51
|
Black HS. A Synopsis of the Associations of Oxidative Stress, ROS, and Antioxidants with Diabetes Mellitus. Antioxidants (Basel) 2022; 11:2003. [PMID: 36290725 PMCID: PMC9598123 DOI: 10.3390/antiox11102003] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 07/30/2023] Open
Abstract
The Greek physician, Aretaios, coined the term "diabetes" in the 1st Century A.D. "Mellitus" arose from the observation that the urine exhibits a sweetness due to its elevated glucose levels. Diabetes mellitus (DM) accounted for 6.7 million deaths globally in 2021 with expenditures of USD 966 billion. Mortality is predicted to rise nearly 10-fold by 2030. Oxidative stress, an imbalance between the generation and removal of reactive oxygen species (ROS), is implicated in the pathophysiology of diabetes. Whereas ROS are generated in euglycemic, natural insulin-regulated glucose metabolism, levels are regulated by factors that regulate cellular respiration, e.g., the availability of NAD-linked substrates, succinate, and oxygen; and antioxidant enzymes that maintain the cellular redox balance. Only about 1-2% of total oxygen consumption results in the formation of superoxide anion and hydrogen peroxide under normal reduced conditions. However, under hyperglycemic conditions, about 10% of the respiratory oxygen consumed may be lost as free radicals. Under hyperglycemic conditions, the two-reaction polyol pathway is activated. Nearly 30% of blood glucose can flux through this pathway-a major path contributing to NADH/NAD+ redox imbalance. Under these conditions, protein glycation and lipid peroxidation increase, and inflammatory cytokines are formed, leading to the further formation of ROS. As mitochondria are the major site of intracellular ROS, these organelles are subject to the deleterious effects of ROS themselves and eventually become dysfunctional-a milestone in Metabolic Syndrome (MetS) of which insulin resistance and diabetes predispose to cardiovascular disease.
Collapse
Affiliation(s)
- Homer S Black
- Department of Dermatology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
52
|
Yap KN, Wong HS, Ramanathan C, Rodriguez-Wagner CA, Roberts MD, Freeman DA, Buffenstein R, Zhang Y. Naked mole-rat and Damaraland mole-rat exhibit lower respiration in mitochondria, cellular and organismal levels. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148582. [PMID: 35667393 DOI: 10.1016/j.bbabio.2022.148582] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Naked mole-rats (NMR) and Damaraland mole-rats (DMR) exhibit extraordinary longevity for their body size, high tolerance to hypoxia and oxidative stress and high reproductive output; these collectively defy the concept that life-history traits should be negatively correlated. However, when life-history traits share similar underlying physiological mechanisms, these may be positively associated with each other. We propose that one such potential common mechanism might be the bioenergetic properties of mole-rats. Here, we aim to characterize the bioenergetic properties of two African mole-rats. We adopted a top-down perspective measuring the bioenergetic properties at the organismal, cellular, and molecular level in both species and the biological significance of these properties were compared with the same measures in Siberian hamsters and C57BL/6 mice, chosen for their similar body size to the mole-rat species. We found mole-rats shared several bioenergetic properties that differed from their comparison species, including low basal metabolic rates, a high dependence on glycolysis rather than on oxidative phosphorylation for ATP production, and low proton conductance across the mitochondrial inner membrane. These shared mole-rat features could be a result of evolutionary adaptation to tolerating variable oxygen atmospheres, in particular hypoxia, and may in turn be one of the molecular mechanisms underlying their extremely long lifespans.
Collapse
Affiliation(s)
- Kang Nian Yap
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, United States of America; Department of Biology, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Hoi Shan Wong
- Calico Life Sciences LLC, South San Francisco, CA 94080, United States of America
| | - Chidambaram Ramanathan
- College of Health Sciences, University of Memphis, Memphis, TN 38152, United States of America
| | | | - Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, AL 36849, United States of America
| | - David A Freeman
- Department of Biological Science, University of Memphis, Memphis, TN 38152, United States of America
| | - Rochelle Buffenstein
- Calico Life Sciences LLC, South San Francisco, CA 94080, United States of America.
| | - Yufeng Zhang
- College of Health Sciences, University of Memphis, Memphis, TN 38152, United States of America.
| |
Collapse
|
53
|
Chang S, Tat J, China SP, Kalyanaraman H, Zhuang S, Chan A, Lai C, Radic Z, Abdel-Rahman EA, Casteel DE, Pilz RB, Ali SS, Boss GR. Cobinamide is a strong and versatile antioxidant that overcomes oxidative stress in cells, flies, and diabetic mice. PNAS NEXUS 2022; 1:pgac191. [PMID: 36276587 PMCID: PMC9578022 DOI: 10.1093/pnasnexus/pgac191] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/12/2022] [Indexed: 01/29/2023]
Abstract
Increased oxidative stress underlies a variety of diseases, including diabetes. Here, we show that the cobalamin/vitamin B12 analog cobinamide is a strong and multifaceted antioxidant, neutralizing superoxide, hydrogen peroxide, and peroxynitrite, with apparent rate constants of 1.9 × 108, 3.7 × 104, and 6.3 × 106 M-1 s-1, respectively, for cobinamide with the cobalt in the +2 oxidation state. Cobinamide with the cobalt in the +3 oxidation state yielded apparent rate constants of 1.1 × 108 and 8.0 × 102 M-1 s-1 for superoxide and hydrogen peroxide, respectively. In mammalian cells and Drosophila melanogaster, cobinamide outperformed cobalamin and two well-known antioxidants, imisopasem manganese and manganese(III)tetrakis(4-benzoic acid)porphyrin, in reducing oxidative stress as evidenced by: (i) decreased mitochondrial superoxide and return of the mitochondrial membrane potential in rotenone- and antimycin A-exposed H9c2 rat cardiomyocytes; (ii) reduced JNK phosphorylation in hydrogen-peroxide-treated H9c2 cells; (iii) increased growth in paraquat-exposed COS-7 fibroblasts; and (iv) improved survival in paraquat-treated flies. In diabetic mice, cobinamide administered in the animals' drinking water completely prevented an increase in lipid and protein oxidation, DNA damage, and fibrosis in the heart. Cobinamide is a promising new antioxidant that has potential use in diseases with heightened oxidative stress.
Collapse
Affiliation(s)
| | | | | | | | - Shunhui Zhuang
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adriano Chan
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Cassandra Lai
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zoran Radic
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Engy A Abdel-Rahman
- Tumor Biology Research Program, Children’s Cancer Hospital, Cairo 57357, Egypt,Pharmacology Department, Faculty of Medicine, Assuit University, Assuit 71515, Egypt
| | - Darren E Casteel
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Renate B Pilz
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
54
|
Yan X, He M, Huang H, Wang Q, Hu Y, Wang X, Jin M, Wang Y, Xia Y, Li Y, Chen G, Cheng J, Jia J. Endogenous H 2S targets mitochondria to promote continual phagocytosis of erythrocytes by microglia after intracerebral hemorrhage. Redox Biol 2022; 56:102442. [PMID: 35998432 PMCID: PMC9420393 DOI: 10.1016/j.redox.2022.102442] [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] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/21/2022] [Accepted: 08/11/2022] [Indexed: 11/21/2022] Open
Abstract
Hematoma clearance, which is achieved largely by phagocytosis of erythrocytes in the hemorrhagic brain, limits injury and facilitates recovery following intracerebral hemorrhage (ICH). Efficient phagocytosis critically depends on the capacity of a single phagocyte to phagocytize dead cells continually. However, the mechanism underlying continual phagocytosis following ICH remains unclear. We aimed to investigate the mechanism in this study. By using ICH models, we found that the gasotransmitter hydrogen sulfide (H2S) is an endogenous modulator of continual phagocytosis following ICH. The expression of the H2S synthase cystathionine β-synthase (CBS) and CBS-derived H2S were elevated in brain-resident phagocytic microglia following ICH, which consequently promoted continual phagocytosis of erythrocytes by microglia. Microglia-specific deletion of CBS delayed spontaneous hematoma clearance via an H2S-mediated mechanism following ICH. Mechanistically, oxidation of CBS-derived endogenous H2S by sulfide-quinone oxidoreductase initiated reverse electron transfer at mitochondrial complex I, leading to superoxide production. Complex I-derived superoxide, in turn, activated uncoupling protein 2 (UCP2) to promote microglial phagocytosis of erythrocytes. Functionally, complex I and UCP2 were required for spontaneous hematoma clearance following ICH. Moreover, hyperhomocysteinemia, an established risk factor for stroke, impaired ICH-enhanced CBS expression and delayed hematoma resolution, while supplementing exogenous H2S accelerated hematoma clearance in mice with hyperhomocysteinemia. The results suggest that the microglial CBS-H2S-complex I axis is critical to continual phagocytosis following ICH and can be targeted to treat ICH. CBS-derived H2S is elevated in brain-resident phagocytic microglia following ICH. CBS-derived H2S promotes continual erythrophagocytosis and hematoma clearance. CBS-derived H2S promotes microglial phagocytosis via complex I-derived ROS. Hyperhomocysteinemia inhibits CBS expression to delay hematoma resolution. The CBS-H2S-complex I axis can be targeted to treat ICH.
Collapse
Affiliation(s)
- Xiaoling Yan
- Clinical Research Center of Neurological Disease of the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases & Institute of Neuroscience, Soochow University, Suzhou, 215123, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Meijun He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Hui Huang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Qi Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Yu Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiaoying Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Meng Jin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yi Wang
- Clinical Research Center of Neurological Disease of the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases & Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Yiqing Xia
- Clinical Research Center of Neurological Disease of the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases & Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Yi Li
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China
| | - Gang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China.
| | - Jian Cheng
- Clinical Research Center of Neurological Disease of the Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases & Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| | - Jia Jia
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
55
|
Mal P, Mukherjee T, Upadhyay AK, Mohanty S, Pattnaik AK. Connecting the dots between inflammatory cascades of obesity and COVID-19 in light of mortal consequences-a review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:57040-57053. [PMID: 35731430 PMCID: PMC9213647 DOI: 10.1007/s11356-022-21461-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/10/2022] [Indexed: 06/15/2023]
Abstract
Obesity is a term that has recently been referred to describe a condition in which a person has become a diseased vessel. Obesity's internal pathology is too mysterious as it has a close resemblance with fatal diseases pathology. Obesity and coronavirus disease 2019 (COVID-19) are simultaneous epidemics declared by many organizations after observing their rampage in the recent world. Oxidative stress, cytokine storm, interleukin, and their contribution to the internal adipocyte environment implicated in the cascades of inflammatory pathology are portrayed here. Major determinants like angiotensin-converting enzyme 2 (ACE2) and renin-angiotensin-aldosterone system (RAAS) axis are highly sensitive molecular factors. Data from various countries suggested a clinical overview of how greater body mass index (BMI) is related to greater COVID-19 risk. It also gives insight into how obese individuals are obligately getting admitted and combating COVID-19 in intensive care unit including children less than 13 years of age under ultimate therapeutic options. There are numerous studies currently taking place for finding a cure for obesity which are mainly focused on natural resources and novel therapies like photobiomodulation (PBM) consisting of laser treatment, infrared treatment, etc. as current pharmacological treatments are reported to have fatal adverse effects. Finally, it is discussed how attenuating obesity will be a solution for future combat strategy. This review gives light on the areas of coagulation, inflammatory parameters, cardiometabolic complications, endothelial dysfunctions, immunological infirmity due to COVID-19 in obese individuals. A conceptual outline about correlation between the inflammatory pathophysiological steps triggering the aggravation of fatal consequences has been drawn in this review.
Collapse
Affiliation(s)
- Payel Mal
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Tuhin Mukherjee
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Abhay K Upadhyay
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Satyajit Mohanty
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Ashok K Pattnaik
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India.
| |
Collapse
|
56
|
Sato A, Shiraishi Y, Kimura T, Osaki A, Kagami K, Ido Y, Adachi T. Resistance to Obesity in SOD1 Deficient Mice with a High-Fat/High-Sucrose Diet. Antioxidants (Basel) 2022; 11:antiox11071403. [PMID: 35883894 PMCID: PMC9312060 DOI: 10.3390/antiox11071403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Abstract
Metabolic syndrome (Mets) is an important condition because it may cause stroke and heart disease in the future. Reactive oxygen species (ROSs) influence the pathogenesis of Mets; however, the types of ROSs and their localization remain largely unknown. In this study, we investigated the effects of SOD1, which localize to the cytoplasm and mitochondrial intermembrane space and metabolize superoxide anion, on Mets using SOD1 deficient mice (SOD1−/−). SOD1−/− fed on a high-fat/high-sucrose diet (HFHSD) for 24 weeks showed reduced body weight gain and adipose tissue size compared to wild-type mice (WT). Insulin secretion was dramatically decreased in SOD1−/− fed on HFHSD even though blood glucose levels were similar to WT. Ambulatory oxygen consumption was accelerated in SOD1−/− with HFHSD; however, ATP levels of skeletal muscle were somewhat reduced compared to WT. Reflecting the reduced ATP, the expression of phosphorylated AMPK (Thr 172) was more robust in SOD1−/−. SOD1 is involved in the ATP production mechanism in mitochondria and may contribute to visceral fat accumulation by causing insulin secretion and insulin resistance.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of Internal Medicine, Division of Cardiovascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.S.); (T.K.); (A.O.); (K.K.); (Y.I.)
| | - Yasunaga Shiraishi
- Division of Environmental Medicine, National Defense Medical College Research Institute, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan;
| | - Toyokazu Kimura
- Department of Internal Medicine, Division of Cardiovascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.S.); (T.K.); (A.O.); (K.K.); (Y.I.)
| | - Ayumu Osaki
- Department of Internal Medicine, Division of Cardiovascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.S.); (T.K.); (A.O.); (K.K.); (Y.I.)
| | - Kazuki Kagami
- Department of Internal Medicine, Division of Cardiovascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.S.); (T.K.); (A.O.); (K.K.); (Y.I.)
| | - Yasuo Ido
- Department of Internal Medicine, Division of Cardiovascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.S.); (T.K.); (A.O.); (K.K.); (Y.I.)
| | - Takeshi Adachi
- Department of Internal Medicine, Division of Cardiovascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.S.); (T.K.); (A.O.); (K.K.); (Y.I.)
- Correspondence: or ; Tel.: +81-4-2995-1597
| |
Collapse
|
57
|
Lewandowski D, Sander CL, Tworak A, Gao F, Xu Q, Skowronska-Krawczyk D. Dynamic lipid turnover in photoreceptors and retinal pigment epithelium throughout life. Prog Retin Eye Res 2022; 89:101037. [PMID: 34971765 PMCID: PMC10361839 DOI: 10.1016/j.preteyeres.2021.101037] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022]
Abstract
The retinal pigment epithelium-photoreceptor interphase is renewed each day in a stunning display of cellular interdependence. While photoreceptors use photosensitive pigments to convert light into electrical signals, the RPE supports photoreceptors in their function by phagocytizing shed photoreceptor tips, regulating the blood retina barrier, and modulating inflammatory responses, as well as regenerating the 11-cis-retinal chromophore via the classical visual cycle. These processes involve multiple protein complexes, tightly regulated ligand-receptors interactions, and a plethora of lipids and protein-lipids interactions. The role of lipids in maintaining a healthy interplay between the RPE and photoreceptors has not been fully delineated. In recent years, novel technologies have resulted in major advancements in understanding several facets of this interplay, including the involvement of lipids in phagocytosis and phagolysosome function, nutrient recycling, and the metabolic dependence between the two cell types. In this review, we aim to integrate the complex role of lipids in photoreceptor and RPE function, emphasizing the dynamic exchange between the cells as well as discuss how these processes are affected in aging and retinal diseases.
Collapse
Affiliation(s)
- Dominik Lewandowski
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Christopher L Sander
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA; Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Aleksander Tworak
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Fangyuan Gao
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Qianlan Xu
- Department of Physiology and Biophysics, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Dorota Skowronska-Krawczyk
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA.
| |
Collapse
|
58
|
Ho JQ, Abramowitz MK. Clinical Consequences of Metabolic Acidosis-Muscle. Adv Chronic Kidney Dis 2022; 29:395-405. [PMID: 36175077 DOI: 10.1053/j.ackd.2022.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/10/2022] [Accepted: 04/25/2022] [Indexed: 01/25/2023]
Abstract
Metabolic acidosis is common in people with chronic kidney disease and can contribute to functional decline, morbidity, and mortality. One avenue through which metabolic acidosis can result in these adverse clinical outcomes is by negatively impacting skeletal muscle; this can occur through several pathways. First, metabolic acidosis promotes protein degradation and impairs protein synthesis, which lead to muscle breakdown. Second, metabolic acidosis hinders mitochondrial function, which decreases oxidative phosphorylation and reduces energy production. Third, metabolic acidosis directly limits muscle contraction. The purpose of this review is to examine the specific mechanisms of each pathway through which metabolic acidosis affects muscle, the impact of metabolic acidosis on physical function, and the effect of treating metabolic acidosis on functional outcomes.
Collapse
Affiliation(s)
- Jim Q Ho
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Matthew K Abramowitz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY; Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY; Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
59
|
Sies H, Belousov VV, Chandel NS, Davies MJ, Jones DP, Mann GE, Murphy MP, Yamamoto M, Winterbourn C. Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat Rev Mol Cell Biol 2022; 23:499-515. [PMID: 35190722 DOI: 10.1038/s41580-022-00456-z] [Citation(s) in RCA: 609] [Impact Index Per Article: 304.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 02/06/2023]
Abstract
'Reactive oxygen species' (ROS) is a generic term that defines a wide variety of oxidant molecules with vastly different properties and biological functions that range from signalling to causing cell damage. Consequently, the description of oxidants needs to be chemically precise to translate research on their biological effects into therapeutic benefit in redox medicine. This Expert Recommendation article pinpoints key issues associated with identifying the physiological roles of oxidants, focusing on H2O2 and O2.-. The generic term ROS should not be used to describe specific molecular agents. We also advocate for greater precision in measurement of H2O2, O2.- and other oxidants, along with more specific identification of their signalling targets. Future work should also consider inter-organellar communication and the interactions of redox-sensitive signalling targets within organs and whole organisms, including the contribution of environmental exposures. To achieve these goals, development of tools that enable site-specific and real-time detection and quantification of individual oxidants in cells and model organisms are needed. We also stress that physiological O2 levels should be maintained in cell culture to better mimic in vivo redox reactions associated with specific cell types. Use of precise definitions and analytical tools will help harmonize research among the many scientific disciplines working on the common goal of understanding redox biology.
Collapse
Affiliation(s)
- Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Faculty of Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | - Vsevolod V Belousov
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Navdeep S Chandel
- Division of Pulmonary & Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Christine Winterbourn
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|
60
|
Hunter JP, Faro LL, Rozenberg K, Dengu F, Ogbemudia A, Weissenbacher A, Mulvey JF, Knijff L, Gopalakrishnan K, Ploeg RJ. Assessment of Mitochondrial Function and Oxygen Consumption Measured During Ex Vivo Normothermic Machine Perfusion of Injured Pig Kidneys Helps to Monitor Organ Viability. Transpl Int 2022; 35:10420. [PMID: 35711321 PMCID: PMC9194576 DOI: 10.3389/ti.2022.10420] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/10/2022] [Indexed: 12/01/2022]
Abstract
Donor kidney assessment may improve organ utilisation. Normothermic Machine Perfusion (NMP) has the potential to facilitate this advance. The mechanism of action is not yet determined and we aimed to assess mitochondrial function during NMP. Anaesthetised pigs (n = 6) had one kidney clamped for 60 min. The healthy contralateral kidney was removed and underwent NMP for 8 h (healthy control (HC), n = 6). Following 60 min warm ischaemia the injured kidney underwent HMP for 24 h, followed by NMP for 8 h (n = 6). Mitochondria were extracted from fresh tissue for analysis. Injured kidneys were analysed as two separate groups (IMa, n = 3 and IMb, n = 3). Renal resistance was higher (0.39ï, ± 0.29 vs. 1.65ï, ± 0.85; p = 0.01) and flow was lower (55ï, ± 28 vs. 7ï, ± 4; p = 0.03) during HMP in IMb than IMa. NMP blood flow was higher in IMa versus IMb (2-way ANOVA; p < 0.001) After 60 min NMP, O2 consumption was significantly lower in IMb versus IMa (p ≤ 0.002). State-3 respiration was significantly different between the groups (37ï, ± 19 vs. 24ï, ± 14 vs. 10ï, ± 8; nmolO2/min/mg; p = 0.049). Lactate levels were significantly lower in IMa versus IMb (p = 0.028). Mitochondrial respiration levels during NMP may be suggestive of kidney viability. Oxygen consumption, renal blood flow and lactate can differentiate severity of kidney injury during NMP.
Collapse
Affiliation(s)
- James P. Hunter
- Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
- *Correspondence: James P. Hunter,
| | - Letizia Lo Faro
- Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Kaithlyn Rozenberg
- Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Fungai Dengu
- Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Anne Ogbemudia
- Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Annemarie Weissenbacher
- Department of Visceral, Transplantation and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - John F. Mulvey
- Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Laura Knijff
- Leiden University Medical Center, Leiden, Netherlands
| | | | - Rutger J. Ploeg
- Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
61
|
Gu SX, Dayal S. Redox Mechanisms of Platelet Activation in Aging. Antioxidants (Basel) 2022; 11:995. [PMID: 35624860 PMCID: PMC9137594 DOI: 10.3390/antiox11050995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
Aging is intrinsically linked with physiologic decline and is a major risk factor for a broad range of diseases. The deleterious effects of advancing age on the vascular system are evidenced by the high incidence and prevalence of cardiovascular disease in the elderly. Reactive oxygen species are critical mediators of normal vascular physiology and have been shown to gradually increase in the vasculature with age. There is a growing appreciation for the complexity of oxidant and antioxidant systems at the cellular and molecular levels, and accumulating evidence indicates a causal association between oxidative stress and age-related vascular disease. Herein, we review the current understanding of mechanistic links between oxidative stress and thrombotic vascular disease and the changes that occur with aging. While several vascular cells are key contributors, we focus on oxidative changes that occur in platelets and their mediation in disease progression. Additionally, we discuss the impact of comorbid conditions (i.e., diabetes, atherosclerosis, obesity, cancer, etc.) that have been associated with platelet redox dysregulation and vascular disease pathogenesis. As we continue to unravel the fundamental redox mechanisms of the vascular system, we will be able to develop more targeted therapeutic strategies for the prevention and management of age-associated vascular disease.
Collapse
Affiliation(s)
- Sean X. Gu
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06511, USA;
| | - Sanjana Dayal
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa City VA Healthcare System, Iowa City, IA 52246, USA
| |
Collapse
|
62
|
Alberici LC, Oliveira HCF. Mitochondrial Adaptive Responses to Hypertriglyceridemia and Bioactive Lipids. Antioxid Redox Signal 2022; 36:953-968. [PMID: 34409856 DOI: 10.1089/ars.2021.0180] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Significance: Altered plasma triglyceride metabolism and changes in dietary fatty acid types and levels are major contributors to the development of metabolic and cardiovascular diseases such as fatty liver disease, obesity, diabetes, and atherosclerosis. Lipid accumulation in visceral adipose tissue and ectopically in other organs, as well as lipid-induced redox imbalance, is connected to mitochondrial dysfunction in a range of oxidative stress-associated metabolic and degenerative disorders. Recent Advances: Successful mitochondrial adaptive responses in the context of hypertriglyceridemia and dietary bioactive polyunsaturated fatty acids contribute to increase body energy expenditure and reduce oxidative stress, thus allowing several cell types to cope with metabolic challenges and stresses. These responses include mitochondrial redox signaling, mild uncoupling, and changes in network dynamic behavior. Critical Issues: Mitochondrial bioenergetics and redox changes in a lipid overload context are relatively well characterized. However, the turning point between adaptive and maladaptive mitochondrial responses remains a critical issue to be elucidated. In addition, the relationship between changes in fusion/fission machinery and mitochondrial function is less well understood. Future Directions: The effective mitochondrial responses described here support the research for new drug design and diet or nutraceutical formulations targeting mitochondrial mild uncoupling and effective quality control as putative strategies for cardiometabolic diseases. Antioxid. Redox Signal. 36, 953-968.
Collapse
Affiliation(s)
- Luciane C Alberici
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Helena C F Oliveira
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
63
|
Sivertsson E, Friederich-Persson M, Persson P, Nangaku M, Hansell P, Palm F. Thyroid hormone increases oxygen metabolism causing intrarenal tissue hypoxia; a pathway to kidney disease. PLoS One 2022; 17:e0264524. [PMID: 35239685 PMCID: PMC8893624 DOI: 10.1371/journal.pone.0264524] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 02/11/2022] [Indexed: 01/10/2023] Open
Abstract
The proposed mechanisms for the development of nephropathy are many, complex and often overlapping. Although recent literature strongly supports a role of kidney hypoxia as an independent pathway to nephropathy, the evidence remains inconclusive since the role of hypoxia is difficult to differentiate from confounding factors such as hyperglycemia, hypertension and oxidative stress. By increasing kidney oxygen consumption using triiodothyronine (T3) and, thus, avoiding these confounding factors, the aim of the present study was to investigate renal hypoxia per se as a causal pathway for the development of nephropathy. Healthy Sprague-Dawley rats were treated with T3 (10 μg/kg/day) and the angiotensin II AT1-receptor antagonist candesartan (1 mg/kg in drinking water) to eliminate effects of T3-induced renin release; and compared to a candesartan treated control group. After 7 weeks of treatment in vivo kidney function, oxygen metabolism and mitochondrial function were evaluated. T3 did not affect glomerular filtration rate or renal blood flow, but increased total kidney oxygen consumption resulting in cortical hypoxia. Nephropathy, demonstrated as albuminuria and tubulointerstitial fibrosis, developed in T3-treated animals. Mitochondria uncoupling mediated by uncoupling protein 2 and the adenosine nucleotide transporter was demonstrated as a mechanism causing the increased kidney oxygen consumption. Importantly, blood glucose levels, mean arterial blood pressure and oxidative stress levels were not affected by T3. In conclusion, the present study provides further evidence for increased kidney oxygen consumption causing intrarenal tissue hypoxia, as a causal pathway for development of nephropathy.
Collapse
Affiliation(s)
- Ebba Sivertsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Patrik Persson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Peter Hansell
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Fredrik Palm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
64
|
Activation of UCP2 by anethole trithione suppresses neuroinflammation after intracerebral hemorrhage. Acta Pharmacol Sin 2022; 43:811-828. [PMID: 34183754 PMCID: PMC8976076 DOI: 10.1038/s41401-021-00698-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/14/2021] [Indexed: 02/06/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating disease, in which neuroinflammation substantially contributes to brain injury. Uncoupling protein 2 (UCP2) is a member of the mitochondrial anion carrier family, which uncouples oxidative phosphorylation from ATP synthesis by facilitating proton leak across the mitochondrial inner membrane. UCP2 has been reported to modulate inflammation. In this study we investigated whether and how UCP2 modulated neuroinflammation through microglia/macrophages following ICH in vitro and in vivo. We used an in vitro neuroinflammation model in murine BV2 microglia to mimic microglial activation following ICH. ICH in vivo model was established in mice through collagenase infusion into the left striatum. ICH mice were treated with anetholetrithione (ADT, 50 mg· kg-1 ·d-1, ip) or the classical protonophoric uncoupler FCCP (injected into hemorrhagic striatum). We showed that the expression and mitochondrial location of microglial UCP2 were not changed in both in vitro and in vivo ICH models. Knockdown of UCP2 exacerbated neuroinflammation in BV2 microglia and mouse ICH models, suggesting that endogenous UCP2 inhibited neuroinflammation and therefore played a protective role following ICH. ADT enhanced mitochondrial ROS production thus inducing mitochondrial uncoupling and activating UCP2 in microglia. ADT robustly suppressed neuroinflammation, attenuated brain edema and improved neurological deficits following ICH, and these effects were countered by striatal knockdown of UCP2. ADT enhanced AMP-activated protein kinase (AMPK) activation in the hemorrhagic brain, which was abrogated by striatal knockdown of UCP2. Moreover, striatal knockdown of AMPK abolished the suppression of neuroinflammation by ADT following ICH. On the other hand, FCCP-induced mitochondrial uncoupling was independent of UCP2 in microglia; and striatal knockdown of UCP2 did not abrogate the suppression of neuroinflammation by FCCP in ICH mice. In conclusion, the uncoupling activity is essential for suppression of neuroinflammation by UCP2. We prove for the first time the concept that activators of endogenous UCP2 such as anetholetrithione are a new class of uncouplers with translational significance.
Collapse
|
65
|
Regulation of Aging and Longevity by Ion Channels and Transporters. Cells 2022; 11:cells11071180. [PMID: 35406743 PMCID: PMC8997527 DOI: 10.3390/cells11071180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022] Open
Abstract
Despite significant advances in our understanding of the mechanisms that underlie age-related physiological decline, our ability to translate these insights into actionable strategies to extend human healthspan has been limited. One of the major reasons for the existence of this barrier is that with a few important exceptions, many of the proteins that mediate aging have proven to be undruggable. The argument put forth here is that the amenability of ion channels and transporters to pharmacological manipulation could be leveraged to develop novel therapeutic strategies to combat aging. This review delves into the established roles for ion channels and transporters in the regulation of aging and longevity via their influence on membrane excitability, Ca2+ homeostasis, mitochondrial and endolysosomal function, and the transduction of sensory stimuli. The goal is to provide the reader with an understanding of emergent themes, and prompt further investigation into how the activities of ion channels and transporters sculpt the trajectories of cellular and organismal aging.
Collapse
|
66
|
Oo SM, Oo HK, Takayama H, Ishii KA, Takeshita Y, Goto H, Nakano Y, Kohno S, Takahashi C, Nakamura H, Saito Y, Matsushita M, Okamatsu-Ogura Y, Saito M, Takamura T. Selenoprotein P-mediated reductive stress impairs cold-induced thermogenesis in brown fat. Cell Rep 2022; 38:110566. [PMID: 35354056 DOI: 10.1016/j.celrep.2022.110566] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 02/03/2022] [Accepted: 03/03/2022] [Indexed: 02/08/2023] Open
Abstract
Reactive oxygen species (ROS) activate uncoupler protein 1 (UCP1) in brown adipose tissue (BAT) under physiological cold exposure and noradrenaline (NA) stimulation to increase thermogenesis. However, the endogenous regulator of ROS in activated BAT and its role in pathological conditions remain unclear. We show that serum levels of selenoprotein P (SeP; encoded by SELENOP) negatively correlate with BAT activity in humans. Physiological cold exposure downregulates Selenop in BAT. Selenop knockout mice show higher rectal temperatures and UCP1 sulfenylation during cold exposure. SeP treatment to brown adipocytes eliminated the NA-induced mitochondrial ROS by upregulating glutathione peroxidase 4 and impaired cellular thermogenesis. A high-fat/high-sucrose diet elevates serum SeP levels and diminishes the elevated NA-induced thermogenesis in BAT-Selenop KO mice. Therefore, SeP is the intrinsic factor inducing reductive stress that impairs thermogenesis in BAT and may be a potential therapeutic target for obesity and diabetes.
Collapse
Affiliation(s)
- Swe Mar Oo
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Hein Ko Oo
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Hiroaki Takayama
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan; Life Sciences Division, Engineering and Technology Department, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kiyo-Aki Ishii
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan; Department of System Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Yumie Takeshita
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Hisanori Goto
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Yujiro Nakano
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Susumu Kohno
- Division of Oncology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Chiaki Takahashi
- Division of Oncology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroyuki Nakamura
- Department of Environmental and Preventive Medicine, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yoshiro Saito
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | | | - Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Masayuki Saito
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|
67
|
Nguyen NT, Nguyen TT, Park KS. Oxidative Stress Related to Plasmalemmal and Mitochondrial Phosphate Transporters in Vascular Calcification. Antioxidants (Basel) 2022; 11:antiox11030494. [PMID: 35326144 PMCID: PMC8944874 DOI: 10.3390/antiox11030494] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 12/04/2022] Open
Abstract
Inorganic phosphate (Pi) is essential for maintaining cellular function but excess of Pi leads to serious complications, including vascular calcification. Accumulating evidence suggests that oxidative stress contributes to the pathogenic progression of calcific changes. However, the molecular mechanism underlying Pi-induced reactive oxygen species (ROS) generation and its detrimental consequences remain unclear. Type III Na+-dependent Pi cotransporter, PiT-1/-2, play a significant role in Pi uptake of vascular smooth muscle cells. Pi influx via PiT-1/-2 increases the abundance of PiT-1/-2 and depolarization-activated Ca2+ entry due to its electrogenic properties, which may lead to Ca2+ and Pi overload and oxidative stress. At least four mitochondrial Pi transporters are suggested, among which the phosphate carrier (PiC) is known to be mainly involved in mitochondrial Pi uptake. Pi transport via PiC may induce hyperpolarization and superoxide generation, which may lead to mitochondrial dysfunction and endoplasmic reticulum stress, together with generation of cytosolic ROS. Increase in net influx of Ca2+ and Pi and their accumulation in the cytosol and mitochondrial matrix synergistically increases oxidative stress and osteogenic differentiation, which could be prevented by suppressing either Ca2+ or Pi overload. Therapeutic strategies targeting plasmalemmal and mitochondrial Pi transports can protect against Pi-induced oxidative stress and vascular calcification.
Collapse
Affiliation(s)
- Nhung Thi Nguyen
- Department of Physiology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
- Mitohormesis Research Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
- Medical Doctor Program, College of Health Sciences, VinUniversity, Hanoi 12406, Vietnam
| | - Tuyet Thi Nguyen
- Department of Physiology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
- Internal Medicine Residency Program, College of Health Sciences, VinUniversity, Hanoi 12406, Vietnam
- Correspondence: (T.T.N.); (K.-S.P.); Tel.: +84-247-108-9779 (T.T.N.); +82-33-741-0294 (K.-S.P.)
| | - Kyu-Sang Park
- Department of Physiology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
- Mitohormesis Research Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
- Correspondence: (T.T.N.); (K.-S.P.); Tel.: +84-247-108-9779 (T.T.N.); +82-33-741-0294 (K.-S.P.)
| |
Collapse
|
68
|
Aljohani AS, Ahmed AA, Althwab SA, Alkhamiss AS, Rasheed Z, Fernández N, Al Abdulmonem W. Gene expression of glutathione S-transferase alpha, glutathione S-transferase rho, glutathione peroxidase, uncoupling protein 2, cytochrome P450 1A, heat shock protein 70 in liver of Oreochromis niloticus upon exposure of microcystin-LR, microcystin-RR and toxic cyanobacteria crude. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
69
|
Raghavan S, Kundumani-Sridharan V, Kumar S, White CW, Das KC. Thioredoxin Prevents Loss of UCP2 in Hyperoxia via MKK4-p38 MAPK-PGC1α Signaling and Limits Oxygen Toxicity. Am J Respir Cell Mol Biol 2022; 66:323-336. [PMID: 34890296 PMCID: PMC8937245 DOI: 10.1165/rcmb.2021-0219oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/18/2021] [Indexed: 11/24/2022] Open
Abstract
Administration of high concentrations of oxygen (hyperoxia) is one of few available options to treat acute hypoxemia-related respiratory failure, as seen in the current coronavirus disease (COVID-19) pandemic. Although hyperoxia can cause acute lung injury through increased production of superoxide anion (O2•-), the choice of high-concentration oxygen administration has become a necessity in critical care. The objective of this study was to test the hypothesis that UCP2 (uncoupling protein 2) has a major function of reducing O2•- generation in the lung in ambient air or in hyperoxia. Lung epithelial cells and wild-type; UCP2-/-; or transgenic, hTrx overexpression-bearing mice (Trx-Tg) were exposed to hyperoxia and O2•- generation was measured by using electron paramagnetic resonance, and lung injury was measured by using histopathologic analysis. UCP2 expression was analyzed by using RT-PCR analysis, Western blotting analysis, and RNA interference. The signal transduction pathways leading to loss of UCP2 expression were analyzed by using IP, phosphoprotein analysis, and specific inhibitors. UCP2 mRNA and protein expression were acutely decreased in hyperoxia, and these decreases were associated with a significant increase in O2•- production in the lung. Treatment of cells with rhTrx (recombinant human thioredoxin) or exposure of Trx-Tg mice prevented the loss of UCP2 protein and decreased O2•- generation in the lung. Trx is also required to maintain UCP2 expression in normoxia. Loss of UCP2 in UCP2-/- mice accentuated lung injury in hyperoxia. Trx activates the MKK4-p38MAPK (p38 mitogen-activated protein kinase)-PGC1α (PPARγ [peroxisome proliferator-activated receptor γ] coactivator 1α) pathway, leading to rescue of UCP2 and decreased O2•- generation in hyperoxia. Loss of UCP2 in hyperoxia is a major mechanism of O2•- production in the lung in hyperoxia. rhTrx can protect against lung injury in hyperoxia due to rescue of the loss of UCP2.
Collapse
Affiliation(s)
- Somasundaram Raghavan
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Texas Tech University, Lubbock, Texas; and
| | - Venkatesh Kundumani-Sridharan
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Texas Tech University, Lubbock, Texas; and
| | - Sudhir Kumar
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Texas Tech University, Lubbock, Texas; and
| | - Carl W. White
- Department of Pediatrics, Children’s Hospital, University of Colorado Health Sciences Center, University of Colorado, Aurora, Colorado
| | - Kumuda C. Das
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Texas Tech University, Lubbock, Texas; and
| |
Collapse
|
70
|
van Rensburg D, Lindeque Z, Harvey BH, Steyn SF. Reviewing the mitochondrial dysfunction paradigm in rodent models as platforms for neuropsychiatric disease research. Mitochondrion 2022; 64:82-102. [DOI: 10.1016/j.mito.2022.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/22/2022] [Accepted: 03/15/2022] [Indexed: 12/19/2022]
|
71
|
Zheng X, Narayanan S, Xu C, Eliasson Angelstig S, Grünler J, Zhao A, Di Toro A, Bernardi L, Mazzone M, Carmeliet P, Del Sole M, Solaini G, Forsberg EA, Zhang A, Brismar K, Schiffer TA, Rajamand Ekberg N, Botusan IR, Palm F, Catrina SB. Repression of hypoxia-inducible factor-1 contributes to increased mitochondrial reactive oxygen species production in diabetes. eLife 2022; 11:70714. [PMID: 35164902 PMCID: PMC8846593 DOI: 10.7554/elife.70714] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 01/27/2022] [Indexed: 12/19/2022] Open
Abstract
Background: Excessive production of mitochondrial reactive oxygen species (ROS) is a central mechanism for the development of diabetes complications. Recently, hypoxia has been identified to play an additional pathogenic role in diabetes. In this study, we hypothesized that ROS overproduction was secondary to the impaired responses to hypoxia due to the inhibition of hypoxia-inducible factor-1 (HIF-1) by hyperglycemia. Methods: The ROS levels were analyzed in the blood of healthy subjects and individuals with type 1 diabetes after exposure to hypoxia. The relation between HIF-1, glucose levels, ROS production and its functional consequences were analyzed in renal mIMCD-3 cells and in kidneys of mouse models of diabetes. Results: Exposure to hypoxia increased circulating ROS in subjects with diabetes, but not in subjects without diabetes. High glucose concentrations repressed HIF-1 both in hypoxic cells and in kidneys of animals with diabetes, through a HIF prolyl-hydroxylase (PHD)-dependent mechanism. The impaired HIF-1 signaling contributed to excess production of mitochondrial ROS through increased mitochondrial respiration that was mediated by Pyruvate dehydrogenase kinase 1 (PDK1). The restoration of HIF-1 function attenuated ROS overproduction despite persistent hyperglycemia, and conferred protection against apoptosis and renal injury in diabetes. Conclusions: We conclude that the repression of HIF-1 plays a central role in mitochondrial ROS overproduction in diabetes and is a potential therapeutic target for diabetic complications. These findings are timely since the first PHD inhibitor that can activate HIF-1 has been newly approved for clinical use. Funding: This work was supported by grants from the Swedish Research Council, Stockholm County Research Council, Stockholm Regional Research Foundation, Bert von Kantzows Foundation, Swedish Society of Medicine, Kung Gustaf V:s och Drottning Victorias Frimurarestifelse, Karolinska Institute’s Research Foundations, Strategic Research Programme in Diabetes, and Erling-Persson Family Foundation for S-B.C.; grants from the Swedish Research Council and Swedish Heart and Lung Foundation for T.A.S.; and ERC consolidator grant for M.M.
Collapse
Affiliation(s)
- Xiaowei Zheng
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Sampath Narayanan
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Cheng Xu
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Jacob Grünler
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Allan Zhao
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Alessandro Di Toro
- Centre for Inherited Cardiovascular Diseases, IRCCS Foundation University Hospital Policlinico San Matteo, Pavia, Italy
| | - Luciano Bernardi
- Folkälsan Research Center, Folkälsan Institute of Genetics, University of Helsinki, Helsinki, Finland
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB); Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Katholieke Universiteit (KU) Leuven; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Marianna Del Sole
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Elisabete A Forsberg
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ao Zhang
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Kerstin Brismar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Tomas A Schiffer
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Neda Rajamand Ekberg
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Endocrinology and Diabetes, Karolinska University Hospital, Stockholm, Sweden.,Center for Diabetes, Academic Specialist Centrum, Stockholm, Sweden
| | - Ileana Ruxandra Botusan
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Endocrinology and Diabetes, Karolinska University Hospital, Stockholm, Sweden.,Center for Diabetes, Academic Specialist Centrum, Stockholm, Sweden
| | - Fredrik Palm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Endocrinology and Diabetes, Karolinska University Hospital, Stockholm, Sweden.,Center for Diabetes, Academic Specialist Centrum, Stockholm, Sweden
| |
Collapse
|
72
|
Abstract
Mitochondria of all tissues convert various metabolic substrates into two forms of energy: ATP and heat. Historically, the primary focus of research in mitochondrial bioenergetics was on the mechanisms of ATP production, while mitochondrial thermogenesis received significantly less attention. Nevertheless, mitochondrial heat production is crucial for the maintenance of body temperature, regulation of the pace of metabolism, and prevention of oxidative damage to mitochondria and the cell. In addition, mitochondrial thermogenesis has gained significance as a pharmacological target for treating metabolic disorders. Mitochondria produce heat as the result of H+ leak across their inner membrane. This review provides a critical assessment of the current field of mitochondrial H+ leak and thermogenesis, with a focus on the molecular mechanisms involved in the function and regulation of uncoupling protein 1 and the ADP/ATP carrier, the two proteins that mediate mitochondrial H+ leak.
Collapse
Affiliation(s)
- Ambre M. Bertholet
- Department of Physiology, University of California San Francisco, 600 16 Street, San Francisco, CA 94158, USA,Department of Physiology, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA,Corresponding authors: ,
| | - Yuriy Kirichok
- Department of Physiology, University of California, San Francisco, California, USA;
| |
Collapse
|
73
|
Hirschenson J, Melgar-Bermudez E, Mailloux RJ. The Uncoupling Proteins: A Systematic Review on the Mechanism Used in the Prevention of Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11020322. [PMID: 35204205 PMCID: PMC8868465 DOI: 10.3390/antiox11020322] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/12/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Mitochondrial uncoupling proteins (UCP) 1-3 fulfill many physiological functions, ranging from non-shivering thermogenesis (UCP1) to glucose-stimulated insulin release (GSIS) and satiety signaling (UCP2) and muscle fuel metabolism (UCP3). Several studies have suggested that UCPs mediate these functions by facilitating proton return to the matrix. This would decrease protonic backpressure on the respiratory chain, lowering the production of hydrogen peroxide (H2O2), a second messenger. However, controlling mitochondrial H2O2 production to prevent oxidative stress by activating these leaks through these proteins is still enthusiastically debated. This is due to compelling evidence that UCP2/3 fulfill other function(s) and the inability to reproduce findings that UCP1-3 use inducible leaks to control reactive oxygen species (ROS) production. Further, other studies have found that UCP2/3 may serve as Ca2+. Therefore, we performed a systematic review aiming to summarize the results collected on the topic. A literature search using a list of curated keywords in Pubmed, BIOSIS Citation Index and Scopus was conducted. Potentially relevant references were screened, duplicate references eliminated, and then literature titles and abstracts were evaluated using Rayyan software. A total of 1101 eligible studies were identified for the review. From this total, 416 studies were evaluated based on our inclusion criteria. In general, most studies identified a role for UCPs in preventing oxidative stress, and in some cases, this may be related to the induction of leaks and lowering protonic backpressure on the respiratory chain. However, some studies also generated evidence that UCP2/3 may mitigate oxidative stress by transporting Ca2+ into the matrix, exporting lipid hydroperoxides, or by transporting C-4 metabolites. Additionally, some showed that activating UCP1 or 3 can increase mitochondrial ROS production, even though there is still augmented protection from oxidative stress. Conclusion: Overall, most available studies demonstrate that UCPs, particularly UCP2/3, prevent oxidative stress. However, the mechanism utilized to do so remains elusive and raises the question that UCP2/3 should be renamed, since they may still not be true “uncoupling proteins”.
Collapse
|
74
|
Muroi H, Hori K, Tokutake Y, Hakamata Y, Kawabata F, Toyomizu M, Kikusato M. Oleuropein suppresses mitochondrial reactive oxygen species generation possibly via an activation of transient receptor potential V1 and sirtuin-1 in cultured chicken muscle cells. Anim Sci J 2022; 93:e13677. [PMID: 35029000 DOI: 10.1111/asj.13677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/16/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022]
Abstract
This study investigated the intracellular mechanism governing the effects of oleuropein (OLE), a phenolic compound of Olea europaea, on mRNA expression of avian uncoupling protein (avUCP) and mitochondrial biogenesis-related factors, and reactive oxygen species (mitROS) generation in a primary cultured chicken muscle cells. The OLE-treated cells exhibited increases in Avucp and ATP5a1z expression and a decrease in mitROS generation (p < 0.05), while the effects was canceled by sirtuin-1 (SIRT1) or transient receptor potential vanilloid 1 (TRPV1) inhibitors, EX-527 or BCTC, respectively. Intracellular Ca2+ concentration was significantly increased by OLE, while the induction was canceled by BCTC. The study also found that TRPV1 was expressed in the cell membrane and endoplasmic reticulum (ER), and Ca2+ could be released from ER in the OLE-treated cells. The OLE-treated cells exhibited increases in the phosphorylation ratio of AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) protein content. EX-527 and BCTC inhibitors canceled the effects of OLE on p-AMPK ratio and PGC-1α content, while EX-527 SIRT did not change PGC-1α content. The results suggest that the OLE effects may be due to Ca2+ release, possibly from TRPV1 at ER, and increased p-AMPK ratio, followed by SIRT1 activation and PGC-1α protein expression.
Collapse
Affiliation(s)
- Hikaru Muroi
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Kouhei Hori
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yukako Tokutake
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yuki Hakamata
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Fuminori Kawabata
- Physiology of Domestic Animals, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Japan
| | - Masaaki Toyomizu
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Motoi Kikusato
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
75
|
Ertuglu L, Yildiz A, Gamboa J, Ikizler TA. Skeletal muscle energetics in patients with moderate to advanced kidney disease. Kidney Res Clin Pract 2022; 41:14-21. [PMID: 35108768 PMCID: PMC8816417 DOI: 10.23876/j.krcp.21.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/05/2021] [Indexed: 11/04/2022] Open
Abstract
Sarcopenia, defined as decrease in muscle function and mass, is common in patients with moderate to advanced chronic kidney disease (CKD) and is associated with poor clinical outcomes. Muscle mitochondrial dysfunction is proposed as one of the mechanisms underlying sarcopenia. Patients with moderate to advanced CKD have decreased muscle mitochondrial content and oxidative capacity along with suppressed activity of various mitochondrial enzymes such as mitochondrial electron transport chain complexes and pyruvate dehydrogenase, leading to impaired energy production. Other mitochondrial abnormalities found in this population include defective beta-oxidation of fatty acids and mitochondrial DNA mutations. These changes are noticeable from the early stages of CKD and correlate with severity of the disease. Damage induced by uremic toxins, oxidative stress, and systemic inflammation has been implicated in the development of mitochondrial dysfunction in CKD patients. Given that mitochondrial function is an important determinant of physical activity and performance, its modulation is a potential therapeutic target for sarcopenia in patients with kidney disease. Coenzyme Q, nicotinamide, and cardiolipin-targeted peptides have been tested as therapeutic interventions in early studies. Aerobic exercise, a well-established strategy to improve muscle function and mass in healthy adults, is not as effective in patients with advanced kidney disease. This might be due to reduced expression or impaired activation of peroxisome proliferator-activated receptor-gamma coactivator 1α, the master regulator of mitochondrial biogenesis. Further studies are needed to broaden our understanding of the pathogenesis of mitochondrial dysfunction and to develop mitochondrial-targeted therapies for prevention and treatment of sarcopenia in patients with CKD.
Collapse
Affiliation(s)
- Lale Ertuglu
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Abdulmecit Yildiz
- Division of Nephrology, Department of Medicine, Uludag University, Bursa, Turkey
| | - Jorge Gamboa
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - T. Alp Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Health Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
- Correspondence: T. Alp Ikizler Division of Nephrology, Vanderbilt University Medical Center, 1161 21st Avenue South, S-3223 Medical Center North, Nashville 37232, TN, USA. E-mail:
| |
Collapse
|
76
|
Xun Z, Wipf P, McMurray CT. XJB-5-131 Is a Mild Uncoupler of Oxidative Phosphorylation. J Huntingtons Dis 2022; 11:141-151. [PMID: 35404288 PMCID: PMC9798833 DOI: 10.3233/jhd-220539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Mitochondria (MT) are energy "powerhouses" of the cell and the decline in their function from oxidative damage is strongly correlated in many diseases. To suppress oxygen damage, we have developed and applied XJB-5-131 as a targeted platform for neutralizing reactive oxygen species (ROS) directly in MT. Although the beneficial activity of XJB-5-131 is well documented, the mechanism of its protective effects is not yet fully understood. OBJECTIVE Here, we elucidate the mechanism of protection for XJB-5-131, a mitochondrial targeted antioxidant and electron scavenger. METHODS The Seahorse Flux Analyzer was used to probe the respiratory states of isolated mouse brain mitochondria treated with XJB-5-131 compared to controls. RESULTS Surprisingly, there is no direct impact of XJB-5-131 radical scavenger on the electron flow through the electron transport chain. Rather, XJB-5-131 is a mild uncoupler of oxidative phosphorylation. The nitroxide moiety in XJB-5-131 acts as a superoxide dismutase mimic, which both extracts or donates electrons during redox reactions. The electron scavenging activity of XJB-5-131 prevents the leakage of electrons and reduces formation of superoxide anion, thereby reducing ROS. CONCLUSION We show here that XJB-5-131 is a mild uncoupler of oxidative phosphorylation in MT. The mild uncoupling property of XJB-5-131 arises from its redox properties, which exert a protective effect by reducing ROS-induced damage without sacrificing energy production. Because mitochondrial decline is a common and central feature of toxicity, the favorable properties of XJB-5-131 are likely to be useful in treating Huntington's disease and a wide spectrum of neurodegenerative diseases for which oxidative damage is a key component. The mild uncoupling properties of XJB-5-131 suggest a valuable mechanism of action for the design of clinically effective antioxidants.
Collapse
Affiliation(s)
- Zhiyin Xun
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cynthia T. McMurray
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA,Correspondence to: Cynthia T. McMurray, Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA. Tel.: +1 510 486 6526; Fax: +1 510 486 6880;
| |
Collapse
|
77
|
Williams PT. Quantile-specific heritability of 8-isoprostane and the modulating effects of smoking, alcohol, cardiovascular disease and diabetes on 8-isoprostane-gene interactions. Free Radic Biol Med 2022; 178:262-270. [PMID: 34883250 PMCID: PMC10101173 DOI: 10.1016/j.freeradbiomed.2021.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Urinary 8-isoprostane provides a significantly heritable measure of oxidative stress. Prior reports suggest that genetic variants may modulate oxidative stress due to smoking, other environmental factors, and disease. Alternatively, these apparent modulations may reflect a dependence of genetic effects on 8-isoprostane concentrations. METHOD To test whether genetic effects on 8-isoprostane concentrations are quantile-dependent, quantile-specific offspring-parent (βOP) and full-sib regression slopes (βFS) were estimated by applying quantile regression to the age- and sex-adjusted creatinine-standardized urinary 8-isoprostane concentrations of Framingham Heart Study families. Quantile-specific heritabilities were calculated as h2 = 2βOP/(1+rspouse) and h2 = {(1+8rspouseβFS)0.5-1}/(2rspouse)). RESULTS Spouse 8-isoprostane concentrations were weakly concordant (rspouse = 0.06). 8-isoprostane heritability (h2±SE) increased significantly with increasing percentiles of its distribution (Plinear trend = 0.0009, Pquadratic trend = 0.0007, Pcubic trend = 0.003) when estimated from βOP, and when estimated from βFS (Plinear trend = 0.005, Pquadratic trend = 0.09, Pcubic trend = 0.06). Compared to the 10th percentile, βOP-estimated h2 was over 22-fold greater at the 90th percentile (Pdifference = 9.2 × 10-5), and 5.3-fold greater when estimated from βFS (Pdifference = 0.004). Significantly higher 8-isoprostane heritability in smokers than nonsmokers (0.352 ± 0.147 vs. 0.061 ± 0.036, Pdifference = 0.01), and heavier than lighter drinkers (0.449 ± 0.216 vs. 0.078 ± 0.037, Pdifference = 0.01) were eliminated when corrected for the higher 8-isoprostane concentrations of the smokers and heavier drinkers. CONCLUSION Heritability of oxidative stress as measured by 8-isoprostane is quantile-dependent, which may contribute to the larger reported effects on oxidative stress by UCP2 -866G > A, IL6 -572C > G and LTA 252A > G polymorphisms in smokers than nonsmokers, by the UCP2 -866G > A polymorphism in coronary heart disease patients, by the ESRRG rs1890552 A > G polymorphism in type 2 diabetics, by the CYBA 242C > T polymorphism after exercise training, by the PLIN 11482G > A/14995A > T haplotype before weight loss, and by the CYBA -930A > G and GSTP1 I105V haplotypes in patients with pulmonary edema.
Collapse
Affiliation(s)
- Paul T Williams
- Lawrence Berkeley National Laboratory, Molecular Biophysics & Integrated Bioimaging Division, 1 Cyclotron Road, Berkeley, CA, 94720, USA.
| |
Collapse
|
78
|
Wu ZS, Huang SM, Wang YC. Palmitate Enhances the Efficacy of Cisplatin and Doxorubicin against Human Endometrial Carcinoma Cells. Int J Mol Sci 2021; 23:ijms23010080. [PMID: 35008502 PMCID: PMC8744704 DOI: 10.3390/ijms23010080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
Endometrial cancer is the most common gynecological cancer worldwide. At present there is no effective screening test for its early detection and no curative treatment for women with advanced-stage or recurrent disease. Overexpression of fatty acid synthase is a common molecular feature of a subgroup of sex steroid-related cancers associated with poor prognoses, including endometrial cancers. Disruption of this fatty acid synthesis leads to cell apoptosis, making it a potential therapeutic target. The saturated fatty acid palmitate reportedly induces lipotoxicity and cell death by inducing oxidative stress in many cell types. Here, we explored the effects of palmitate combined with doxorubicin or cisplatin in the HEC-1-A and RL95-2 human endometrial cancer cell lines. The results showed that physiological concentrations of exogenous palmitate significantly increased cell cycle arrest, DNA damage, autophagy, and apoptosis in both RL95-2 and HEC-1-A cells. It also increased the chemosensitivity of both cell types. Notably, we did not observe that palmitate lipotoxicity reflected increased levels of reactive oxygen species, suggesting palmitate acts via a different mechanism in endometrial cancer. This study thus provides a potential therapeutic strategy in which palmitate is used as an adjuvant in the treatment of endometrial cancer.
Collapse
Affiliation(s)
- Zih-Syuan Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei City 114, Taiwan; (Z.-S.W.); (S.-M.H.)
| | - Shih-Ming Huang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei City 114, Taiwan; (Z.-S.W.); (S.-M.H.)
- Department of Biochemistry, National Defense Medical Center, Taipei City 114, Taiwan
| | - Yu-Chi Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei City 114, Taiwan; (Z.-S.W.); (S.-M.H.)
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114, Taiwan
- Correspondence:
| |
Collapse
|
79
|
Shi S, Wang L, van der Laan LJW, Pan Q, Verstegen MMA. Mitochondrial Dysfunction and Oxidative Stress in Liver Transplantation and Underlying Diseases: New Insights and Therapeutics. Transplantation 2021; 105:2362-2373. [PMID: 33577251 PMCID: PMC9005104 DOI: 10.1097/tp.0000000000003691] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 12/06/2022]
Abstract
Mitochondria are essential organelles for cellular energy and metabolism. Like with any organ, the liver highly depends on the function of these cellular powerhouses. Hepatotoxic insults often lead to an impairment of mitochondrial activity and an increase in oxidative stress, thereby compromising the metabolic and synthetic functions. Mitochondria play a critical role in ATP synthesis and the production or scavenging of free radicals. Mitochondria orchestrate many cellular signaling pathways involved in the regulation of cell death, metabolism, cell division, and progenitor cell differentiation. Mitochondrial dysfunction and oxidative stress are closely associated with ischemia-reperfusion injury during organ transplantation and with different liver diseases, including cholestasis, steatosis, viral hepatitis, and drug-induced liver injury. To develop novel mitochondria-targeting therapies or interventions, a better understanding of mitochondrial dysfunction and oxidative stress in hepatic pathogenesis is very much needed. Therapies targeting mitochondria impairment and oxidative imbalance in liver diseases have been extensively studied in preclinical and clinical research. In this review, we provide an overview of how oxidative stress and mitochondrial dysfunction affect liver diseases and liver transplantation. Furthermore, we summarize recent developments of antioxidant and mitochondria-targeted interventions.
Collapse
Affiliation(s)
- Shaojun Shi
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Ling Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
80
|
Mitochondrial Uncoupling Proteins (UCPs) as Key Modulators of ROS Homeostasis: A Crosstalk between Diabesity and Male Infertility? Antioxidants (Basel) 2021; 10:antiox10111746. [PMID: 34829617 PMCID: PMC8614977 DOI: 10.3390/antiox10111746] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/14/2022] Open
Abstract
Uncoupling proteins (UCPs) are transmembrane proteins members of the mitochondrial anion transporter family present in the mitochondrial inner membrane. Currently, six homologs have been identified (UCP1-6) in mammals, with ubiquitous tissue distribution and multiple physiological functions. UCPs are regulators of key events for cellular bioenergetic metabolism, such as membrane potential, metabolic efficiency, and energy dissipation also functioning as pivotal modulators of ROS production and general cellular redox state. UCPs can act as proton channels, leading to proton re-entry the mitochondrial matrix from the intermembrane space and thus collapsing the proton gradient and decreasing the membrane potential. Each homolog exhibits its specific functions, from thermogenesis to regulation of ROS production. The expression and function of UCPs are intimately linked to diabesity, with their dysregulation/dysfunction not only associated to diabesity onset, but also by exacerbating oxidative stress-related damage. Male infertility is one of the most overlooked diabesity-related comorbidities, where high oxidative stress takes a major role. In this review, we discuss in detail the expression and function of the different UCP homologs. In addition, the role of UCPs as key regulators of ROS production and redox homeostasis, as well as their influence on the pathophysiology of diabesity and potential role on diabesity-induced male infertility is debated.
Collapse
|
81
|
Albracht-Schulte K, Wilson S, Johnson P, Pahlavani M, Ramalingam L, Goonapienuwala B, Kalupahana NS, Festuccia WT, Scoggin S, Kahathuduwa CN, Moustaid-Moussa N. Sex-Dependent Effects of Eicosapentaenoic Acid on Hepatic Steatosis in UCP1 Knockout Mice. Biomedicines 2021; 9:1549. [PMID: 34829779 PMCID: PMC8615653 DOI: 10.3390/biomedicines9111549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/26/2022] Open
Abstract
Visceral obesity may be a driving factor in nonalcoholic fatty liver disease (NAFLD) development. Previous studies have shown that the omega-3 polyunsaturated fatty acid, eicosapentaenoic acid (EPA), ameliorates obesity in high-fat (HF) fed male, C57Bl/6 mice at thermoneutral conditions, independent of uncoupling protein 1 (UCP1). Our goals herein were to investigate sex-dependent mechanisms of EPA in the livers of wild type (WT) and UCP1 knockout (KO) male and female mice fed a HF diet (45% kcal fat; WT-HF, KO-HF) with or without supplementation of 36 g/kg EPA (WT-EPA, KO-EPA). KO significantly increased body weight in males, with no significant reductions with EPA in the WT or KO groups. In females, there were no significant differences in body weight among KO groups and no effects of EPA. In males, liver TGs were significantly higher in the KO-HF group and reduced with EPA, which was not observed in females. Accordingly, gene and protein markers of mitochondrial oxidation, peroxisomal biogenesis and oxidation, as well as metabolic futile cycles were sex-dependently impacted by KO and EPA supplementation. These findings suggest a genotypic difference in response to dietary EPA supplementation on the livers of male and female mice with diet-induced obesity and housed at thermoneutrality.
Collapse
Affiliation(s)
- Kembra Albracht-Schulte
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Savanna Wilson
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Paige Johnson
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Mandana Pahlavani
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Latha Ramalingam
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Bimba Goonapienuwala
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Nishan S. Kalupahana
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
- Department of Physiology, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - William T. Festuccia
- Department of Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil;
| | - Shane Scoggin
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| | - Chanaka N. Kahathuduwa
- Texas Tech University Health Sciences Center, Department of Laboratory Sciences and Primary Care, Lubbock, TX 79430, USA;
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.A.-S.); (S.W.); (P.J.); (M.P.); (L.R.); (B.G.); (N.S.K.); (S.S.)
| |
Collapse
|
82
|
González-García P, Barriocanal-Casado E, Díaz-Casado ME, López-Herrador S, Hidalgo-Gutiérrez A, López LC. Animal Models of Coenzyme Q Deficiency: Mechanistic and Translational Learnings. Antioxidants (Basel) 2021; 10:antiox10111687. [PMID: 34829558 PMCID: PMC8614664 DOI: 10.3390/antiox10111687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/21/2021] [Accepted: 10/23/2021] [Indexed: 12/16/2022] Open
Abstract
Coenzyme Q (CoQ) is a vital lipophilic molecule that is endogenously synthesized in the mitochondria of each cell. The CoQ biosynthetic pathway is complex and not completely characterized, and it involves at least thirteen catalytic and regulatory proteins. Once it is synthesized, CoQ exerts a wide variety of mitochondrial and extramitochondrial functions thank to its redox capacity and its lipophilicity. Thus, low levels of CoQ cause diseases with heterogeneous clinical symptoms, which are not always understood. The decreased levels of CoQ may be primary caused by defects in the CoQ biosynthetic pathway or secondarily associated with other diseases. In both cases, the pathomechanisms are related to the CoQ functions, although further experimental evidence is required to establish this association. The conventional treatment for CoQ deficiencies is the high doses of oral CoQ10 supplementation, but this therapy is not effective for some specific clinical presentations, especially in those involving the nervous system. To better understand the CoQ biosynthetic pathway, the biological functions linked to CoQ and the pathomechanisms of CoQ deficiencies, and to improve the therapeutic outcomes of this syndrome, a variety of animal models have been generated and characterized in the last decade. In this review, we show all the animal models available, remarking on the most important outcomes that each model has provided. Finally, we also comment some gaps and future research directions related to CoQ metabolism and how the current and novel animal models may help in the development of future research studies.
Collapse
Affiliation(s)
- Pilar González-García
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
- Correspondence: (P.G.-G.); (L.C.L.)
| | - Eliana Barriocanal-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - María Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Sergio López-Herrador
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
| | - Luis C. López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; (E.B.-C.); (M.E.D.-C.); (S.L.-H.); (A.H.-G.)
- Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain
- Correspondence: (P.G.-G.); (L.C.L.)
| |
Collapse
|
83
|
The Adrenergic Nerve Network in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:271-294. [PMID: 34664245 DOI: 10.1007/978-3-030-73119-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
The central and autonomic nervous systems interact and converge to build up an adrenergic nerve network capable of promoting cancer. While a local adrenergic sympathetic innervation in peripheral solid tumors influences cancer and stromal cell behavior, the brain can participate to the development of cancer through an intermixed dysregulation of the sympathoadrenal system, adrenergic neurons, and the hypothalamo-pituitary-adrenal axis. A deeper understanding of the adrenergic nerve circuitry within the brain and tumors and its interactions with the microenvironment should enable elucidation of original mechanisms of cancer and novel therapeutic strategies.
Collapse
|
84
|
Bucher M, Kadam L, Ahuna K, Myatt L. Differences in Glycolysis and Mitochondrial Respiration between Cytotrophoblast and Syncytiotrophoblast In-Vitro: Evidence for Sexual Dimorphism. Int J Mol Sci 2021; 22:ijms221910875. [PMID: 34639216 PMCID: PMC8509198 DOI: 10.3390/ijms221910875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 01/18/2023] Open
Abstract
In the placenta the proliferative cytotrophoblast cells fuse into the terminally differentiated syncytiotrophoblast layer which undertakes several energy-intensive functions including nutrient uptake and transfer and hormone synthesis. We used Seahorse glycolytic and mitochondrial stress tests on trophoblast cells isolated at term from women of healthy weight to evaluate if cytotrophoblast (CT) and syncytiotrophoblast (ST) have different bioenergetic strategies, given their different functions. Whereas there are no differences in basal glycolysis, CT have significantly greater glycolytic capacity and reserve than ST. In contrast, ST have significantly higher basal, ATP-coupled and maximal mitochondrial respiration and spare capacity than CT. Consequently, under stress conditions CT can increase energy generation via its higher glycolytic capacity whereas ST can use its higher and more efficient mitochondrial respiration capacity. We have previously shown that with adverse in utero conditions of diabetes and obesity trophoblast respiration is sexually dimorphic. We found no differences in glycolytic parameters between sexes and no difference in mitochondrial respiration parameters other than increases seen upon syncytialization appear to be greater in females. There were differences in metabolic flexibility, i.e., the ability to use glucose, glutamine, or fatty acids, seen upon syncytialization between the sexes with increased flexibility in female trophoblast suggesting a better ability to adapt to changes in nutrient supply.
Collapse
|
85
|
Sunil C, Zheng X, Yang Z, Cui K, Su Y, Xu B. Antifatigue effects of Hechong (Tylorrhynchus heterochaetus) through modulation of Nrf2/ARE- mediated antioxidant signaling pathway. Food Chem Toxicol 2021; 157:112589. [PMID: 34601041 DOI: 10.1016/j.fct.2021.112589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 12/29/2022]
Abstract
This study was to evaluate the antifatigue effect of T. heterochaetus and explore the underlying mechanism of action. T. heterochaetus extract was treated to mice for 28 days. On the 28th day, after weight loaded swimming test. The levels of antioxidant enzymes and levels of pro- and anti-inflammatory cytokines in the liver and muscles of exercised mice were evaluated. mRNA and protein expression levels of Nrf2, SOD, HO-1, and Keap-1 were evaluated using RT-PCR and western blot analysis. The low (2.70 mg/0.5 ml/20 g) and medium (5.41 mg/0.5 ml/20 g) dose enhanced the activities of antioxidant enzymes like SOD, CAT and GPx in the liver and skeletal muscle thereby enhancing the antifatigue effect. The low and medium doses showed good anti-inflammatory effects by evaluating the levels of pro and anti-inflammatory cytokines such as TNF-α, IL-1β, IL-6, and IL-10 both in the liver and skeletal muscle. Furthermore, RT-PCR and western blot analysis showed increased expression of HO-1, SOD, Nrf2, and decreased expression of Keap-1 gene and proteins in liver and skeletal muscle of T. heterochaetus treated group mice. The current results indicate that T. heterochaetus exert the antifatigue effect through attenuating oxidative stress injury and inflammatory responses through the Nrf2/ARE-mediated signaling pathway.
Collapse
Affiliation(s)
- Christudas Sunil
- Food Science and Technology Programme, BNU-HKBU United International College, Zhuhai, Guangdong, 519087, China
| | - Xuchu Zheng
- Food Science and Technology Programme, BNU-HKBU United International College, Zhuhai, Guangdong, 519087, China
| | - Ziqing Yang
- Food Science and Technology Programme, BNU-HKBU United International College, Zhuhai, Guangdong, 519087, China
| | - Kuopeng Cui
- Estuarine Fisheries Research Institute of Doumen, Zhuhai, Guangdong, 519000, China
| | - Yuepeng Su
- Estuarine Fisheries Research Institute of Doumen, Zhuhai, Guangdong, 519000, China
| | - Baojun Xu
- Food Science and Technology Programme, BNU-HKBU United International College, Zhuhai, Guangdong, 519087, China.
| |
Collapse
|
86
|
Ritchie DJ, Friesen CR. Invited review: Thermal effects on oxidative stress in vertebrate ectotherms. Comp Biochem Physiol A Mol Integr Physiol 2021; 263:111082. [PMID: 34571153 DOI: 10.1016/j.cbpa.2021.111082] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/07/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
Human-induced climate change is occurring rapidly. Ectothermic organisms are particularly vulnerable to these temperature changes due to their reliance on environmental temperature. The extent of ectothermic thermal adaptation and plasticity in the literature is well documented; however, the role of oxidative stress in these processes needs more attention. Oxidative stress occurs when reactive oxygen species, generated mainly through aerobic respiration, overwhelm antioxidant defences and damage crucial biomolecules. The effects of oxidative damage include the alteration of life-history traits and reductions in whole-organism fitness. Here we review the literature addressing experimental temperature effects on oxidative stress in vertebrate ectotherms. Acute and acclimation temperature treatments produce distinctly different results and highlight the role of phylogeny and thermal adaptation in shaping oxidative stress responses. Acute treatments on organisms adapted to stable environments generally produced significant oxidative stress responses, whilst organisms adapted to variable conditions exhibited capacity to cope with temperature changes and mitigate oxidative stress. In acclimation treatments, the temperature treatments higher than optimal temperatures tended to produce significantly less oxidative stress than lower temperatures in reptiles, whilst in some eurythermal fish species, no oxidative stress response was observed. These results highlight the importance of phylogeny and adaptation to past environmental conditions for temperature-dependent oxidative stress responses. We conclude with recommendations on experimental procedures to investigate these phenomena with reference to thermal plasticity, adaptation and biogeographic variation that provide the most significant benefits to adaptable populations. These results have potential conservation ramifications as they may shed light on the physiological effects of temperature alterations in some vertebrate ectotherms.
Collapse
Affiliation(s)
- Daniel J Ritchie
- School of Earth, Atmospheric and Life Sciences, The University of Wollongong, 2522 Wollongong, New South Wales, Australia
| | - Christopher R Friesen
- School of Earth, Atmospheric and Life Sciences, The University of Wollongong, 2522 Wollongong, New South Wales, Australia; School of Life and Environmental Sciences, University of Sydney, Heydon-Laurence Bldg A08, Science Road, Sydney, NSW 2006, Australia.
| |
Collapse
|
87
|
Targeting Reactive Oxygen Species Capacity of Tumor Cells with Repurposed Drug as an Anticancer Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8532940. [PMID: 34539975 PMCID: PMC8443364 DOI: 10.1155/2021/8532940] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
Accumulating evidence shows that elevated levels of reactive oxygen species (ROS) are associated with cancer initiation, growth, and response to therapies. As concentrations increase, ROS influence cancer development in a paradoxical way, either triggering tumorigenesis and supporting the proliferation of cancer cells at moderate levels of ROS or causing cancer cell death at high levels of ROS. Thus, ROS can be considered an attractive target for therapy of cancer and two apparently contradictory but virtually complementary therapeutic strategies for the regulation of ROS to treat cancer. Despite tremendous resources being invested in prevention and treatment for cancer, cancer remains a leading cause of human deaths and brings a heavy burden to humans worldwide. Chemotherapy remains the key treatment for cancer therapy, but it produces harmful side effects. Meanwhile, the process of de novo development of new anticancer drugs generally needs increasing cost, long development cycle, and high risk of failure. The use of ROS-based repurposed drugs may be one of the promising ways to overcome current cancer treatment challenges. In this review, we briefly introduce the source and regulation of ROS and then focus on the status of repurposed drugs based on ROS regulation for cancer therapy and propose the challenges and direction of ROS-mediated cancer treatment.
Collapse
|
88
|
A Role of Stress Sensor Nrf2 in Stimulating Thermogenesis and Energy Expenditure. Biomedicines 2021; 9:biomedicines9091196. [PMID: 34572382 PMCID: PMC8472024 DOI: 10.3390/biomedicines9091196] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/01/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
During chronic cold stress, thermogenic adipocytes generate heat through uncoupling of mitochondrial respiration from ATP synthesis. Recent discovery of various dietary phytochemicals, endogenous metabolites, synthetic compounds, and their molecular targets for stimulating thermogenesis has provided promising strategies to treat or prevent obesity and its associated metabolic diseases. Nuclear factor E2 p45-related factor 2 (Nrf2) is a stress response protein that plays an important role in obesity and metabolisms. However, both Nrf2 activation and Nrf2 inhibition can suppress obesity and metabolic diseases. Here, we summarized and discussed conflicting findings of Nrf2 activities accounting for part of the variance in thermogenesis and energy metabolism. We also discussed the utility of Nrf2-activating mechanisms for their potential applications in stimulating energy expenditure to prevent obesity and improve metabolic deficits.
Collapse
|
89
|
Sinenko SA, Starkova TY, Kuzmin AA, Tomilin AN. Physiological Signaling Functions of Reactive Oxygen Species in Stem Cells: From Flies to Man. Front Cell Dev Biol 2021; 9:714370. [PMID: 34422833 PMCID: PMC8377544 DOI: 10.3389/fcell.2021.714370] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS), superoxide anion and hydrogen peroxide, are generated as byproducts of oxidative phosphorylation in the mitochondria or via cell signaling-induced NADPH oxidases in the cytosol. In the recent two decades, a plethora of studies established that elevated ROS levels generated by oxidative eustress are crucial physiological mediators of many cellular and developmental processes. In this review, we discuss the mechanisms of ROS generation and regulation, current understanding of ROS functions in the maintenance of adult and embryonic stem cells, as well as in the process of cell reprogramming to a pluripotent state. Recently discovered cell-non-autonomous ROS functions mediated by growth factors are crucial for controlling cell differentiation and cellular immune response in Drosophila. Importantly, many physiological functions of ROS discovered in Drosophila may allow for deciphering and understanding analogous processes in human, which could potentially lead to the development of novel therapeutic approaches in ROS-associated diseases treatment.
Collapse
Affiliation(s)
- Sergey A Sinenko
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | | | - Andrey A Kuzmin
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Alexey N Tomilin
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
90
|
Ardalan A, Sowlati-Hashjin S, Oduwoye H, Uwumarenogie SO, Karttunen M, Smith MD, Jelokhani-Niaraki M. Biphasic Proton Transport Mechanism for Uncoupling Proteins. J Phys Chem B 2021; 125:9130-9144. [PMID: 34365794 DOI: 10.1021/acs.jpcb.1c04766] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
It has been suggested that uncoupling proteins (UCPs) transport protons via interconversion between two conformational states: one in the "cytoplasmic state" and the other in the "matrix state". Matrix and cytoplasmic salt-bridge networks are key controllers of these states. This study proposes a mechanism for proton transport in tetrameric UCP2, with focus on the role of the matrix network. Eleven mutants were prepared to disrupt (K → Q or D → N mutations) or alter (K → D and D → K mutations) the salt-bridges in the matrix network. Proteins were recombinantly expressed in Escherichia coli membrane, reconstituted in model lipid membranes, and their structures and functions were analyzed by gel electrophoresis, circular dichroism spectroscopy, fluorescence assays, as well as molecular dynamics simulations. It is shown that the UCP2 matrix network contains five salt-bridges (rather than the previously reported three), and the matrix network can regulate the proton transport by holding the protein's transmembrane helices in close proximity, limiting the movement of the activator fatty acid(s). A biphasic two-state molecular model is proposed for proton transport in tetrameric (a dimer of stable dimers) UCP2, in which all the monomers are functional, and monomers in each dimer are in the same transport mode. Purine nucleotide (e.g., ATP) can occlude the internal pore of the monomeric units of UCP tetramers via interacting with positive residues at or in the proximity of the matrix network (K38, K141, K239, R88, R185, and R279) and prevent switching between cytoplasmic and matrix states, thus inhibiting the proton transport. This study provides new insights into the mechanism of proton transport and regulation in UCPs.
Collapse
Affiliation(s)
- Afshan Ardalan
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
| | - Shahin Sowlati-Hashjin
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 3K7, Canada.,The Center for Advanced Materials and Biomaterials Research, The University of Western Ontario, London, Ontario N6K 3K7, Canada
| | - Habib Oduwoye
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
| | - Stephanie O Uwumarenogie
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
| | - Mikko Karttunen
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 3K7, Canada.,The Center for Advanced Materials and Biomaterials Research, The University of Western Ontario, London, Ontario N6K 3K7, Canada.,Department of Physics and Astronomy, The University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Matthew D Smith
- Department of Biology, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
| | - Masoud Jelokhani-Niaraki
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
| |
Collapse
|
91
|
Arulkumaran N, Pollen SJ, Tidswell R, Gaupp C, Peters VBM, Stanzani G, Snow TAC, Duchen MR, Singer M. Selective mitochondrial antioxidant MitoTEMPO reduces renal dysfunction and systemic inflammation in experimental sepsis. Br J Anaesth 2021; 127:577-586. [PMID: 34332740 DOI: 10.1016/j.bja.2021.05.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/04/2021] [Accepted: 05/27/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Excess mitochondrial reactive oxygen species (mROS) in sepsis is associated with organ failure, in part by generating inflammation through the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome. We determined the impact of a mitochondrial-targeted antioxidant (MitoTEMPO) on mitochondrial dysfunction in renal proximal tubular epithelial cells, peritoneal immune cell function ex vivo, and organ dysfunction in a rat model of sepsis. METHODS The effects of MitoTEMPO were assessed ex vivo using adenosine triphosphate and lipopolysaccharide-stimulated rat peritoneal immune cells and fresh rat kidney slices exposed to serum from septic rats. We assessed mROS production and phagocytotic capacity (flow cytometry), mitochondrial functionality (multiphoton imaging, respirometry), and NLRP3 inflammasome activation in cell culture. The effect of MitoTEMPO on organ dysfunction was evaluated in a rat model of faecal peritonitis. RESULTS MitoTEMPO decreased septic serum-induced mROS (P<0.001) and maintained normal reduced nicotinamide adenine dinucleotide redox state (P=0.02) and mitochondrial membrane potential (P<0.001) in renal proximal tubular epithelial cells ex vivo. In lipopolysaccharide-stimulated peritoneal immune cells, MitoTEMPO abrogated the increase in mROS (P=0.006) and interleukin-1β (IL-1β) (P=0.03) without affecting non-mitochondrial oxygen consumption or the phagocytotic-induced respiratory burst (P>0.05). In vivo, compared with untreated septic animals, MitoTEMPO reduced systemic IL-1β (P=0.01), reduced renal oxidative stress as determined by urine isoprostane levels (P=0.04), and ameliorated renal dysfunction (reduced serum urea (P<0.001) and creatinine (P=0.05). CONCLUSIONS Reduction of mROS by a mitochondria-targeted antioxidant reduced IL-1β, and protected mitochondrial, cellular, and organ functionality after septic insults.
Collapse
Affiliation(s)
- Nishkantha Arulkumaran
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK.
| | - Sean J Pollen
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Robert Tidswell
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Charlotte Gaupp
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Vera B M Peters
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Giacomo Stanzani
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Timothy A C Snow
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Michael R Duchen
- Department of Cell and Development Biology, University College London, London, UK
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| |
Collapse
|
92
|
Botto L, Bulbarelli A, Lonati E, Cazzaniga E, Tassotti M, Mena P, Del Rio D, Palestini P. Study of the Antioxidant Effects of Coffee Phenolic Metabolites on C6 Glioma Cells Exposed to Diesel Exhaust Particles. Antioxidants (Basel) 2021; 10:antiox10081169. [PMID: 34439417 PMCID: PMC8388867 DOI: 10.3390/antiox10081169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/12/2021] [Accepted: 07/19/2021] [Indexed: 01/17/2023] Open
Abstract
The contributing role of environmental factors to the development of neurodegenerative diseases has become increasingly evident. Here, we report that exposure of C6 glioma cells to diesel exhaust particles (DEPs), a major constituent of urban air pollution, causes intracellular reactive oxygen species (ROS) production. In this scenario, we suggest employing the possible protective role that coffee phenolic metabolites may have. Coffee is a commonly consumed hot beverage and a major contributor to the dietary intake of (poly) phenols. Taking into account physiological concentrations, we analysed the effects of two different coffee phenolic metabolites mixes consisting of compounds derived from bacterial metabolization reactions or phase II conjugations, as well as caffeic acid. The results showed that these mixes were able to counteract DEP-induced oxidative stress. The cellular components mediating the downregulation of ROS included extracellular signal-regulated kinase 1/2 (ERK1/2), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and uncoupling protein 2 (UCP2). Contrary to coffee phenolic metabolites, the treatment with N-acetylcysteine (NAC), a known antioxidant, was found to be ineffective in preventing the DEP exposure oxidant effect. These results revealed that coffee phenolic metabolites could be promising candidates to protect against some adverse health effects of daily exposure to air pollution.
Collapse
Affiliation(s)
- Laura Botto
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.B.); (A.B.); (E.L.); (E.C.)
| | - Alessandra Bulbarelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.B.); (A.B.); (E.L.); (E.C.)
- POLARIS Centre, University of Milano-Bicocca, 20126 Milano, Italy
- Bicocca Center of Science and Technology for Food, University of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Elena Lonati
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.B.); (A.B.); (E.L.); (E.C.)
- Bicocca Center of Science and Technology for Food, University of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Emanuela Cazzaniga
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.B.); (A.B.); (E.L.); (E.C.)
- Bicocca Center of Science and Technology for Food, University of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Michele Tassotti
- Human Nutrition Unit, Department of Food and Drug, University of Parma, 43121 Parma, Italy; (M.T.); (P.M.); (D.D.R.)
| | - Pedro Mena
- Human Nutrition Unit, Department of Food and Drug, University of Parma, 43121 Parma, Italy; (M.T.); (P.M.); (D.D.R.)
| | - Daniele Del Rio
- Human Nutrition Unit, Department of Food and Drug, University of Parma, 43121 Parma, Italy; (M.T.); (P.M.); (D.D.R.)
- School of Advanced Studies on Food and Nutrition, University of Parma, 43121 Parma, Italy
| | - Paola Palestini
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.B.); (A.B.); (E.L.); (E.C.)
- POLARIS Centre, University of Milano-Bicocca, 20126 Milano, Italy
- Bicocca Center of Science and Technology for Food, University of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
- Correspondence:
| |
Collapse
|
93
|
Wasyluk W, Nowicka-Stążka P, Zwolak A. Heart Metabolism in Sepsis-Induced Cardiomyopathy-Unusual Metabolic Dysfunction of the Heart. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18147598. [PMID: 34300048 PMCID: PMC8303349 DOI: 10.3390/ijerph18147598] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/26/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022]
Abstract
Due to the need for continuous work, the heart uses up to 8% of the total energy expenditure. Due to the relatively low adenosine triphosphate (ATP) storage capacity, the heart's work is dependent on its production. This is possible due to the metabolic flexibility of the heart, which allows it to use numerous substrates as a source of energy. Under normal conditions, a healthy heart obtains approximately 95% of its ATP by oxidative phosphorylation in the mitochondria. The primary source of energy is fatty acid oxidation, the rest of the energy comes from the oxidation of pyruvate. A failed heart is characterised by a disturbance in these proportions, with the contribution of individual components as a source of energy depending on the aetiology and stage of heart failure. A unique form of cardiac dysfunction is sepsis-induced cardiomyopathy, characterised by a significant reduction in energy production and impairment of cardiac oxidation of both fatty acids and glucose. Metabolic disorders appear to contribute to the pathogenesis of cardiac dysfunction and therefore are a promising target for future therapies. However, as many aspects of the metabolism of the failing heart remain unexplained, this issue requires further research.
Collapse
Affiliation(s)
- Weronika Wasyluk
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-093 Lublin, Poland; (P.N.-S.); (A.Z.)
- Doctoral School, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence:
| | - Patrycja Nowicka-Stążka
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-093 Lublin, Poland; (P.N.-S.); (A.Z.)
| | - Agnieszka Zwolak
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-093 Lublin, Poland; (P.N.-S.); (A.Z.)
| |
Collapse
|
94
|
He J, Liu R, Zheng W, Guo H, Yang Y, Zhao R, Yao W. High ambient temperature exposure during late gestation disrupts glycolipid metabolism and hepatic mitochondrial function tightly related to gut microbial dysbiosis in pregnant mice. Microb Biotechnol 2021; 14:2116-2129. [PMID: 34272826 PMCID: PMC8449678 DOI: 10.1111/1751-7915.13893] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/03/2021] [Indexed: 12/11/2022] Open
Abstract
As global warming intensifies, emerging evidence has demonstrated high ambient temperature during pregnancy negatively affects maternal physiology with compromised pregnant outcomes; however, little is known about the roles of gut microbiota and its underlying mechanisms in this process. Here, for the first time, we explored the potential mechanisms of gut microbiota involved in the disrupted glycolipid metabolism via hepatic mitochondrial function. Our results indicate heat stress (HS) reduces fat and protein contents and serum levels of insulin and triglyceride (TG), while increases that of non-esterified fatty acid (NEFA), β-hydroxybutyric acid (B-HBA), creatinine and blood urea nitrogen (BUN) (P < 0.05). Additionally, HS downregulates both mitochondrial genes (mtDNA) and nuclear encoding mitochondrial functional genes with increasing serum levels of malondialdehyde (MDA) and 8-hydroxydeoxyguanosine (8-OHdG) (P < 0.05). Regarding microbial response, HS boosts serum levels of lipopolysaccharide (LPS) (P < 0.05) and alters β-diversity (ANOSIM, P < 0.01), increasing the proportions of Escherichia-Shigella, Acinetobacter and Klebsiella (q < 0.05), while reducing that of Ruminiclostridium, Blautia, Lachnospiraceae_NK4A136_group, Clostridium VadinBB60 and Muribaculaceae (q < 0.05). PICRUSt analysis predicts that HS upregulates 11 KEGG pathways, mainly including bile secretion and bacterial invasion of epithelial cells. The collective results suggest that microbial dysbiosis due to late gestational HS has strong associations with damaged hepatic mitochondrial function and disrupted metabolic profiles.
Collapse
Affiliation(s)
- Jianwen He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.,Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Riliang Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Weijiang Zheng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Huiduo Guo
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yunnan Yang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ruqian Zhao
- Key Lab of Animal Physiology and Biochemistry, Nanjing Agricultural University, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Nanjing, 210095, China
| | - Wen Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.,Key Lab of Animal Physiology and Biochemistry, Nanjing Agricultural University, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Nanjing, 210095, China
| |
Collapse
|
95
|
Decker ST, Kwon OS, Zhao J, Hoidal JR, Heuckstadt T, Richardson RS, Sanders KA, Layec G. Skeletal muscle mitochondrial adaptations induced by long-term cigarette smoke exposure. Am J Physiol Endocrinol Metab 2021; 321:E80-E89. [PMID: 34121449 PMCID: PMC8321829 DOI: 10.1152/ajpendo.00544.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022]
Abstract
Because patients with chronic obstructive pulmonary disease (COPD) are often physically inactive, it is still unclear whether the lower respiratory capacity in the locomotor muscles of these patients is due to cigarette smoking per se or is secondary to physical deconditioning. Accordingly, the purpose of this study was to examine mitochondrial alterations in the quadriceps muscle of 10 mice exposed to 8 mo of cigarette smoke, a sedentary mouse model of emphysema, and 9 control mice, using immunoblotting, spectrophotometry, and high-resolution respirometry in permeabilized muscle fibers. Mice exposed to smoke displayed a twofold increase in the oxidative stress marker, 4-HNE, (P < 0.05) compared with control mice. This was accompanied by significant decrease in protein expression of UCP3 (65%), ANT (58%), and mitochondrial complexes II-V (∼60%-75%). In contrast, maximal ADP-stimulated respiration with complex I and II substrates (CON: 23.6 ± 6.6 and SMO: 19.2 ± 8.2 ρM·mg-1·s-1) or octanoylcarnitine (CON: 21.8 ± 9.0 and SMO: 16.5 ± 6.6 ρM·mg-1·s-1) measured in permeabilized muscle fibers, as well as citrate synthase activity, were not significantly different between groups. Collectively, our findings revealed that sedentary mice exposed to cigarette smoke for 8 mo, which is typically associated with pulmonary inflammation and emphysema, exhibited a preserved mitochondrial respiratory capacity for various substrates, including fatty acid, in the skeletal muscle. However, the mitochondrial adaptations induced by cigarette smoke favored the development of chronic oxidative stress, which can indirectly contribute to augment the susceptibility to muscle fatigue and exercise intolerance.NEW & NOTEWORTHY It is unclear whether the exercise intolerance and skeletal muscle mitochondrial dysfunction observed in patients with COPD is due to cigarette smoke exposure, per se, or if they are secondary consequences to inactivity. Herein, while long-term exposure to cigarette smoke induces oxidative stress and an altered skeletal muscle phenotype, cigarette smoke does not directly contribute to mitochondrial dysfunction. With this evidence, we demonstrate the critical role of physical inactivity in cigarette smoke-related skeletal muscle dysfunction.
Collapse
Affiliation(s)
- Stephen T Decker
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Oh-Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, Connecticut
- UConn Center on Aging and Department of Orthopaedic Surgery, University of Connecticut, School of Medicine, Farmington, Connecticut
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, Utah
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Jia Zhao
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, Utah
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - John R Hoidal
- Department of Internal Medicine, Pulmonary Division, University of Utah, Salt Lake City, Utah
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Thomas Heuckstadt
- Department of Internal Medicine, Pulmonary Division, University of Utah, Salt Lake City, Utah
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Russell S Richardson
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, Utah
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Karl A Sanders
- Department of Internal Medicine, Pulmonary Division, University of Utah, Salt Lake City, Utah
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Gwenael Layec
- Department of Kinesiology, University of Massachusetts Amherst, Amherst, Massachusetts
- Institute of Applied Life Science, University of Massachusetts Amherst, Amherst, Massachusetts
- Geriatric Research, Education, and Clinical Center, George E. Wahlen VA Medical Center, Salt Lake City, Utah
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| |
Collapse
|
96
|
McGuirk S, Audet-Delage Y, Annis MG, Xue Y, Vernier M, Zhao K, St-Louis C, Minarrieta L, Patten DA, Morin G, Greenwood CM, Giguère V, Huang S, Siegel PM, St-Pierre J. Resistance to different anthracycline chemotherapeutics elicits distinct and actionable primary metabolic dependencies in breast cancer. eLife 2021; 10:65150. [PMID: 34181531 PMCID: PMC8238502 DOI: 10.7554/elife.65150] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/12/2021] [Indexed: 12/18/2022] Open
Abstract
Chemotherapy resistance is a critical barrier in cancer treatment. Metabolic adaptations have been shown to fuel therapy resistance; however, little is known regarding the generality of these changes and whether specific therapies elicit unique metabolic alterations. Using a combination of metabolomics, transcriptomics, and functional genomics, we show that two anthracyclines, doxorubicin and epirubicin, elicit distinct primary metabolic vulnerabilities in human breast cancer cells. Doxorubicin-resistant cells rely on glutamine to drive oxidative phosphorylation and de novo glutathione synthesis, while epirubicin-resistant cells display markedly increased bioenergetic capacity and mitochondrial ATP production. The dependence on these distinct metabolic adaptations is revealed by the increased sensitivity of doxorubicin-resistant cells and tumor xenografts to buthionine sulfoximine (BSO), a drug that interferes with glutathione synthesis, compared with epirubicin-resistant counterparts that are more sensitive to the biguanide phenformin. Overall, our work reveals that metabolic adaptations can vary with therapeutics and that these metabolic dependencies can be exploited as a targeted approach to treat chemotherapy-resistant breast cancer.
Collapse
Affiliation(s)
- Shawn McGuirk
- Department of Biochemistry, Faculty of Medicine, McGill University, Montreal, Canada.,Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Yannick Audet-Delage
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada.,Ottawa Institute of Systems Biology, Ottawa, Canada
| | - Matthew G Annis
- Goodman Cancer Research Centre, McGill University, Montreal, Canada.,Department of Medicine, Faculty of Medicine, McGill University, Montreal, Canada
| | - Yibo Xue
- Department of Biochemistry, Faculty of Medicine, McGill University, Montreal, Canada.,Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Mathieu Vernier
- Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Kaiqiong Zhao
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada.,Lady Davis Institute, Jewish General Hospital, Montreal, Canada
| | - Catherine St-Louis
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada.,Ottawa Institute of Systems Biology, Ottawa, Canada
| | - Lucía Minarrieta
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada.,Ottawa Institute of Systems Biology, Ottawa, Canada
| | - David A Patten
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada.,Ottawa Institute of Systems Biology, Ottawa, Canada
| | - Geneviève Morin
- Department of Biochemistry, Faculty of Medicine, McGill University, Montreal, Canada.,Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Celia Mt Greenwood
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada.,Lady Davis Institute, Jewish General Hospital, Montreal, Canada.,Department of Human Genetics, McGill University, Montreal, Canada.,Gerald Bronfman Department of Oncology, Montreal, Canada
| | - Vincent Giguère
- Department of Biochemistry, Faculty of Medicine, McGill University, Montreal, Canada.,Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Sidong Huang
- Department of Biochemistry, Faculty of Medicine, McGill University, Montreal, Canada.,Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University, Montreal, Canada.,Department of Medicine, Faculty of Medicine, McGill University, Montreal, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Faculty of Medicine, McGill University, Montreal, Canada.,Goodman Cancer Research Centre, McGill University, Montreal, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada.,Ottawa Institute of Systems Biology, Ottawa, Canada
| |
Collapse
|
97
|
Pancreatic Islets Exhibit Dysregulated Adaptation of Insulin Secretion after Chronic Epinephrine Exposure. Curr Issues Mol Biol 2021; 43:240-250. [PMID: 34071501 PMCID: PMC8929152 DOI: 10.3390/cimb43010020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 01/11/2023] Open
Abstract
Chronic adrenergic stimulation is the dominant factor in impairment of the β-cell function. Sustained adrenergic exposure generates dysregulated insulin secretion in fetal sheep. Similar results have been shown in Min6 under the elevated epinephrine condition, but impairments after adrenergic removal are still unknown and a high rate of proliferation in Min6 has been ignored. Therefore, we incubated primary rats' islets with half maximal inhibitory concentrations of epinephrine for three days, then determined their insulin secretion responsiveness and related signals two days after removal of adrenaline via radioimmunoassay and qPCR. Insulin secretion was not different between the exposure group (1.07 ± 0.04 ng/islet/h) and control (1.23 ± 0.17 ng/islet/h), but total islet insulin content after treatment (5.46 ± 0.87 ng/islet/h) was higher than control (3.17 ± 0.22 ng/islet/h, p < 0.05), and the fractional insulin release was 36% (p < 0.05) lower after the treatment. Meanwhile, the mRNA expression of Gαs, Gαz and Gβ1-2 decreased by 42.8% 19.4% and 24.8%, respectively (p < 0.05). Uncoupling protein 2 (Ucp2), sulphonylurea receptor 1 (Sur1) and superoxide dismutase 2 (Sod2) were significantly reduced (38.5%, 23.8% and 53.8%, p < 0.05). Chronic adrenergic exposure could impair insulin responsiveness in primary pancreatic islets. Decreased G proteins and Sur1 expression affect the regulation of insulin secretion. In conclusion, the sustained under-expression of Ucp2 and Sod2 may further change the function of β-cell, which helps to understand the long-term adrenergic adaptation of pancreatic β-cell.
Collapse
|
98
|
Álvarez-Artime A, García-Soler B, Sainz RM, Mayo JC. Emerging Roles for Browning of White Adipose Tissue in Prostate Cancer Malignant Behaviour. Int J Mol Sci 2021; 22:5560. [PMID: 34074045 PMCID: PMC8197327 DOI: 10.3390/ijms22115560] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
In addition to its well-known role as an energy repository, adipose tissue is one of the largest endocrine organs in the organism due to its ability to synthesize and release different bioactive molecules. Two main types of adipose tissue have been described, namely white adipose tissue (WAT) with a classical energy storage function, and brown adipose tissue (BAT) with thermogenic activity. The prostate, an exocrine gland present in the reproductive system of most mammals, is surrounded by periprostatic adipose tissue (PPAT) that contributes to maintaining glandular homeostasis in conjunction with other cell types of the microenvironment. In pathological conditions such as the development and progression of prostate cancer, adipose tissue plays a key role through paracrine and endocrine signaling. In this context, the role of WAT has been thoroughly studied. However, the influence of BAT on prostate tumor development and progression is unclear and has received much less attention. This review tries to bring an update on the role of different factors released by WAT which may participate in the initiation, progression and metastasis, as well as to compile the available information on BAT to discuss and open a new field of knowledge about the possible protective role of BAT in prostate cancer.
Collapse
Affiliation(s)
- Alejandro Álvarez-Artime
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario s/n, 33011 Oviedo, Spain
| | - Belén García-Soler
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
| | - Rosa María Sainz
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario s/n, 33011 Oviedo, Spain
| | - Juan Carlos Mayo
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario s/n, 33011 Oviedo, Spain
| |
Collapse
|
99
|
Bo B, Li S, Zhou K, Wei J. The Regulatory Role of Oxygen Metabolism in Exercise-Induced Cardiomyocyte Regeneration. Front Cell Dev Biol 2021; 9:664527. [PMID: 33937268 PMCID: PMC8083961 DOI: 10.3389/fcell.2021.664527] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022] Open
Abstract
During heart failure, the heart is unable to regenerate lost or damaged cardiomyocytes and is therefore unable to generate adequate cardiac output. Previous research has demonstrated that cardiac regeneration can be promoted by a hypoxia-related oxygen metabolic mechanism. Numerous studies have indicated that exercise plays a regulatory role in the activation of regeneration capacity in both healthy and injured adult cardiomyocytes. However, the role of oxygen metabolism in regulating exercise-induced cardiomyocyte regeneration is unclear. This review focuses on the alteration of the oxygen environment and metabolism in the myocardium induced by exercise, including the effects of mild hypoxia, changes in energy metabolism, enhanced elimination of reactive oxygen species, augmentation of antioxidative capacity, and regulation of the oxygen-related metabolic and molecular pathway in the heart. Deciphering the regulatory role of oxygen metabolism and related factors during and after exercise in cardiomyocyte regeneration will provide biological insight into endogenous cardiac repair mechanisms. Furthermore, this work provides strong evidence for exercise as a cost-effective intervention to improve cardiomyocyte regeneration and restore cardiac function in this patient population.
Collapse
Affiliation(s)
- Bing Bo
- Kinesiology Department, School of Physical Education, Henan University, Kaifeng, China.,Sports Reform and Development Research Center, School of Physical Education, Henan University, Kaifeng, China
| | - Shuangshuang Li
- Kinesiology Department, School of Physical Education, Henan University, Kaifeng, China
| | - Ke Zhou
- Kinesiology Department, School of Physical Education, Henan University, Kaifeng, China.,Sports Reform and Development Research Center, School of Physical Education, Henan University, Kaifeng, China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| |
Collapse
|
100
|
Fu M, Sun Y, Kenry, Zhang M, Zhou H, Shen W, Hu Y, Zhu Q. A dual-rotator fluorescent probe for analyzing the viscosity of mitochondria and blood. Chem Commun (Camb) 2021; 57:3508-3511. [PMID: 33690772 DOI: 10.1039/d1cc00519g] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A novel mitochondria-targeting molecular rotator FD was designed to visualize changes in viscosity under hypoxic conditions in vitro and in vivo. Importantly, FD can be used to detect changes in the blood viscosity of liver cancer and liver cirrhosis patients, and also rehabilitation of liver disease.
Collapse
Affiliation(s)
- Manlin Fu
- College of Biotechnology and Bioengineering & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China.
| | | | | | | | | | | | | | | |
Collapse
|