1
|
Ghaffari-Bohlouli P, Jafari H, Okoro OV, Alimoradi H, Nie L, Jiang G, Kakkar A, Shavandi A. Gas Therapy: Generating, Delivery, and Biomedical Applications. SMALL METHODS 2024; 8:e2301349. [PMID: 38193272 DOI: 10.1002/smtd.202301349] [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: 10/05/2023] [Revised: 12/11/2023] [Indexed: 01/10/2024]
Abstract
Oxygen (O2), nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H2S), and hydrogen (H2) with direct effects, and carbon dioxide (CO2) with complementary effects on the condition of various diseases are known as therapeutic gases. The targeted delivery and in situ generation of these therapeutic gases with controllable release at the site of disease has attracted attention to avoid the risk of gas poisoning and improve their performance in treating various diseases such as cancer therapy, cardiovascular therapy, bone tissue engineering, and wound healing. Stimuli-responsive gas-generating sources and delivery systems based on biomaterials that enable on-demand and controllable release are promising approaches for precise gas therapy. This work highlights current advances in the design and development of new approaches and systems to generate and deliver therapeutic gases at the site of disease with on-demand release behavior. The performance of the delivered gases in various biomedical applications is then discussed.
Collapse
Affiliation(s)
- Pejman Ghaffari-Bohlouli
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, H3A 0B8, Canada
| | - Hafez Jafari
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Oseweuba Valentine Okoro
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Houman Alimoradi
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Lei Nie
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
- College of Life Sciences, Xinyang Normal University, Xinyang, 464000, China
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, H3A 0B8, Canada
| | - Amin Shavandi
- 3BIO-BioMatter, École polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| |
Collapse
|
2
|
Geiger M, Gorica E, Mohammed SA, Mongelli A, Mengozi A, Delfine V, Ruschitzka F, Costantino S, Paneni F. Epigenetic Network in Immunometabolic Disease. Adv Biol (Weinh) 2024; 8:e2300211. [PMID: 37794610 DOI: 10.1002/adbi.202300211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Although a large amount of data consistently shows that genes affect immunometabolic characteristics and outcomes, epigenetic mechanisms are also heavily implicated. Epigenetic changes, including DNA methylation, histone modification, and noncoding RNA, determine gene activity by altering the accessibility of chromatin to transcription factors. Various factors influence these alterations, including genetics, lifestyle, and environmental cues. Moreover, acquired epigenetic signals can be transmitted across generations, thus contributing to early disease traits in the offspring. A closer investigation is critical in this aspect as it can help to understand the underlying molecular mechanisms further and gain insights into potential therapeutic targets for preventing and treating diseases arising from immuno-metabolic dysregulation. In this review, the role of chromatin alterations in the transcriptional modulation of genes involved in insulin resistance, systemic inflammation, macrophage polarization, endothelial dysfunction, metabolic cardiomyopathy, and nonalcoholic fatty liver disease (NAFLD), is discussed. An overview of emerging chromatin-modifying drugs and the importance of the individual epigenetic profile for personalized therapeutic approaches in patients with immuno-metabolic disorders is also presented.
Collapse
Affiliation(s)
- Martin Geiger
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Shafeeq Ahmed Mohammed
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessandro Mengozi
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Valentina Delfine
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- Department of Research and Education, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| |
Collapse
|
3
|
Li H, Zheng J, Wu Y, Zhou H, Zeng S, Li Q. Dendrobium officinale polysaccharide decreases podocyte injury in diabetic nephropathy by regulating IRS-1/AKT signal and promoting mitophagy. Aging (Albany NY) 2023; 15:10291-10306. [PMID: 37812195 PMCID: PMC10599763 DOI: 10.18632/aging.205075] [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: 04/18/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUNDS High glucose (HG) caused oxidative stress and mitochondrial dysfunction, resulting in insulin resistance in podocytes, a key mechanism of diabetic nephropathy. Dendrobium officinale polysaccharide (DOP) was able to improve insulin resistance and antioxidant capability. OBJECTIVE The purpose of this study is to explore the mechanism by which DOP decreases the podocyte injury induced by HG. METHODS MPC5 cells were treated with HG, DOP, and IRS-1/2 inhibitor NT157. Afterwards, glucose consumption, generations of ROS and MDA were measured using the detection kits. Mitophagy was monitored using both MtphagTracyker and LysoTracker. The mitochondrial membrane potential was evaluated by JC-1 staining. DOP was also used in a mouse model of diabetes, with the measurements of urine albumin, blood creatinine and blood urea nitrogen. RESULTS Treatment with DOP suppressed the HG-induced reduction of glucose consumption, the phosphorylation of IRS-1 (phospho Y632), AKT (phospho Ser473 and Thr308) and Nephrin. In addition, HG-induced augment of ROS and MDA, formation of γ-H2A.X foci and translocation of AKT to nucleus were inhibited by DOP. DOP enhanced mitophagy, which was associated with decreased mitochondrial membrane potential and ROS production. DOP conferred protective effect on podocyte in the diabetic mouse by reducing the albumin/creatinine ratio and blood urea nitrogen, and restoring Nephrin expression in podocytes. CONCLUSIONS DOP alleviates HG-induced podocyte injuryby regulating IRS-1/AKT signal and promoting mitophagy.
Collapse
Affiliation(s)
- Huahua Li
- Department of Geriatric, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Furong, Changsha 410005, P.R. China
| | - Jin Zheng
- Department of Geriatric, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Furong, Changsha 410005, P.R. China
| | - Yacen Wu
- Department of Rehabilitation, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Furong, Changsha 410005, P.R. China
| | - Hong Zhou
- Department of Geriatric, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Furong, Changsha 410005, P.R. China
| | - Suli Zeng
- Department of Geriatric, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Furong, Changsha 410005, P.R. China
| | - Quanqing Li
- Department of Geriatric, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Furong, Changsha 410005, P.R. China
| |
Collapse
|
4
|
Antioxidant Phytochemicals as Potential Therapy for Diabetic Complications. Antioxidants (Basel) 2023; 12:antiox12010123. [PMID: 36670985 PMCID: PMC9855127 DOI: 10.3390/antiox12010123] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/10/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
The global prevalence of diabetes continues to increase partly due to rapid urbanization and an increase in the aging population. Consequently, this is associated with a parallel increase in the prevalence of diabetic vascular complications which significantly worsen the burden of diabetes. For these diabetic vascular complications, there is still an unmet need for safe and effective alternative/adjuvant therapeutic interventions. There is also an increasing urge for therapeutic options to come from natural products such as plants. Hyperglycemia-induced oxidative stress is central to the development of diabetes and diabetic complications. Furthermore, oxidative stress-induced inflammation and insulin resistance are central to endothelial damage and the progression of diabetic complications. Human and animal studies have shown that polyphenols could reduce oxidative stress, hyperglycemia, and prevent diabetic complications including diabetic retinopathy, diabetic nephropathy, and diabetic peripheral neuropathy. Part of the therapeutic effects of polyphenols is attributed to their modulatory effect on endogenous antioxidant systems. This review attempts to summarize the established effects of polyphenols on endogenous antioxidant systems from the literature. Moreover, potential therapeutic strategies for harnessing the potential benefits of polyphenols for diabetic vascular complications are also discussed.
Collapse
|
5
|
Ghiarone T, Castorena-Gonzalez JA, Foote CA, Ramirez-Perez FI, Ferreira-Santos L, Cabral-Amador FJ, de la Torre R, Ganga RR, Wheeler AA, Manrique-Acevedo C, Padilla J, Martinez-Lemus LA. ADAM17 cleaves the insulin receptor ectodomain on endothelial cells and causes vascular insulin resistance. Am J Physiol Heart Circ Physiol 2022; 323:H688-H701. [PMID: 36018759 PMCID: PMC9512115 DOI: 10.1152/ajpheart.00039.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 11/22/2022]
Abstract
Inflammation and vascular insulin resistance are hallmarks of type 2 diabetes (T2D). However, several potential mechanisms causing abnormal endothelial insulin signaling in T2D need further investigation. Evidence indicates that the activity of ADAM17 (a disintegrin and metalloproteinase-17) and the presence of insulin receptor (IR) in plasma are increased in subjects with T2D. Accordingly, we hypothesized that in T2D, increased ADAM17 activity sheds the IR ectodomain from endothelial cells and impairs insulin-induced vasodilation. We used small visceral arteries isolated from a cross-sectional study of subjects with and without T2D undergoing bariatric surgery, human cultured endothelial cells, and recombinant proteins to test our hypothesis. Here, we demonstrate that arteries from subjects with T2D had increased ADAM17 expression, reduced presence of tissue inhibitor of metalloproteinase-3 (TIMP3), decreased extracellular IRα, and impaired insulin-induced vasodilation versus those from subjects without T2D. In vitro, active ADAM17 cleaved the ectodomain of the IRβ subunit. Endothelial cells with ADAM17 overexpression or exposed to the protein kinase-C activator, PMA, had increased ADAM17 activity, decreased IRα presence on the cell surface, and increased IR shedding. Moreover, pharmacological inhibition of ADAM17 with TAPI-0 rescued PMA-induced IR shedding and insulin-signaling impairments in endothelial cells and insulin-stimulated vasodilation in human arteries. In aggregate, our findings suggest that ADAM17-mediated shedding of IR from the endothelial surface impairs insulin-mediated vasodilation. Thus, we propose that inhibition of ADAM17 sheddase activity should be considered a strategy to restore vascular insulin sensitivity in T2D.NEW & NOTEWORTHY To our knowledge, this is the first study to investigate the involvement of ADAM17 in causing impaired insulin-induced vasodilation in T2D. We provide evidence that ADAM17 activity is increased in the vasculature of patients with T2D and support the notion that ADAM17-mediated shedding of endothelial IRα ectodomains is a novel mechanism causing vascular insulin resistance. Our results highlight that targeting ADAM17 activity may be a potential therapeutic strategy to correct vascular insulin resistance in T2D.
Collapse
Affiliation(s)
- Thaysa Ghiarone
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Jorge A Castorena-Gonzalez
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Department of Pharmacology, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Christopher A Foote
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri
| | | | | | | | - Rama R Ganga
- Department of Surgery, University of Missouri, Columbia, Missouri
| | - Andrew A Wheeler
- Department of Surgery, University of Missouri, Columbia, Missouri
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri
| |
Collapse
|
6
|
Pánico P, Velasco M, Salazar AM, Picones A, Ortiz-Huidobro RI, Guerrero-Palomo G, Salgado-Bernabé ME, Ostrosky-Wegman P, Hiriart M. Is Arsenic Exposure a Risk Factor for Metabolic Syndrome? A Review of the Potential Mechanisms. Front Endocrinol (Lausanne) 2022; 13:878280. [PMID: 35651975 PMCID: PMC9150370 DOI: 10.3389/fendo.2022.878280] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/11/2022] [Indexed: 12/14/2022] Open
Abstract
Exposure to arsenic in drinking water is a worldwide health problem. This pollutant is associated with increased risk of developing chronic diseases, including metabolic diseases. Metabolic syndrome (MS) is a complex pathology that results from the interaction between environmental and genetic factors. This condition increases the risk of developing type 2 diabetes, cardiovascular diseases, and cancer. The MS includes at least three of the following signs, central obesity, impaired fasting glucose, insulin resistance, dyslipidemias, and hypertension. Here, we summarize the existing evidence of the multiple mechanisms triggered by arsenic to developing the cardinal signs of MS, showing that this pollutant could contribute to the multifactorial origin of this pathology.
Collapse
Affiliation(s)
- Pablo Pánico
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Myrian Velasco
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ana María Salazar
- Department of Genomic Medicine and Environmental Toxicology. Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Arturo Picones
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rosa Isela Ortiz-Huidobro
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gabriela Guerrero-Palomo
- Department of Genomic Medicine and Environmental Toxicology. Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Manuel Eduardo Salgado-Bernabé
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Patricia Ostrosky-Wegman
- Department of Genomic Medicine and Environmental Toxicology. Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marcia Hiriart
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- *Correspondence: Marcia Hiriart,
| |
Collapse
|
7
|
The Blood-Brain Barrier, Oxidative Stress, and Insulin Resistance. Antioxidants (Basel) 2021; 10:antiox10111695. [PMID: 34829566 PMCID: PMC8615183 DOI: 10.3390/antiox10111695] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
The blood–brain barrier (BBB) is a network of specialized endothelial cells that regulates substrate entry into the central nervous system (CNS). Acting as the interface between the periphery and the CNS, the BBB must be equipped to defend against oxidative stress and other free radicals generated in the periphery to protect the CNS. There are unique features of brain endothelial cells that increase the susceptibility of these cells to oxidative stress. Insulin signaling can be impacted by varying levels of oxidative stress, with low levels of oxidative stress being necessary for signaling and higher levels being detrimental. Insulin must cross the BBB in order to access the CNS, levels of which are important in peripheral metabolism as well as cognition. Any alterations in BBB transport due to oxidative stress at the BBB could have downstream disease implications. In this review, we cover the interactions of oxidative stress at the BBB, how insulin signaling is related to oxidative stress, and the impact of the BBB in two diseases greatly affected by oxidative stress and insulin resistance: diabetes mellitus and Alzheimer’s disease.
Collapse
|
8
|
Akoumianakis I, Badi I, Douglas G, Chuaiphichai S, Herdman L, Akawi N, Margaritis M, Antonopoulos AS, Oikonomou EK, Psarros C, Galiatsatos N, Tousoulis D, Kardos A, Sayeed R, Krasopoulos G, Petrou M, Schwahn U, Wohlfart P, Tennagels N, Channon KM, Antoniades C. Insulin-induced vascular redox dysregulation in human atherosclerosis is ameliorated by dipeptidyl peptidase 4 inhibition. Sci Transl Med 2021; 12:12/541/eaav8824. [PMID: 32350133 DOI: 10.1126/scitranslmed.aav8824] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 10/01/2019] [Accepted: 04/01/2020] [Indexed: 12/12/2022]
Abstract
Recent clinical trials have revealed that aggressive insulin treatment has a neutral effect on cardiovascular risk in patients with diabetes despite improved glycemic control, which may suggest confounding direct effects of insulin on the human vasculature. We studied 580 patients with coronary atherosclerosis undergoing coronary artery bypass surgery (CABG), finding that high endogenous insulin was associated with reduced nitric oxide (NO) bioavailability ex vivo in vessels obtained during surgery. Ex vivo experiments with human internal mammary arteries and saphenous veins obtained from 94 patients undergoing CABG revealed that both long-acting insulin analogs and human insulin triggered abnormal responses of post-insulin receptor substrate 1 downstream signaling ex vivo, independently of systemic insulin resistance status. These abnormal responses led to reduced NO bioavailability, activation of NADPH oxidases, and uncoupling of endothelial NO synthase. Treatment with an oral dipeptidyl peptidase 4 inhibitor (DPP4i) in vivo or DPP4i administered to vessels ex vivo restored physiological insulin signaling, reversed vascular insulin responses, reduced vascular oxidative stress, and improved endothelial function in humans. The detrimental effects of insulin on vascular redox state and endothelial function as well as the insulin-sensitizing effect of DPP4i were also validated in high-fat diet-fed ApoE-/- mice treated with DPP4i. High plasma DPP4 activity and high insulin were additively related with higher cardiac mortality in patients with coronary atherosclerosis undergoing CABG. These findings may explain the inability of aggressive insulin treatment to improve cardiovascular outcomes, raising the question whether vascular insulin sensitization with DPP4i should precede initiation of insulin treatment and continue as part of a long-term combination therapy.
Collapse
Affiliation(s)
- Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Ileana Badi
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Gillian Douglas
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Surawee Chuaiphichai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Laura Herdman
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Nadia Akawi
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Marios Margaritis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Alexios S Antonopoulos
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Evangelos K Oikonomou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Costas Psarros
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | | | - Dimitris Tousoulis
- First Cardiology Clinic, Athens University Medical School, Athens 115 27, Greece
| | - Attila Kardos
- Milton Keynes University Hospital NHS Foundation Trust and Faculty of Life Sciences, University of Buckingham, Buckingham MK6 5LD, UK
| | - Rana Sayeed
- Cardiothoracic Surgery Department, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - George Krasopoulos
- Cardiothoracic Surgery Department, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Mario Petrou
- Cardiothoracic Surgery Department, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Uwe Schwahn
- Sanofi Aventis Deutschland GmbH, Frankfurt D-65926, Germany
| | | | | | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK.
| |
Collapse
|
9
|
Zamani B, Sheikhi A, Namazi N, Larijani B, Azadbakht L. The Effects of Supplementation with Probiotic on Biomarkers of Oxidative Stress in Adult Subjects: a Systematic Review and Meta-analysis of Randomized Trials. Probiotics Antimicrob Proteins 2021; 12:102-111. [PMID: 30666617 DOI: 10.1007/s12602-018-9500-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Previous studies have supposed that probiotic supplementation led to a positive effect on different health outcomes. Furthermore, several studies indicated that probiotics supplementation improved antioxidant status, while some studies did not indicate these effects. Hence, current systematic review and meta-analysis study was conducted to determine the effect of probiotic supplementation on some oxidative stress biomarkers among adult subjects. We searched four electronic databases PubMed, SCOPUS, ISI Web of Science, and the Cochrane Library till November 2017. Clinical trials that compared the effects of probiotic supplementation with the control group were included. A random-effect model was used to pool weighted mean difference (WMD). Finding of 11 included studies (n = 577) indicated that probiotic supplementation increased total antioxidant capacity (TAC) (WMD 77.30 mmol/L; 95% confidence interval [CI] 2.60, 152.01; I2 = 88.3%) and reduced malondialdehyde (MDA) (WMD - 0.31 μmol/L; 95% CI - 0.54, - 0.08; I2 = 71.5%) significantly compared to the control group. However, its effects on glutathione (GSH) was not significant (WMD = 19.32 μmol/L; 95% CI - 18.70, 57.33; I2 = 64.9%). The current meta-analysis revealed that probiotic supplementation may result in increasing TAC and lowering MDA, which improve antioxidant status. However, due to high heterogeneity, findings should be interpreted with caution. Further investigations are required to elucidate the effect of supplementation with probiotics on biomarkers of antioxidants.
Collapse
Affiliation(s)
- Behzad Zamani
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P.O. Box: 1416643931, Tehran, Iran
| | - Ali Sheikhi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P.O. Box: 1416643931, Tehran, Iran
| | - Nazli Namazi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Azadbakht
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P.O. Box: 1416643931, Tehran, Iran. .,Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
10
|
Masi S, Ambrosini S, Mohammed SA, Sciarretta S, Lüscher TF, Paneni F, Costantino S. Epigenetic Remodeling in Obesity-Related Vascular Disease. Antioxid Redox Signal 2021; 34:1165-1199. [PMID: 32808539 DOI: 10.1089/ars.2020.8040] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The prevalence of obesity and cardiometabolic phenotypes is alarmingly increasing across the globe and is associated with atherosclerotic vascular complications and high mortality. In spite of multifactorial interventions, vascular residual risk remains high in this patient population, suggesting the need for breakthrough therapies. The mechanisms underpinning obesity-related vascular disease remain elusive and represent an intense area of investigation. Recent Advances: Epigenetic modifications-defined as environmentally induced chemical changes of DNA and histones that do not affect DNA sequence-are emerging as a potent modulator of gene transcription in the vasculature and might significantly contribute to the development of obesity-induced endothelial dysfunction. DNA methylation and histone post-translational modifications cooperate to build complex epigenetic signals, altering transcriptional networks that are implicated in redox homeostasis, mitochondrial function, vascular inflammation, and perivascular fat homeostasis in patients with cardiometabolic disturbances. Critical Issues: Deciphering the epigenetic landscape in the vasculature is extremely challenging due to the complexity of epigenetic signals and their function in regulating transcription. An overview of the most important epigenetic pathways is required to identify potential molecular targets to treat or prevent obesity-related endothelial dysfunction and atherosclerotic disease. This would enable the employment of precision medicine approaches in this setting. Future Directions: Current and future research efforts in this field entail a better definition of the vascular epigenome in obese patients as well as the unveiling of novel, cell-specific chromatin-modifying drugs that are able to erase specific epigenetic signals that are responsible for maladaptive transcriptional alterations and vascular dysfunction in obese patients. Antioxid. Redox Signal. 34, 1165-1199.
Collapse
Affiliation(s)
- Stefano Masi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Sebastiano Sciarretta
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Heart Division, Royal Brompton and Harefield Hospital Trust, National Heart & Lung Institute, Imperial College, London, United Kingdom
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| |
Collapse
|
11
|
|
12
|
Askari M, Mozaffari H, Darooghegi Mofrad M, Jafari A, Surkan PJ, Amini MR, Azadbakht L. Effects of garlic supplementation on oxidative stress and antioxidative capacity biomarkers: A systematic review and meta-analysis of randomized controlled trials. Phytother Res 2021; 35:3032-3045. [PMID: 33484037 DOI: 10.1002/ptr.7021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 12/30/2020] [Accepted: 01/03/2021] [Indexed: 01/24/2023]
Abstract
Evidence suggests that garlic supplementation may have an effect on oxidative stress by augmenting the rate of enzymatic and non-enzymatic antioxidants and diminishing pro-oxidant enzymes. Given inconsistencies across studies, we aimed to systematically review the current literature and quantify the effects of garlic supplementation on oxidative stress. We conducted a systematic search with multiple databases (Scopus, PubMed, and Web of Science) to find relevant articles published prior to October 2020. Results were reported as bias-corrected standardized mean difference (Hedges' g) with 95% confidence intervals (CI) using random-effects models. Cochrane's Q and I squared (I2 ) tests were used to determine heterogeneity among the studies included. Twelve randomized controlled trials (RCTs) were included. Garlic doses ranged from 80 to 4,000 mg/day, and intervention duration varied between 2 and 24 weeks. Garlic supplementation increased serum level of total antioxidant capacity (TAC) (Hedges' g: 2.77, 95% CI: 1.37 to 4.17, p < 0.001) and superoxide dismutase (SOD) (Hedges' g: 13.76, 95% CI: 4.24 to 23.29, p = 0.004), while it reduced the malondialdehyde serum level (MDA) (Hedges' g: -1.94, 95% CI: -3.17 to -0.70, p = 0.002). Due to limited data available, glutathione (GSH) was not considered for the current meta-analysis. The nonlinear dose-response effect of garlic supplementation was not observed with regard to serum TAC and MDA levels (TAC: p-nonlinearity = 0.398; MDA: p-nonlinearity = 0.488). Garlic supplementation appears to improve serum levels of TAC, MDA, and SOD. Garlic supplementation may be useful to reduce oxidative stress and related diseases. Future studies with large sample sizes and longer duration are required to confirm these findings.
Collapse
Affiliation(s)
- Mohammadreza Askari
- Department of Community Nutrition, School of Nutritional Science and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Hadis Mozaffari
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, British Columbia, Canada
| | - Manije Darooghegi Mofrad
- Department of Community Nutrition, School of Nutritional Science and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Jafari
- Department of Community Nutrition, School of Nutritional Science and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Pamela J Surkan
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Mohammad Reza Amini
- Department of Clinical Nutrition, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Azadbakht
- Department of Community Nutrition, School of Nutritional Science and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.,Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Yang Y, Huang Z, Li LL. Advanced nitric oxide donors: chemical structure of NO drugs, NO nanomedicines and biomedical applications. NANOSCALE 2021; 13:444-459. [PMID: 33403376 DOI: 10.1039/d0nr07484e] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Nitric oxide (NO), as an endogenous diatomic molecule, plays a key regulatory role in many physiological and pathological processes. This diatomic free radical has been shown to affect different physiological and cellular functions and participates in many regulatory functions ranging from changing the cardiovascular system to regulating neuronal functions. Thus, NO gas therapy as an emerging and promising treatment method has attracted increasing attention in the treatment of various pathological diseases. As is known, the physiological and pathological regulation of NO depends mainly on its location, exposure time and released dosage. However, NO gas lacks effective accumulation and controlled long-term gas releasing capacity at specific sites, resulting in limited therapeutic efficacy and potential side effects. Thus, researchers have developed various NO donors, but eventually found that it is still difficult to control the long-term release of NO. Inspired by the self-assembly properties of nanomaterials, researchers have realized that nanomaterials can be used to support NO donors to form nanomedicine to achieve spatial and temporal controlled release of NO. In this review, according to the history of the medicinal development of NO, we first summarize the chemical design of NO donors, NO prodrugs, and NO-conjugated drugs. Then, NO nanomedicines formed by various nanomaterials and NO donors depending on nanotechnology are highlighted. Finally, the biomedical applications of NO nanomedicine with optimized properties are summarized.
Collapse
Affiliation(s)
- Yueqi Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P. R. China. and Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, P. R. China.
| | - Zhangjian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Li-Li Li
- Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, P. R. China.
| |
Collapse
|
14
|
Zhou H, Tu Q, Zhang Y, Xie HQ, Shuai QY, Huang XC, Fu J, Cao Z. Shear stress improves the endothelial progenitor cell function via the CXCR7/ERK pathway axis in the coronary artery disease cases. BMC Cardiovasc Disord 2020; 20:403. [PMID: 32894067 PMCID: PMC7487552 DOI: 10.1186/s12872-020-01681-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Dysfunction in the late Endothelial Progenitor Cells (EPCs) is responsible for endothelial repair in patients with Coronary Artery Disease (CAD), and the shear stress is beneficial for EPCs function. However, the impact of shear stress on the capacity of EPCs in CAD patients has not been elucidated yet. The C-X-C chemokine receptor 7/extracellular signal-regulated kinase (CXCR7)/(ERK) pathways are identified to regulate EPCs function in CAD patients. Here, we hypothesize that shear stress upregulates the CXCR7/ERK pathways, which restore the EPCs function in CAD patients. METHODS The human Peripheral Blood Mononuclear Cells (PBMCs) were collected from healthy adults and CAD patients and then used for EPCs cultivation. The Lv-siRNA for human CXCR7 was transfected into induced EPCs isolated from the CAD patients. Meanwhile, the EPCs from CAD patients were subjected to shear stress generated by a biomimetic device. Next, the cell viability, migration, tube formation, and apoptosis were detected by CCK-8, Transwell assay, Matrigel, and flow cytometry, respectively. Also, the CXCR7/ERK pathways in human EPCs were analyzed by Western blotting and qRT-PCR. RESULT Compared to the EPCs collected from normal adults, the CAD patient-derived EPCs showed reduced in vitro vasculogenic capacity. Also, the level of CXCR7 in CAD patient-derived EPCs was significantly reduced compared to the EPCs of healthy subjects. Meanwhile, the extracellular signal-regulated kinase (ERK), which represents a CXCR7 downstream signaling pathway, had decreased phosphorylation level. The shear stress treatment augmented the CXCR7 expression and also elevated ERK phosphorylation, which is comparable to the up-regulation of CAD patient-derived EPCs function. Further, the small interfering RNA (siRNA)-mediated CXCR7 knockdown diminished the enhanced migration, adhesion, and tube formation capacity of shear stress treated CAD patient-derived EPCs. CONCLUSION Up-regulation of the CXCR7/ERK pathways by shear stress can be a promising new target in enhancing the vasculogenic ability of CAD patient-derived EPCs.
Collapse
Affiliation(s)
- Hua Zhou
- Department of Medical Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Qiang Tu
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Yan Zhang
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Hua Qiang Xie
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Qing Yun Shuai
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Xiao Chuan Huang
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Jie Fu
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Zheng Cao
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| |
Collapse
|
15
|
Down-regulation of the insulin signaling pathway by SHC may correlate with congenital heart disease in Chinese populations. Clin Sci (Lond) 2020; 134:349-358. [PMID: 31971563 DOI: 10.1042/cs20190255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND/AIMS Congenital heart disease (CHD) is one of the most common and severe congenital defects. The incidence of fetal cardiac malformation is increased in the context of maternal gestational diabetes mellitus (GDM). Therefore, we wanted to determine whether abnormalities in the insulin signaling pathway are associated with the occurrence of nonsyndromic CHD (ns-CHD). METHODS We used digital gene expression profiling (DGE) of right atrial myocardial tissue samples from eight ns-CHD patients and four controls. The genes potentially associated with CHD were validated by real-time fluorescence quantitative PCR analysis of right atrial myocardial tissues from 37 patients and 10 controls and the H9C2 cell line. RESULTS The results showed that the insulin signaling pathway, which is mediated by the SHC gene family, was inhibited in the ns-CHD patients. The expression levels of five genes (PTPRF, SHC4, MAP2K2, MKNK2, and ELK1) in the pathway were significantly down-regulated in the patients' atrial tissues (P<0.05 for all). In vitro, the H9C2 cells cultured in high glucose (33 mmol/l) expressed less SHC4, MAP2K2, and Elk-1 than those cultured in low glucose (25 mmol/l). Furthermore, the high glucose concentration down-regulated the 25 genes associated with blood vessel development based on Gene Ontology (GO) term enrichment analyses of RNA-seq data. CONCLUSION We considered that changes in the insulin signaling pathway mediated by SHC might be involved in the heart development process. This mechanism might account for the increase in the incidence of fetal cardiac malformations in the context of GDM.
Collapse
|
16
|
Abstract
Endothelial cell (EC) metabolism is important for health and disease. Metabolic pathways, such as glycolysis, fatty acid oxidation, and amino acid metabolism, determine vasculature formation. These metabolic pathways have different roles in securing the production of energy and biomass and the maintenance of redox homeostasis in vascular migratory tip cells, proliferating stalk cells, and quiescent phalanx cells, respectively. Emerging evidence demonstrates that perturbation of EC metabolism results in EC dysfunction and vascular pathologies. Here, we summarize recent insights into EC metabolic pathways and their deregulation in vascular diseases. We further discuss the therapeutic implications of targeting EC metabolism in various pathologies.
Collapse
Affiliation(s)
- Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; ,
| | - Anil Kumar
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; ,
| | - Peter Carmeliet
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; , .,Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven B-3000, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven B-3000, Belgium
| |
Collapse
|
17
|
Jun TJ, Jelani AM, Omar J, Rahim RA, Yaacob NM. Serum Anti-Müllerian Hormone in Polycystic Ovary Syndrome and its Relationship with Insulin Resistance, Lipid Profile and Adiponectin. Indian J Endocrinol Metab 2020; 24:191-195. [PMID: 32699789 PMCID: PMC7333742 DOI: 10.4103/ijem.ijem_305_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 11/29/2019] [Accepted: 12/16/2019] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVES This study was done to estimate serum anti-Müllerian hormone (AMH) level in polycystic ovary syndrome (PCOS) patients and to correlate serum AMH level with insulin resistance, lipid profile, and adiponectin levels. MATERIALS AND METHODS A cross-sectional study was conducted at Hospital Universiti Sains Malaysia (Hospital USM), Health Campus, Kubang Kerian, Kelantan, Malaysia. Thirty newly diagnosed patients with PCOS attending gynecology clinic between July 2016 and April 2017 were recruited. Fasting venous blood samples were collected from the subjects. Serum AMH, insulin, adiponectin, triglycerides, high-density lipoprotein cholesterol (HDL-C), and plasma glucose levels were measured, and insulin resistance was calculated based on homeostasis model of assessment-insulin resistance (HOMA-IR). The serum AMH level was estimated, and the correlation of serum AMH level with the metabolic parameters was analyzed. RESULTS The median of serum AMH levels in women with PCOS was 6.8 ng/mL (interquartile range: 7.38 ng/mL). There was a significant negative correlation between serum AMH and HOMA-IR or triglyceride levels (r = -0.49, P = 0.006 and r = -0.55, P = 0.002, respectively). A significant positive correlation was observed between serum AMH and serum HDL-C or serum adiponectin levels (r = 0.56, P = 0.001 and r = 0.44, P = 0.014, respectively) in all study subjects. CONCLUSION The serum AMH level is associated with HOMA-IR, triglycerides, HDL-C, and adiponectin levels, and hence it may be used as a potential cardiometabolic risk marker in women with PCOS.
Collapse
Affiliation(s)
- Thien Jun Jun
- Department of Pathology, Sarawak General Hospital, Kuching, Sarawak, Malaysia
| | - Aniza Mohammed Jelani
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Julia Omar
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Rahimah Abdul Rahim
- Department of Obstetric and Gynaecology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Najib Majdi Yaacob
- Unit of Biostatistic and Research Methodology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
18
|
Sun L, Liu YL, Ye F, Xie JW, Zeng JW, Qin L, Xue J, Wang YT, Guo KM, Ma MM, Tang YB, Li XY, Gao M. Free fatty acid-induced H 2O 2 activates TRPM2 to aggravate endothelial insulin resistance via Ca 2+-dependent PERK/ATF4/TRB3 cascade in obese mice. Free Radic Biol Med 2019; 143:288-299. [PMID: 31445205 DOI: 10.1016/j.freeradbiomed.2019.08.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 08/16/2019] [Accepted: 08/19/2019] [Indexed: 01/12/2023]
Abstract
Transient Receptor Potential Melastatin-2 (TRPM2) is a nonselective cation channel mediating Ca2+ influx in response to oxidative stress. Given that insulin resistance-related endothelial dysfunction in obesity attributes to fatty-acid-induced reactive oxygen species (ROS) overproduction, in this study, we addressed the possible role of TRPM2 in obesity-related endothelial insulin resistance and the underlying mechanisms. Whole-cell patch clamp technique, intracellular Ca2+ concentration measurement, western blot, vasorelaxation assay, and high-fat diet (HFD)-induced obese model were employed to assess the relationship between TRPM2 and endothelial insulin response. We found that both the expression and activity of TRPM2 were higher in endothelial cells of obese mice. Palmitate rose a cationic current in endothelial cells which was inhibited or enlarged by TRPM2 knockdown or overexpression. Silencing of TRPM2 remarkably improved insulin-induced endothelial Akt activation, nitric oxide synthase (eNOS) phosphorylation and nitric oxide (NO) production, while TRPM2 overexpression resulted in the opposite effects. Furthermore, TRPM2-mediated Ca2+ entry, CaMKII activation and the following activation of PERK/ATF4/TRB3 cascade were involved in the mechanism of obesity or palmitate-induced endothelial insulin resistance. Notably, in vivo study, knockdown of TRPM2 with adeno-associated virus harboring short-hairpin RNA (shRNA) against TRPM2 alleviated endothelial insulin resistance and ameliorated endothelium-dependent vasodilatation in obese mice. Thus, these results suggest that TRPM2-activated Ca2+ signaling is necessary to induce insulin resistance-related endothelial dysfunction in obesity. Downregulation or pharmacological inhibition of TRPM2 channels may lead to the development of effective drugs for treatment of endothelial dysfunction associated with oxidative stress state.
Collapse
Affiliation(s)
- Lu Sun
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China; Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yan-Li Liu
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Fang Ye
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing-Wen Xie
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Jia-Wei Zeng
- Department of Pharmacy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li Qin
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Jing Xue
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Yi-Ting Wang
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Kai-Min Guo
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ming-Ming Ma
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yong-Bo Tang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiao-Yan Li
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
| | - Min Gao
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
| |
Collapse
|
19
|
Pikkemaat M, Andersson T, Melander O, Chalmers J, Rådholm K, Bengtsson Boström K. C-peptide predicts all-cause and cardiovascular death in a cohort of individuals with newly diagnosed type 2 diabetes. The Skaraborg diabetes register. Diabetes Res Clin Pract 2019; 150:174-183. [PMID: 30878389 DOI: 10.1016/j.diabres.2019.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/30/2019] [Accepted: 03/07/2019] [Indexed: 02/02/2023]
Abstract
AIMS To study the association between baseline level of C-peptide and all-cause death, cardiovascular death and cardiovascular complications among persons with newly diagnosed type 2 diabetes. METHODS The Skaraborg Diabetes Register contains data on baseline C-peptide concentrations among 398 persons <65 years with newly diagnosed type 2 diabetes 1996-1998. National registries were used to determine all-cause death, cardiovascular death and incidence of myocardial infarction and ischemic stroke until 31 December 2014. The association between baseline C-peptide and outcomes were evaluated with adjustment for multiple confounders by Cox regression analysis. Missing data were handled by multiple imputation. RESULTS In the imputed and fully adjusted model there was a significant association between 1 nmol/l increase in C-peptide concentration and all-cause death (HR 2.20, 95% CI 1.49-3.25, p < 0.001, number of events = 104), underlying cardiovascular death (HR 2.69, 1.49-4.85, p = 0.001, n = 35) and the composite outcome of underlying cardiovascular death, myocardial infarction or ischemic stroke (HR 1.61, 1.06-2.45, p = 0.027, n = 90). CONCLUSIONS Elevated C-peptide levels at baseline in persons with newly diagnosed type 2 diabetes are associated with increased risk of all-cause and cardiovascular mortality. C-peptide might be used to identify persons at high risk of cardiovascular complications and premature death.
Collapse
Affiliation(s)
- Miriam Pikkemaat
- Husensjö Health Care Center, Helsingborg, Sweden; Center for Primary Health Care Research, Department of Clinical Sciences in Malmö, Lund University, Sweden.
| | - Tobias Andersson
- Närhälsan Norrmalm Health Centre, Skövde, Sweden; Department of Public Health and Community Medicine/Primary Health Care, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Olle Melander
- Department of Medicine, Malmö University Hospital, Lund University, Sweden
| | - John Chalmers
- The George Institute for Global Health, Faculty of Medicine, UNSW Sydney, Australia
| | - Karin Rådholm
- The George Institute for Global Health, Faculty of Medicine, UNSW Sydney, Australia; Division of Community Medicine, Primary Care, Department of Medicine and Health Sciences, Faculty of Health Sciences, Linköping University, Department of Local Care West, County Council of Östergötland, Linköping, Sweden
| | - Kristina Bengtsson Boström
- Department of Public Health and Community Medicine/Primary Health Care, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden; R&D Center Skaraborg Primary Care, Skövde, Sweden
| |
Collapse
|
20
|
Guzmán DC, Brizuela NO, Herrera MO, Peraza AV, Juárez-Olguín H, Mejía GB. Insulin plus zinc induces a favorable biochemical response effects on oxidative damage and dopamine levels in rat brain. Int J Biol Macromol 2019; 132:230-235. [PMID: 30928372 DOI: 10.1016/j.ijbiomac.2019.03.200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 01/27/2023]
Abstract
The aim was to determine the effect of zinc (Zn) and insulin on oxidative stress and levels of dopamine in brain of rats. Wistar rats were treated either with zinc alone or combined with insulin during 10 days. After the last dose blood glucose was measured. Their brains were extracted to measure H2O2, Ca+2, Mg+2 ATPase, glutathione (GSH), lipid peroxidation (Tbars) and Dopamine. Zn does not possess anti-glycemic effect like Insulin however, it is noticeable that the combination of Insulin plus Zn induces a major glucose reduction (p < 0.0001) than Insulin alone. In cerebellum/medulla oblongata, the groups treated with Insulin and Zn show a significantly increase in dopamine (p < 0.005). Insulin plus Zn reduced GSH level in cortex. Insulin plus Zn reduced level of H2O2 in Striatum and in cerebellum/medulla oblongata. Lipid peroxidation was significantly reduced by the administration of Insulin as in the combination of Insulin and Zn in all regions (p < 0.0001). In cerebellum medulla oblongata, ATPase activity showed an increase only in the group treated with Insulin + Zn. CONCLUSION: These results suggest that the use of insulin plus Zn produce favorable changes on oxidative stress and this as consequence on the levels of dopamine.
Collapse
Affiliation(s)
- David Calderón Guzmán
- Laboratory of Neurosciences, Instituto Nacional de Pediatría (INP), Mexico City, Mexico
| | - Norma Osnaya Brizuela
- Laboratory of Neurosciences, Instituto Nacional de Pediatría (INP), Mexico City, Mexico
| | | | | | - Hugo Juárez-Olguín
- Laboratory of Pharmacology, INP, Mexico City, Mexico; Department of Pharmacology, Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico.
| | | |
Collapse
|
21
|
Ross M, Lithgow H, Hayes L, Florida-James G. Potential Cellular and Biochemical Mechanisms of Exercise and Physical Activity on the Ageing Process. Subcell Biochem 2019; 91:311-338. [PMID: 30888658 DOI: 10.1007/978-981-13-3681-2_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Exercise in young adults has been consistently shown to improve various aspects of physiological and psychological health but we are now realising the potential benefits of exercise with advancing age. Specifically, exercise improves cardiovascular, musculoskeletal, and metabolic health through reductions in oxidative stress, chronic low-grade inflammation and modulating cellular processes within a variety of tissues. In this this chapter we will discuss the effects of acute and chronic exercise on these processes and conditions in an ageing population, and how physical activity affects our vasculature, skeletal muscle function, our immune system, and cardiometabolic risk in older adults.
Collapse
Affiliation(s)
- Mark Ross
- School of Applied Science, Edinburgh Napier University, Edinburgh, Scotland, UK.
| | - Hannah Lithgow
- School of Applied Science, Edinburgh Napier University, Edinburgh, Scotland, UK
| | - Lawrence Hayes
- Active Ageing Research Group, University of Cumbria, Lancaster, UK
| | | |
Collapse
|
22
|
Pathophysiological mechanisms of diabetic cardiomyopathy and the therapeutic potential of epigallocatechin-3-gallate. Biomed Pharmacother 2018; 109:2155-2172. [PMID: 30551473 DOI: 10.1016/j.biopha.2018.11.086] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/14/2018] [Accepted: 11/20/2018] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular complications are considered one of the leading causes of morbidity and mortality among diabetic patients. Diabetic cardiomyopathy (DCM) is a type of cardiovascular damage presents in diabetic patients independent of the coexistence of ischemic heart disease or hypertension. It is characterized by impaired diastolic relaxation time, myocardial dilatation and hypertrophy and reduced systolic and diastolic functions of the left ventricle. Molecular mechanisms underlying these pathological changes in the diabetic heart are most likely multifactorial and include, but not limited to, oxidative/nitrosative stress, increased advanced glycation end products, mitochondrial dysfunction, inflammation and cell death. The aim of this review is to address the major molecular mechanisms implicated in the pathogenesis of DCM. In addition, this review provides studies conducted to determine the pharmacological effects of (-)-epigallocatechin-3-gallate (EGCG), the major polyphenol in green tea, focusing on its therapeutic potential against the processes involved in the pathogenesis and progression of DCM. EGCG has been shown to exert several potential therapeutic properties both in vitro and in vivo. Given its therapeutic potential, EGCG might be a promising drug candidate to decrease the morbidity and mortality associated with DCM and other diabetes complications.
Collapse
|
23
|
Saeedi Borujeni MJ, Esfandiary E, Baradaran A, Valiani A, Ghanadian M, Codoñer-Franch P, Basirat R, Alonso-Iglesias E, Mirzaei H, Yazdani A. Molecular aspects of pancreatic β-cell dysfunction: Oxidative stress, microRNA, and long noncoding RNA. J Cell Physiol 2018; 234:8411-8425. [PMID: 30565679 DOI: 10.1002/jcp.27755] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/23/2018] [Indexed: 02/06/2023]
Abstract
Metabolic syndrome is known as a frequent precursor of type 2 diabetes mellitus (T2D). This disease could affect 8% of the people worldwide. Given that pancreatic β-cell dysfunction and loss have central roles in the initiation and progression of the disease, the understanding of cellular and molecular pathways associated with pancreatic β-cell dysfunction can provide more information about the underlying pathways involved in T2D. Multiple lines evidence indicated that oxidative stress, microRNA, and long noncoding RNA play significant roles in various steps of diseases. Oxidative stress is one of the important factors involved in T2D pathogenesis. This could affect the function and survival of the β cell via activation or inhibition of several processes and targets, such as receptor-signal transduction, enzyme activity, gene expression, ion channel transport, and apoptosis. Besides oxidative stress, microRNAs and noncoding RNAs have emerged as epigenetic regulators that could affect pancreatic β-cell dysfunction. These molecules exert their effects via targeting a variety of cellular and molecular pathways involved in T2D pathogenesis. Here, we summarized the molecular aspects of pancreatic β-cell dysfunction. Moreover, we highlighted the roles of oxidative stress, microRNAs, and noncoding RNAs in pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Mohammad Javad Saeedi Borujeni
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ebrahim Esfandiary
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azar Baradaran
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Valiani
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mustafa Ghanadian
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pilar Codoñer-Franch
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - Reyhane Basirat
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Amid Yazdani
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
24
|
Epigenetic processing in cardiometabolic disease. Atherosclerosis 2018; 281:150-158. [PMID: 30290963 DOI: 10.1016/j.atherosclerosis.2018.09.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/24/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Abstract
Albeit a consistent body of evidence supports the notion that genes influence cardiometabolic features and outcomes, the "non-genetic regulation" of this process is gaining increasing attention. Plastic chemical changes of DNA/histone complexes - known as epigenetic changes - critically determine gene activity by rapidly modifying chromatin accessibility to transcription factors. In this review, we describe the emerging role of chromatin modifications as fine tuners of gene transcription in adipogenesis, insulin resistance, macrophage polarization, immuno-metabolism, endothelial dysfunction and metabolic cardiomyopathy. Epigenetic processing participates in the dynamic interplay among different organs in the cardiometabolic patient. DNA methylation and post-translational histone modifications in both visceral and subcutaneous adipose tissue enable the transcription of genes implicated in lipo- and adipogenesis, inflammation and insulin resistance. Along the same line, complex networks of chromatin modifying enzymes are responsible for impaired nitric oxide bioavailability and defective insulin signalling in the vasculature, thus leading to reduced capillary recruitment and insulin delivery in the liver, skeletal muscle and adipose tissue. Furthermore, changes in methylation status of IL-4, IFNγ and Forkhead box P3 (Foxp3) gene loci are crucial for the polarization of immune cells, thus leading to adipose tissue inflammation and atherosclerosis. Cell-specific epigenetic information could advance our understanding of cardiometabolic processes, thus leading to individualized risk assessment and personalized therapeutic approaches in patients with cardiometabolic disturbances. The development of new chromatin modifying drugs indicates that targeting epigenetic changes is a promising approach to reduce the burden of cardiovascular disease in this setting.
Collapse
|
25
|
Oikonomou EK, Antoniades C. Immunometabolic Regulation of Vascular Redox State: The Role of Adipose Tissue. Antioxid Redox Signal 2018; 29:313-336. [PMID: 28657335 DOI: 10.1089/ars.2017.7017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE Vascular oxidative stress plays a crucial role in atherogenesis and cardiovascular disease (CVD). Recent evidence suggests that vascular redox state is under the control of complex pathophysiological mechanisms, ranging from inflammation to obesity and insulin resistance (IR). Recent Advances: Adipose tissue (AT) is now recognized as a dynamic endocrine and paracrine organ that secretes several bioactive molecules, called adipokines. AT has recently been shown to regulate vascular redox state in both an endocrine and a paracrine manner through the secretion of adipokines, therefore providing a mechanistic link for the association between obesity, IR, inflammation, and vascular disease. Importantly, AT behaves as a sensor of cardiovascular oxidative stress, modifying its secretory profile in response to cardiovascular oxidative injury. CRITICAL ISSUES The present article presents an up-to-date review of the association between AT and vascular oxidative stress. We focus on the effects of individual adipokines on modulating reactive oxygen species production and scavenging in the vascular wall. In addition, we highlight how inflammation, obesity, and IR alter the biology and secretome of AT leading to a more pro-oxidant phenotype with a particular focus on the local regulatory mechanisms of perivascular AT driven by vascular oxidation. FUTURE DIRECTIONS The complex and dynamic biology of AT, as well as its importance in the regulation of vascular redox state, provides numerous opportunities for the development of novel, targeted treatments in the management of CVD. Therapeutic modulation of AT biology could improve vascular redox state affecting vascular disease pathogenesis. Antioxid. Redox Signal. 29, 313-336.
Collapse
Affiliation(s)
- Evangelos K Oikonomou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford , Oxford, United Kingdom
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford , Oxford, United Kingdom
| |
Collapse
|
26
|
Zhang H, Li Y, Mao Z, Liu X, Zhang X, Yang K, Liu R, Qian X, Zhang H, Jiang J, Zhang G, Wang C. Sex-specific associations of serum uric acid with metabolic syndrome in Chinese rural population: The RuralDiab study. Clin Chim Acta 2018; 480:119-125. [DOI: 10.1016/j.cca.2018.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 02/03/2018] [Accepted: 02/05/2018] [Indexed: 12/22/2022]
|
27
|
Abstract
Chronic diseases are defined diseases whose symptoms last for at least six months and tend to worsen over time. In Europe, they cause at least 86% of deaths. In this speculative unifying model I set a new hypothesis for the etiology of the majority of chronic diseases. The main aim is to put order and observe our organism in a systemic way, connecting pathologies we now see as disconnected phenomena, with the conceptual frameworks of complex systems and network medicine. Chronic diseases could be caused by a first unsolved acute infection. In case the pathogen cannot be completely eliminated, it becomes a persistent infectious. After the acute episode, some mild symptoms will occur and probably disappear; the chronic disease will remain latent over time. It will manifest even after years or decades, in the presence of another acute infection, a particular stress, trauma, or another event. The presence of the persistent infectious elicits changes in the immune and systemic regulation, and these processes degenerate over time. They will assume their rules and patterns, being independent from the initial stimulus. The key to understand the dynamics and individuality of chronic diseases is the immune system and its networks. The immune mechanisms that can lead to the persistent response are mainly the switch from the Th1 to the Th2 immunity and the molecular mimicry. The first persistent infectious will also modify the susceptibility to other pathogens, facilitating new infections and new consequent persistent infectious. From the immune point of view, our organism is divided into three compartments: the outer one, which comprehend all the surfaces in contact with the environment, the intermediate one, which comprehend the internal organs and tissues, and the innermost one, comprehending the Central Nervous System and the adluminal compartment of the seminiferous tubule. The immune key-role is played respectively by the mucosa-associated lymphoid tissue, the endothelium, the blood-brain barrier and blood-testis barrier. The chronic diseases follow a progressive scheme, involving the three compartments from the outer to the innermost one. The primer microorganism at the origin of the majority of diseases could be streptococcus, or staphylococcus. Both cause acute in children, with a great variability of responses and symptoms, and both cause molecular mimicry. This model can be tested and proved in more ways, I propose here some of them. It could pave the way to a radical change in our comprehension and therapeutic approaches to chronic diseases.
Collapse
|
28
|
Eelen G, de Zeeuw P, Treps L, Harjes U, Wong BW, Carmeliet P. Endothelial Cell Metabolism. Physiol Rev 2018; 98:3-58. [PMID: 29167330 PMCID: PMC5866357 DOI: 10.1152/physrev.00001.2017] [Citation(s) in RCA: 330] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 06/19/2017] [Accepted: 06/22/2017] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) are more than inert blood vessel lining material. Instead, they are active players in the formation of new blood vessels (angiogenesis) both in health and (life-threatening) diseases. Recently, a new concept arose by which EC metabolism drives angiogenesis in parallel to well-established angiogenic growth factors (e.g., vascular endothelial growth factor). 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3-driven glycolysis generates energy to sustain competitive behavior of the ECs at the tip of a growing vessel sprout, whereas carnitine palmitoyltransferase 1a-controlled fatty acid oxidation regulates nucleotide synthesis and proliferation of ECs in the stalk of the sprout. To maintain vascular homeostasis, ECs rely on an intricate metabolic wiring characterized by intracellular compartmentalization, use metabolites for epigenetic regulation of EC subtype differentiation, crosstalk through metabolite release with other cell types, and exhibit EC subtype-specific metabolic traits. Importantly, maladaptation of EC metabolism contributes to vascular disorders, through EC dysfunction or excess angiogenesis, and presents new opportunities for anti-angiogenic strategies. Here we provide a comprehensive overview of established as well as newly uncovered aspects of EC metabolism.
Collapse
Affiliation(s)
- Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Pauline de Zeeuw
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Ulrike Harjes
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Brian W Wong
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium; and Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| |
Collapse
|
29
|
Insulin Resistance and Its Association with Metabolic Syndrome in Korean Children. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8728017. [PMID: 29457038 PMCID: PMC5804402 DOI: 10.1155/2017/8728017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/21/2017] [Accepted: 12/03/2017] [Indexed: 01/27/2023]
Abstract
Background This study investigated the association between insulin resistance (IR) and metabolic syndrome (MetS) in children. Methods A cross-sectional study involving 1036 healthy children aged between 7 and 13 years was conducted. Homeostasis model assessment of insulin resistance (HOMA-IR) was calculated as an index of IR. Participants were classified according to the HOMA-IR quartiles. Results Incremental, linear trends were found in age (p < 0.001), body mass index (BMI) (p < 0.001), body fat (p < 0.001), waist circumference (p < 0.001), resting blood pressures (BP) (p < 0.001), triglycerides (TG) (p < 0.001), total cholesterol (TC) (p < 0.001), high density lipoprotein-cholesterol (HDL-C) (p < 0.001), FBG (p < 0.001), and insulin (<0.001) according to incremental HOMA-IR categories (from the 1st to 4th quartile). Compared with children in the 1st HOMA-IR quartile, children in the 4th HOMA-IR quartile had significantly higher odd ratios (ORs) of abnormalities in systolic (p = 0.051) and diastolic BP (p = 0.005), FBG (p < 0.001), TG (p < 0.001), TC (p = 0.016), and HDL-C (p = 0.006) even after adjustments for age, gender, BMI, and body fat percentage. Children in the 3rd HOMA-IR quartile had significant abnormalities in FBG (p < 0.001), TG (p = 0.001), and HDL-C (p = 0.010) even after adjustments for the covariates. Conclusion The current findings suggest that IR is significantly associated with the clustering of MetS risk factors in children in Korea.
Collapse
|
30
|
Abstract
The review presents modern information on the development of disorders of endothelium-dependent vascular reactivity in diabetes mellitus (DM) type 2. In type 2 DM, disorders of endothelium-dependent vascular reactivity associated with hyperglycemia and oxidative stress, manifesting by a reduced vascular response to vasodilators and pressor (paradoxical) vascular reactions to them, directly associated with cardiovascular events are observed.
Collapse
|
31
|
Natarajan G, Perriotte-Olson C, Bhinderwala F, Powers R, Desouza CV, Talmon GA, Yuhang J, Zimmerman MC, Kabanov AV, Saraswathi V. Nanoformulated copper/zinc superoxide dismutase exerts differential effects on glucose vs lipid homeostasis depending on the diet composition possibly via altered AMPK signaling. Transl Res 2017; 188:10-26. [PMID: 28867395 PMCID: PMC5819896 DOI: 10.1016/j.trsl.2017.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 12/17/2022]
Abstract
Evidence suggests that superoxide dismutase 1 (SOD1) promotes glucose vs lipid metabolism depending on the diet type. We recently reported that nanoformulated SOD1 (Nano) improved lipid metabolism without altering glucose homeostasis in high-fat (HF) diet-fed mice. Here, we sought to determine the effects and potential mechanisms of Nano in modulating glucose and lipid homeostasis in mice fed a normal chow diet (CD) vs HF diet. Mice were fed a CD or a HF diet (45%) for 10 wk and injected with Nano once every 2 days for 15 days. The fasting glucose level was lower (P < 0.05) in CD + Nano-treated mice compared to control. Conversely, blood glucose was not altered but serum triglycerides were lower in HF + Nano-treated mice. Genes involved in fatty acid synthesis were reduced by Nano in the skeletal muscle of CD but not of HF diet-fed mice. Adenosine monophosphate-activated protein kinase (AMPK), which promotes both glucose and lipid metabolism depending on the fuel availability, is activated by Nano in CD-fed mice. Moreover, Nano increased phosphorylation of ACC, a downstream target of AMPK, in both CD and HF diet-fed mice. Nano increased mitochondrial respiration in C2C12 myocytes in the presence of glucose or fatty acid, and this effect is inhibited by Compound C, an AMPK inhibitor. Our data suggest that Nano promotes glucose and lipid metabolism in CD and HF diet-fed mice, respectively, and this effect is mediated partly via AMPK signaling.
Collapse
Affiliation(s)
- Gopalakrishnan Natarajan
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Neb
| | - Curtis Perriotte-Olson
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Neb
| | - Fatema Bhinderwala
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Neb; Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Neb
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Neb; Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Neb
| | - Cyrus V Desouza
- VA Nebraska-Western Iowa Health Care System, Omaha, Neb; Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Neb
| | - Geoffrey A Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Neb
| | - Jiang Yuhang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Matthew C Zimmerman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Neb
| | - Alexander V Kabanov
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Viswanathan Saraswathi
- Department of Internal Medicine/Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, Neb; VA Nebraska-Western Iowa Health Care System, Omaha, Neb.
| |
Collapse
|
32
|
Huang LY, Yen IC, Tsai WC, Ahmetaj-Shala B, Chang TC, Tsai CS, Lee SY. Rhodiola crenulata Attenuates High Glucose Induced Endothelial Dysfunction in Human Umbilical Vein Endothelial Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:1201-1216. [DOI: 10.1142/s0192415x17500665] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Rhodiola crenulata root extract (RCE), a traditional Chinese medicine, has been shown to regulate glucose and lipid metabolism via the AMPK pathway in high glucose (HG) conditions. However, the effect of RCE on HG-induced endothelial dysfunction remains unclear. The present study was designed to examine the effects and mechanisms of RCE against hyperglycemic insult in endothelial cells. Human umbilical vein endothelial cells (HUVECs) were pretreated with or without RCE and then exposed to 33[Formula: see text]mM HG medium. The cell viability, nitrite production, oxidative stress markers, and vasoactive factors, as well as the mechanisms underlying RCE action, were then investigated. We found that RCE significantly improved cell death, nitric oxide (NO) defects, and oxidative stress in HG conditions. In addition, RCE significantly decreased the HG-induced vasoactive markers, including endothelin-1 (ET-1), fibronectin, and vascular endothelial growth factor (VEGF). However, the RCE-restored AMPK-Akt-eNOS-NO axis and cell viability were abolished by the presence of an AMPK inhibitor. These findings suggested that the protective effects of RCE were associated with the AMPK-Akt-eNOS-NO signaling pathway. In conclusion, we showed that RCE protected endothelial cells from hyperglycemic insult and demonstrated its potential for use as a treatment for endothelial dysfunction in diabetes mellitus.
Collapse
Affiliation(s)
- Li-Yen Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - I-Chuan Yen
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Cheng Tsai
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | | | - Tsu-Chung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Sung Tsai
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Taoyuan Armed General Forces General Hospital, Taoyuan, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, Taipei, Taiwan
| | - Shih-Yu Lee
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
33
|
Haberzettl P, O’Toole TE, Bhatnagar A, Conklin DJ. Exposure to Fine Particulate Air Pollution Causes Vascular Insulin Resistance by Inducing Pulmonary Oxidative Stress. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1830-1839. [PMID: 27128347 PMCID: PMC5132639 DOI: 10.1289/ehp212] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 09/28/2015] [Accepted: 04/12/2016] [Indexed: 05/19/2023]
Abstract
BACKGROUND Epidemiological evidence suggests that exposure to ambient air fine particulate matter (PM2.5) increases the risk of developing type 2 diabetes and cardiovascular disease. However, the mechanisms underlying these effects of PM2.5 remain unclear. OBJECTIVES We tested the hypothesis that PM2.5 exposure decreases vascular insulin sensitivity by inducing pulmonary oxidative stress. METHODS Mice fed control (10-13% kcal fat) and high-fat (60% kcal fat, HFD) diets, treated with 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl (TEMPOL) or mice overexpressing lung-specific extracellular superoxide dismutase (ecSOD) were exposed to HEPA-filtered air or to concentrated PM2.5 (CAP) for 9 or 30 days, and changes in systemic and organ-specific insulin sensitivity and inflammation were measured. RESULTS In control diet-fed mice, exposure to CAP for 30 days decreased insulin-stimulated Akt phosphorylation in lung, heart, and aorta but not in skeletal muscle, adipose tissue, and liver and did not affect adiposity or systemic glucose tolerance. In HFD-fed mice, 30-day CAP exposure suppressed insulin-stimulated endothelial nitric oxide synthase (eNOS) phosphorylation in skeletal muscle and increased adipose tissue inflammation and systemic glucose intolerance. In control diet-fed mice, a 9-day CAP exposure was sufficient to suppress insulin-stimulated Akt and eNOS phosphorylation and to decrease IκBα (inhibitor of the transcription factor NF-κB levels in the aorta. Treatment with the antioxidant TEMPOL or lung-specific overexpression of ecSOD prevented CAP-induced vascular insulin resistance and inflammation. CONCLUSIONS Short-term exposure to PM2.5 induces vascular insulin resistance and inflammation triggered by a mechanism involving pulmonary oxidative stress. Suppression of vascular insulin signaling by PM2.5 may accelerate the progression to systemic insulin resistance, particularly in the context of diet-induced obesity. Citation: Haberzettl P, O'Toole TE, Bhatnagar A, Conklin DJ. 2016. Exposure to fine particulate air pollution causes vascular insulin resistance by inducing pulmonary oxidative stress. Environ Health Perspect 124:1830-1839; http://dx.doi.org/10.1289/EHP212.
Collapse
Affiliation(s)
- Petra Haberzettl
- Address correspondence to P. Haberzettl, Diabetes and Obesity Center, Delia Baxter Building II, 580 S. Preston St., Room 404F, University of Louisville, Louisville, KY 40202 USA. Telephone: (502) 852-4235. E-mail:
| | | | | | | |
Collapse
|
34
|
Zhao W, Wu C, Li S, Chen X. Adiponectin protects palmitic acid induced endothelial inflammation and insulin resistance via regulating ROS/IKKβ pathways. Cytokine 2016; 88:167-176. [DOI: 10.1016/j.cyto.2016.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 12/24/2022]
|
35
|
Gao L, Zhao YC, Liang Y, Lin XH, Tan YJ, Wu DD, Li XZ, Ye BZ, Kong FQ, Sheng JZ, Huang HF. The impaired myocardial ischemic tolerance in adult offspring of diabetic pregnancy is restored by maternal melatonin treatment. J Pineal Res 2016; 61:340-52. [PMID: 27299979 DOI: 10.1111/jpi.12351] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/13/2016] [Indexed: 01/01/2023]
Abstract
Diabetic pregnancy, with ever increasing prevalence, adversely affects embryogenesis and increases vasculometabolic disorder risks in adult offspring. However, it remains poorly understood whether maternal diabetes increases the offspring's susceptibility to heart injuries in adulthood. In this study, we observed that cardiac function and structure were comparable between adult offspring born to diabetic mice and their counterparts born to nondiabetic mice at baseline. However, in response to myocardial ischemia/reperfusion (MIR), diabetic mother offspring exhibited augmented infarct size, cardiac dysfunction, and myocardial apoptosis compared with control, in association with exaggerated activation of mitochondria- and endoplasmic reticulum (ER) stress-mediated apoptosis pathways and oxidative stress. Molecular analysis showed that the impaired myocardial ischemic tolerance in diabetic mother offspring was mainly attributable to blunted cardiac insulin receptor substrate (IRS)-1/Akt signaling. Furthermore, the effect of maternal melatonin administration on offspring's response to MIR was determined, and the results indicated that melatonin treatment in diabetic dams during pregnancy significantly improved the tolerance to MIR injury in their offspring, via restoring cardiac IRS-1/Akt signaling. Taken together, these data suggest that maternal diabetes predisposes offspring to augmented MIR injury in adulthood, and maternal melatonin supplementation during diabetic pregnancy may hold promise for improving myocardial ischemic tolerance in the offspring.
Collapse
Affiliation(s)
- Ling Gao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi-Chao Zhao
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Liang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xian-Hua Lin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ya-Jing Tan
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dan-Dan Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin-Zhu Li
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo-Zhi Ye
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Fan-Qi Kong
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jian-Zhong Sheng
- The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, China
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - He-Feng Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- The Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, China.
- Institute of Embryo-Fetal Original Adult Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
36
|
Takata K, Imaizumi S, Zhang B, Miura SI, Saku K. Stabilization of high-risk plaques. Cardiovasc Diagn Ther 2016; 6:304-21. [PMID: 27500090 DOI: 10.21037/cdt.2015.10.03] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The prevalence of atherosclerotic cardiovascular diseases (ASCVDs) is increasing globally and they have become the leading cause of death in most countries. Numerous experimental and clinical studies have been conducted to identify major risk factors and effective control strategies for ASCVDs. The development of imaging modalities with the ability to determine the plaque composition enables us to further identify high-risk plaque and evaluate the effectiveness of different treatment strategies. While intensive lipid-lowering by statins can stabilize or even regress plaque by various mechanisms, such as the reduction of lipid accumulation in a necrotic lipid core, the reduction of inflammation, and improvement of endothelial function, there are still considerable residual risks that need to be understood. We reviewed important findings regarding plaque vulnerability and some encouraging emerging approaches for plaque stabilization.
Collapse
Affiliation(s)
- Kohei Takata
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Satoshi Imaizumi
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Bo Zhang
- Department of Biochemistry, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Shin-Ichiro Miura
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Keijiro Saku
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| |
Collapse
|
37
|
Yang F, Li M, Liu Y, Wang T, Feng Z, Cui H, Gu N. Glucose and magnetic-responsive approach toward in situ nitric oxide bubbles controlled generation for hyperglycemia theranostics. J Control Release 2016; 228:87-95. [PMID: 26951926 DOI: 10.1016/j.jconrel.2016.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/23/2016] [Accepted: 03/01/2016] [Indexed: 01/01/2023]
Abstract
Stimuli-responsive devices that deliver drugs or imaging contrast agents in spatial-, temporal- and dosage-controlled fashions have emerged as the most promising and valuable platform for targeted and controlled drug delivery. However, implementing high performance of these functions in one single delivery carrier remains extremely challenging. Herein, we have developed a sequential strategy for developing glucose and magnetic-responsive microvesicle delivery system, which regulates the glucose levels and spatiotemporally controls the generation of nitric oxide gas free bubbles. It is observed that such injectable microvesicles loaded with enzyme and magnetic nanoparticles can firstly regulate hyperglycemic level based on the enzymatic reactions between glucose oxidase and glucose. In a sequential manner, concomitant magnetic field stimuli enhance the shell permeability while prompts the reaction between H2O2 and l-arginine to generate the gasotransmitters nitric oxide, which can be imaged by ultrasound and further delivered for diabetic nephropathy therapy. Therefore, magnetic microvesicles with glucose oxidase may be designed as a novel theranostic approach for restoring glucose homeostasis and spatiotemporally control NO release for maintaining good overall diabetic health.
Collapse
Affiliation(s)
- Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Mingxi Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yang Liu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Tuantuan Wang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhenqiang Feng
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Huating Cui
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
38
|
Obesity-Related Diseases and Syndromes: Insulin Resistance, Type 2 Diabetes Mellitus, Non-alcoholic Fatty Liver Disease, Cardiovascular Disease, and Metabolic Syndrome. Obesity (Silver Spring) 2016. [DOI: 10.1007/978-3-319-39409-1_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
39
|
Molina MN, Ferder L, Manucha W. Emerging Role of Nitric Oxide and Heat Shock Proteins in Insulin Resistance. Curr Hypertens Rep 2015; 18:1. [DOI: 10.1007/s11906-015-0615-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
40
|
Santilli F, D'Ardes D, Davì G. Oxidative stress in chronic vascular disease: From prediction to prevention. Vascul Pharmacol 2015; 74:23-37. [DOI: 10.1016/j.vph.2015.09.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/04/2015] [Accepted: 09/08/2015] [Indexed: 12/14/2022]
|
41
|
Costantino S, Paneni F, Cosentino F. Ageing, metabolism and cardiovascular disease. J Physiol 2015; 594:2061-73. [PMID: 26391109 DOI: 10.1113/jp270538] [Citation(s) in RCA: 290] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/04/2015] [Indexed: 12/31/2022] Open
Abstract
Age is one of the major risk factors associated with cardiovascular disease (CVD). About one-fifth of the world population will be aged 65 or older by 2030, with an exponential increase in CVD prevalence. It is well established that environmental factors (overnutrition, smoking, pollution, sedentary lifestyles) may lead to premature defects in mitochondrial functionality, insulin signalling, endothelial homeostasis and redox balance, fostering early senescent features. Over the last few years, molecular investigations have unveiled common signalling networks which may link the ageing process with deterioration of cardiovascular homeostasis and metabolic disturbances, namely insulin resistance. These different processes seem to be highly interconnected and their interplay may favour adverse vascular and cardiac phenotypes responsible for myocardial infarction, stroke and heart failure. In the present review, we carefully describe novel molecular cues underpinning ageing, metabolism and CVD. In particular, we describe a dynamic interplay between emerging pathways such as FOXOs, AMPK, SIRT1, p66(Shc) , JunD and NF-kB. This overview will provide the background for attractive molecular targets to prevent age-driven pathology in the vasculature and the heart.
Collapse
Affiliation(s)
- Sarah Costantino
- Cardiology Unit, Department of Medicine Solna, Karolinska University Hospital, Stockholm, Sweden
| | - Francesco Paneni
- Cardiology Unit, Department of Medicine Solna, Karolinska University Hospital, Stockholm, Sweden
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine Solna, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
42
|
Lee KP, Kim JE, Park WH. Cytoprotective effect of rhamnetin on miconazole-induced H9c2 cell damage. Nutr Res Pract 2015; 9:586-91. [PMID: 26634046 PMCID: PMC4667198 DOI: 10.4162/nrp.2015.9.6.586] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 05/28/2015] [Accepted: 06/29/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND/OBJECTIVES Reactive oxygen species (ROS) formation is closely related to miconazole-induced heart dysfunction. Although rhamnetin has antioxidant effects, it remained unknown whether it can protect against miconazole-induced cardiomyocyte apoptosis. Thus, we investigated the effects of rhamnetin on miconazole-stimulated H9c2 cell apoptosis. MATERIALS/METHODS Cell morphology was observed by inverted microscope and cell viability was determined using a WelCount™ cell proliferation assay kit. Miconazole-induced ROS production was evaluated by fluorescence-activated cell sorting with 6-carboxy-2',7'-dichlorofluoroscein diacetate (H2DCF-DA) stain. Immunoblot analysis was used to determine apurinic/apyrimidinic endonuclease 1 (APE/Ref-1) and cleaved cysteine-aspartic protease (caspase) 3 expression. NADPH oxidase levels were measured using real-time polymerase chain reaction. RESULTS Miconazole (3 and 10 µM) induced abnormal morphological changes and cell death in H9c2 cells. Rhamnetin enhanced the viability of miconazole (3 µM)-treated cells in a dose-dependent manner. Rhamnetin (1 and 3 µM) treatment downregulated cleaved caspase 3 and upregulated APE/Ref-1 expression in miconazole-stimulated cells. Additionally, rhamnetin significantly reduced ROS generation. CONCLUSIONS Our data suggest that rhamnetin may have cytoprotective effects in miconazole-stimulated H9c2 cardiomyocytes via ROS inhibition. This effect most likely occurs through the upregulation of APE/Ref-1 and attenuation of hydrogen peroxide levels.
Collapse
Affiliation(s)
- Kang Pa Lee
- Department of Medical Science, School of Medicine, Konkuk University, Seoul 143-701, Korea
| | - Jai-Eun Kim
- Department of Pathology, College of Korean Medicine, Dongguk University, Gyeonggi-Do 410-820, Korea
| | - Won-Hwan Park
- Department of Diagnosis, College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-Do 410-820, Korea
| |
Collapse
|
43
|
Du YP, Song GY, Wang FJ, Ren LP, Liu YQ, Zhang YN, Qi HQ, Ding HX. Effect of oxymatrine on insulin resistance in patients with type 2 diabetes mellitus. Shijie Huaren Xiaohua Zazhi 2015; 23:2555-2561. [DOI: 10.11569/wcjd.v23.i16.2555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the influence of oxymatrine on insulin resistance in patients with type 2 diabetes mellitus and the mechanism involved.
METHODS: This was a prospective randomized controlled clinical study. Patients with type 2 diabetes mellitus were divided into either an oxymatrine treatment group or an untreated group. Glucose oxidase method was used to detect serum fasting blood glucose (FBG) and fasting insulin (FINS). The homeostasis model assessment of insulin resistance (HOMA-IR) and homeostasis model assessment of insulin sensitivity index (HOMA-ISI) were also detected. ELISA was carried out to detect the serum levels of reactive oxygen species (ROS) and tumor necrosis factor-α (TNF-α). Western blot was used to detect the expression of protein kinase B (AKT), p-AKT, glycogen synthase kinase-3α/β (GSK3α/β), and p-GSK3α/β proteins.
RESULTS: FBG, FINS and HOMA-IR significantly decreased and HOMA-ISI increased in the oxymatrine treatment group compared with the untreated group (P < 0.05). Serum levels of ROS and TNF-α in the oxymatrine treatment group decreased significantly compared with the untreated group (P < 0.05). Western blot analysis showed that the total protein levels of AKT and GSK3α/β were unchanged (P > 0.05), but the expression of p-AKT and p-GSK3α/β significantly increased in the oxymatrine treatment group compared with the untreated group (P < 0.05).
CONCLUSION: Oxymatrine can reduce FBG and FINS, and improve insulin resistance by reducing the production of serum ROS and TNF-α and by influencing the photophosphorylation of key proteins (such as AKT and GSK3α/β) in the insulin resistance-related signaling pathways.
Collapse
|
44
|
Cross-sectional and longitudinal associations between serum uric acid and metabolic syndrome: Results from Fangchenggang Area Male Health and Examination Survey in China. Clin Chim Acta 2015; 446:226-30. [DOI: 10.1016/j.cca.2015.04.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/14/2015] [Accepted: 04/16/2015] [Indexed: 11/22/2022]
|
45
|
García-Fontana B, Morales-Santana S, Longobardo V, Reyes-García R, Rozas-Moreno P, García-Salcedo JA, Muñoz-Torres M. Relationship between Proinflammatory and Antioxidant Proteins with the Severity of Cardiovascular Disease in Type 2 Diabetes Mellitus. Int J Mol Sci 2015; 16:9469-83. [PMID: 25923078 PMCID: PMC4463599 DOI: 10.3390/ijms16059469] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/01/2015] [Accepted: 04/08/2015] [Indexed: 02/05/2023] Open
Abstract
Type 2 diabetes mellitus patients are at significant risk of cardiovascular disease, however, the pathophysiology of these complications is complex and incompletely known in this population. The aim of this study was to compare the serum proteome of patients with type 2 diabetes mellitus presenting or not presenting cardiovascular disease with non-diabetic subjects to find essential proteins related to these cardiovascular complications. This cross-sectional study compares the serum proteome by a combination of protein depletion with 2D-DIGE (2-dimension Difference Gel Electrophoresis) methodology. The proteins differentially expressed were identified by MALDI TOF/TOF (Matrix-assisted laser desorption/ionization and Time-Of-Flight ion detector) or LC-MS/MS (Liquid Chromatography coupled to Mass-Mass Spectrometry). Type 2 diabetes mellitus patients with cardiovascular disease showed higher expression of plasma retinol binding protein and glutathione peroxidase-3 compared to those without cardiovascular disease and non-diabetic controls. These results show that proteins related to the inflammatory and redox state appear to play an important role in the pathogenesis of the cardiovascular disease in the type 2 diabetes mellitus patients.
Collapse
Affiliation(s)
- Beatriz García-Fontana
- Bone Metabolic Unit, Endocrinology Division (RETICEF), Instituto de Investigación Biosanitaria (Ibs) Granada, University Hospital San Cecilio, Granada 18012, Spain.
| | - Sonia Morales-Santana
- Bone Metabolic Unit, Endocrinology Division (RETICEF), Instituto de Investigación Biosanitaria (Ibs) Granada, University Hospital San Cecilio, Granada 18012, Spain.
- Proteomic Research Service, Instituto de Investigación Biosanitaria (Ibs) Granada, University Hospital San Cecilio, Granada 18012, Spain.
| | - Victoria Longobardo
- Proteomic Research Service, Institute of Parasitology and Biomedicine "López Neyra" (C.S.I.C.), Granada 18016, Spain.
| | - Rebeca Reyes-García
- Bone Metabolic Unit, Endocrinology Division (RETICEF), Instituto de Investigación Biosanitaria (Ibs) Granada, University Hospital San Cecilio, Granada 18012, Spain.
| | - Pedro Rozas-Moreno
- Endocrinology Division, Ciudad Real General Hospital, Ciudad Real 13005, Spain.
| | - José Antonio García-Salcedo
- Infectious Diseases Unit, Instituto de Investigación Biosanitaria (Ibs) Granada, University Hospital San Cecilio, Av. Dr. Olóriz 16, Granada 18012, Spain.
| | - Manuel Muñoz-Torres
- Bone Metabolic Unit, Endocrinology Division (RETICEF), Instituto de Investigación Biosanitaria (Ibs) Granada, University Hospital San Cecilio, Granada 18012, Spain.
- Endocrinology Unit, University Hospital San Cecilio, Av. Dr. Olóriz 16, Granada 18012, Spain.
| |
Collapse
|