital of Nanjing Medical University Nanjing China
- Jiangsu Key Laboratory of Pediatrics Nanjing Medical University Nanjing China
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University Nanjing China
| | - Jie Yin
- Department of Nephrology Children's Hospital of Nanjing Medical University Nanjing China
- Jiangsu Key Laboratory of Pediatrics Nanjing Medical University Nanjing China
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University Nanjing China
| | - Yue Zhang
- Department of Nephrology Children's Hospital of Nanjing Medical University Nanjing China
- Jiangsu Key Laboratory of Pediatrics Nanjing Medical University Nanjing China
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University Nanjing China
| | - Hongbing Chen
- Department of Clinical Laboratory Children's Hospital of Nanjing Medical University Nanjing China
| | - Songming Huang
- Department of Nephrology Children's Hospital of Nanjing Medical University Nanjing China
- Jiangsu Key Laboratory of Pediatrics Nanjing Medical University Nanjing China
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University Nanjing China
| | - Zhanjun Jia
- Department of Nephrology Children's Hospital of Nanjing Medical University Nanjing China
- Jiangsu Key Laboratory of Pediatrics Nanjing Medical University Nanjing China
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University Nanjing China
| | - Aihua Zhang
- Department of Nephrology Children's Hospital of Nanjing Medical University Nanjing China
- Jiangsu Key Laboratory of Pediatrics Nanjing Medical University Nanjing China
- Nanjing Key Laboratory of Pediatrics Children's Hospital of Nanjing Medical University Nanjing China
| |
Collapse
|
33
|
Jin H, Ko YS, Park SW, Kim HJ. P2Y 2R activation by ATP induces oxLDL-mediated inflammasome activation through modulation of mitochondrial damage in human endothelial cells. Free Radic Biol Med 2019; 136:109-117. [PMID: 30959169 DOI: 10.1016/j.freeradbiomed.2019.04.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/22/2019] [Accepted: 04/01/2019] [Indexed: 11/30/2022]
Abstract
Oxidative stress and the related inflammatory responses are closely associated with many diseases including cardiovascular diseases such as atherosclerosis. Especially, mitochondrial damage and inflammasome activation have been reported to be directly involved in atherogenesis. In addition, we previously reported that endothelial cells (ECs) exposed to oxidized LDL (oxLDL) release ATP, which activates P2Y2R, resulting in the expression of receptors for advanced glycation end products and adhesion molecules that are involved in the pathogenesis of atherosclerosis. Therefore, it is expected that P2Y2R activation by ATP released under inflammatory conditions may be linked to the inflammasome-mediated pathogenesis of cardiovascular diseases such as atherosclerosis. However, the exact association remains unclear. Thus, in this study, we investigated the role of P2Y2R in oxLDL-mediated inflammasome activation and the related atherosclerotic pathogenesis in ECs. ECs stimulated with oxLDL demonstrated increased intracellular production and extracellular secretion of ATP. In addition, mitochondrial reactive oxygen species (mtROS) production and mitochondrial DNA (mtDNA) expression and cytosolic release were increased in ECs stimulated with oxLDL or the P2Y2R agonists ATP and UTP. Moreover, caspase-1 activity and IL-1β production were increased in ECs stimulated with oxLDL, ATP or UTP through the modulation of mtROS production and mtDNA expression, in a P2Y2R-dependent manner. Furthermore, TLR-9 and NF-κB activation was increased in ECs in response to oxLDL, ATP or UTP, in a mtDNA-dependent manner. Taken together, our findings suggest that P2Y2R activation by ATP is involved in oxLDL-mediated inflammasome activation and subsequent IL-1β production through the modulation of mtROS-mtDNA induction and the TLR9-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Hana Jin
- Department of Pharmacology, College of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju, 52727, South Korea
| | - Young Shin Ko
- Department of Pharmacology, College of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju, 52727, South Korea
| | - Sang Won Park
- Department of Pharmacology, College of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju, 52727, South Korea
| | - Hye Jung Kim
- Department of Pharmacology, College of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju, 52727, South Korea.
| |
Collapse
|
34
|
Zhao RZ, Jiang S, Zhang L, Yu ZB. Mitochondrial electron transport chain, ROS generation and uncoupling (Review). Int J Mol Med 2019; 44:3-15. [PMID: 31115493 PMCID: PMC6559295 DOI: 10.3892/ijmm.2019.4188] [Citation(s) in RCA: 526] [Impact Index Per Article: 87.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 04/19/2019] [Indexed: 12/18/2022] Open
Abstract
The mammalian mitochondrial electron transport chain (ETC) includes complexes I-IV, as well as the electron transporters ubiquinone and cytochrome c. There are two electron transport pathways in the ETC: Complex I/III/IV, with NADH as the substrate and complex II/III/IV, with succinic acid as the substrate. The electron flow is coupled with the generation of a proton gradient across the inner membrane and the energy accumulated in the proton gradient is used by complex V (ATP synthase) to produce ATP. The first part of this review briefly introduces the structure and function of complexes I-IV and ATP synthase, including the specific electron transfer process in each complex. Some electrons are directly transferred to O2 to generate reactive oxygen species (ROS) in the ETC. The second part of this review discusses the sites of ROS generation in each ETC complex, including sites IF and IQ in complex I, site IIF in complex II and site IIIQo in complex III, and the physiological and pathological regulation of ROS. As signaling molecules, ROS play an important role in cell proliferation, hypoxia adaptation and cell fate determination, but excessive ROS can cause irreversible cell damage and even cell death. The occurrence and development of a number of diseases are closely related to ROS overproduction. Finally, proton leak and uncoupling proteins (UCPS) are discussed. Proton leak consists of basal proton leak and induced proton leak. Induced proton leak is precisely regulated and induced by UCPs. A total of five UCPs (UCP1-5) have been identified in mammalian cells. UCP1 mainly plays a role in the maintenance of body temperature in a cold environment through non-shivering thermogenesis. The core role of UCP2-5 is to reduce oxidative stress under certain conditions, therefore exerting cytoprotective effects. All diseases involving oxidative stress are associated with UCPs.
Collapse
Affiliation(s)
- Ru-Zhou Zhao
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Shuai Jiang
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lin Zhang
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zhi-Bin Yu
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
35
|
Kaur P, Choudhury D. Insulin Promotes Wound Healing by Inactivating NFkβP50/P65 and Activating Protein and Lipid Biosynthesis and alternating Pro/Anti-inflammatory Cytokines Dynamics. Biomol Concepts 2019; 10:11-24. [DOI: 10.1515/bmc-2019-0002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/04/2019] [Indexed: 02/07/2023] Open
Abstract
AbstractFour hundred and twenty-two million people have diabetes due to excess free body glucose in their body fluids. Diabetes leads to various problems including retinopathy, neuropathy, arthritis, damage blood vessels etc; it also causes a delay in wound healing. Insufficiency of insulin is the main reason for diabetes-I and systemic insulin treatment is a remedy. The perspective of the potential use of insulin/insulin based drugs to treat chronic wounds in diabetic conditions is focused on in this review. At the site of the wound, TNF-ɑ, IFN-ϒ, IL-1β and IL-6 pro-inflammatory cytokines cause the generation of free radicals, leading to inflammation which becomes persistent in diabetes. Insulin induces expression of IL-4/IL-13, IL-10 anti-inflammatory cytokines etc which further down-regulates NFkβP50/P65 assembly. Insulin shifts the equilibrium towards NFkβP50/P50 which leads to down-regulation of inflammatory cytokines such as IL-6, IL-10 etc through STAT6, STAT3 and c-Maf activation causing nullification of an inflammatory condition. Insulin also promotes protein and lipid biosynthesis which indeed promotes wound recovery. Here, in this article, the contributions of insulin in controlling wound tissue microenvironments and remodulation of tissue have been summarised, which may be helpful to develop novel insulin-based formulation(s) for effective treatment of wounds in diabetic conditions.
Collapse
Affiliation(s)
- Pawandeep Kaur
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala-147004, Punjab, India
| | - Diptiman Choudhury
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala-147004, Punjab, India
| |
Collapse
|
36
|
Zhao J, Xu SZ, Liu J. Fibrinopeptide A induces C-reactive protein expression through the ROS-ERK1/2/p38-NF-κB signal pathway in the human umbilical vascular endothelial cells. J Cell Physiol 2019; 234:13481-13492. [PMID: 30633345 DOI: 10.1002/jcp.28027] [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] [Received: 09/21/2018] [Accepted: 11/30/2018] [Indexed: 12/25/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall. Inflammation causes endothelial injury and dysfunction, which is an initial step of atherosclerosis. Fibrinopeptide A (FPA) is a biomarker of the activation of the coagulation system, and a high concentration of FPA in the blood occurs in patients with ischemic cardiocerebrovascular diseases. The present research observed that FPA stimulated the generation of C-reactive protein (CRP), IL-1β, and IL-6 in human umbilical vascular endothelial cells (HUVECs); and anti-IL-1 β and anti-IL-6 neutralizing antibodies did not alter FPA-induced CRP expression in HUVECs. The subchronic administration of FPA into rats increased the plasma FPA and CRP levels. Further studies showed that FPA stimulated superoxide anion generation, activated ERK1/2 and p38, promoted nuclear factor κB (NF-κB) nuclear translocation, and raised the NF-κB level in the nuclei of HUVECs. Antioxidant N-acetylcysteine (NAC), complex II inhibitor thenoyltrifluoroacetone (TTFA), and NADPH oxidase inhibitor diphenyleneiodonium (DPI) inhibited FPA-stimulated generation of superoxide anion, and NAC reduced FPA-induced expressions of the phosphorylated ERK1/2 and p38. NAC, TTFA, DPI, inhibitors of ERK1/2, p38, and NF-κB all downregulated FPA-induced CRP expression. These results indicate that FPA induces CRP expression in HUVECs via the ROS-ERK1/2/p38-NF-κB signal pathway. Moreover, this is the first report that FPA produces a proinflammatory effect on the vascular endothelial cells.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Shou-Zhu Xu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, China.,Department of Public Health, Shaanxi University of Chinese Medicine, Xianyang City, China
| | - Juntian Liu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
37
|
del Carmen Baez M, Tarán M, Moya M, de la Paz Scribano Parada M. Oxidative Stress in Metabolic Syndrome: Experimental Model of Biomarkers. MODULATION OF OXIDATIVE STRESS IN HEART DISEASE 2019:313-338. [DOI: 10.1007/978-981-13-8946-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
38
|
Bansal S, Kare PK, Tripathi AK, Madhu SV. Advanced Glycation End Products: A Potential Contributor of Oxidative Stress for Cardio-Vascular Problems in Diabetes. OXIDATIVE STRESS IN HEART DISEASES 2019:437-459. [DOI: 10.1007/978-981-13-8273-4_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
39
|
Dorighello GG, Paim BA, Leite ACR, Vercesi AE, Oliveira HC. Spontaneous experimental atherosclerosis in hypercholesterolemic mice advances with ageing and correlates with mitochondrial reactive oxygen species. Exp Gerontol 2018; 109:47-50. [DOI: 10.1016/j.exger.2017.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/21/2016] [Accepted: 02/13/2017] [Indexed: 10/20/2022]
|
40
|
T MM, Anand T, Khanum F. Attenuation of cytotoxicity induced by tBHP in H9C2 cells by Bacopa monniera and Bacoside A. PATHOPHYSIOLOGY 2018; 25:143-149. [DOI: 10.1016/j.pathophys.2018.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 02/08/2018] [Accepted: 03/21/2018] [Indexed: 12/20/2022] Open
|
41
|
Yin J, Zhuang J, Lv S, Mu Y. Study on a 65-mer peptide mimetic enzyme with GPx and SOD dual function. J Mol Recognit 2018; 31:e2714. [PMID: 29656411 DOI: 10.1002/jmr.2714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/14/2018] [Accepted: 02/21/2018] [Indexed: 11/06/2022]
Abstract
Excessive reactive oxygen species (ROS) levels are harmful to the body. The peroxidase, GPx, and the superoxide dismutase, SOD, are important antioxidant enzymes for preventing ROS-induced damage. Se-CuZn-65P is an enzyme mimetic with dual GPx and SOD antioxidant function. However, currently, its production is mainly based on the cysteine auxotrophic expression technique, which is inefficient, expensive, and time consuming. In this study, we combined protein engineering and the chemical mutation method to synthesize Se-CuZn-65P. The DNA sequence encoding the 65 amino acid peptide with the desired sequence transformations to incorporate the SOD and the GPx catalytic sites was cloned and expressed in a soluble protein expression vector. The protein yield increased up to 152 mg/L, which is 10 times higher than in previous studies. The SOD and GPx activity of Se-CuZn-65P was high (1181 U/mg and 753 U/μmol, respectively). The binding constant of glutathione was 5.6 × 104 L·mol-1 , which shows that Se-CuZn-65P efficiently catalyzed hydrogen peroxide reduction by glutathione. Mitochondrial damage experiments confirmed the double protective role of the Se-CuZn-65P peptide and demonstrated functional synergy between the SOD and the GPx domains, which indicates its potential to be used in the treatment of ROS-related diseases. Our research may give a new thought to increase the yield of mimic.
Collapse
Affiliation(s)
- Juxin Yin
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou, China
| | - Jianjian Zhuang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| | - Shaowu Lv
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, China
| | - Ying Mu
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou, China
| |
Collapse
|
42
|
Huang M, Wei R, Wang Y, Su T, Li P, Chen X. The uremic toxin hippurate promotes endothelial dysfunction via the activation of Drp1-mediated mitochondrial fission. Redox Biol 2018; 16:303-313. [PMID: 29573704 PMCID: PMC5953222 DOI: 10.1016/j.redox.2018.03.010] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 12/31/2022] Open
Abstract
The accumulation of uremic toxins in chronic kidney disease (CKD) induces inflammation, oxidative stress and endothelial dysfunction, which is a key step in atherosclerosis. Accumulating evidence indicates increased mitochondrial fission is a contributing mechanism for impaired endothelial function. Hippurate, a uremic toxin, has been reported to be involved in cardiovascular diseases. Here, we assessed the endothelial toxicity of hippurate and the contribution of altered mitochondrial dynamics to hippurate-induced endothelial dysfunction. Treatment of human aortic endothelial cells with hippurate reduced the expression of endothelial nitric oxide synthase (eNOS) and increased the expression of intercellular cell adhesion molecule-1 (ICAM-1) and von Willebrand factor (vWF). The mechanisms of hippurate-induced endothelial dysfunction in vitro depended on the activation of Dynamin-related protein 1 (Drp1)-mediated mitochondrial fission and overproduction of mitochondrial reactive oxygen species (mitoROS). In a rat model in which CKD was induced by 5/6 nephrectomy (CKD rat), we observed increased oxidative stress, impaired endothelium-dependent vasodilation, and elevated soluble biomarkers of endothelial dysfunction (ICAM-1 and vWF). Similarly, endothelial dysfunction was identified in healthy rats treated with disease-relevant concentrations of hippurate. In aortas of CKD rats and hippurate-treated rats, we observed an increase in Drp1 protein levels and mitochondrial fission. Inhibition of Drp1 improved endothelial function in both rat models. These results indicate that hippurate, by itself, can cause endothelial dysfunction. Increased mitochondrial fission plays an active role in hippurate-induced endothelial dysfunction via an increase in mitoROS.
Collapse
Affiliation(s)
- Mengjie Huang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China
| | - Ribao Wei
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China.
| | - Yang Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China
| | - Tingyu Su
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China
| | - Ping Li
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease, Beijing 100853,People's Republic of China
| |
Collapse
|
43
|
Eshaq RS, Aldalati AMZ, Alexander JS, Harris NR. Diabetic retinopathy: Breaking the barrier. PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2017; 24:229-241. [PMID: 28732591 PMCID: PMC5711541 DOI: 10.1016/j.pathophys.2017.07.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 06/26/2017] [Accepted: 07/04/2017] [Indexed: 12/28/2022]
Abstract
Diabetic retinopathy (DR) remains a major complication of diabetes and a leading cause of blindness among adults worldwide. DR is a progressive disease affecting both type I and type II diabetic patients at any stage of the disease, and targets the retinal microvasculature. DR results from multiple biochemical, molecular and pathophysiological changes to the retinal vasculature, which affect both microcirculatory functions and ultimately photoreceptor function. Several neural, endothelial, and support cell (e.g., pericyte) mechanisms are altered in a pathological fashion in the hyperglycemic environment during diabetes that can disturb important cell surface components in the vasculature producing the features of progressive DR pathophysiology. These include loss of the glycocalyx, blood-retinal barrier dysfunction, increased expression of inflammatory cell markers and adhesion of blood leukocytes and platelets. Included in this review is a discussion of modifications that occur at or near the surface of the retinal vascular endothelial cells, and the consequences of these alterations on the integrity of the retina.
Collapse
Affiliation(s)
- Randa S Eshaq
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center -Shreveport, 1501 Kings Highway, Shreveport, LA 71130, United States
| | - Alaa M Z Aldalati
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center -Shreveport, 1501 Kings Highway, Shreveport, LA 71130, United States
| | - J Steven Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center -Shreveport, 1501 Kings Highway, Shreveport, LA 71130, United States
| | - Norman R Harris
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center -Shreveport, 1501 Kings Highway, Shreveport, LA 71130, United States.
| |
Collapse
|
44
|
Interleukin-6 deficiency facilitates myocardial dysfunction during high fat diet-induced obesity by promoting lipotoxicity and inflammation. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3128-3141. [DOI: 10.1016/j.bbadis.2017.08.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/10/2017] [Accepted: 08/22/2017] [Indexed: 12/28/2022]
|
45
|
Hernandez-Lopez R, Chavez-Gonzalez A, Torres-Barrera P, Moreno-Lorenzana D, Lopez-DiazGuerrero N, Santiago-German D, Isordia-Salas I, Smadja D, C. Yoder M, Majluf-Cruz A, Alvarado-Moreno JA. Reduced proliferation of endothelial colony-forming cells in unprovoked venous thromboembolic disease as a consequence of endothelial dysfunction. PLoS One 2017; 12:e0183827. [PMID: 28910333 PMCID: PMC5598948 DOI: 10.1371/journal.pone.0183827] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 08/11/2017] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Venous thromboembolic disease (VTD) is a public health problem. We recently reported that endothelial colony-forming cells (ECFCs) derived from endothelial cells (EC) (ECFC-ECs) from patients with VTD have a dysfunctional state. For this study, we proposed that a dysfunctional status of these cells generates a reduction of its proliferative ability, which is also associated with senescence and reactive oxygen species (ROS). METHODS AND RESULTS Human mononuclear cells (MNCs) were obtained from peripheral blood from 40 healthy human volunteers (controls) and 50 patients with VTD matched by age (20-50 years) and sex to obtain ECFCs. We assayed their proliferative ability with plasma of patients and controls and supernatants of cultures from ECFC-ECs, senescence-associated β-galactosidase (SA-β-gal), ROS, and expression of ephrin-B2/Eph-B4 receptor. Compared with cells from controls, cells from VTD patients showed an 8-fold increase of ECFCs that emerged 1 week earlier, reduced proliferation at long term (39%) and, in passages 4 and 10, a highly senescent rate (30±1.05% vs. 91.3±15.07%, respectively) with an increase of ROS and impaired expression of ephrin-B2/Eph-4 genes. Proliferation potential of cells from VTD patients was reduced in endothelial medium [1.4±0.22 doubling population (DP)], control plasma (1.18±0.31 DP), or plasma from VTD patients (1.65±0.27 DP). CONCLUSIONS As compared with controls, ECFC-ECs from individuals with VTD have higher oxidative stress, proliferation stress, cellular senescence, and low proliferative potential. These findings suggest that patients with a history of VTD are ECFC-ECs dysfunctional that could be associated to permanent risk for new thrombotic events.
Collapse
Affiliation(s)
- Rubicel Hernandez-Lopez
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Posgrado en Biologia Experimental, Universidad Autonoma Metropolitana, Iztapalapa. Mexico City, Mexico
| | - Antonieta Chavez-Gonzalez
- Unidad de Investigacion Medica en Enfermedades Oncologicas, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Patricia Torres-Barrera
- Unidad de Investigacion Medica en Enfermedades Oncologicas, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Dafne Moreno-Lorenzana
- Unidad de Investigacion Medica en Enfermedades Oncologicas, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Norma Lopez-DiazGuerrero
- Posgrado en Biologia Experimental, Universidad Autonoma Metropolitana, Iztapalapa. Mexico City, Mexico
| | | | - Irma Isordia-Salas
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - David Smadja
- Paris Descartes University, INSERM UMR-S 1140, Faculté de Pharmacie de Paris, Paris, France
- AP-HP, Hôpital Européen Georges Pompidou, Hematology department, Paris, France
| | - Mervin C. Yoder
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Abraham Majluf-Cruz
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - J. Antonio Alvarado-Moreno
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
46
|
S. J, M.S. SS, R. A, Shanmuganathan G, Mozhi SS, S. ND. Potential antioxidant and cytoprotective effects of essential oil extracted from Cymbopogon citratus on OxLDL and H 2 O 2 LDL induced Human Peripheral Blood Mononuclear Cells (PBMC). FOOD SCIENCE AND HUMAN WELLNESS 2017. [DOI: 10.1016/j.fshw.2017.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
47
|
Haymana C, Aydoğdu A, Soykut B, Erdem O, Ibrahimov T, Dinc M, Meric C, Basaran Y, Sonmez A, Azal O. Oxidative stress status in congenital hypogonadism: an appraisal. Toxicol Mech Methods 2017; 27:451-457. [DOI: 10.1080/15376516.2017.1320693] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- C. Haymana
- Department of Endocrinology and Metabolism, Gulhane Training and Research Hospital, Ankara, Turkey
| | - A. Aydoğdu
- Department of Endocrinology and Metabolism, Gulhane Training and Research Hospital, Ankara, Turkey
| | - B. Soykut
- Department of Pharmaceutical Toxicology, Gulhane Training and Research Hospital, Ankara, Turkey
| | - O. Erdem
- Department of Pharmaceutical Toxicology, Gulhane Training and Research Hospital, Ankara, Turkey
| | - T. Ibrahimov
- Department of Endocrinology and Metabolism, Gulhane Training and Research Hospital, Ankara, Turkey
| | - M. Dinc
- Department of Endocrinology and Metabolism, Gulhane Training and Research Hospital, Ankara, Turkey
| | - C. Meric
- Department of Endocrinology and Metabolism, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Y. Basaran
- Department of Endocrinology and Metabolism, Gulhane Training and Research Hospital, Ankara, Turkey
| | - A. Sonmez
- Department of Endocrinology and Metabolism, Gulhane Training and Research Hospital, Ankara, Turkey
| | - O. Azal
- Department of Endocrinology and Metabolism, Gulhane Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
48
|
Santos L, Davel AP, Almeida TIR, Almeida MR, Soares EA, Fernandes GJM, Magalhães SF, Barauna VG, Garcia JAD. Soy milk versus simvastatin for preventing atherosclerosis and left ventricle remodeling in LDL receptor knockout mice. ACTA ACUST UNITED AC 2017; 50:e5854. [PMID: 28225891 PMCID: PMC5333721 DOI: 10.1590/1414-431x20165854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/07/2016] [Indexed: 02/02/2023]
Abstract
Functional food intake has been highlighted as a strategy for the prevention of cardiovascular diseases by reducing risk factors. In this study, we compared the effects of oral treatment with soy milk and simvastatin on dyslipidemia, left ventricle remodeling and atherosclerotic lesion of LDL receptor knockout mice (LDLr-/-) fed a hyperlipidic diet. Forty 3-month old male LDLr-/- mice were distributed into four groups: control group (C), in which animals received standard diet; HL group, in which animals were fed a hyperlipidic diet; HL+SM or HL+S groups, in which animals were submitted to a hyperlipidic diet plus soy milk or simvastatin, respectively. After 60 days, both soy milk and simvastatin treatment prevented dyslipidemia, atherosclerotic lesion progression and left ventricle hypertrophy in LDLr-/- mice. These beneficial effects of soy milk and simvastatin were associated with reduced oxidative stress and inflammatory state in the heart and aorta caused by the hyperlipidic diet. Treatment with soy milk was more effective in preventing HDLc reduction and triacylglycerol and VLDLc increase. On the other hand, simvastatin was more effective in preventing an increase in total cholesterol, LDLc and superoxide production in aorta, as well as CD40L both in aorta and left ventricle of LDLr-/-. In conclusion, our results suggest a cardioprotective effect of soy milk in LDLr-/- mice comparable to the well-known effects of simvastatin.
Collapse
Affiliation(s)
- L Santos
- Unidade Acadêmica de Serra Talhada, Universidade Federal Rural de Pernambuco, Serra Talhada, PE, Brasil.,Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade de Campinas, Campinas, SP, Brasil
| | - A P Davel
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade de Campinas, Campinas, SP, Brasil
| | - T I R Almeida
- Instituto Federal do Sul de Minas, Muzambinho, MG, Brasil
| | - M R Almeida
- Instituto Federal do Sul de Minas, Muzambinho, MG, Brasil
| | - E A Soares
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, MG, Brasil
| | - G J M Fernandes
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas, MG, Brasil
| | - S F Magalhães
- Departmento de Biomedicina, Universidade José do Rosário Vellano, Alfenas, MG, Brasil
| | - V G Barauna
- Departamento de Ciências Fisiológicas, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| | - J A D Garcia
- Departmento de Tecnologia, Ciência e Educação, Instituto Federal do Sul de Minas, Machado, MG, Brasil.,Departamento de Fisiologia, Universidade José do Rosário Vellano, Alfenas, MG, Brasil
| |
Collapse
|
49
|
Bernardes N, Ayyappan P, De Angelis K, Bagchi A, Akolkar G, da Silva Dias D, Belló-Klein A, Singal PK. Excessive consumption of fructose causes cardiometabolic dysfunctions through oxidative stress and inflammation. Can J Physiol Pharmacol 2017; 95:1078-1090. [PMID: 28187269 DOI: 10.1139/cjpp-2016-0663] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A rapid rise in obesity, as well as physical inactivity, in industrialized countries is associated with fructose-consumption-mediated metabolic syndrome having a strong association with cardiovascular disease. Although insulin resistance is thought to be at the core, visceral obesity, hypertension, and hypertriglyceridemia are also considered important components of this metabolic disorder. In addition, various other abnormalities such as inflammation, oxidative stress, and elevated levels of uric acid are also part of this syndrome. Lifestyle changes through improved physical activity, as well as nutrition, are important approaches to minimize metabolic syndrome and its deleterious effects.
Collapse
Affiliation(s)
- Nathalia Bernardes
- a Universidade Nove de Julho, Diretoria de Mestrado, Av. Francisco Matatazzo, 612, 10 andar, Centro de Pos Graduacao Stricto Sensu, Barra Funda, Sao Paulo, Brazil
| | - Prathapan Ayyappan
- b Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Katia De Angelis
- a Universidade Nove de Julho, Diretoria de Mestrado, Av. Francisco Matatazzo, 612, 10 andar, Centro de Pos Graduacao Stricto Sensu, Barra Funda, Sao Paulo, Brazil
| | - Ashim Bagchi
- b Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Gauri Akolkar
- b Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Danielle da Silva Dias
- a Universidade Nove de Julho, Diretoria de Mestrado, Av. Francisco Matatazzo, 612, 10 andar, Centro de Pos Graduacao Stricto Sensu, Barra Funda, Sao Paulo, Brazil
| | - Adriane Belló-Klein
- c Laboratory of Cardiovascular Physiology, Institute of Basic Health Science (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Pawan K Singal
- b Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
50
|
Ellulu MS. Obesity, cardiovascular disease, and role of vitamin C on inflammation: a review of facts and underlying mechanisms. Inflammopharmacology 2017; 25:313-328. [PMID: 28168552 DOI: 10.1007/s10787-017-0314-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/23/2017] [Indexed: 02/07/2023]
Abstract
Obesity means the accumulation of excessive fat that may interfere with the maintenance of optimal state of health. Obesity causes cardiac and vascular disease through well-known mediators such as hypertension, type-2 diabetes mellitus, and dyslipidemia, but there are evidences for other mediators such as chronic inflammation, oxidative stress, and thrombosis. The decreased levels of antioxidants factors and nitric oxide predispose to further cardiovascular adverse events. To reduce the risks, antioxidants can help by neutralizing the free radicals and protecting from damage by donating electrons. Having the capacity, vitamin C protects from oxidative stress, prevention of non-enzymatic glycosylation of proteins, and enhances arterial dilation through its effect on nitric oxide release. It also decreases lipid peroxidation, and alleviates inflammation. The anti-inflammatory property of vitamin C could be attributed to ability to modulate the NF-kB DNA binding activity and down-regulation in the hepatic mRNA expression for the interleukins and tumor factors.
Collapse
Affiliation(s)
- Mohammed S Ellulu
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang, Selangor, Malaysia. .,Clinical Nutrition Specialist, Gaza, Palestine.
| |
Collapse
|