1
|
Zhang Z, Zhao L, Wang J, Chen H, Lin Y, Wang F, Wang L, Chen J, Liu J, Zhang X. Luteolin, as a bidirectional ROS regulator, elevates mouse beige adipocyte browning. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159620. [PMID: 40311840 DOI: 10.1016/j.bbalip.2025.159620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/11/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
In beige adipocytes, UCP1-dependent thermogenesis can be driven by intracellular reactive oxygen species (ROS) generation. While ROS elevation also induces mast cell activation, serotonin synthesis and release from mast cells inhibits beige progenitor cell proliferation and browning. As a natural antioxidant and mast cell stabilizer, luteolin promotes adipocyte thermogenesis and inhibits mast cell activation. Thus, to activate adipocyte thermogenesis, how luteolin regulates ROS level in beige adipocytes and mast cells needs to be further investigated. In this study, mouse subcutaneous stromal vascular fraction (SVF) cells are induced to differentiate into beige adipocytes, and mouse bone marrow-derived mast cells (BMMCs) are activated with hydrogen peroxide (H2O2). Intracellular ROS level is augmented in differentiated beige adipocytes and H2O2-activated BMMCs, and H2O2-activated BMMCs inhibited brown differentiation of SVF cells and thermogenesis of beige adipocytes. In beige adipocytes, unlike synthetic antioxidant N-acetylcysteine (NAC), luteolin elevates the expression of thermogenic and beige-selective marker genes and intracellular ROS generation. Contrarily, luteolin inhibits H2O2-induced mast cell activation and ROS generation. Further, luteolin partially reverses the inhibitory effects of H2O2-activated BMMCs on the brown differentiation of SVF cells and the thermogenesis of beige adipocytes. Molecular mechanistic studies demonstrate that luteolin regulates intracellular ROS level in beige adipocytes and mast cells via the nuclear factor erythroid 2-related factor 2 (Nrf2)/Catalase pathway. Altogether, as a ROS regulator, luteolin contrarily affects intracellular ROS generation in beige adipocytes and mast cells, and hence elevates adipocyte browning.
Collapse
Affiliation(s)
- Zhixin Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Linli Zhao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Jiahui Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Hao Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Yan Lin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Fangbin Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Lu Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Juan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China.
| | - Jian Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China; Engineering Research Center of Bioprocess, Ministry of Education, Hefei University of Technology, Hefei, Anhui 230009, China.
| | - Xian Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China.
| |
Collapse
|
2
|
Szabó A, Váncsa S, Hegyi P, Kói T, Ács J, Hermánné RJ, Ács N, Szarvas T, Nyirády P, Kopa Z. Assessing the efficacy of varicocelectomy, antioxidants, FSH treatment, and lifestyle modifications on sperm DNA fragmentation: a systematic review and meta-analysis. Sci Rep 2025; 15:10118. [PMID: 40128318 PMCID: PMC11933313 DOI: 10.1038/s41598-025-93267-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
Infertility affects 15% of couples in developed countries, 50% accounting for male origin. In 2021, sperm DNA fragmentation (SDF) testing was incorporated into male fertility evaluations as an evidence-based functional test. We aimed to assess the effect of interventions that could potentially improve SDF. A random-effect meta-analysis was performed with a systematic search in three databases. Pooled mean differences (MD) with 95% confidence intervals (CI) were calculated. Initially, 36,531 articles were found, of which 86 papers with over 8,000 patients were included. Three months following varicocelectomy, the decrease in SDF was -6.74% (CI: -9.40, -4.08) compared to preoperative data of patients, whereas it was -12.39% (CI: -22.41, -2.36) after six months and -10.06% (CI: -22.69, + 2.56) at twelve months. A dose-dependent effect could also be observed in grade II and III varicoceles. The overall SDF decrease at three months was -4.27% (CI: -6.11, -2.43) for antioxidants, -4.51% (CI: -6.81, -2.20) for combined antioxidant therapy and -3.36% (CI: -4.44, -2.28) for monotherapy. In terms of follicle-stimulating hormone treatment (FSH) at three months, the change in SDF was -6.66% (CI: -9.64, -3.69). For lifestyle modifications, a change of -3.24% (CI: -5.33, -1.16) was observed at three months. Other interventions are narratively summarized in the systematic review. Of the interventions reviewed, varicocelectomy is the most effective in reducing SDF at six months, FSH treatment might be useful, antioxidants appear questionable, and lifestyle interventions require more studies of similar designs to draw firm conclusions.
Collapse
Affiliation(s)
- Anett Szabó
- Department of Urology, Semmelweis University, Üllői út 78/b, H-1082, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Szilárd Váncsa
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - Tamás Kói
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Stochastics, Institute of Mathematics, Budapest University of Technology and Economics, Budapest, Hungary
| | - Júlia Ács
- Department of Urology, Semmelweis University, Üllői út 78/b, H-1082, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Réka Juhász Hermánné
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Dietetics and Nutrition Sciences, Semmelweis University, Budapest, Hungary
| | - Nándor Ács
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Tibor Szarvas
- Department of Urology, Semmelweis University, Üllői út 78/b, H-1082, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Urology, University of Duisburg-Essen and German Cancer Consortium, Essen, Germany
| | - Péter Nyirády
- Department of Urology, Semmelweis University, Üllői út 78/b, H-1082, Budapest, Hungary
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zsolt Kopa
- Department of Urology, Semmelweis University, Üllői út 78/b, H-1082, Budapest, Hungary.
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
3
|
Lee J, Roh JL. Ferroptosis: iron release mechanisms in the bioenergetic process. Cancer Metastasis Rev 2025; 44:36. [PMID: 40000477 DOI: 10.1007/s10555-025-10252-8] [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: 11/15/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
Ferroptosis, an iron-dependent form of cell death, has been the focus of extensive research over the past decade, leading to the elucidation of key molecules and mechanisms involved in this process. While several studies have highlighted iron sources for the Fenton reaction, the predominant mechanism for iron release in ferroptosis has been identified as ferritinophagy, which occurs in response to iron starvation. However, much of the existing literature has concentrated on lipid peroxidation rather than on the mechanisms of iron release. This review proposes three distinct mechanisms of iron mobilization: ferritinophagy, reductive pathways with selective gating of ferritin pores, and quinone-mediated iron mobilization. Notably, the latter two mechanisms operate independently of iron starvation and rely primarily on reductants such as NADH and O2•-. The inhibition of the respiratory chain, particularly under the activation of α-ketoglutarate dehydrogenase, leads to the accumulation of these reductants, which in turn promotes iron release from ferritin and indirectly inhibits AMP-activated protein kinase through excessive iron levels. In this work, we delineate the intricate relationship between iron mobilization and bioenergetic processes under conditions of oxidative stress. Furthermore, this review aims to enhance the understanding of the connections between ferroptosis and these mechanisms.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do, 13496, Republic of Korea
- Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do, 13496, Republic of Korea.
- Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea.
| |
Collapse
|
4
|
Mladenov M, Sazdova I, Hadzi-Petrushev N, Konakchieva R, Gagov H. The Role of Reductive Stress in the Pathogenesis of Endocrine-Related Metabolic Diseases and Cancer. Int J Mol Sci 2025; 26:1910. [PMID: 40076537 PMCID: PMC11899626 DOI: 10.3390/ijms26051910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Reductive stress (RS), characterized by excessive accumulation of reducing equivalents such as NADH and NADPH, is emerging as a key factor in metabolic disorders and cancer. While oxidative stress (OS) has been widely studied, RS and its complex interplay with endocrine regulation remain less understood. This review explores molecular circuits of bidirectional crosstalk between metabolic hormones and RS, focusing on their role in diabetes, obesity, cardiovascular diseases, and cancer. RS disrupts insulin secretion and signaling, exacerbates metabolic inflammation, and contributes to adipose tissue dysfunction, ultimately promoting insulin resistance. In cardiovascular diseases, RS alters vascular smooth muscle cell function and myocardial metabolism, influencing ischemia-reperfusion injury outcomes. In cancer, RS plays a dual role: it enhances tumor survival by buffering OS and promoting metabolic reprogramming, yet excessive RS can trigger proteotoxicity and mitochondrial dysfunction, leading to apoptosis. Recent studies have identified RS-targeting strategies, including redox-modulating therapies, nanomedicine, and drug repurposing, offering potential for novel treatments. However, challenges remain, particularly in distinguishing physiological RS from pathological conditions and in overcoming therapy-induced resistance. Future research should focus on developing selective RS biomarkers, optimizing therapeutic interventions, and exploring the role of RS in immune and endocrine regulation.
Collapse
Affiliation(s)
- Mitko Mladenov
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (M.M.); (N.H.-P.)
- Department of Fundamental and Applied Physiology, Russian States Medical University, 117997 Moscow, Russia
| | - Iliyana Sazdova
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 1164 Sofia, Bulgaria;
| | - Nikola Hadzi-Petrushev
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (M.M.); (N.H.-P.)
| | - Rossitza Konakchieva
- Department of Cell and Developmental Biology, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 1164 Sofia, Bulgaria;
| | - Hristo Gagov
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 1164 Sofia, Bulgaria;
| |
Collapse
|
5
|
Zhang S, Wang N, Gao Z, Gao J, Wang X, Xie H, Wang CY, Zhang S. Reductive stress: The key pathway in metabolic disorders induced by overnutrition. J Adv Res 2025:S2090-1232(25)00031-1. [PMID: 39805424 DOI: 10.1016/j.jare.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/04/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The balance of redox states is crucial for maintaining physiological homeostasis. For decades, the focus has been mainly on the concept of oxidative stress, which is involved in the mechanism of almost all diseases. However, robust evidence has highlighted that reductive stress, the other side of the redox spectrum, plays a pivotal role in the development of various diseases, particularly those related to metabolism and cardiovascular health. AIM OF REVIEW In this review, we present an extensive array of evidence for the occurrence of reductive stress and its significant implications mainly in metabolic and cardiovascular diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW Reductive stress is defined as a shift in the cellular redox balance towards a more reduced state, characterized by an excess of endogenous reductants (such as NADH, NADPH, and GSH) over their oxidized counterparts (NAD+, NADP+, and GSSG). While oxidative stress has been the predominant mechanism studied in obesity, metabolic disorders, and cardiovascular diseases, growing evidence underscores the critical role of reductive stress. This review discusses how reductive stress contributes to metabolic and cardiovascular pathologies, emphasizing its effects on key cellular processes. For example, excessive NADH accumulation can disrupt mitochondrial function by impairing the electron transport chain, leading to decreased ATP production and increased production of reactive oxygen species. In the endoplasmic reticulum (ER), an excess of reductive equivalents hampers protein folding, triggering ER stress and activating the unfolded protein response, which can lead to insulin resistance and compromised cellular homeostasis. Furthermore, we explore how excessive antioxidant supplementation can exacerbate reductive stress by further shifting the redox balance, potentially undermining the beneficial effects of exercise, impairing cardiovascular health, and aggravating metabolic disorders, particularly in obese individuals. This growing body of evidence calls for a reevaluation of the role of reductive stress in disease pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Shiyi Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichao Gao
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Gao
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohui Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Xie
- Institute of Translational Medicine, Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Cong-Yi Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shu Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Wang W, Arreola M, Mathews T, DeVilbiss A, Zhao Z, Martin-Sandoval M, Mohammed A, Benegiamo G, Awani A, Goeminne L, Dever D, Nakauchi Y, Porteus MH, Pavel-Dinu M, Al-Herz W, Auwerx J, Morrison SJ, Weinacht KG. Failure of metabolic checkpoint control during late-stage granulopoiesis drives neutropenia in reticular dysgenesis. Blood 2024; 144:2718-2734. [PMID: 39378586 PMCID: PMC11830988 DOI: 10.1182/blood.2024024123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/30/2024] [Accepted: 08/24/2024] [Indexed: 10/10/2024] Open
Abstract
ABSTRACT Cellular metabolism is highly dynamic during hematopoiesis, yet the regulatory networks that maintain metabolic homeostasis during differentiation are incompletely understood. Herein, we have studied the grave immunodeficiency syndrome reticular dysgenesis caused by loss of mitochondrial adenylate kinase 2 (AK2) function. By coupling single-cell transcriptomics in samples from patients with reticular dysgenesis with a CRISPR model of this disorder in primary human hematopoietic stem cells, we found that the consequences of AK2 deficiency for the hematopoietic system are contingent on the effective engagement of metabolic checkpoints. In hematopoietic stem and progenitor cells, including early granulocyte precursors, AK2 deficiency reduced mechanistic target of rapamycin (mTOR) signaling and anabolic pathway activation. This conserved nutrient homeostasis and maintained cell survival and proliferation. In contrast, during late-stage granulopoiesis, metabolic checkpoints were ineffective, leading to a paradoxical upregulation of mTOR activity and energy-consuming anabolic pathways such as ribonucleoprotein synthesis in AK2-deficient cells. This caused nucleotide imbalance, including highly elevated adenosine monophosphate and inosine monophosphate levels, the depletion of essential substrates such as NAD+ and aspartate, and ultimately resulted in proliferation arrest and demise of the granulocyte lineage. Our findings suggest that even severe metabolic defects can be tolerated with the help of metabolic checkpoints but that the failure of such checkpoints in differentiated cells results in a catastrophic loss of homeostasis.
Collapse
Affiliation(s)
- Wenqing Wang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Martin Arreola
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Thomas Mathews
- Children’s Medical Center Research Institute, The University of Texas Southwestern, Dallas, TX
| | - Andrew DeVilbiss
- Children’s Medical Center Research Institute, The University of Texas Southwestern, Dallas, TX
| | - Zhiyu Zhao
- Children’s Medical Center Research Institute, The University of Texas Southwestern, Dallas, TX
| | - Misty Martin-Sandoval
- Children’s Medical Center Research Institute, The University of Texas Southwestern, Dallas, TX
| | - Abdulvasey Mohammed
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Giorgia Benegiamo
- Laboratory for Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Avni Awani
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Ludger Goeminne
- Laboratory for Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Daniel Dever
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Yusuke Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Matthew H. Porteus
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Mara Pavel-Dinu
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Waleed Al-Herz
- Department of Pediatrics, College of Medicine, Kuwait University, Safat, Kuwait
| | - Johan Auwerx
- Laboratory for Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Sean J. Morrison
- Children’s Medical Center Research Institute, The University of Texas Southwestern, Dallas, TX
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Katja G. Weinacht
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
7
|
Kunwar A, Aishwarya J. "Reductive stress" the overlooked side of cellular redox modulation in cancer: opportunity for design of next generation redox chemotherapeutics. Free Radic Res 2024; 58:782-795. [PMID: 39604822 DOI: 10.1080/10715762.2024.2433988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
The last three decades of redox biology research have been dominated by the term "oxidative stress" since it was first coined by Helmut Sies to represent a form of cellular redox modulation characterized by redox imbalance toward overproduction of oxidants. Almost every pathological condition, including cancer, has been linked with oxidative stress and so forth; targeting oxidative stress became the strategy for the new drug discovery with anticancer drugs aiming to selectively induce oxidative stress in cancerous cells while antioxidants aiming to prevent carcinogenesis as prophylactic agents. Time has now come to realize, how harmful the other side of the cellular redox spectrum, "reductive stress," characterized by redox imbalance toward the accumulation of reducing equivalents, maybe during carcinogenesis, and to tap its potential for the design of next-generation chemotherapeutic agents. Adjuvants-causing reductive stress may also work synergistically with radiation therapy under hypoxia to achieve better tumor control. Keeping this evolving field into account, the present review provides a current understating of the role of reductive stress in carcinogenesis, the status of reductive stress-based chemotherapeutic agents with particular emphasis on sulfhydryl and selenium-containing compounds and the gap areas that need to be addressed in future.
Collapse
Affiliation(s)
- Amit Kunwar
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - J Aishwarya
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
- Advanced Centre for Treatment, Research and Education in Cancer, Mumbai, India
| |
Collapse
|
8
|
Tejeda‐Chavez HR, Montero S, Saavedra‐Molina A, Lemus M, Tejeda‐Luna JB, Roces de Alvarez‐Buylla E. Reductive stress in mitochondria isolated from the carotid body of type 1 diabetic male Wistar rats. Physiol Rep 2024; 12:e70016. [PMID: 39294856 PMCID: PMC11410552 DOI: 10.14814/phy2.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 09/21/2024] Open
Abstract
The carotid body (CB) senses changes in arterial O2 partial pressure (pO2) and glucose levels; therefore, it is key for the detection of hypoxia and hypoglycemia. The CB has been suggested to detect pO2 through an increase in reactive oxygen species (ROS) in the mitochondria. However, the mechanism protecting the chemoreceptor cells and their mitochondria from ROS and hyperglycemia is poorly understood. Here we measured glutathione levels in CB mitochondria of control and in streptozotocin (STZ)-induced type 1 diabetic male Wistar rats. We found a dramatic reduction in total glutathione from 11.45 ± 1.30 μmol/mg protein in control rats to 1.45 ± 0.31 μmol/mg protein in diabetic rats. However, the ratio of reduced to oxidized glutathione, a measure of the redox index, was increased in diabetic rats compared to controls. We conclude that the mitochondria of CB chemoreceptor cells in type 1 diabetic male Wistar rats were likely under glutathione-reducing stress.
Collapse
Affiliation(s)
| | - Sergio Montero
- Faculty of MedicineColima of UniversityColimaMexico
- Department of Neuroendocrinology, University Center of Biomedical ResearchColima UniversityColimaMexico
| | | | - Monica Lemus
- Department of Neuroendocrinology, University Center of Biomedical ResearchColima UniversityColimaMexico
| | | | | |
Collapse
|
9
|
Liu W, Zou H, You D, Zhang H, Xu L. Sodium Houttuybonate Promotes the Browning of White Adipose Tissue by Inhibiting Ferroptosis via the AMPK-NRF2-HO1 Pathway. Antioxidants (Basel) 2024; 13:1057. [PMID: 39334717 PMCID: PMC11428211 DOI: 10.3390/antiox13091057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
The rising prevalence of obesity has resulted in an increased demand for innovative and effective treatment strategies. Houttuynia cordata Thunb. (H. cordata) has demonstrated promising potential in preventing obesity. However, the mechanism underlying the anti-obesity effects of H. cordata and its bioactive component, sodium houttuybonate (SH), remains unclear. Our study reveals that SH treatment promotes the browning of inguinal white adipose tissue (iWAT) and prevents the obesity induced by a high-fat diet. SH significantly mitigates ferroptosis by upregulating glutathione peroxidase 4 (Gpx4) and decreasing malondialdehyde (MDA) levels, while also enhancing superoxide dismutase (SOD) levels. Furthermore, SH promotes the phosphorylation of AMP-activated protein kinase (AMPK), which subsequently increases the expression of nuclear factor erythroid 2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1) in the iWAT. However, the effects of SH were attenuated by ML385, an Nrf2 inhibitor. Collectively, our findings suggest that SH induces iWAT browning and prevents diet-induced obesity primarily through the AMPK/NRF2/HO-1 pathway by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Endocrinology, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, China
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Huren Zou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Danming You
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Lingling Xu
- Department of Endocrinology, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, China
| |
Collapse
|
10
|
Tamir TY, Chaudhary S, Li AX, Trojan SE, Flower CT, Vo P, Cui Y, Davis JC, Mukkamala RS, Venditti FN, Hillis AL, Toker A, Vander Heiden MG, Spinelli JB, Kennedy NJ, Davis RJ, White FM. Structural and systems characterization of phosphorylation on metabolic enzymes identifies sex-specific metabolic reprogramming in obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.609894. [PMID: 39257804 PMCID: PMC11383994 DOI: 10.1101/2024.08.28.609894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Coordination of adaptive metabolism through cellular signaling networks and metabolic response is essential for balanced flow of energy and homeostasis. Post-translational modifications such as phosphorylation offer a rapid, efficient, and dynamic mechanism to regulate metabolic networks. Although numerous phosphorylation sites have been identified on metabolic enzymes, much remains unknown about their contribution to enzyme function and systemic metabolism. In this study, we stratify phosphorylation sites on metabolic enzymes based on their location with respect to functional and dimerization domains. Our analysis reveals that the majority of published phosphosites are on oxidoreductases, with particular enrichment of phosphotyrosine (pY) sites in proximity to binding domains for substrates, cofactors, active sites, or dimer interfaces. We identify phosphosites altered in obesity using a high fat diet (HFD) induced obesity model coupled to multiomics, and interrogate the functional impact of pY on hepatic metabolism. HFD induced dysregulation of redox homeostasis and reductive metabolism at the phosphoproteome and metabolome level in a sex-specific manner, which was reversed by supplementing with the antioxidant butylated hydroxyanisole (BHA). Partial least squares regression (PLSR) analysis identified pY sites that predict HFD or BHA induced changes of redox metabolites. We characterize predictive pY sites on glutathione S-transferase pi 1 (GSTP1), isocitrate dehydrogenase 1 (IDH1), and uridine monophosphate synthase (UMPS) using CRISPRi-rescue and stable isotope tracing. Our analysis revealed that sites on GSTP1 and UMPS inhibit enzyme activity while the pY site on IDH1 induces activity to promote reductive carboxylation. Overall, our approach provides insight into the convergence points where cellular signaling fine-tunes metabolism.
Collapse
Affiliation(s)
- Tigist Y Tamir
- Koch Institute for Integrative Cancer Research
- Center for Precision Cancer Medicine
- Department of Biological Engineering
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shreya Chaudhary
- Koch Institute for Integrative Cancer Research
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Annie X Li
- Koch Institute for Integrative Cancer Research
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sonia E Trojan
- Koch Institute for Integrative Cancer Research
- Department of Biology
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cameron T Flower
- Koch Institute for Integrative Cancer Research
- Center for Precision Cancer Medicine
- Program in Computational and Systems Biology
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Paula Vo
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yufei Cui
- Koch Institute for Integrative Cancer Research
- Department of Biological Engineering
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeffrey C Davis
- Koch Institute for Integrative Cancer Research
- Department of Biology
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rachit S Mukkamala
- Koch Institute for Integrative Cancer Research
- Department of Biological Engineering
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Francesca N Venditti
- Koch Institute for Integrative Cancer Research
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alissandra L Hillis
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alex Toker
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research
- Center for Precision Cancer Medicine
- Department of Biology
- Massachusetts Institute of Technology, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jessica B Spinelli
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Norman J Kennedy
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Forest M White
- Koch Institute for Integrative Cancer Research
- Center for Precision Cancer Medicine
- Department of Biological Engineering
- Program in Computational and Systems Biology
- Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
11
|
Heo HS, Kim YE, Lee JH. Antioxidant activity of Jeju lava seawater through translocation of Nrf2 in human fibroblast. Food Sci Biotechnol 2024; 33:2653-2661. [PMID: 39144193 PMCID: PMC11319678 DOI: 10.1007/s10068-023-01510-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 11/20/2023] [Accepted: 12/17/2023] [Indexed: 08/16/2024] Open
Abstract
Reactive oxygen species (ROS) are associated with various pathological conditions, including atherosclerosis and cancer. Photoaging, mainly caused by UVB-induced ROS, accelerates skin aging and collagen degradation. Nuclear factor erythroid 2-related factor 2 (Nrf2) regulates antioxidant enzymes and has demonstrated protective effects against chronic diseases. Jeju lava seawater (JLS), which is rich in minerals, is attracting attention for its health benefits. The current study investigates the antioxidant properties of JLS in human dermal fibroblasts (HDFs). experiments were conducted by culturing HDFs in JLS with different water hardness levels and irradiating UVB. The results show that JLS does not affect HDF viability, especially at high water hardness. JLS treatment enhances collagen production and upregulates Nrf2 and antioxidant enzymes such as NQO1 and HO-1. This mechanism involves the translocation of Nrf2 to the cell nucleus. JLS shows promise as an antioxidant, potentially mitigating the effects of oxidative stress and promoting collagen synthesis.
Collapse
Affiliation(s)
- Hee Sun Heo
- Department of Food Science and Biotechnology, College of Life Sciences, CHA University, Gyeonggi-do, 11160 Republic of Korea
| | | | - Jong Hun Lee
- Department of Food Science and Biotechnology, College of Bio-Nano Technology, Gachon University, Gyeonggi-do, 13120 Republic of Korea
| |
Collapse
|
12
|
Ward NP, Yoon SJ, Flynn T, Sherwood AM, Olley MA, Madej J, DeNicola GM. Mitochondrial respiratory function is preserved under cysteine starvation via glutathione catabolism in NSCLC. Nat Commun 2024; 15:4244. [PMID: 38762605 PMCID: PMC11102494 DOI: 10.1038/s41467-024-48695-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/03/2024] [Indexed: 05/20/2024] Open
Abstract
Cysteine metabolism occurs across cellular compartments to support diverse biological functions and prevent the induction of ferroptosis. Though the disruption of cytosolic cysteine metabolism is implicated in this form of cell death, it is unknown whether the substantial cysteine metabolism resident within the mitochondria is similarly pertinent to ferroptosis. Here, we show that despite the rapid depletion of intracellular cysteine upon loss of extracellular cystine, cysteine-dependent synthesis of Fe-S clusters persists in the mitochondria of lung cancer cells. This promotes a retention of respiratory function and a maintenance of the mitochondrial redox state. Under these limiting conditions, we find that glutathione catabolism by CHAC1 supports the mitochondrial cysteine pool to sustain the function of the Fe-S proteins critical to oxidative metabolism. We find that disrupting Fe-S cluster synthesis under cysteine restriction protects against the induction of ferroptosis, suggesting that the preservation of mitochondrial function is antagonistic to survival under starved conditions. Overall, our findings implicate mitochondrial cysteine metabolism in the induction of ferroptosis and reveal a mechanism of mitochondrial resilience in response to nutrient stress.
Collapse
Affiliation(s)
- Nathan P Ward
- Department of Metabolism & Physiology, Moffitt Cancer Center, Tampa, FL, USA.
| | - Sang Jun Yoon
- Department of Metabolism & Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Tyce Flynn
- Department of Metabolism & Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Amanda M Sherwood
- Department of Metabolism & Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Maddison A Olley
- Department of Metabolism & Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Juliana Madej
- Department of Metabolism & Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Gina M DeNicola
- Department of Metabolism & Physiology, Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
13
|
Kishi S, Nagasu H, Kidokoro K, Kashihara N. Oxidative stress and the role of redox signalling in chronic kidney disease. Nat Rev Nephrol 2024; 20:101-119. [PMID: 37857763 DOI: 10.1038/s41581-023-00775-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/21/2023]
Abstract
Chronic kidney disease (CKD) is a major public health concern, underscoring a need to identify pathogenic mechanisms and potential therapeutic targets. Reactive oxygen species (ROS) are derivatives of oxygen molecules that are generated during aerobic metabolism and are involved in a variety of cellular functions that are governed by redox conditions. Low levels of ROS are required for diverse processes, including intracellular signal transduction, metabolism, immune and hypoxic responses, and transcriptional regulation. However, excess ROS can be pathological, and contribute to the development and progression of chronic diseases. Despite evidence linking elevated levels of ROS to CKD development and progression, the use of low-molecular-weight antioxidants to remove ROS has not been successful in preventing or slowing disease progression. More recent advances have enabled evaluation of the molecular interactions between specific ROS and their targets in redox signalling pathways. Such studies may pave the way for the development of sophisticated treatments that allow the selective control of specific ROS-mediated signalling pathways.
Collapse
Affiliation(s)
- Seiji Kishi
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Hajime Nagasu
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Kengo Kidokoro
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Naoki Kashihara
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| |
Collapse
|
14
|
Hine C, Patel AK, Ponti AK. Diet-Modifiable Redox Alterations in Ageing and Cancer. Subcell Biochem 2024; 107:129-172. [PMID: 39693023 PMCID: PMC11753504 DOI: 10.1007/978-3-031-66768-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
With ageing comes some of life's best and worst moments. Those lucky enough to live out into the seventh, eighth, and nineth decades and perhaps beyond have more opportunities to experience the wonders and joys of the world. As the world's population shifts towards more and more of these individuals, this is something to be celebrated. However, it is not without negative consequences. Advanced age also ushers in health decline and the burden of non-communicable diseases such as cancer, heart disease, stroke, and organ function decay. Thus, alleviating or at least dampening the severity of ageing as a whole, as well as these individual age-related disorders will enable the improvement in lifespan and healthspan. In the following chapter, we delve into hypothesised causes of ageing and experimental interventions that can be taken to slow their progression. We also highlight cellular and subcellular mechanisms of ageing with a focus on protein thiol oxidation and posttranslational modifications that impact cellular homeostasis and the advent and progression of ageing-related cancers. By having a better understanding of the mechanisms of ageing, we can hopefully develop effective, safe, and efficient therapeutic modalities that can be used prophylactically and/or concurrent to the onset of ageing.
Collapse
Affiliation(s)
- Christopher Hine
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA.
| | - Anand Kumar Patel
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Cardiovascular Genetics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - András K Ponti
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| |
Collapse
|
15
|
Bauzá-Thorbrügge M, Peris E, Zamani S, Micallef P, Paul A, Bartesaghi S, Benrick A, Wernstedt Asterholm I. NRF2 is essential for adaptative browning of white adipocytes. Redox Biol 2023; 68:102951. [PMID: 37931470 PMCID: PMC10652207 DOI: 10.1016/j.redox.2023.102951] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/08/2023] Open
Abstract
White adipose tissue browning, defined by accelerated mitochondrial metabolism and biogenesis, is considered a promising mean to treat or prevent obesity-associated metabolic disturbances. We hypothesize that redox stress acutely leads to increased production of reactive oxygen species (ROS), which activate electrophile sensor nuclear factor erythroid 2-Related Factor 2 (NRF2) that over time results in an adaptive adipose tissue browning process. To test this, we have exploited adipocyte-specific NRF2 knockout mice and cultured adipocytes and analyzed time- and dose-dependent effect of NAC and lactate treatment on antioxidant expression and browning-like processes. We found that short-term antioxidant treatment with N-acetylcysteine (NAC) induced reductive stress as evident from increased intracellular NADH levels, increased ROS-production, reduced oxygen consumption rate (OCR), and increased NRF2 levels in white adipocytes. In contrast, and in line with our hypothesis, longer-term NAC treatment led to a NRF2-dependent browning response. Lactate treatment elicited similar effects as NAC, and mechanistically, these NRF2-dependent adipocyte browning responses in vitro were mediated by increased heme oxygenase-1 (HMOX1) activity. Moreover, this NRF2-HMOX1 axis was also important for β3-adrenergic receptor activation-induced adipose tissue browning in vivo. In conclusion, our findings show that administration of exogenous antioxidants can affect biological function not solely through ROS neutralization, but also through reductive stress. We also demonstrate that NRF2 is essential for white adipose tissue browning processes.
Collapse
Affiliation(s)
- Marco Bauzá-Thorbrügge
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Eduard Peris
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Shabnam Zamani
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Peter Micallef
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Alexandra Paul
- Department of Biology and Biological Engineering, Division of Chemical Biology, Chalmers University of Technology, Gothenburg, Sweden; The Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Stefano Bartesaghi
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Benrick
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; School of Health Sciences, University of Skövde, Skövde, Sweden
| | - Ingrid Wernstedt Asterholm
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
16
|
Jia D, Tian Z, Wang R. Exercise mitigates age-related metabolic diseases by improving mitochondrial dysfunction. Ageing Res Rev 2023; 91:102087. [PMID: 37832607 DOI: 10.1016/j.arr.2023.102087] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023]
Abstract
The benefits of regular physical activity are related to delaying and reversing the onset of ageing and age-related disorders, including cardiomyopathy, neurodegenerative diseases, cancer, obesity, diabetes, and fatty liver diseases. However, the molecular mechanisms of the benefits of exercise or physical activity on ageing and age-related disorders remain poorly understood. Mitochondrial dysfunction is implicated in the pathogenesis of ageing and age-related metabolic diseases. Mitochondrial health is an important mediator of cellular function. Therefore, exercise alleviates metabolic diseases in individuals with advancing ageing and age-related diseases by the remarkable promotion of mitochondrial biogenesis and function. Exerkines are identified as signaling moieties released in response to exercise. Exerkines released by exercise have potential roles in improving mitochondrial dysfunction in response to age-related disorders. This review comprehensive summarizes the benefits of exercise in metabolic diseases, linking mitochondrial dysfunction to the onset of age-related diseases. Using relevant examples utilizing this approach, the possibility of designing therapeutic interventions based on these molecular mechanisms is addressed.
Collapse
Affiliation(s)
- Dandan Jia
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
17
|
Vardar Acar N, Özgül RK. A big picture of the mitochondria-mediated signals: From mitochondria to organism. Biochem Biophys Res Commun 2023; 678:45-61. [PMID: 37619311 DOI: 10.1016/j.bbrc.2023.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/02/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Mitochondria, well-known for years as the powerhouse and biosynthetic center of the cell, are dynamic signaling organelles beyond their energy production and biosynthesis functions. The metabolic functions of mitochondria, playing an important role in various biological events both in physiological and stress conditions, transform them into important cellular stress sensors. Mitochondria constantly communicate with the rest of the cell and even from other cells to the organism, transmitting stress signals including oxidative and reductive stress or adaptive signals such as mitohormesis. Mitochondrial signal transduction has a vital function in regulating integrity of human genome, organelles, cells, and ultimately organism.
Collapse
Affiliation(s)
- Neşe Vardar Acar
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - R Köksal Özgül
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
18
|
Ito H, Shoji Y, Matsumoto KI, Fukuhara K, Nakanishi I. Anti-cancer Effect of a Planar Catechin Analog through the Decrease in Mitochondrial Membrane Potential. ACS Med Chem Lett 2023; 14:1478-1481. [PMID: 37849552 PMCID: PMC10577690 DOI: 10.1021/acsmedchemlett.3c00328] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/28/2023] [Indexed: 10/19/2023] Open
Abstract
Catechin is one of the best-known antioxidants and is reported to have some favorable physiological activities, including anti-cancer effects. We previously synthesized a catechin analog, planar catechin, which showed a 10-fold larger radical scavenging activity than (+)-catechin. However, the physiological effects of the planar catechin have remained unclear. In this study, we examined cytotoxicity and mitochondrial membrane potential after planar catechin treatment using a rat normal gastric mucosal cell line, RGM1, and its chemically induced cancer-like cell line, RGK1. Interestingly, the planar catechin showed remarkable cytotoxicity compared to (+)-catechin, with cancer cell specificity. Furthermore, the decrease in the mitochondrial membrane potential of cancer cells was observed at specific concentrations of the planar catechin. These results indicate that the planar catechin, possessing higher antioxidant activity, induces its anti-cancer effect through a decrease in the mitochondrial membrane potential and thus can be a promising agent for cancer treatment.
Collapse
Affiliation(s)
- Hiromu Ito
- Quantum
RedOx Chemistry Team, Institute for Quantum Life Science (iQLS), Quantum
Life and Medical Science Directorate (QLMS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| | - Yoshimi Shoji
- Quantum
RedOx Chemistry Team, Institute for Quantum Life Science (iQLS), Quantum
Life and Medical Science Directorate (QLMS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
- Quantitative
RedOx Sensing Group, Department of Radiation Regulatory Science Research,
Institute for Radiological Science (NIRS), Quantum Life and Medical
Science Directorate (QLMS), National Institutes
for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| | - Ken-ichiro Matsumoto
- Quantitative
RedOx Sensing Group, Department of Radiation Regulatory Science Research,
Institute for Radiological Science (NIRS), Quantum Life and Medical
Science Directorate (QLMS), National Institutes
for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| | - Kiyoshi Fukuhara
- Division
of Medicinal Chemistry, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Ikuo Nakanishi
- Quantum
RedOx Chemistry Team, Institute for Quantum Life Science (iQLS), Quantum
Life and Medical Science Directorate (QLMS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba-shi, Chiba 263-8555, Japan
| |
Collapse
|
19
|
Panico G, Fasciolo G, Migliaccio V, De Matteis R, Lionetti L, Napolitano G, Agnisola C, Venditti P, Lombardi A. 1,3-Butanediol Administration Increases β-Hydroxybutyrate Plasma Levels and Affects Redox Homeostasis, Endoplasmic Reticulum Stress, and Adipokine Production in Rat Gonadal Adipose Tissue. Antioxidants (Basel) 2023; 12:1471. [PMID: 37508009 PMCID: PMC10376816 DOI: 10.3390/antiox12071471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Ketone bodies (KBs) are an alternative energy source under starvation and play multiple roles as signaling molecules regulating energy and metabolic homeostasis. The mechanism by which KBs influence visceral white adipose tissue physiology is only partially known, and our study aimed to shed light on the effects they exert on such tissue. To this aim, we administered 1,3-butanediol (BD) to rats since it rapidly enhances β-hydroxybutyrate serum levels, and we evaluated the effect it induces within 3 h or after 14 days of treatment. After 14 days of treatment, rats showed a decrease in body weight gain, energy intake, gonadal-WAT (gWAT) weight, and adipocyte size compared to the control. BD exerted a pronounced antioxidant effect and directed redox homeostasis toward reductive stress, already evident within 3 h after its administration. BD lowered tissue ROS levels and oxidative damage to lipids and proteins and enhanced tissue soluble and enzymatic antioxidant capacity as well as nuclear erythroid factor-2 protein levels. BD also reduced specific mitochondrial maximal oxidative capacity and induced endoplasmic reticulum stress as well as interrelated processes, leading to changes in the level of adipokines/cytokines involved in inflammation, macrophage infiltration into gWAT, adipocyte differentiation, and lipolysis.
Collapse
Affiliation(s)
- Giuliana Panico
- Department of Biology, University of Naples Federico II, Complesso Monte Sant'Angelo Via Cintia 26, 80126 Napoli, Italy
| | - Gianluca Fasciolo
- Department of Biology, University of Naples Federico II, Complesso Monte Sant'Angelo Via Cintia 26, 80126 Napoli, Italy
| | - Vincenzo Migliaccio
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Rita De Matteis
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Lillà Lionetti
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Gaetana Napolitano
- Department of Science and Technology, Parthenope University of Naples, 80143 Naples, Italy
| | - Claudio Agnisola
- Department of Biology, University of Naples Federico II, Complesso Monte Sant'Angelo Via Cintia 26, 80126 Napoli, Italy
| | - Paola Venditti
- Department of Biology, University of Naples Federico II, Complesso Monte Sant'Angelo Via Cintia 26, 80126 Napoli, Italy
| | - Assunta Lombardi
- Department of Biology, University of Naples Federico II, Complesso Monte Sant'Angelo Via Cintia 26, 80126 Napoli, Italy
| |
Collapse
|
20
|
Vardar Acar N, Özgül RK. The bridge between cell survival and cell death: reactive oxygen species-mediated cellular stress. EXCLI JOURNAL 2023; 22:520-555. [PMID: 37534225 PMCID: PMC10390897 DOI: 10.17179/excli2023-6221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 08/04/2023]
Abstract
As a requirement of aerobic metabolism, regulation of redox homeostasis is indispensable for the continuity of living homeostasis and life. Since the stability of the redox state is necessary for the maintenance of the biological functions of the cells, the balance between the pro-oxidants, especially ROS and the antioxidant capacity is kept in balance in the cells through antioxidant defense systems. The pleiotropic transcription factor, Nrf2, is the master regulator of the antioxidant defense system. Disruption of redox homeostasis leads to oxidative and reductive stress, bringing about multiple pathophysiological conditions. Oxidative stress characterized by high ROS levels causes oxidative damage to biomolecules and cell death, while reductive stress characterized by low ROS levels disrupt physiological cell functions. The fact that ROS, which were initially attributed as harmful products of aerobic metabolism, at the same time function as signal molecules at non-toxic levels and play a role in the adaptive response called mithormesis points out that ROS have a dose-dependent effect on cell fate determination. See also Figure 1(Fig. 1).
Collapse
Affiliation(s)
- Nese Vardar Acar
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Riza Köksal Özgül
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
21
|
In vitro effects of vitamins C and E on adipocyte function and redox status in obesity. PHARMANUTRITION 2022. [DOI: 10.1016/j.phanu.2022.100315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
22
|
Therapeutic strategies for liver diseases based on redox control systems. Biomed Pharmacother 2022; 156:113764. [DOI: 10.1016/j.biopha.2022.113764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
|
23
|
Bauzá-Thorbrügge M, Banke E, Chanclón B, Peris E, Wu Y, Musovic S, Jönsson C, Strålfors P, Rorsman P, Olofsson CS, Asterholm IW. Adipocyte-specific ablation of the Ca 2+ pump SERCA2 impairs whole-body metabolic function and reveals the diverse metabolic flexibility of white and brown adipose tissue. Mol Metab 2022; 63:101535. [PMID: 35760318 PMCID: PMC9287368 DOI: 10.1016/j.molmet.2022.101535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) transports Ca2+ from the cytosol into the ER and is essential for appropriate regulation of intracellular Ca2+ homeostasis. The objective of this study was to test the hypothesis that SERCA pumps are involved in the regulation of white adipocyte hormone secretion and other aspects of adipose tissue function and that this control is disturbed in obesity-induced type-2 diabetes. METHODS SERCA expression was measured in isolated human and mouse adipocytes as well as in whole mouse adipose tissue by Western blot and RT-qPCR. To test the significance of SERCA2 in adipocyte functionality and whole-body metabolism, we generated adipocyte-specific SERCA2 knockout mice. The mice were metabolically phenotyped by glucose tolerance and tracer studies, histological analyses, measurements of glucose-stimulated insulin release in isolated islets, and gene/protein expression analyses. We also tested the effect of pharmacological SERCA inhibition and genetic SERCA2 ablation in cultured adipocytes. Intracellular and mitochondrial Ca2+ levels were recorded with dual-wavelength ratio imaging and mitochondrial function was assessed by Seahorse technology. RESULTS We demonstrate that SERCA2 is downregulated in white adipocytes from patients with obesity and type-2 diabetes as well as in adipocytes from diet-induced obese mice. SERCA2-ablated adipocytes display disturbed Ca2+ homeostasis associated with upregulated ER stress markers and impaired hormone release. These adipocyte alterations are linked to mild lipodystrophy, reduced adiponectin levels, and impaired glucose tolerance. Interestingly, adipocyte-specific SERCA2 ablation leads to increased glucose uptake in white adipose tissue while glucose uptake is reduced in brown adipose tissue. This dichotomous effect on glucose uptake is due to differently regulated mitochondrial function. In white adipocytes, SERCA2 deficiency triggers an adaptive increase in FGF21, increased mitochondrial UCP1 levels, and increased oxygen consumption rate (OCR). In contrast, brown SERCA2 null adipocytes display reduced OCR despite increased mitochondrial content and UCP1 levels compared to wild type controls. CONCLUSIONS Our data suggest causal links between reduced white adipocyte SERCA2 levels, deranged adipocyte Ca2+ homeostasis, adipose tissue dysfunction and type-2 diabetes.
Collapse
Affiliation(s)
- Marco Bauzá-Thorbrügge
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Elin Banke
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Belén Chanclón
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Eduard Peris
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Yanling Wu
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Saliha Musovic
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Cecilia Jönsson
- Department of Biomedical and Clinical Sciences, Linköping University, SE-58185 Linköping, Sweden.
| | - Peter Strålfors
- Department of Biomedical and Clinical Sciences, Linköping University, SE-58185 Linköping, Sweden.
| | - Patrik Rorsman
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX4 7LE, UK.
| | - Charlotta S Olofsson
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Ingrid Wernstedt Asterholm
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| |
Collapse
|
24
|
Silva SB, Honorato-Sampaio K, Costa SP, Domingues TE, da Cruz TMM, Rodrigues CM, Costa KB, Dos Santos JM, da Silva Lage VK, Gaiad TP, Santos AP, Dias-Peixoto MF, Coimbra CC, Dos Reis AM, Szawka RE, Figueiredo PHS, Costa HS, Oliveira MX, Mendonça VA, Lacerda ACR. The superior beneficial effects of exercise training versus hormone replacement therapy on skeletal muscle of ovariectomized rats. Sci Rep 2022; 12:8764. [PMID: 35610295 PMCID: PMC9130272 DOI: 10.1038/s41598-022-12739-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/06/2022] [Indexed: 12/31/2022] Open
Abstract
Previous studies have highlighted the positive effects of Estradiol (E2) replacement therapy and physical exercise on skeletal muscle during menopause. However, the comparison effects of exercise training (ET) and estradiol replacement therapy during menopause on skeletal muscle have not been investigated to date. This study aimed to compare the effects of endurance exercise training versus E2 replacement therapy on mitochondrial density, redox status, and inflammatory biomarkers in the skeletal muscle of ovariectomized rats. Thirty female Wistar rats (12-week-old) were randomly assigned into three groups: Untrained ovariectomized rats (UN-OVX, n = 10); untrained ovariectomized rats treated with estradiol replacement therapy (E2-OVX); and, trained ovariectomized rats (TR-OVX). After ovariectomy, the E2-OVX rats were treated subcutaneously with E2 (implanted Silastic® capsule containing 360 μg of 17β-estradiol/mL) while the TR-OVX group performed an exercise training protocol (50–70% of maximal running speed on a treadmill, 60 min/day, 5 days/week for 8 weeks). After euthanasia, the soleus muscle was processed for histological and biochemical evaluations. Only exercise prevented the reduction of maximal oxygen consumption and increased mechanical efficiency (ME). While mitochondrial muscle density, total antioxidant capacity (FRAP), catalase (CAT) activity, and interleukin 10 levels were higher in TR-OVX, only OVX-E2 presented higher CAT activity and lower interleukin 6 levels. Endurance exercise training compared with E2 replacement therapy maintains the aerobic capacity improving the ME of OVX rats. In addition, only endurance exercise training raises the skeletal muscle mitochondrial content and tends to balance the redox and inflammatory status in the skeletal muscle of OVX rats.
Collapse
Affiliation(s)
- Sara Barros Silva
- Programa de Pós-Graduação em Reabilitação e Desempenho Funcional (PPGReab), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Campus JK-Highway MGT-367-Km 583, N°. 5000-Alto da Jacuba, Diamantina, 39100-000, Brazil.,Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil
| | - Kinulpe Honorato-Sampaio
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Sociedade Brasileira de Fisiologia (SBFis), Diamantina, Brazil.,Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Minas Gerais, Brazil
| | - Sabrina Paula Costa
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil
| | - Talita Emanuela Domingues
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Sociedade Brasileira de Fisiologia (SBFis), Diamantina, Brazil
| | - Timilly Mayra Martins da Cruz
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil
| | - Cíntia Maria Rodrigues
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Sociedade Brasileira de Fisiologia (SBFis), Diamantina, Brazil
| | - Karine Beatriz Costa
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Sociedade Brasileira de Fisiologia (SBFis), Diamantina, Brazil
| | - Jousielle Márcia Dos Santos
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Sociedade Brasileira de Fisiologia (SBFis), Diamantina, Brazil
| | - Vanessa Kelly da Silva Lage
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Sociedade Brasileira de Fisiologia (SBFis), Diamantina, Brazil
| | - Thais Peixoto Gaiad
- Programa de Pós-Graduação em Reabilitação e Desempenho Funcional (PPGReab), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Campus JK-Highway MGT-367-Km 583, N°. 5000-Alto da Jacuba, Diamantina, 39100-000, Brazil.,Faculdade de Ciências Biológicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Minas Gerais, Brazil
| | - Ana Paula Santos
- Programa de Pós-Graduação em Reabilitação e Desempenho Funcional (PPGReab), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Campus JK-Highway MGT-367-Km 583, N°. 5000-Alto da Jacuba, Diamantina, 39100-000, Brazil.,Faculdade de Ciências Biológicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Minas Gerais, Brazil
| | - Marco Fabrício Dias-Peixoto
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Sociedade Brasileira de Fisiologia (SBFis), Diamantina, Brazil.,Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Faculdade de Ciências Biológicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Minas Gerais, Brazil
| | - Cândido Celso Coimbra
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Adelina Martha Dos Reis
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Raphael Escorsim Szawka
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Pedro Henrique Scheidt Figueiredo
- Programa de Pós-Graduação em Reabilitação e Desempenho Funcional (PPGReab), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Campus JK-Highway MGT-367-Km 583, N°. 5000-Alto da Jacuba, Diamantina, 39100-000, Brazil.,Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Faculdade de Ciências Biológicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Minas Gerais, Brazil
| | - Henrique Silveira Costa
- Programa de Pós-Graduação em Reabilitação e Desempenho Funcional (PPGReab), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Campus JK-Highway MGT-367-Km 583, N°. 5000-Alto da Jacuba, Diamantina, 39100-000, Brazil.,Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Faculdade de Ciências Biológicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Minas Gerais, Brazil
| | - Murilo Xavier Oliveira
- Programa de Pós-Graduação em Reabilitação e Desempenho Funcional (PPGReab), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Campus JK-Highway MGT-367-Km 583, N°. 5000-Alto da Jacuba, Diamantina, 39100-000, Brazil.,Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Faculdade de Ciências Biológicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Minas Gerais, Brazil
| | - Vanessa Amaral Mendonça
- Programa de Pós-Graduação em Reabilitação e Desempenho Funcional (PPGReab), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Campus JK-Highway MGT-367-Km 583, N°. 5000-Alto da Jacuba, Diamantina, 39100-000, Brazil.,Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Sociedade Brasileira de Fisiologia (SBFis), Diamantina, Brazil.,Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil.,Faculdade de Ciências Biológicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Minas Gerais, Brazil
| | - Ana Cristina Rodrigues Lacerda
- Programa de Pós-Graduação em Reabilitação e Desempenho Funcional (PPGReab), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Campus JK-Highway MGT-367-Km 583, N°. 5000-Alto da Jacuba, Diamantina, 39100-000, Brazil. .,Centro Integrado de Pós-Graduação e Pesquisa em Saúde (CIPq-Saúde), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil. .,Programa Multicêntrico de Pós-graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Sociedade Brasileira de Fisiologia (SBFis), Diamantina, Brazil. .,Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Brazil. .,Faculdade de Ciências Biológicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, Minas Gerais, Brazil.
| |
Collapse
|
25
|
Cen C, Zhang K, Fu J, Wu X, Wu J, Zheng Y, Zhang Y. Odor-producing response pattern by four typical freshwater algae under stress: Acute microplastic exposure as an example. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 821:153350. [PMID: 35077797 DOI: 10.1016/j.scitotenv.2022.153350] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
Algae-induced odor problems in water have been repeatedly occurred concerns for drinking water quality. However, present researches mostly focus on the odor-producing pattern of algae in normal growth, and there is scarce discussion on those under stress. Microplastics (MPs) pollution have been global concern for their negative ecological impacts and frequently co-occurs with odor-producing algal bloom in freshwaters. Thus, this study aimed to elucidate the effects and mechanisms of MPs as an environmental stress on algal odorant production for good illustration of odor-producing response pattern under stress. Variation in MP size (polystyrene microspheres; 100 nm, 1000 nm and 10 μm) had significant effects on odorant formation (β-cycloidal, 2-methylisopropanol, 2,4-heptandienal and 2,4-decadienal) by four freshwater algae (Microcystis aeruginosa, Pseudanabaena sp., Cyclotella meneghiniana and Melosira varians). The size ratio of MPs over cells (SRMC) was proposed to categorize the size-ratio dependent effects on the algal odorant production. Interestingly, when SRMC was in the range of 0.1-1, there were always promoting effects; when SRMC < 0.1 or SRMC > 1, there exhibited inhibiting effects, and the inhibiting effects of SRMC < 0.1 were far more severe than those of SRMC > 1. The promotion on odorant production in the SRMC range of 0.1-1 was mainly attributed to the increase in cellular yield, which was related to the increased odorant precursors derived from the oxidation products of reactive oxygen species (ROS). Alternatively, the inhibition of odorant production caused by MPs with SRMC < 0.1 was the results of simultaneously inhibiting cellular density and cellular yield, which might be attributed to the cellular internalization of MPs, inducing the extensive toxic effects. This study illustrated the possibilities of MPs in impairing the esthetics of the source water and provided guidance for the future algal odor issues under stress.
Collapse
Affiliation(s)
- Cheng Cen
- College of Civil Engineering and Architecture, Zhejiang University, Hangzhou 310058, China
| | - Kejia Zhang
- College of Civil Engineering and Architecture, Zhejiang University, Hangzhou 310058, China.
| | - Jie Fu
- School of Environmental Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaogang Wu
- College of Civil Engineering and Architecture, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Wu
- College of Civil Engineering and Architecture, Zhejiang University, Hangzhou 310058, China
| | - Yingying Zheng
- College of Civil Engineering and Architecture, Zhejiang University, Hangzhou 310058, China
| | - Yibo Zhang
- School of Environmental Science and Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
26
|
Agarwal A, Maldonado Rosas I, Anagnostopoulou C, Cannarella R, Boitrelle F, Munoz LV, Finelli R, Durairajanayagam D, Henkel R, Saleh R. Oxidative Stress and Assisted Reproduction: A Comprehensive Review of Its Pathophysiological Role and Strategies for Optimizing Embryo Culture Environment. Antioxidants (Basel) 2022; 11:antiox11030477. [PMID: 35326126 PMCID: PMC8944628 DOI: 10.3390/antiox11030477] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress (OS) due to an imbalance between reactive oxygen species (ROS) and antioxidants has been established as an important factor that can negatively affect the outcomes of assisted reproductive techniques (ARTs). Excess ROS exert their pathological effects through damage to cellular lipids, organelles, and DNA, alteration of enzymatic function, and apoptosis. ROS can be produced intracellularly, from immature sperm, oocytes, and embryos. Additionally, several external factors may induce high ROS production in the ART setup, including atmospheric oxygen, CO2 incubators, consumables, visible light, temperature, humidity, volatile organic compounds, and culture media additives. Pathological amounts of ROS can also be generated during the cryopreservation-thawing process of gametes or embryos. Generally, these factors can act at any stage during ART, from gamete preparation to embryo development, till the blastocyst stage. In this review, we discuss the in vitro conditions and environmental factors responsible for the induction of OS in an ART setting. In addition, we describe the effects of OS on gametes and embryos. Furthermore, we highlight strategies to ameliorate the impact of OS during the whole human embryo culture period, from gametes to blastocyst stage.
Collapse
Affiliation(s)
- Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH 44195, USA; (R.F.); (R.H.)
- Correspondence:
| | | | | | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy;
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Florence Boitrelle
- Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, 78300 Poissy, France;
- Department BREED, UVSQ, INRAE, Paris Saclay University, 78350 Jouy-en-Josas, France
| | - Lina Villar Munoz
- Citmer Reproductive Medicine, IVF LAB, Mexico City 11520, Mexico; (I.M.R.); (L.V.M.)
| | - Renata Finelli
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH 44195, USA; (R.F.); (R.H.)
| | - Damayanthi Durairajanayagam
- Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia;
| | - Ralf Henkel
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH 44195, USA; (R.F.); (R.H.)
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W2 1NY, UK
- Department of Medical Bioscience, University of the Western Cape, Bellville, Cape Town 7530, South Africa
- LogixX Pharma, Theale RG7 4AB, UK
| | - Ramadan Saleh
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Sohag University, Sohag 82524, Egypt;
- Ajyal IVF Center, Ajyal Hospital, Sohag 82524, Egypt
| |
Collapse
|
27
|
Vardar Acar N, Dursun A, Aygün D, Gürses Cila HE, Lay İ, Gülbakan B, Özgül RK. An investigation of different intracellular parameters for Inborn Errors of Metabolism: Cellular stress, antioxidant response and autophagy. Free Radic Biol Med 2022; 179:190-199. [PMID: 34974126 DOI: 10.1016/j.freeradbiomed.2021.12.312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 11/24/2022]
Abstract
Oxidative stress is associated with various disease pathologies including Inborn Errors of Metabolism (IEMs), among the most important causes of childhood morbidity and mortality. At least as much as oxidative stress in cells, reductive stress poses a danger to the disruption of cell homeostasis. p62/SQSTM1, protects cells from stress by activation of Nrf2/Keap1 and autophagy pathways. In this study, we tested the role of cellular stress, mitochondrial dysfunction and autophagy via Nrf2/Keap1/p62 pathway in the pathophysiology of three main groups of IEMs. Our results showed that antioxidant and oxidant capacity alone would not be sufficient to reflect the true clinical picture of these diseases. ATP, ROS and mitochondrial membrane potantial (MMP) measurements demonstrated increased cellular stress and bioenergetic imbalance in methylmalonic acidemia (MMA), indicating mild mitochondrial dysfunction. In isovaleric acidemia (IVA), no major change was detected in ATP, ROS and MMP values. Propionic acidemia (PA), mitochondrial diseases (MIT) and mucopolysaccharidosis IV (MPS IV) might point out mitohormesis to cope with chronic reductive stress. Induction of Nrf2/Keap1/p62 pathway and increased expression of HMOX1 were detected in all IEMs. LC3B-II and p62 expression results indicated an impaired autophagic flux in MIT and MPS IV and an induction of autophagic flux in MMA, PA and IVA, but also partial expression of Beclin1, enables autophagy activation, was detected in all IEMs. We conclude that individual diagnosis and treatments are of great importance in IEMs. In addition, we assume that the application of therapeutic antioxidant or preventive treatments without determining the cellular stress status in IEMs may disrupt the sensitive oxidant-antioxidant balance in the cell, leading to the potential to further disrupt the clinical picture, especially in patients with reductive stress. To the best of our knowledge, this is the first study to simultaneously relate IEMs with cellular stress, mitochondrial dysfunction, and autophagy.
Collapse
Affiliation(s)
- Neşe Vardar Acar
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ali Dursun
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Damla Aygün
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - H Esra Gürses Cila
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - İncilay Lay
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Basri Gülbakan
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - R Köksal Özgül
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
28
|
Zhang L, Kim SH, Park KH, Zhi-Wei Y, Jie Z, Townsend DM, Tew KD. Glutathione S-Transferase P Influences Redox Homeostasis and Response to Drugs that Induce the Unfolded Protein Response in Zebrafish. J Pharmacol Exp Ther 2021; 377:121-132. [PMID: 33514607 PMCID: PMC8047768 DOI: 10.1124/jpet.120.000417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/25/2021] [Indexed: 01/21/2023] Open
Abstract
We have created a novel glutathione S-transferase π1 (gstp1) knockout (KO) zebrafish model and used it for comparative analyses of redox homeostasis and response to drugs that cause endoplasmic reticulum (ER) stress and induce the unfolded protein response (UPR). Under basal conditions, gstp1 KO larvae had higher expression of antioxidant nuclear factor erythroid 2-related factor 2 (Nrf2) accompanied by a more reduced larval environment and a status consistent with reductive stress. Compared with wild type, various UPR markers were decreased in KO larvae, but treatment with drugs that induce ER stress caused greater toxicities and increased expression of Nrf2 and UPR markers in KO. Tunicamycin and 02-{2,4-dinitro-5-[4-(N-methylamino)benzoyloxy]phenyl}1-(N,N-dimethylamino)diazen-1-ium-1,2-diolate (PABA/nitric oxide) activated inositol-requiring protein-1/X-box binding protein 1 pathways, whereas thapsigargin caused greater activation of protein kinase-like ER kinase/activating transcription factor 4/CHOP pathways. These results suggest that this teleost model is useful for predicting how GSTP regulates organismal management of oxidative/reductive stress and is a determinant of response to drug-induced ER stress and the UPR. SIGNIFICANCE STATEMENT: A new zebrafish model has been created to study the importance of glutathione S-transferase π1 in development, redox homeostasis, and response to drugs that enact cytotoxicity through endoplasmic reticulum stress and induction of the unfolded protein response.
Collapse
Affiliation(s)
- Leilei Zhang
- Leilei Zhang, Seok-Hyung Kim, Ki-Hoon Park, Zhi-wei Ye, Jie Zhang, Danyelle M. Townsend, Kenneth D. Tew Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., Z.Y., J.Z., K.D.T.), Division of Nephrology, Department of Medicine (S.-H.K., K.-H.P.), and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Seok-Hyung Kim
- Leilei Zhang, Seok-Hyung Kim, Ki-Hoon Park, Zhi-wei Ye, Jie Zhang, Danyelle M. Townsend, Kenneth D. Tew Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., Z.Y., J.Z., K.D.T.), Division of Nephrology, Department of Medicine (S.-H.K., K.-H.P.), and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Ki-Hoon Park
- Leilei Zhang, Seok-Hyung Kim, Ki-Hoon Park, Zhi-wei Ye, Jie Zhang, Danyelle M. Townsend, Kenneth D. Tew Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., Z.Y., J.Z., K.D.T.), Division of Nephrology, Department of Medicine (S.-H.K., K.-H.P.), and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Ye Zhi-Wei
- Leilei Zhang, Seok-Hyung Kim, Ki-Hoon Park, Zhi-wei Ye, Jie Zhang, Danyelle M. Townsend, Kenneth D. Tew Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., Z.Y., J.Z., K.D.T.), Division of Nephrology, Department of Medicine (S.-H.K., K.-H.P.), and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Zhang Jie
- Leilei Zhang, Seok-Hyung Kim, Ki-Hoon Park, Zhi-wei Ye, Jie Zhang, Danyelle M. Townsend, Kenneth D. Tew Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., Z.Y., J.Z., K.D.T.), Division of Nephrology, Department of Medicine (S.-H.K., K.-H.P.), and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Danyelle M Townsend
- Leilei Zhang, Seok-Hyung Kim, Ki-Hoon Park, Zhi-wei Ye, Jie Zhang, Danyelle M. Townsend, Kenneth D. Tew Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., Z.Y., J.Z., K.D.T.), Division of Nephrology, Department of Medicine (S.-H.K., K.-H.P.), and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Kenneth D Tew
- Leilei Zhang, Seok-Hyung Kim, Ki-Hoon Park, Zhi-wei Ye, Jie Zhang, Danyelle M. Townsend, Kenneth D. Tew Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., Z.Y., J.Z., K.D.T.), Division of Nephrology, Department of Medicine (S.-H.K., K.-H.P.), and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
29
|
Zhou Y, Li H, Xia N. The Interplay Between Adipose Tissue and Vasculature: Role of Oxidative Stress in Obesity. Front Cardiovasc Med 2021; 8:650214. [PMID: 33748199 PMCID: PMC7969519 DOI: 10.3389/fcvm.2021.650214] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases (CVDs) rank the leading cause of morbidity and mortality globally. Obesity and its related metabolic syndrome are well-established risk factors for CVDs. Therefore, understanding the pathophysiological role of adipose tissues is of great importance in maintaining cardiovascular health. Oxidative stress, characterized by excessive formation of reactive oxygen species, is a common cellular stress shared by obesity and CVDs. While plenty of literatures have illustrated the vascular oxidative stress, very few have discussed the impact of oxidative stress in adipose tissues. Adipose tissues can communicate with vascular systems, in an endocrine and paracrine manner, through secreting several adipocytokines, which is largely dysregulated in obesity. The aim of this review is to summarize current understanding of the relationship between oxidative stress in obesity and vascular endothelial dysfunction. In this review, we briefly describe the possible causes of oxidative stress in obesity, and the impact of obesity-induced oxidative stress on adipose tissue function. We also summarize the crosstalk between adipose tissue and vasculature mediated by adipocytokines in vascular oxidative stress. In addition, we highlight the potential target mediating adipose tissue oxidative stress.
Collapse
Affiliation(s)
- Yawen Zhou
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
30
|
Aydos LR, do Amaral LA, Jacobowski AC, de Souza RS, Parisotto EB, de Menezes MB, Junior FFB, Fernandes ES, Silva IS, Portugal LC, Oliveira CG, Masuko GTS, Cavalheiro LF, Nazário CED, Dos Santos EF, Macedo MLR. Buriti pulp oil did not improve high-fat diet-induced metabolic disorders in c57bl/6 mice. J Anim Physiol Anim Nutr (Berl) 2021; 105:364-375. [PMID: 33226712 DOI: 10.1111/jpn.13473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/19/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022]
Abstract
Metabolic syndrome (MetS) and obesity are growing in many parts of the world, becoming public health problems. It is proposed that foods with functional properties can assist in the treatment of these diseases. Crude buriti pulp oil (BPO) is a food traditionally consumed by residents in the Pantanal, Cerrado and Brazilian Amazon. It is rich in oleic acid, tocopherols and carotenoids, emerging as a potential functional food. Thus, this study aimed to evaluate the effect of the supplementation of BPO on metabolic disorders caused by a high-fat diet. Four groups of C57BL6 mice were used, a lean group with AIN-93M diet and control oil supplementation, an obese group with a high-fat diet and control oil supplementation, and two obese groups with a high-fat diet and BPO supplementation in the amounts of 50 and 100 mg/kg. BPO worsened the metabolic state caused by the high-fat diet, worsening risk factors associated with MetS, as the abdominal circumference and retroperitoneal fat, serum levels of total cholesterol, uric acid, alanine transaminase, glucose and triglycerides, and renal fat, in addition to changes in glycaemic control and oxidative stress markers. C57BL/6 mice fed with a high-fat diet and supplemented with BPO presented a worsening in metabolic risk factors associated with MetS.
Collapse
Affiliation(s)
- Leonardo Recena Aydos
- Postgraduate Program in Health and Development in the Midwest Region, Faculty of Medicine (FAMED), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
- Protein Purification Laboratory and its Biological Functions (LPPFB), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
- Faculty of Medicine, Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| | - Luane Aparecida do Amaral
- Postgraduate Program in Health and Development in the Midwest Region, Faculty of Medicine (FAMED), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
- Faculty of Medicine, Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| | - Ana Cristina Jacobowski
- Protein Purification Laboratory and its Biological Functions (LPPFB), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
- Faculty of Pharmaceutical Sciences, Food, and Nutrition (FACFAN), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| | - Roberta Serafim de Souza
- Postgraduate Program in Health and Development in the Midwest Region, Faculty of Medicine (FAMED), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
- Protein Purification Laboratory and its Biological Functions (LPPFB), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
- Faculty of Medicine, Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| | - Eduardo Benedetti Parisotto
- Faculty of Pharmaceutical Sciences, Food, and Nutrition (FACFAN), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| | - Mariana Biava de Menezes
- Faculty of Pharmaceutical Sciences, Food, and Nutrition (FACFAN), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| | - Felipe Francisco Bittencourt Junior
- Faculty of Biological and Health Sciences, University Center of Grande Dourados (UNIGRAN), Dourados - MS, Brazil
- Clinical analysis laboratory, University Center of Grande Dourados (UNIGRAN), Dourados - MS, Brazil
| | - Emely Schuindt Fernandes
- Faculty of Biological and Health Sciences, University Center of Grande Dourados (UNIGRAN), Dourados - MS, Brazil
- Clinical analysis laboratory, University Center of Grande Dourados (UNIGRAN), Dourados - MS, Brazil
| | - Iandara Schettert Silva
- Faculty of Medicine, Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
- Laboratory of Experimental Disease Models, Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| | - Luciane Candeloro Portugal
- Bioscience Institute (INBIO), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| | - Cláudio Gonçalves Oliveira
- Bioscience Institute (INBIO), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| | | | | | | | - Elisvânia Freitas Dos Santos
- Faculty of Pharmaceutical Sciences, Food, and Nutrition (FACFAN), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| | - Maria Lígia Rodrigues Macedo
- Protein Purification Laboratory and its Biological Functions (LPPFB), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
- Faculty of Pharmaceutical Sciences, Food, and Nutrition (FACFAN), Federal University of Mato Grosso do Sul (UFMS), Campo Grande - MS, Brazil
| |
Collapse
|
31
|
Jersin RÅ, Tallapragada DSP, Madsen A, Skartveit L, Fjære E, McCann A, Lawrence-Archer L, Willems A, Bjune JI, Bjune MS, Våge V, Nielsen HJ, Thorsen HL, Nedrebø BG, Busch C, Steen VM, Blüher M, Jacobson P, Svensson PA, Fernø J, Rydén M, Arner P, Nygård O, Claussnitzer M, Ellingsen S, Madsen L, Sagen JV, Mellgren G, Dankel SN. Role of the Neutral Amino Acid Transporter SLC7A10 in Adipocyte Lipid Storage, Obesity, and Insulin Resistance. Diabetes 2021; 70:680-695. [PMID: 33408126 DOI: 10.2337/db20-0096] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 12/14/2020] [Indexed: 11/13/2022]
Abstract
Elucidation of mechanisms that govern lipid storage, oxidative stress, and insulin resistance may lead to improved therapeutic options for type 2 diabetes and other obesity-related diseases. Here, we find that adipose expression of the small neutral amino acid transporter SLC7A10, also known as alanine-serine-cysteine transporter-1 (ASC-1), shows strong inverse correlates with visceral adiposity, insulin resistance, and adipocyte hypertrophy across multiple cohorts. Concordantly, loss of Slc7a10 function in zebrafish in vivo accelerates diet-induced body weight gain and adipocyte enlargement. Mechanistically, SLC7A10 inhibition in human and murine adipocytes decreases adipocyte serine uptake and total glutathione levels and promotes reactive oxygen species (ROS) generation. Conversely, SLC7A10 overexpression decreases ROS generation and increases mitochondrial respiratory capacity. RNA sequencing revealed consistent changes in gene expression between human adipocytes and zebrafish visceral adipose tissue following loss of SLC7A10, e.g., upregulation of SCD (lipid storage) and downregulation of CPT1A (lipid oxidation). Interestingly, ROS scavenger reduced lipid accumulation and attenuated the lipid-storing effect of SLC7A10 inhibition. These data uncover adipocyte SLC7A10 as a novel important regulator of adipocyte resilience to nutrient and oxidative stress, in part by enhancing glutathione levels and mitochondrial respiration, conducive to decreased ROS generation, lipid accumulation, adipocyte hypertrophy, insulin resistance, and type 2 diabetes.
Collapse
Affiliation(s)
- Regine Å Jersin
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Divya Sri Priyanka Tallapragada
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - André Madsen
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Linn Skartveit
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Even Fjære
- Institute of Marine Research, Bergen, Norway
| | | | - Laurence Lawrence-Archer
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Aron Willems
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Jan-Inge Bjune
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Mona S Bjune
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Villy Våge
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
- Center of Health Research, Førde Hospital Trust, Førde, Norway
| | | | | | - Bjørn Gunnar Nedrebø
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haugesund Hospital, Haugesund, Norway
| | | | - Vidar M Steen
- NORMENT, K.G. Jebsen Center for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. E. Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Matthias Blüher
- Clinic for Endocrinology and Nephrology, Medical Research Center, Leipzig, Germany
| | - Peter Jacobson
- Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Per-Arne Svensson
- Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Johan Fernø
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Peter Arner
- Department of Medicine (H7), Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ottar Nygård
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Melina Claussnitzer
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Ståle Ellingsen
- Institute of Marine Research, Bergen, Norway
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Lise Madsen
- Institute of Marine Research, Bergen, Norway
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jørn V Sagen
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
- Bergen Stem Cell Consortium, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Simon N Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
32
|
Antioxidants Targeting Mitochondrial Oxidative Stress: Promising Neuroprotectants for Epilepsy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6687185. [PMID: 33299529 PMCID: PMC7710440 DOI: 10.1155/2020/6687185] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022]
Abstract
Mitochondria are major sources of reactive oxygen species (ROS) within the cell and are especially vulnerable to oxidative stress. Oxidative damage to mitochondria results in disrupted mitochondrial function and cell death signaling, finally triggering diverse pathologies such as epilepsy, a common neurological disease characterized with aberrant electrical brain activity. Antioxidants are considered as promising neuroprotective strategies for epileptic condition via combating the deleterious effects of excessive ROS production in mitochondria. In this review, we provide a brief discussion of the role of mitochondrial oxidative stress in the pathophysiology of epilepsy and evidences that support neuroprotective roles of antioxidants targeting mitochondrial oxidative stress including mitochondria-targeted antioxidants, polyphenols, vitamins, thiols, and nuclear factor E2-related factor 2 (Nrf2) activators in epilepsy. We point out these antioxidative compounds as effectively protective approaches for improving prognosis. In addition, we specially propose that these antioxidants exert neuroprotection against epileptic impairment possibly by modulating cell death interactions, notably autophagy-apoptosis, and autophagy-ferroptosis crosstalk.
Collapse
|
33
|
Tanaka M, Vécsei L. Monitoring the Redox Status in Multiple Sclerosis. Biomedicines 2020; 8:E406. [PMID: 33053739 PMCID: PMC7599550 DOI: 10.3390/biomedicines8100406] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Worldwide, over 2.2 million people suffer from multiple sclerosis (MS), a multifactorial demyelinating disease of the central nervous system. MS is characterized by a wide range of motor, autonomic, and psychobehavioral symptoms, including depression, anxiety, and dementia. The blood, cerebrospinal fluid, and postmortem brain samples of MS patients provide evidence on the disturbance of reduction-oxidation (redox) homeostasis, such as the alterations of oxidative and antioxidative enzyme activities and the presence of degradation products. This review article discusses the components of redox homeostasis, including reactive chemical species, oxidative enzymes, antioxidative enzymes, and degradation products. The reactive chemical species cover frequently discussed reactive oxygen/nitrogen species, infrequently featured reactive chemicals such as sulfur, carbonyl, halogen, selenium, and nucleophilic species that potentially act as reductive, as well as pro-oxidative stressors. The antioxidative enzyme systems cover the nuclear factor erythroid-2-related factor 2 (NRF2)-Kelch-like ECH-associated protein 1 (KEAP1) signaling pathway. The NRF2 and other transcriptional factors potentially become a biomarker sensitive to the initial phase of oxidative stress. Altered components of the redox homeostasis in MS were discussed in search of a diagnostic, prognostic, predictive, and/or therapeutic biomarker. Finally, monitoring the battery of reactive chemical species, oxidative enzymes, antioxidative enzymes, and degradation products helps to evaluate the redox status of MS patients to expedite the building of personalized treatment plans for the sake of a better quality of life.
Collapse
Affiliation(s)
- Masaru Tanaka
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary;
- Department of Neurology, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - László Vécsei
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary;
- Department of Neurology, Interdisciplinary Excellence Centre, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
34
|
Man AWC, Zhou Y, Xia N, Li H. Perivascular Adipose Tissue as a Target for Antioxidant Therapy for Cardiovascular Complications. Antioxidants (Basel) 2020; 9:E574. [PMID: 32630640 PMCID: PMC7402161 DOI: 10.3390/antiox9070574] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/23/2020] [Accepted: 06/27/2020] [Indexed: 12/12/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is the connective tissue surrounding most of the systemic blood vessels. PVAT is now recognized as an important endocrine tissue that maintains vascular homeostasis. Healthy PVAT has anticontractile, anti-inflammatory, and antioxidative roles. Vascular oxidative stress is an important pathophysiological event in cardiometabolic complications of obesity, type 2 diabetes, and hypertension. Accumulating data from both humans and experimental animal models suggests that PVAT dysfunction is potentially linked to cardiovascular diseases, and associated with augmented vascular inflammation, oxidative stress, and arterial remodeling. Reactive oxygen species produced from PVAT can be originated from mitochondria, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, and uncoupled endothelial nitric oxide synthase. PVAT can also sense vascular paracrine signals and response by secreting vasoactive adipokines. Therefore, PVAT may constitute a novel therapeutic target for the prevention and treatment of cardiovascular diseases. In this review, we summarize recent findings on PVAT functions, ROS production, and oxidative stress in different pathophysiological settings and discuss the potential antioxidant therapies for cardiovascular diseases by targeting PVAT.
Collapse
Affiliation(s)
| | | | | | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (A.W.C.M.); (Y.Z.); (N.X.)
| |
Collapse
|
35
|
Tjahjono E, McAnena AP, Kirienko NV. The evolutionarily conserved ESRE stress response network is activated by ROS and mitochondrial damage. BMC Biol 2020; 18:74. [PMID: 32600387 PMCID: PMC7322875 DOI: 10.1186/s12915-020-00812-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 06/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Mitochondrial dysfunction causes or contributes to a wide variety of pathologies, including neurodegenerative diseases, cancer, metabolic diseases, and aging. Cells actively surveil a number of mitochondrial readouts to ensure that cellular homeostasis is maintained. Results In this article, we characterize the role of the ethanol and stress response element (ESRE) pathway in mitochondrial surveillance and show that it is robustly activated when the concentration of reactive oxygen species (ROS) in the cell increases. While experiments were mostly performed in Caenorhabditis elegans, we observed similar gene activation profile in human cell lines. The linear relationship between ROS and ESRE activation differentiates ESRE from known mitochondrial surveillance pathways, such as the mitochondrial unfolded protein response (UPRmt), which monitor mitochondrial protein import. The ability of the ESRE network to be activated by increased ROS allows the cell to respond to oxidative and reductive stresses. The ESRE network works in tandem with other mitochondrial surveillance mechanisms as well, in a fashion that suggests a partially redundant hierarchy. For example, mutation of the UPRmt pathway results in earlier and more robust activation of the ESRE pathway. Interestingly, full expression of ATFS-1, a key transcription factor for the UPRmt, requires the presence of an ESRE motif in its promoter region. Conclusion The ESRE pathway responds to mitochondrial damage by monitoring ROS levels. This response is conserved in humans. The ESRE pathway is activated earlier when other mitochondrial surveillance pathways are unavailable during mitochondrial crises, potentially to mitigate stress and restore health. However, the exact mechanisms of pathway activation and crosstalk remain to be elucidated. Ultimately, a better understanding of this network, and its role in the constellation of mitochondrial and cellular stress networks, will improve healthspan.
Collapse
Affiliation(s)
- Elissa Tjahjono
- Department of BioSciences, Rice University, 6100 Main St, MS140, Houston, TX, 77005, USA
| | - Aidan P McAnena
- Department of BioSciences, Rice University, 6100 Main St, MS140, Houston, TX, 77005, USA
| | - Natalia V Kirienko
- Department of BioSciences, Rice University, 6100 Main St, MS140, Houston, TX, 77005, USA.
| |
Collapse
|
36
|
Ma WX, Li CY, Tao R, Wang XP, Yan LJ. Reductive Stress-Induced Mitochondrial Dysfunction and Cardiomyopathy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5136957. [PMID: 32566086 PMCID: PMC7277050 DOI: 10.1155/2020/5136957] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/26/2020] [Accepted: 05/12/2020] [Indexed: 02/05/2023]
Abstract
The goal of this review was to summarize reported studies focusing on cellular reductive stress-induced mitochondrial dysfunction, cardiomyopathy, dithiothreitol- (DTT-) induced reductive stress, and reductive stress-related free radical reactions published in the past five years. Reductive stress is considered to be a double-edged sword in terms of antioxidation and disease induction. As many underlying mechanisms are still unclear, further investigations are obviously warranted. Nonetheless, reductive stress is thought to be caused by elevated levels of cellular reducing power such as NADH, glutathione, and NADPH; and this area of research has attracted increasing attention lately. Albeit, we think there is a need to conduct further studies in identifying more indicators of the risk assessment and prevention of developing heart damage as well as exploring more targets for cardiomyopathy treatment. Hence, it is expected that further investigation of underlying mechanisms of reductive stress-induced mitochondrial dysfunction will provide novel insights into therapeutic approaches for ameliorating reductive stress-induced cardiomyopathy.
Collapse
Affiliation(s)
- Wei-Xing Ma
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center (UNTHSC), Fort Worth, Texas 76107, USA
- Qingdao University of Science and Technology, 266042 Qingdao, Shandong, China
| | - Chun-Yan Li
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center (UNTHSC), Fort Worth, Texas 76107, USA
- Shantou University Medical College, 515041 Shantou, Guangdong, China
| | - Ran Tao
- Qingdao Municipal Center for Disease Control & Prevention, 266034 Qingdao, Shandong, China
| | - Xin-Ping Wang
- Qingdao University of Science and Technology, 266042 Qingdao, Shandong, China
| | - Liang-Jun Yan
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center (UNTHSC), Fort Worth, Texas 76107, USA
| |
Collapse
|
37
|
Di Vincenzo A, Tana C, El Hadi H, Pagano C, Vettor R, Rossato M. Antioxidant, Anti-Inflammatory, and Metabolic Properties of Tocopherols and Tocotrienols: Clinical Implications for Vitamin E Supplementation in Diabetic Kidney Disease. Int J Mol Sci 2019; 20:ijms20205101. [PMID: 31618817 PMCID: PMC6834186 DOI: 10.3390/ijms20205101] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 12/23/2022] Open
Abstract
Diabetes mellitus is a metabolic disorder characterized by the development of vascular complications associated with high morbidity and mortality and the consequent relevant costs for the public health systems. Diabetic kidney disease is one of these complications that represent the main cause of end-stage renal disease in Western countries. Hyperglycemia, inflammation, and oxidative stress contribute to its physiopathology, and several investigations have been performed to evaluate the role of antioxidant supplementation as a complementary approach for the prevention and control of diabetes and associated disturbances. Vitamin E compounds, including different types of tocopherols and tocotrienols, have been considered as a treatment to tackle major cardiovascular outcomes in diabetic subjects, but often with conflicting or even negative results. However, their effects on diabetic nephropathy are even less clear, despite several intervention studies that showed the improvement of renal parameters after supplementation in patients with diabetic kidney disease. Then we performed a review of the literature about the role of vitamin E supplementation on diabetic nephropathy, also describing the underlying antioxidant, anti-inflammatory, and metabolic mechanisms to evaluate the possible use of tocopherols and tocotrienols in clinical practice.
Collapse
Affiliation(s)
- Angelo Di Vincenzo
- Department of Medicine-DIMED, Clinica Medica 3, Center for the Study and Integrated Management of Obesity, University-Hospital of Padova, 35100 Padova, Italy.
| | - Claudio Tana
- Internal Medicine and Critical Subacute Care Unit, Medicine Geriatric-Rehabilitation Department, and Department of Medicine and Surgery, University-Hospital of Parma, 43126 Parma, Italy.
| | - Hamza El Hadi
- Department of Medicine-DIMED, Clinica Medica 3, Center for the Study and Integrated Management of Obesity, University-Hospital of Padova, 35100 Padova, Italy.
- Department of Medicine, Klinikum Rheine, 48431 Rheine, Germany.
| | - Claudio Pagano
- Department of Medicine-DIMED, Clinica Medica 3, Center for the Study and Integrated Management of Obesity, University-Hospital of Padova, 35100 Padova, Italy.
| | - Roberto Vettor
- Department of Medicine-DIMED, Clinica Medica 3, Center for the Study and Integrated Management of Obesity, University-Hospital of Padova, 35100 Padova, Italy.
| | - Marco Rossato
- Department of Medicine-DIMED, Clinica Medica 3, Center for the Study and Integrated Management of Obesity, University-Hospital of Padova, 35100 Padova, Italy.
| |
Collapse
|