1
|
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
|
2
|
Martinez-Canton M, Galvan-Alvarez V, Martin-Rincon M, Calbet JAL, Gallego-Selles A. Unlocking peak performance: The role of Nrf2 in enhancing exercise outcomes and training adaptation in humans. Free Radic Biol Med 2024; 224:168-181. [PMID: 39151836 DOI: 10.1016/j.freeradbiomed.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/30/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024]
Abstract
Since the discovery of the nuclear factor erythroid-derived 2-like 2 (Nrf2) transcription factor thirty years ago, it has been shown that it regulates more than 250 genes involved in a multitude of biological processes, including redox balance, mitochondrial biogenesis, metabolism, detoxification, cytoprotection, inflammation, immunity, autophagy, cell differentiation, and xenobiotic metabolism. In skeletal muscle, Nrf2 signalling is primarily activated in response to perturbation of redox balance by reactive oxygen species or electrophiles. Initial investigations into human skeletal muscle Nrf2 responses to exercise, dating back roughly a decade, have consistently indicated that exercise-induced ROS production stimulates Nrf2 signalling. Notably, recent studies employing Nrf2 knockout mice have revealed impaired skeletal muscle contractile function characterised by reduced force output and increased fatigue susceptibility compared to wild-type counterparts. These deficiencies partially stem from diminished basal mitochondrial respiratory capacity and an impaired capacity to upregulate specific mitochondrial proteins in response to training, findings corroborated by inducible muscle-specific Nrf2 knockout models. In humans, baseline Nrf2 expression in skeletal muscle correlates with maximal oxygen uptake and high-intensity exercise performance. This manuscript delves into the mechanisms underpinning Nrf2 signalling in response to acute exercise in human skeletal muscle, highlighting the involvement of ROS, antioxidants and Keap1/Nrf2 signalling in exercise performance. Furthermore, it explores Nrf2's role in mediating adaptations to chronic exercise and its impact on overall exercise performance. Additionally, the influence of diet and certain supplements on basal Nrf2 expression and its role in modulating acute and chronic exercise responses are briefly addressed.
Collapse
Affiliation(s)
- Miriam Martinez-Canton
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, Las Palmas de Gran Canaria, 35017, Spain; Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Paseo Blas Cabrera Felipe "Físico" s/n, 35017, Las Palmas de Gran Canaria, Spain
| | - Victor Galvan-Alvarez
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, Las Palmas de Gran Canaria, 35017, Spain; Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Paseo Blas Cabrera Felipe "Físico" s/n, 35017, Las Palmas de Gran Canaria, Spain
| | - Marcos Martin-Rincon
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, Las Palmas de Gran Canaria, 35017, Spain; Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Paseo Blas Cabrera Felipe "Físico" s/n, 35017, Las Palmas de Gran Canaria, Spain
| | - Jose A L Calbet
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, Las Palmas de Gran Canaria, 35017, Spain; Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Paseo Blas Cabrera Felipe "Físico" s/n, 35017, Las Palmas de Gran Canaria, Spain; Department of Physical Performance, The Norwegian School of Sport Sciences, Postboks, 4014 Ulleval Stadion, 0806, Oslo, Norway; School of Kinesiology, Faculty of Education, The University of British Columbia, Vancouver, BC, Canada.
| | - Angel Gallego-Selles
- Department of Physical Education, University of Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, Las Palmas de Gran Canaria, 35017, Spain; Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria, Paseo Blas Cabrera Felipe "Físico" s/n, 35017, Las Palmas de Gran Canaria, Spain.
| |
Collapse
|
3
|
Liu L, de Leeuw K, van Goor H, Westra J. The Role of Antioxidant Transcription Factor Nrf2 and Its Activating Compounds in Systemic Lupus Erythematosus. Antioxidants (Basel) 2024; 13:1224. [PMID: 39456477 PMCID: PMC11504041 DOI: 10.3390/antiox13101224] [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: 08/31/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease in which kidney involvement, so-called lupus nephritis (LN), is common and one of the most severe manifestations. Oxidative stress (OS) may play a role in the pathogenesis of LN through the exacerbation of inflammation and immune cell dysfunction/dysregulation. Nuclear factor erythroid 2-related factor 2 (Nrf2), also known as nuclear factor erythroid-derived 2-like 2, is a transcription factor that in humans is encoded by the NFE2L2 gene and is regarded as a central regulator of the antioxidative response. Nrf2-activating compounds have been shown to alleviate oxidative stress in cells and tissues of lupus-prone mice. Although the precise mechanisms of Nrf2 activation on the immune system in SLE remain to be elucidated, Nrf2-activating compounds are considered novel therapeutical options to suppress OS and thereby might alleviate disease activity in SLE, especially in LN. This review therefore summarizes the role of the Nrf2 signaling pathway in the pathogenesis of SLE with LN and describes compounds modulating this pathway as potential additional clinical interventions.
Collapse
Affiliation(s)
- Lu Liu
- Department of Rheumatology and Clinical Immunology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Karina de Leeuw
- Department of Rheumatology and Clinical Immunology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Johanna Westra
- Department of Rheumatology and Clinical Immunology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
4
|
Hecht F, Zocchi M, Alimohammadi F, Harris IS. Regulation of antioxidants in cancer. Mol Cell 2024; 84:23-33. [PMID: 38029751 PMCID: PMC10843710 DOI: 10.1016/j.molcel.2023.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023]
Abstract
Scientists in this field often joke, "If you don't have a mechanism, say it's ROS." Seemingly connected to every biological process ever described, reactive oxygen species (ROS) have numerous pleiotropic roles in physiology and disease. In some contexts, ROS act as secondary messengers, controlling a variety of signaling cascades. In other scenarios, they initiate damage to macromolecules. Finally, in their worst form, ROS are deadly to cells and surrounding tissues. A set of molecules with detoxifying abilities, termed antioxidants, is the direct counterpart to ROS. Notably, antioxidants exist in the public domain, touted as a "cure-all" for diseases. Research has disproved many of these claims and, in some cases, shown the opposite. Of all the diseases, cancer stands out in its paradoxical relationship with antioxidants. Although the field has made numerous strides in understanding the roles of antioxidants in cancer, many questions remain.
Collapse
Affiliation(s)
- Fabio Hecht
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Marco Zocchi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Fatemeh Alimohammadi
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Isaac S Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
5
|
Lin DW, Hsu YC, Chang CC, Hsieh CC, Lin CL. Insights into the Molecular Mechanisms of NRF2 in Kidney Injury and Diseases. Int J Mol Sci 2023; 24:6053. [PMID: 37047024 PMCID: PMC10094034 DOI: 10.3390/ijms24076053] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Redox is a constant phenomenon in organisms. From the signaling pathway transduction to the oxidative stress during the inflammation and disease process, all are related to reduction-oxidation (redox). Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor targeting many antioxidant genes. In non-stressed conditions, NRF2 maintains the hemostasis of redox with housekeeping work. It expresses constitutively with basal activity, maintained by Kelch-like-ECH-associated protein 1 (KEAP1)-associated ubiquitination and degradation. When encountering stress, it can be up-regulated by several mechanisms to exert its anti-oxidative ability in diseases or inflammatory processes to protect tissues and organs from further damage. From acute kidney injury to chronic kidney diseases, such as diabetic nephropathy or glomerular disease, many results of studies have suggested that, as a master of regulating redox, NRF2 is a therapeutic option. It was not until the early termination of the clinical phase 3 trial of diabetic nephropathy due to heart failure as an unexpected side effect that we renewed our understanding of NRF2. NRF2 is not just a simple antioxidant capacity but has pleiotropic activities, harmful or helpful, depending on the conditions and backgrounds.
Collapse
Affiliation(s)
- Da-Wei Lin
- Department of Internal Medicine, St. Martin de Porres Hospital, Chiayi 600, Taiwan;
| | - Yung-Chien Hsu
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| | - Cheng-Chih Chang
- Department of Surgery, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (C.-C.C.); (C.-C.H.)
| | - Ching-Chuan Hsieh
- Department of Surgery, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (C.-C.C.); (C.-C.H.)
| | - Chun-Liang Lin
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Kidney Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| |
Collapse
|
6
|
Yan R, Tan F, Wang J, Zhao N. Conformation and dynamics of an active filament in crowded media. J Chem Phys 2023; 158:114905. [PMID: 36948796 DOI: 10.1063/5.0142559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
The structural and dynamical properties of active filamentous objects under macromolecular crowding have a great relevance in biology. By means of Brownian dynamics simulations, we perform a comparative study for the conformational change and diffusion dynamics of an active chain in pure solvents and in crowded media. Our result shows a robust compaction-to-swelling conformational change with the augment of the Péclet number. The presence of crowding facilitates self-trapping of monomers and, thus, reinforces the activity mediated compaction. In addition, the efficient collisions between the self-propelled monomers and crowders induce a coil-to-globulelike transition, indicated by a marked change of the Flory scaling exponent of the gyration radius. Moreover, the diffusion dynamics of the active chain in crowded solutions demonstrates activity-enhanced subdiffusion. The center of mass diffusion manifests rather new scaling relations with respect to both the chain length and Péclet number. The interplay of chain activity and medium crowding provides a new mechanism to understand the non-trivial properties of active filaments in complex environments.
Collapse
Affiliation(s)
- Ran Yan
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Fei Tan
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Jingli Wang
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Nanrong Zhao
- College of Chemistry, Sichuan University, Chengdu 610064, China
| |
Collapse
|
7
|
Weiss-Sadan T, Ge M, Hayashi M, Gohar M, Yao CH, de Groot A, Harry S, Carlin A, Fischer H, Shi L, Wei TY, Adelmann CH, Wolf K, Vornbäumen T, Dürr BR, Takahashi M, Richter M, Zhang J, Yang TY, Vijay V, Fisher DE, Hata AN, Haigis MC, Mostoslavsky R, Bardeesy N, Papagiannakopoulos T, Bar-Peled L. NRF2 activation induces NADH-reductive stress, providing a metabolic vulnerability in lung cancer. Cell Metab 2023; 35:487-503.e7. [PMID: 36841242 PMCID: PMC9998367 DOI: 10.1016/j.cmet.2023.01.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/15/2022] [Accepted: 01/26/2023] [Indexed: 02/27/2023]
Abstract
Multiple cancers regulate oxidative stress by activating the transcription factor NRF2 through mutation of its negative regulator, KEAP1. NRF2 has been studied extensively in KEAP1-mutant cancers; however, the role of this pathway in cancers with wild-type KEAP1 remains poorly understood. To answer this question, we induced NRF2 via pharmacological inactivation of KEAP1 in a panel of 50+ non-small cell lung cancer cell lines. Unexpectedly, marked decreases in viability were observed in >13% of the cell lines-an effect that was rescued by NRF2 ablation. Genome-wide and targeted CRISPR screens revealed that NRF2 induces NADH-reductive stress, through the upregulation of the NAD+-consuming enzyme ALDH3A1. Leveraging these findings, we show that cells treated with KEAP1 inhibitors or those with endogenous KEAP1 mutations are selectively vulnerable to Complex I inhibition, which impairs NADH oxidation capacity and potentiates reductive stress. Thus, we identify reductive stress as a metabolic vulnerability in NRF2-activated lung cancers.
Collapse
Affiliation(s)
- Tommy Weiss-Sadan
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Maolin Ge
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Makiko Hayashi
- Department of Pathology, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; Laura and Isaac Pelmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Magdy Gohar
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Cong-Hui Yao
- Department of Cell Biology, Blavatnik Institute Harvard Medical School, Boston, MA 02115, USA
| | - Adriaan de Groot
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stefan Harry
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alexander Carlin
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hannah Fischer
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lei Shi
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ting-Yu Wei
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Charles H Adelmann
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Cutaneous Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Konstantin Wolf
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Tristan Vornbäumen
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Benedikt R Dürr
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Mariko Takahashi
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Marianne Richter
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Junbing Zhang
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Tzu-Yi Yang
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Vindhya Vijay
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David E Fisher
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Cutaneous Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Aaron N Hata
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute Harvard Medical School, Boston, MA 02115, USA
| | - Raul Mostoslavsky
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Nabeel Bardeesy
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; The MGH Center for Regenerative Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; Laura and Isaac Pelmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Liron Bar-Peled
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; The MGH Center for Regenerative Medicine, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
8
|
Lei T, Zhao C, Yan R, Zhao N. Collective behavior of chiral active particles with anisotropic interactions in a confined space. SOFT MATTER 2023; 19:1312-1329. [PMID: 36723153 DOI: 10.1039/d2sm01402e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Extensive studies so far have indicated that chirality, anisotropic interactions and spatial confinement play important roles in collective dynamics in active matter systems. However, how the overall interplay of these crucial factors affects the novel phases and macroscopic properties remains less explored. Here, using Langevin dynamics simulations, we investigate the self-organization of a chiral active system composed of amphiphilic Janus particles, where the embedded anisotropic interaction orientation is assumed to be either the same or just opposite to the direction of active force. A wealth of dynamic phases are observed including formation of phase separation, clustering state, homogeneous state, spiral vortex flow, swarm and spatiotemporal oscillation. By tuning self-propelled angular speed and anisotropic interaction strength, we identify the non-equilibrium phase diagrams, and reveal the very non-trivial modulation of both vortex and swarm patterns. Intriguingly, we find that strong chirality-alignment-confinement coupling yields a self-driven spatial and temporal organization periodically oscillating between a counterclockwise vortex and a clockwise one. Our work provides a new understanding of the novel self-assembly arising in such a confined system and enables new strategies for achieving ordered dynamic structures.
Collapse
Affiliation(s)
- Ting Lei
- College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Chaonan Zhao
- College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Ran Yan
- College of Chemistry, Sichuan University, Chengdu 610064, China.
| | - Nanrong Zhao
- College of Chemistry, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
9
|
Hu B, Huang Y, Jakobs TC, Kang Q, Lv Z, Liu W, Wang R. Viability of mitochondria-labeled retinal ganglion cells in organotypic retinal explant cultures by two methods. Exp Eye Res 2023; 226:109311. [PMID: 36403849 PMCID: PMC11003390 DOI: 10.1016/j.exer.2022.109311] [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: 08/21/2022] [Revised: 10/09/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022]
Abstract
Retinal explant cultures provide a valuable system to study retinal function in vitro. This study established a new retinal explant culture method to prolong the survival of retinal ganglion cells (RGCs). Explants were prepared in two different ways: with or without optic nerve. Retinas from newborn mice that had received an injection of MitoTracker Red into the contralateral superior colliculus to label axonal mitochondria were cultured as organotypic culture for 7 days in vitro. At several time points during the culture, viability of RGCs was assessed by multi-electrode array recording, and morphology by immunohistochemical methods. During the culture, the thickness of the retinal tissue in both groups gradually decreased, however, the structure of the layers of the retina could be identified. Massive apoptosis in the retinal ganglion cell layer (GCL) appeared on the first day of culture, thereafter the number of apoptotic cells decreased. Glial activation was observed throughout the culture, and there was no difference in morphology between the two groups. RGCs loss was exacerbated on 3rdday of culture, and RGCs loss in retinal explants with preserved optic nerve was significantly lower than in retinas that did not preserve the optic nerve. More and longer-lasting mitochondrial signals were observed in the injured area of the optic nerve-preserving explants. Retinal explants provide an invaluable tool for studying retinal function and developing treatments for ocular diseases. The optic nerve-preserving culture helps preserve the integrity of RGCs. The higher number of mitochondria in the nerve-preserving cultures may help maintain viability of RGCs.
Collapse
Affiliation(s)
- Baoqi Hu
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China; Department of Ophthalmology, The First Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710002, China; Department of Ophthalmology, Xi'an No. 1 Hospital, Xi'an, Shaanxi, 710002, China
| | - Yaoyao Huang
- Department of Ophthalmology, The First Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710002, China; Department of Ophthalmology, Xi'an No. 1 Hospital, Xi'an, Shaanxi, 710002, China; Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Tatjana C Jakobs
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary / Schepens Eye Research Institute, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, United States
| | - Qianyan Kang
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ziwei Lv
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Wenxuan Liu
- Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Rui Wang
- Department of Ophthalmology, The First Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710002, China; Department of Ophthalmology, Xi'an No. 1 Hospital, Xi'an, Shaanxi, 710002, China; Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi, 710002, China.
| |
Collapse
|
10
|
Ogura J. [Association of Abnormal Disulfide Bond Formation with Disease Development and Progression]. YAKUGAKU ZASSHI 2022; 142:1055-1060. [PMID: 36184439 DOI: 10.1248/yakushi.22-00119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
As intermolecular and intramolecular disulfide bridges in proteins play a vital role in the stability of the final protein structure, the disruption of disulfide bridges in proteins may lead to disease development and progression. Therefore, understanding the association of abnormal protein disulfide bond formation with disease development and progression can be useful for developing novel drugs for various diseases. Considering that disulfide-linked protein folding involves redox reactions in the endoplasmic reticulum, this process may be affected by oxidative stress. We hypothesized that oxidative stress-related diseases may be induced by abnormal protein disulfide bond formation. This review introduces the association of abnormal protein disulfide bond formation with disease development and progression.
Collapse
Affiliation(s)
- Jiro Ogura
- Laboratory of Pharmaceutical Sciences, Graduate School of Medicine, Yamagata University.,Department of Pharmacy, Yamagata University Hospital
| |
Collapse
|
11
|
Lei T, Yan R, Zhao N. Biased-angle effect on diffusion dynamics and phase separation in anisotropic active particle system. J Chem Phys 2022; 156:204901. [DOI: 10.1063/5.0090427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
A deep understanding for collective behavior in an active matter system with complex interactions has far-reaching impact in biology. In the present work, we adopt Langevin dynamics simulations to investigate diffusion dynamics and phase separation in an anisotropic active particle system with a tunable biased angle α defined as the deviation between the active force direction and anisotropic orientation. Our results demonstrate that the biased angle can induce super-rotational diffusion dynamics characterized by a power-law relationship between the mean square angle displacement (MSAD) and the time interval Δ t in the form of MSAD ∼ Δ t β with β > 1 and also result in non-trivial phase separation kinetics. As activity is dominant, nucleation time shows a non-monotonic dependence on the biased angle. Moreover, there arises a distinct transition of phase separation, from spinodal decomposition without apparent nucleation time to binodal decomposition with prominent nucleation delay. A significant inhibition effect occurs at right and obtuse angles, where the remarkable super-rotational diffusion prevents particle aggregation, leading to a slow nucleation process. As active force is competitive to anisotropic interactions, the system is almost homogeneous, while, intriguingly, we observe a re-entrant phase separation as a small acute angle is introduced. The prominent super-rotational diffusion under small angles provides an optimum condition for particle adsorption and cluster growth and, thus, accounts for the re-entrance of phase separation. A consistent scenario for the physical mechanism of our observations is achieved by properly considering the modulation of the biased angle on the interplay between activity and anisotropic interactions.
Collapse
Affiliation(s)
- Ting Lei
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Ran Yan
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Nanrong Zhao
- College of Chemistry, Sichuan University, Chengdu 610064, China
| |
Collapse
|
12
|
Madan S, Uttekar B, Chowdhary S, Rikhy R. Mitochondria Lead the Way: Mitochondrial Dynamics and Function in Cellular Movements in Development and Disease. Front Cell Dev Biol 2022; 9:781933. [PMID: 35186947 PMCID: PMC8848284 DOI: 10.3389/fcell.2021.781933] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023] Open
Abstract
The dynamics, distribution and activity of subcellular organelles are integral to regulating cell shape changes during various physiological processes such as epithelial cell formation, cell migration and morphogenesis. Mitochondria are famously known as the powerhouse of the cell and play an important role in buffering calcium, releasing reactive oxygen species and key metabolites for various activities in a eukaryotic cell. Mitochondrial dynamics and morphology changes regulate these functions and their regulation is, in turn, crucial for various morphogenetic processes. In this review, we evaluate recent literature which highlights the role of mitochondrial morphology and activity during cell shape changes in epithelial cell formation, cell division, cell migration and tissue morphogenesis during organism development and in disease. In general, we find that mitochondrial shape is regulated for their distribution or translocation to the sites of active cell shape dynamics or morphogenesis. Often, key metabolites released locally and molecules buffered by mitochondria play crucial roles in regulating signaling pathways that motivate changes in cell shape, mitochondrial shape and mitochondrial activity. We conclude that mechanistic analysis of interactions between mitochondrial morphology, activity, signaling pathways and cell shape changes across the various cell and animal-based model systems holds the key to deciphering the common principles for this interaction.
Collapse
|
13
|
Sengupta P, Dutta S, Alahmar AT. Reductive Stress and Male Infertility. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1391:311-321. [PMID: 36472829 DOI: 10.1007/978-3-031-12966-7_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Male infertility research and clinical advances had vast progress in the last few decades. Strong research evidence underpinned the concepts of oxidative stress (OS)-mediated male reproductive disruptions, which bear answers to several cases of idiopathic male infertility. Antioxidant treatment held the prime solution for OS-mediated male infertility. But excess use of antioxidants is challenged by the research breakthrough that reductive stress also predisposes to male infertility, resolutely instituting that any biological extremes of the redox spectrum are deleterious to male fertility. Superfluity of reducing agents may hinder essential oxidation mechanisms, affecting physiological homeostasis. These mechanisms need to be explicated and updated time and again to identify the fine thread between OS-mediated male infertility treatment and induction of reductive stress. This chapter thus presents the evidence-based concepts pertaining to the antioxidants actions to combat OS-induced male infertility, the mechanism of induction of reductive stress and its impact on male reproduction.
Collapse
Affiliation(s)
- Pallav Sengupta
- Physiology Unit, Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, UAE.
| | - Sulagna Dutta
- School of Medical Sciences, Bharath Institute of Higher Education and Research (BIHER), Chennai, India
| | - Ahmed T Alahmar
- Department of Medical Physiology, College of Medicine, University of Babylon, Hillah, Iraq
| |
Collapse
|
14
|
Jia J, Jin H, Nan D, Yu W, Huang Y. New insights into targeting mitochondria in ischemic injury. Apoptosis 2021; 26:163-183. [PMID: 33751318 DOI: 10.1007/s10495-021-01661-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2021] [Indexed: 12/15/2022]
Abstract
Stroke is the leading cause of adult disability and death worldwide. Mitochondrial dysfunction has been recognized as a marker of neuronal death during ischemic stroke. Maintaining the function of mitochondria is important for improving the survival of neurons and maintaining neuronal function. Damaged mitochondria induce neuronal cell apoptosis by releasing reactive oxygen species (ROS) and pro-apoptotic factors. Mitochondrial fission and fusion processes and mitophagy are of great importance to mitochondrial quality control. This paper reviews the dynamic changes in mitochondria, the roles of mitochondria in different cell types, and related signaling pathways in ischemic stroke. This review describes in detail the role of mitochondria in the process of neuronal injury and protection in cerebral ischemia, and integrates neuroprotective drugs targeting mitochondria in recent years, which may provide a theoretical basis for the progress of treatment of ischemic stroke. The potential of mitochondrial-targeted therapy is also emphasized, which provides valuable insights for clinical research.
Collapse
Affiliation(s)
- Jingjing Jia
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Haiqiang Jin
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Ding Nan
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Weiwei Yu
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China.
| |
Collapse
|
15
|
Hu G, Zhong M, Zhao J, Gao H, Gan L, Zhang H, Zhang S, Fang J. Fluorescent Probes for Imaging Protein Disulfides in Live Organisms. ACS Sens 2021; 6:1384-1391. [PMID: 33721991 DOI: 10.1021/acssensors.1c00049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cellular redox homeostasis is predominantly controlled by the ratio of thiols and disulfides, and reversible thiol-disulfide exchange reactions are fundamental of the biological redox regulation. However, due to the dynamic exchanges of thiols and disulfides, the detection, especially the in situ detection, of protein disulfides (PDS) is challenging. We employ the strategy, i.e., the increase of emission upon an environment-sensitive dye binding to proteins, to design PDS probes and discover a two-photon probe PDSTP590 (S6) that selectively recognizes PDS in live organisms. With the aid of the probe, we further disclose the elevation of PDS in brains of the mouse stroke model.
Collapse
Affiliation(s)
- Guodong Hu
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Miao Zhong
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jintao Zhao
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Hao Gao
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Lu Gan
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China
| | - Hong Zhang
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China
| | - Shengxiang Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
16
|
Health disparities: Intracellular consequences of social determinants of health. Toxicol Appl Pharmacol 2021; 416:115444. [PMID: 33549591 DOI: 10.1016/j.taap.2021.115444] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/27/2021] [Accepted: 01/30/2021] [Indexed: 12/14/2022]
Abstract
Health disparities exist dependent on socioeconomic status, living conditions, race/ethnicity, diet, and exposures to environmental pollutants. Herein, the various exposures contributing to a person's exposome are collectively considered social determinants of health (SDOH), and the SDOH-exposome impacts health more than health care. This review discusses the extent of evidence of the physiologic consequences of these exposures at the intracellular level. We consider how the SDOH-exposome, which captures how individuals live, work and age, induces cell processes that modulate a conceptual "redox rheostat." Like an electrical resistor, the SDOH-exposome, along with genetic predisposition and age, regulate reductive and oxidative (redox) stress circuits and thereby stimulate inflammation. Regardless of the source of the SDOH-exposome that induces chronic inflammation and immunosenescence, the outcome influences cardiometabolic diseases, cancers, infections, sepsis, neurodegeneration and autoimmune diseases. The endogenous redox rheostat is connected with regulatory molecules such as NAD+/NADH and SIRT1 that drive redox pathways. In addition to these intracellular and mitochondrial processes, we discuss how the SDOH-exposome can influence the balance between metabolism and regulation of immune responsiveness involving the two main molecular drivers of inflammation, the NLRP3 inflammasome and NF-κB induction. Mitochondrial and inflammasome activities play key roles in mediating defenses against pathogens and controlling inflammation before diverse cell death pathways are induced. Specifically, pyroptosis, cell death by inflammation, is intimately associated with common disease outcomes that are influenced by the SDOH-exposome. Redox influences on immunometabolism including protein cysteines and ion fluxes are discussed regarding health outcomes. In summary, this review presents a translational research perspective, with evidence from in vitro and in vivo models as well as clinical and epidemiological studies, to outline the intracellular consequences of the SDOH-exposome that drive health disparities in patients and populations. The relevance of this conceptual and theoretical model considering the SARS-CoV-2 pandemic are highlighted. Finally, the case of asthma is presented as a chronic condition that is modified by adverse SDOH exposures and is manifested through the dysregulation of immune cell redox regulatory processes we highlight in this review.
Collapse
|
17
|
George AK, Homme RP, Stanisic D, Tyagi SC, Singh M. Protecting the aging eye with hydrogen sulfide. Can J Physiol Pharmacol 2021; 99:161-170. [PMID: 32721225 DOI: 10.1139/cjpp-2020-0216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Research demonstrates that senescence is associated with tissue and organ dysfunction, and the eye is no exception. Sequelae arising from aging have been well defined as distinct clinical entities and vision impairment has significant psychosocial consequences. Retina and adjacent tissues like retinal pigmented epithelium and choroid are the key structures that are required for visual perception. Any structural and functional changes in retinal layers and blood retinal barrier could lead to age-related macular degeneration, diabetic retinopathy, and glaucoma. Further, there are significant oxygen gradients in the eye that can lead to excessive reactive oxygen species, resulting in endoplasmic reticulum and mitochondrial stress response. These radicals are source of functional and morphological impairment in retinal pigmented epithelium and retinal ganglion cells. Therefore, ocular diseases could be summarized as disturbance in the redox homeostasis. Hyperhomocysteinemia is a risk factor and causes vascular occlusive disease of the retina. Interestingly, hydrogen sulfide (H2S) has been proven to be an effective antioxidant agent, and it can help treat diseases by alleviating stress and inflammation. Concurrent glutamate excitotoxicity, endoplasmic reticulum stress, and microglia activation are also linked to stress; thus, H2S may offer additional interventional strategy. A refined understanding of the aging eye along with H2S biology and pharmacology may help guide newer therapies for the eye.
Collapse
Affiliation(s)
- Akash K George
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Rubens P Homme
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Dragana Stanisic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Mahavir Singh
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
18
|
Kwon Y. Possible Beneficial Effects of N-Acetylcysteine for Treatment of Triple-Negative Breast Cancer. Antioxidants (Basel) 2021; 10:169. [PMID: 33498875 PMCID: PMC7911701 DOI: 10.3390/antiox10020169] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 12/24/2022] Open
Abstract
N-acetylcysteine (NAC) is a widely used antioxidant with therapeutic potential. However, the cancer-promoting effect of NAC observed in some preclinical studies has raised concerns regarding its clinical use. Reactive oxygen species (ROS) can mediate signaling that results in both cancer-promoting and cancer-suppressing effects. The beneficial effect of NAC may depend on whether the type of cancer relies on ROS signaling for its survival and metastasis. Triple-negative breast cancer (TNBC) has aggressive phenotypes and is currently treated with standard chemotherapy as the main systemic treatment option. Particularly, basal-like TNBC cells characterized by inactivated BRCA1 and mutated TP53 produce high ROS levels and rely on ROS signaling for their survival and malignant progression. In addition, the high ROS levels in TNBC cells can mediate the interplay between cancer cells and the tissue microenvironment (TME) to trigger the recruitment and conversion of stromal cells and induce hypoxic responses, thus leading to the creation of cancer-supportive TMEs and increased cancer aggressiveness. However, NAC treatment effectively reduces the ROS production and ROS-mediated signaling that contribute to cell survival, metastasis, and drug resistance in TNBC cells. Therefore, the inclusion of NAC in standard chemotherapy could probably provide additional benefits for TNBC patients.
Collapse
Affiliation(s)
- Youngjoo Kwon
- Department of Food Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
19
|
Kong H, Reczek CR, McElroy GS, Steinert EM, Wang T, Sabatini DM, Chandel NS. Metabolic determinants of cellular fitness dependent on mitochondrial reactive oxygen species. SCIENCE ADVANCES 2020; 6:6/45/eabb7272. [PMID: 33148642 PMCID: PMC7673681 DOI: 10.1126/sciadv.abb7272] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 09/14/2020] [Indexed: 05/21/2023]
Abstract
Mitochondria-derived reactive oxygen species (mROS) are required for the survival, proliferation, and metastasis of cancer cells. The mechanism by which mitochondrial metabolism regulates mROS levels to support cancer cells is not fully understood. To address this, we conducted a metabolism-focused CRISPR-Cas9 genetic screen and uncovered that loss of genes encoding subunits of mitochondrial complex I was deleterious in the presence of the mitochondria-targeted antioxidant mito-vitamin E (MVE). Genetic or pharmacologic inhibition of mitochondrial complex I in combination with the mitochondria-targeted antioxidants, MVE or MitoTEMPO, induced a robust integrated stress response (ISR) and markedly diminished cell survival and proliferation in vitro. This was not observed following inhibition of mitochondrial complex III. Administration of MitoTEMPO in combination with the mitochondrial complex I inhibitor phenformin decreased the leukemic burden in a mouse model of T cell acute lymphoblastic leukemia. Thus, mitochondrial complex I is a dominant metabolic determinant of mROS-dependent cellular fitness.
Collapse
Affiliation(s)
- Hyewon Kong
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Colleen R Reczek
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Gregory S McElroy
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Elizabeth M Steinert
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Tim Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Broad Institute, Cambridge, MA 02142, USA
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Navdeep S Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
20
|
Bellezza I, Riuzzi F, Chiappalupi S, Arcuri C, Giambanco I, Sorci G, Donato R. Reductive stress in striated muscle cells. Cell Mol Life Sci 2020; 77:3547-3565. [PMID: 32072237 PMCID: PMC11105111 DOI: 10.1007/s00018-020-03476-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/17/2020] [Accepted: 02/03/2020] [Indexed: 12/11/2022]
Abstract
Reductive stress is defined as a condition of sustained increase in cellular glutathione/glutathione disulfide and NADH/NAD+ ratios. Reductive stress is emerging as an important pathophysiological event in several diseased states, being as detrimental as is oxidative stress. Occurrence of reductive stress has been documented in several cardiomyopathies and is an important pathophysiological factor particularly in coronary artery disease and myocardial infarction. Excess activation of the transcription factor, Nrf2-the master regulator of the antioxidant response-, consequent in most cases to defective autophagy, can lead to reductive stress. In addition, hyperglycemia-induced activation of the polyol pathway can lead to increased NADH/NAD+ ratio, which might translate into increased levels of hydrogen sulfide-via enhanced activity of cystathionine β-synthase-that would fuel reductive stress through inhibition of mitochondrial complex I. Reductive stress may be either a potential weapon against cancer priming tumor cells to apoptosis or a cancer's ally promoting tumor cell proliferation and making tumor cells resistant to reactive oxygen species-inducing drugs. In non-cancer pathological states reductive stress is definitely harmful paradoxically leading to reactive oxygen species overproduction via excess NADPH oxidase 4 activity. In face of the documented occurrence of reductive stress in several heart diseases, there is much less information about the occurrence and effects of reductive stress in skeletal muscle tissue. In the present review we describe relevant results emerged from studies of reductive stress in the heart and review skeletal muscle conditions in which reductive stress has been experimentally documented and those in which reductive stress might have an as yet unrecognized pathophysiological role. Establishing whether reductive stress has a (patho)physiological role in skeletal muscle will hopefully contribute to answer the question whether antioxidant supplementation to the general population, athletes, and a large cohort of patients (e.g. heart, sarcopenic, dystrophic, myopathic, cancer, and bronco-pulmonary patients) is harmless or detrimental.
Collapse
Affiliation(s)
- Ilaria Bellezza
- Department of Experimental Medicine, Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Francesca Riuzzi
- Department of Experimental Medicine, Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Sara Chiappalupi
- Department of Experimental Medicine, Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine, Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine, Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Guglielmo Sorci
- Department of Experimental Medicine, Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
- Centro Universitario Di Ricerca Sulla Genomica Funzionale, University of Perugia, 06132, Perugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine, Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy.
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy.
| |
Collapse
|
21
|
Tanaka LY, Oliveira PVS, Laurindo FRM. Peri/Epicellular Thiol Oxidoreductases as Mediators of Extracellular Redox Signaling. Antioxid Redox Signal 2020; 33:280-307. [PMID: 31910038 DOI: 10.1089/ars.2019.8012] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Significance: Supracellular redox networks regulating cell-extracellular matrix (ECM) and organ system architecture merge with structural and functional (catalytic or allosteric) properties of disulfide bonds. This review addresses emerging evidence that exported thiol oxidoreductases (TORs), such as thioredoxin, protein disulfide isomerases (PDIs), quiescin sulfhydryl oxidases (QSOX)1, and peroxiredoxins, composing a peri/epicellular (pec)TOR pool, mediate relevant signaling. pecTOR functions depend mainly on kinetic and spatial regulation of thiol-disulfide exchange reactions governed by redox potentials, which are modulated by exported intracellular low-molecular-weight thiols, together conferring signal specificity. Recent Advances: pecTOR redox-modulates several targets including integrins, ECM proteins, surface molecules, and plasma components, although clear-cut documentation of direct effects is lacking in many cases. TOR catalytic pathways, displaying common patterns, culminate in substrate thiol reduction, oxidation, or isomerization. Peroxiredoxins act as redox/peroxide sensors, contrary to PDIs, which are likely substrate-targeted redox modulators. Emerging evidence suggests important pecTOR roles in patho(physio)logical processes, including blood coagulation, vascular remodeling, mechanosensing, endothelial function, immune responses, and inflammation. Critical Issues: Effects of pecPDIs supporting thrombosis/platelet activation have been well documented and reached the clinical arena. Roles of pecPDIA1 in vascular remodeling/mechanosensing are also emerging. Extracellular thioredoxin and pecPDIs redox-regulate immunoinflammation. Routes of TOR externalization remain elusive and appear to involve Golgi-independent routes. pecTORs are particularly accessible drug targets. Future Directions: Further understanding mechanisms of thiol redox reactions and developing assays for assessing pecTOR redox activities remain important research avenues. Also, addressing pecTORs as disease markers and achieving more efficient/specific drugs for pecTOR modulation are major perspectives for diagnostic/therapeutic improvements.
Collapse
Affiliation(s)
- Leonardo Y Tanaka
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Percillia V S Oliveira
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
22
|
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
|
23
|
Abstract
Significance: Reducing equivalents (NAD(P)H and glutathione [GSH]) are essential for maintaining cellular redox homeostasis and for modulating cellular metabolism. Reductive stress induced by excessive levels of reduced NAD+ (NADH), reduced NADP+ (NADPH), and GSH is as harmful as oxidative stress and is implicated in many pathological processes. Recent Advances: Reductive stress broadens our view of the importance of cellular redox homeostasis and the influences of an imbalanced redox niche on biological functions, including cell metabolism. Critical Issues: The distribution of cellular NAD(H), NADP(H), and GSH/GSH disulfide is highly compartmentalized. Understanding how cells coordinate different pools of redox couples under unstressed and stressed conditions is critical for a comprehensive view of redox homeostasis and stress. It is also critical to explore the underlying mechanisms of reductive stress and its biological consequences, including effects on energy metabolism. Future Directions: Future studies are needed to investigate how reductive stress affects cell metabolism and how cells adapt their metabolism to reductive stress. Whether or not NADH shuttles and mitochondrial nicotinamide nucleotide transhydrogenase enzyme can regulate hypoxia-induced reductive stress is also a worthy pursuit. Developing strategies (e.g., antireductant approaches) to counteract reductive stress and its related adverse biological consequences also requires extensive future efforts.
Collapse
Affiliation(s)
- Wusheng Xiao
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Shen L, Ju JJ, Liu Q, Wang SS, Meng H, Ge XQ, Huang WY. Antioxidative and Neuroprotective Effects of the Cytochalasans From Endophytes. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20917308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Six 10-indolyl-cytochalasans, chaetoglobosin F (1), chaetoglobosin Fex (2), chaetoglobosin E (3), cytoglobosin A (4), penochalasin C (5), and isochaetoglobosin D (6), and 2 10-phenyl-cytochalasans, cytochalasin H (7) and 18-methoxycytochalasin J (8) were isolated from 2 plant endophytes, Chaetomiun globosum WQ and Phomopsis sp. IFB-E060, respectively. These cytochalasans were investigated with radical-scavenging activity assay and hydrogen peroxide (H2O2)/ N-methyl-4-phenylpyridinium iodide (MPP+)-induced pheochromocytoma cell line 12 (PC12) cell models, respectively. Results showed that 7 compounds had antioxidative effects except 5, with scavenging 2,2-diphenyl-1-picrylhydrazyl radical effect 1 = 6= 7 > 2> 4 = 3 > 8 and scavenging 2,2-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt radical effect 1 = 6 = 7 > 2 > 3 > 8 > 4. They could also inhibit H2O2/MPP+-induced damage in PC12 cells by increasing cell viability and decreasing lactate dehydrogenase release. Compounds 1, 6, and 7 exhibited the strongest antioxidative potencies, which are more potent than vitamin E. Additionally, antioxidative and neuroprotective effects of 1∼8 showed some structure–activity relationship. These findings would be useful for looking for cytochalasin-related new lead compounds or drugs to prevent and treat Parkinson’s disease.
Collapse
Affiliation(s)
- Li Shen
- Institute of Translational Medicine & Medical College, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, PR China
| | - Jing-Jing Ju
- Institute of Translational Medicine & Medical College, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, PR China
- The Second Affiliated Hospital of Nanjing Medical University, PR China
| | - Qin Liu
- Jiangsu Lixiahe Region Institute of Agricultural Science, Yangzhou, PR China
| | - Sha-Sha Wang
- Institute of Translational Medicine & Medical College, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, PR China
| | - Hao Meng
- Beijing Academy of Science and Technology, PR China
| | - Xiao-Qun Ge
- Institute of Translational Medicine & Medical College, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, PR China
| | - Wu-Yang Huang
- Institute of Translational Medicine & Medical College, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, PR China
- Department of Nutrition and Health, Institute of Agro-Product Processing & Jiangsu Key Laboratory for Horticultural Crop Genetic Improvement, Jiangsu Academy of Agricultural Sciences, Nanjing, PR China
| |
Collapse
|
25
|
Nascimento-Dos-Santos G, de-Souza-Ferreira E, Lani R, Faria CC, Araújo VG, Teixeira-Pinheiro LC, Vasconcelos T, Gonçalo T, Santiago MF, Linden R, Galina A, Petrs-Silva H. Neuroprotection from optic nerve injury and modulation of oxidative metabolism by transplantation of active mitochondria to the retina. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165686. [PMID: 31953215 DOI: 10.1016/j.bbadis.2020.165686] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 01/16/2023]
Abstract
Mitochondrial dysfunctions are linked to a series of neurodegenerative human conditions, including Parkinson's disease, schizophrenia, optic neuropathies, and glaucoma. Recently, a series of studies have pointed mitotherapy - exogenous mitochondria transplant - as a promising way to attenuate the progression of neurologic disorders; however, the neuroprotective and pro-regenerative potentials of isolated mitochondria in vivo have not yet been elucidated. In this present work, we tested the effects of transplants of active (as well-coupled organelles were named), liver-isolated mitochondria on the survival of retinal ganglion cells and axonal outgrowth after optic nerve crush. Our data show that intravitreally transplanted, full active mitochondria incorporate into the retina, improve its oxidative metabolism and electrophysiological activity at 1 day after transplantation. Moreover, mitotherapy increases cell survival in the ganglion cell layer at 14 days, and leads to a higher number of axons extending beyond the injury site at 28 days; effects that are dependent on the organelles' structural integrity. Together, our findings support mitotherapy as a promising approach for future therapeutic interventions upon central nervous system damage.
Collapse
Affiliation(s)
- Gabriel Nascimento-Dos-Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Eduardo de-Souza-Ferreira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Rafael Lani
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Caroline Coelho Faria
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Victor Guedes Araújo
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | | | - Taliane Vasconcelos
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Thaís Gonçalo
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Marcelo Felippe Santiago
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Rafael Linden
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Antonio Galina
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Hilda Petrs-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| |
Collapse
|
26
|
Peroxisomal Hydrogen Peroxide Metabolism and Signaling in Health and Disease. Int J Mol Sci 2019; 20:ijms20153673. [PMID: 31357514 PMCID: PMC6695606 DOI: 10.3390/ijms20153673] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 12/29/2022] Open
Abstract
Hydrogen peroxide (H2O2), a non-radical reactive oxygen species generated during many (patho)physiological conditions, is currently universally recognized as an important mediator of redox-regulated processes. Depending on its spatiotemporal accumulation profile, this molecule may act as a signaling messenger or cause oxidative damage. The focus of this review is to comprehensively evaluate the evidence that peroxisomes, organelles best known for their role in cellular lipid metabolism, also serve as hubs in the H2O2 signaling network. We first briefly introduce the basic concepts of how H2O2 can drive cellular signaling events. Next, we outline the peroxisomal enzyme systems involved in H2O2 metabolism in mammals and reflect on how this oxidant can permeate across the organellar membrane. In addition, we provide an up-to-date overview of molecular targets and biological processes that can be affected by changes in peroxisomal H2O2 metabolism. Where possible, emphasis is placed on the molecular mechanisms and factors involved. From the data presented, it is clear that there are still numerous gaps in our knowledge. Therefore, gaining more insight into how peroxisomes are integrated in the cellular H2O2 signaling network is of key importance to unravel the precise role of peroxisomal H2O2 production and scavenging in normal and pathological conditions.
Collapse
|
27
|
de-Souza-Ferreira E, Rios-Neto IM, Martins EL, Galina A. Mitochondria-coupled glucose phosphorylation develops after birth to modulate H 2 O 2 release and calcium handling in rat brain. J Neurochem 2019; 149:624-640. [PMID: 31001830 DOI: 10.1111/jnc.14705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/20/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022]
Abstract
The adult brain is a high-glucose and oxygen-dependent organ, with an extremely organized network of cells and large energy-consuming synapses. To reach this level of organization, early stages in development must include an efficient control of cellular events and regulation of intracellular signaling molecules and ions such as hydrogen peroxide (H2 O2 ) and calcium (Ca2+ ), but in cerebral tissue, these mechanisms of regulation are still poorly understood. Hexokinase (HK) is the first enzyme in the metabolism of glucose and, when bound to mitochondria (mtHK), it has been proposed to have a role in modulation of mitochondrial H2 O2 generation and Ca2+ handling. Here, we have investigated how mtHK modulates these signals in the mitochondrial context during postnatal development of the mouse brain. Using high-resolution respirometry, western blot analysis, spectrometry and resorufin, and Calcium Green fluorescence assays with brain mitochondria purified postnatally from day 1 to day 60, we demonstrate that brain HK increases its coupling to mitochondria and to oxidative phosphorylation to induce a cycle of ADP entry/ATP exit of the mitochondrial matrix that leads to efficient control over H2 O2 generation and Ca2+ uptake during development until reaching plateau at day 21. This contrasts sharply with the antioxidant enzymes, which do not increase as mitochondrial H2 O2 generation escalates. These results suggest that, as its use of glucose increases, the brain couples HK to mitochondria to improve glucose metabolism, redox balance and Ca2+ signaling during development, positioning mitochondria-bound hexokinase as a hub for intracellular signaling control.
Collapse
Affiliation(s)
- Eduardo de-Souza-Ferreira
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Izac Miranda Rios-Neto
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduarda Lopes Martins
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Antonio Galina
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
28
|
Wang S, Nong X, Yang G. Selenium-Rich Diet Induces Myocardial Structural and Functional Abnormalities by Activating Caspase-9 and Caspase-3 in Gpx-1P198L-Overexpression Transgenic Mice. Med Sci Monit 2019; 25:61-70. [PMID: 30602716 PMCID: PMC6327778 DOI: 10.12659/msm.911120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background Selenium (Se) deficiency and supplementation result in multiple effects. GPx-1 (Pro198Leu) polymorphism is associated with Se deficiency. This study aimed to observe associations between Se-deficiency/supplement and GPx-1-198Leu overexpression in myocardial injuries. Material/Methods GPx-1P198L transgenic (Tg) mice and non-transgenic wild-type (WT) littermates were divided into Control (CON, 0.1–0.2 mg/kg), Se-deficiency (SD, <0.02 mg/kg), and Se-supplement (SS, 0.4 mg/kg) groups. Cardiac functions were observed with animal M-mode echocardiography. Se level was measured using 2,3-diamino Kenai fluorospectrophotometry. Total cardiac GPx activity was also measured. Myocardial histopathology was determined with HE and Masson’s trichrome staining. Caspase-9 and caspase-3 were measured with Western blot analysis. Results In WT Se-deficient mice, cardiac GPx activity was significantly decreased, and was not elevated by overexpression of GPx-1-198Leu gene. Increased GPx activity was observed in WT Se-supplemented mice and Tg Se-supplemented mice (much more). Se deficiency as well as supplementation resulted in cardiac systolic dysfunction, which was not affected by GPx-1-198Leu gene. Se deficiency led to myocardial fibrosis and pathological changes accompanied by increased activation of caspase-9 and caspase-3. Se supplementation significantly reduced pathological changes, as well as caspase-9 and caspase-3 levels in the presence of increased myocardial fibrosis. In Se-deficient mice, GPx-1-198Leu overexpression did not significantly decrease myocardial pathological injuries and fibrosis. In Se-supplemented Tg mice, myocardial fibrosis and caspase-9 level were increased, although pathological injuries and caspase-3 were similar to that in Se-supplemented WT mice. Conclusions Se deficiency as well as supplementation induced myocardial structural and functional abnormalities through activation of caspase-9 and caspase-3 in GPx-1P198L overexpression transgenic mice.
Collapse
Affiliation(s)
- Suqin Wang
- Department of Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China (mainland).,Department of Cardiology, People's Hospital of Henan Province, Zhengzhou, Henan, China (mainland)
| | - Xiting Nong
- Department of Endocrinology, Xi'an Central Hospital, Xi'an, Shaanxi, China (mainland)
| | - Guang Yang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
29
|
Lismont C, Nordgren M, Brees C, Knoops B, Van Veldhoven PP, Fransen M. Peroxisomes as Modulators of Cellular Protein Thiol Oxidation: A New Model System. Antioxid Redox Signal 2019; 30:22-39. [PMID: 28594286 DOI: 10.1089/ars.2017.6997] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS Peroxisomes are ubiquitous, single-membrane-bounded organelles that contain considerable amounts of enzymes involved in the production or breakdown of hydrogen peroxide (H2O2), a key signaling molecule in multiple biological processes and disease states. Despite this, the role of this organelle in cross-compartmental H2O2 signaling remains largely unclear, mainly because of the difficulty to modulate peroxisomal H2O2 production in a selective manner. This study aimed at establishing and validating a cellular model suitable to decipher the complex signaling processes associated with peroxisomal H2O2 release. RESULTS Here, we report the development of a human cell line that can be used to selectively generate H2O2 inside peroxisomes in a time- and dose-controlled manner. In addition, we provide evidence that peroxisome-derived H2O2 can oxidize redox-sensitive cysteine residues in multiple proteins within (e.g., peroxiredoxin-5 [PRDX5]) and outside (e.g., nuclear factor kappa B subunit 1 [NFKB1] and subunit RELA proto-oncogene [RELA], phosphatase and tensin homolog [PTEN], forkhead box O3 [FOXO3], and peroxin 5 [PEX5]) the peroxisomal compartment. Furthermore, we show that the extent of protein oxidation depends on the subcellular location of the target protein and is inversely correlated to catalase activity and cellular glutathione content. Finally, we demonstrate that excessive H2O2 production inside peroxisomes does not induce their selective degradation, at least not under the conditions examined. INNOVATION This study describes for the first time a powerful model system that can be used to examine the role of peroxisome-derived H2O2 in redox-regulated (patho)physiological processes, a research area in need of further investigation and innovative approaches. CONCLUSION Our results provide unambiguous evidence that peroxisomes can serve as regulatory hubs in thiol-based signaling networks.
Collapse
Affiliation(s)
- Celien Lismont
- 1 Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven , Leuven, Belgium
| | - Marcus Nordgren
- 1 Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven , Leuven, Belgium
| | - Chantal Brees
- 1 Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven , Leuven, Belgium
| | - Bernard Knoops
- 2 Group of Animal Molecular and Cellular Biology, Institut des Sciences de la Vie (ISV), Université catholique de Louvain , Louvain-la-Neuve, Belgium
| | - Paul P Van Veldhoven
- 1 Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven , Leuven, Belgium
| | - Marc Fransen
- 1 Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven , Leuven, Belgium
| |
Collapse
|
30
|
Fransen M, Lismont C. Redox Signaling from and to Peroxisomes: Progress, Challenges, and Prospects. Antioxid Redox Signal 2019; 30:95-112. [PMID: 29433327 DOI: 10.1089/ars.2018.7515] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Peroxisomes are organelles that are best known for their role in cellular lipid and hydrogen peroxide (H2O2) metabolism. Emerging evidence suggests that these organelles serve as guardians and modulators of cellular redox balance, and that alterations in their redox metabolism may contribute to aging and the development of chronic diseases such as neurodegeneration, diabetes, and cancer. Recent Advances: H2O2 is an important signaling messenger that controls many cellular processes by modulating protein activity through cysteine oxidation. Somewhat surprisingly, the potential involvement of peroxisomes in H2O2-mediated signaling processes has been overlooked for a long time. However, recent advances in the development of live-cell approaches to monitor and modulate spatiotemporal fluxes in redox species at the subcellular level have opened up new avenues for research in redox biology and boosted interest in the concept of peroxisomes as redox signaling platforms. CRITICAL ISSUES This review first introduces the reader to what is known about the role of peroxisomes in cellular H2O2 production and clearance, with a focus on mammalian cells. Next, it briefly describes the benefits and drawbacks of current strategies used to investigate the complex interplay between peroxisome metabolism and cellular redox state. Furthermore, it integrates and critically evaluates literature dealing with the interrelationship between peroxisomal redox metabolism, cell signaling, and human disease. FUTURE DIRECTIONS As the precise molecular mechanisms underlying many of these associations are still poorly understood, a key focus for future research should be the identification of primary targets for peroxisome-derived H2O2.
Collapse
Affiliation(s)
- Marc Fransen
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven , Leuven, Belgium
| | - Celien Lismont
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven , Leuven, Belgium
| |
Collapse
|
31
|
Haworth NL, Wouters MJ, Hunter MO, Ma L, Wouters MA. Cross-strand disulfides in the hydrogen bonding site of antiparallel β-sheet (aCSDhs): Forbidden disulfides that are highly strained, easily broken. Protein Sci 2018; 28:239-256. [PMID: 30383331 DOI: 10.1002/pro.3545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/30/2018] [Accepted: 10/30/2018] [Indexed: 12/16/2022]
Abstract
Some disulfide bonds perform important structural roles in proteins, but another group has functional roles via redox reactions. Forbidden disulfides are stressed disulfides found in recognizable protein contexts, which currently constitute more than 10% of all disulfides in the PDB. They likely have functional redox roles and constitute a major subset of all redox-active disulfides. The torsional strain of forbidden disulfides is typically higher than for structural disulfides, but not so high as to render them immediately susceptible to reduction under physionormal conditions. Previously we characterized the most abundant forbidden disulfide in the Protein Data Bank, the aCSDn: a canonical motif in which disulfide-bonded cysteine residues are positioned directly opposite each other on adjacent anti-parallel β-strands such that the backbone hydrogen-bonded moieties are directed away from each other. Here we perform a similar analysis for the aCSDh, a less common motif in which the opposed cysteine residues are backbone hydrogen bonded. Oxidation of two Cys in this context places significant strain on the protein system, with the β-chains tilting toward each other to allow disulfide formation. Only left-handed aCSDh conformations are compatible with the inherent right-handed twist of β-sheets. aCSDhs tend to be more highly strained than aCSDns, particularly when both hydrogen bonds are formed. We discuss characterized roles of aCSDh motifs in proteins of the dataset, which include catalytic disulfides in ribonucleotide reductase and ahpC peroxidase as well as a redox-active disulfide in P1 lysozyme, involved in a major conformation change. The dataset also includes many binding proteins.
Collapse
Affiliation(s)
- Naomi L Haworth
- Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia.,Structural & Computational Biology Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Michael J Wouters
- Electricity Section, National Measurement Institute, Lindfield, New South Wales, Australia
| | - Morgan O Hunter
- Bioinformatics, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Lixia Ma
- School of Statistics, Henan University of Economics and Law, Henan Province, China
| | - Merridee A Wouters
- Bioinformatics, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia.,Cancer Data Science, Children's Medical Research Institute, Westmead, New South Wales, Australia
| |
Collapse
|
32
|
Wang RS, Oldham WM, Maron BA, Loscalzo J. Systems Biology Approaches to Redox Metabolism in Stress and Disease States. Antioxid Redox Signal 2018; 29:953-972. [PMID: 29121773 PMCID: PMC6104248 DOI: 10.1089/ars.2017.7256] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 10/12/2017] [Accepted: 11/04/2017] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE All cellular metabolic processes are tied to the cellular redox environment. Therefore, maintaining redox homeostasis is critically important for normal cell function. Indeed, redox stress contributes to the pathobiology of many human diseases. The cellular redox response system is composed of numerous interconnected components, including free radicals, redox couples, protein thiols, enzymes, metabolites, and transcription factors. Moreover, interactions between and among these factors are regulated in time and space. Owing to their complexity, systems biology approaches to the characterization of the cellular redox response system may provide insights into novel homeostatic mechanisms and methods of therapeutic reprogramming. Recent Advances: The emergence and development of systems biology has brought forth a set of innovative technologies that provide new avenues for studying redox metabolism. This article will review these systems biology approaches and their potential application to the study of redox metabolism in stress and disease states. CRITICAL ISSUES Clarifying the scope of biological intermediaries affected by dysregulated redox metabolism requires methods that are suitable for analyzing big datasets as classical methods that do not account for multiple interactions are unlikely to portray the totality of perturbed metabolic systems. FUTURE DIRECTIONS Given the diverse redox microenvironments within cells, it will be important to improve the spatial resolution of omic approaches. Futures studies on the integration of multiple systems-based methods and heterogeneous omics data for redox metabolism are required to accelerate the development of the field of redox systems biology. Antioxid. Redox Signal. 29, 953-972.
Collapse
Affiliation(s)
- Rui-Sheng Wang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - William M. Oldham
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Bradley A. Maron
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Section of Cardiology, Veterans Affairs Boston Healthcare System, West Roxbury, Massachusetts
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
33
|
Affiliation(s)
- Joseph Loscalzo
- From the Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
34
|
Simmen T, Herrera-Cruz MS. Plastic mitochondria-endoplasmic reticulum (ER) contacts use chaperones and tethers to mould their structure and signaling. Curr Opin Cell Biol 2018; 53:61-69. [DOI: 10.1016/j.ceb.2018.04.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/10/2018] [Accepted: 04/30/2018] [Indexed: 12/19/2022]
|
35
|
Epigenetics of Subcellular Structure Functioning in the Origin of Risk or Resilience to Comorbidity of Neuropsychiatric and Cardiometabolic Disorders. Int J Mol Sci 2018; 19:ijms19051456. [PMID: 29757967 PMCID: PMC5983601 DOI: 10.3390/ijms19051456] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/04/2018] [Accepted: 05/09/2018] [Indexed: 12/31/2022] Open
Abstract
Mechanisms controlling mitochondrial function, protein folding in the endoplasmic reticulum (ER) and nuclear processes such as telomere length and DNA repair may be subject to epigenetic cues that relate the genomic expression and environmental exposures in early stages of life. They may also be involved in the comorbid appearance of cardiometabolic (CMD) and neuropsychiatric disorders (NPD) during adulthood. Mitochondrial function and protein folding in the endoplasmic reticulum are associated with oxidative stress and elevated intracellular calcium levels and may also underlie the vulnerability for comorbid CMD and NPD. Mitochondria provide key metabolites such as nicotinamide adenine dinucleotide (NAD+), ATP, α-ketoglutarate and acetyl coenzyme A that are required for many transcriptional and epigenetic processes. They are also a source of free radicals. On the other hand, epigenetic markers in nuclear DNA determine mitochondrial biogenesis. The ER is the subcellular organelle in which secretory proteins are folded. Many environmental factors stop the ability of cells to properly fold proteins and modify post-translationally secretory and transmembrane proteins leading to endoplasmic reticulum stress and oxidative stress. ER functioning may be epigenetically determined. Chronic ER stress is emerging as a key contributor to a growing list of human diseases, including CMD and NPD. Telomere loss causes chromosomal fusion, activation of the control of DNA damage-responses, unstable genome and altered stem cell function, which may underlie the comorbidity of CMD and NPD. The length of telomeres is related to oxidative stress and may be epigenetically programmed. Pathways involved in DNA repair may be epigenetically programmed and may contribute to diseases. In this paper, we describe subcellular mechanisms that are determined by epigenetic markers and their possible relation to the development of increased susceptibility to develop CMD and NPD.
Collapse
|
36
|
Redox crosstalk at endoplasmic reticulum (ER) membrane contact sites (MCS) uses toxic waste to deliver messages. Cell Death Dis 2018; 9:331. [PMID: 29491367 PMCID: PMC5832433 DOI: 10.1038/s41419-017-0033-4] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/29/2017] [Accepted: 10/03/2017] [Indexed: 12/17/2022]
Abstract
Many cellular redox reactions housed within mitochondria, peroxisomes and the endoplasmic reticulum (ER) generate hydrogen peroxide (H2O2) and other reactive oxygen species (ROS). The contribution of each organelle to the total cellular ROS production is considerable, but varies between cell types and also over time. Redox-regulatory enzymes are thought to assemble at a “redox triangle” formed by mitochondria, peroxisomes and the ER, assembling “redoxosomes” that sense ROS accumulations and redox imbalances. The redoxosome enzymes use ROS, potentially toxic by-products made by some redoxosome members themselves, to transmit inter-compartmental signals via chemical modifications of downstream proteins and lipids. Interestingly, important components of the redoxosome are ER chaperones and oxidoreductases, identifying ER oxidative protein folding as a key ROS producer and controller of the tri-organellar membrane contact sites (MCS) formed at the redox triangle. At these MCS, ROS accumulations could directly facilitate inter-organellar signal transmission, using ROS transporters. In addition, ROS influence the flux of Ca2+ ions, since many Ca2+ handling proteins, including inositol 1,4,5 trisphosphate receptors (IP3Rs), SERCA pumps or regulators of the mitochondrial Ca2+ uniporter (MCU) are redox-sensitive. Fine-tuning of these redox and ion signaling pathways might be difficult in older organisms, suggesting a dysfunctional redox triangle may accompany the aging process.
Collapse
|
37
|
Nrf2-Keap1 signaling in oxidative and reductive stress. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:721-733. [PMID: 29499228 DOI: 10.1016/j.bbamcr.2018.02.010] [Citation(s) in RCA: 1221] [Impact Index Per Article: 174.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/25/2018] [Accepted: 02/22/2018] [Indexed: 02/07/2023]
Abstract
Nrf2 and its endogenous inhibitor, Keap1, function as a ubiquitous, evolutionarily conserved intracellular defense mechanism to counteract oxidative stress. Sequestered by cytoplasmic Keap1 and targeted to proteasomal degradation in basal conditions, in case of oxidative stress Nrf2 detaches from Keap1 and translocates to the nucleus, where it heterodimerizes with one of the small Maf proteins. The heterodimers recognize the AREs, that are enhancer sequences present in the regulatory regions of Nrf2 target genes, essential for the recruitment of key factors for transcription. In the present review we briefly introduce the Nrf2-Keap1 system and describe Nrf2 functions, illustrate the Nrf2-NF-κB cross-talk, and highlight the effects of the Nrf2-Keap1 system in the physiology and pathophysiology of striated muscle tissue taking into account its role(s) in oxidative stress and reductive stress.
Collapse
|
38
|
Abstract
Human immunodeficiency virus-1 (HIV-1) is known to interact with multiple host cellular proteins during its replication in the target cell. While many of these host cellular proteins facilitate viral replication, a number of them are reported to inhibit HIV-1 replication at various stages of its life cycle. These host cellular proteins, which are known as restriction factors, constitute an integral part of the host's first line of defence against the viral pathogen. Since the discovery of apolipoprotein B mRNA-editing enzyme 3G (APOBEC3G) as an HIV-1 restriction factor, several human proteins have been identified that exhibit anti-HIV-1 restriction. While each restriction factor employs a distinct mechanism of inhibition, the HIV-1 virus has equally evolved complex counter strategies to neutralize their inhibitory effect. APOBEC3G, tetherin, sterile alpha motif and histidine-aspartate domain 1 (SAMHD1), and trim-5α are some of the best known HIV-1 restriction factors that have been studied in great detail. Recently, six novel restriction factors were discovered that exhibit significant antiviral activity: endoplasmic reticulum α1,2-mannosidase I (ERManI), translocator protein (TSPO), guanylate-binding protein 5 (GBP5), serine incorporator (SERINC3/5) and zinc-finger antiviral protein (ZAP). The focus of this review is to discuss the antiviral mechanism of action of these six restriction factors and provide insights into the probable counter-evasion strategies employed by the HIV-1 virus. The recent discovery of new restriction factors substantiates the complex host-pathogen interactions occurring during HIV-1 pathogenesis and makes it imperative that further investigations are conducted to elucidate the molecular basis of HIV-1 replication.
Collapse
Affiliation(s)
- Dibya Ghimire
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi 110021, India
| | - Madhu Rai
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi 110021, India
| | - Ritu Gaur
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi 110021, India
| |
Collapse
|
39
|
Peroxisomes and Cellular Oxidant/Antioxidant Balance: Protein Redox Modifications and Impact on Inter-organelle Communication. Subcell Biochem 2018; 89:435-461. [PMID: 30378035 DOI: 10.1007/978-981-13-2233-4_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Disturbances in cellular redox balance have been associated with pro-aging mechanisms and increased risk for various chronic disease states. Multiple lines of evidence indicate that peroxisomes are central players in cellular redox metabolism. Nevertheless, the potential role of this organelle as intracellular redox signaling platform has been largely overlooked for a long time. Fortunately, this situation is now changing. This review provides a snapshot of the current progress in the field, with an emphasis on the situation in mammals. We first briefly introduce the basics of redox biology and how reactive oxygen and nitrogen species can drive cellular signaling events. Next, we discuss current evidence linking peroxisome (dys)function to redox signaling, both in health and disease. We also highlight what is currently known about the downstream targets of peroxisome-derived oxidants. In addition, we present an extensive list of proteins that are involved in peroxisome functioning and have been identified as being responsive to oxidative stress in large scale redox proteomics studies. Finally, we address how changes in peroxisomal redox state may impact on functional mechanisms underlying inter-organelle communication. Gaining more insight into these mechanisms is key to our understanding of how peroxisomes are embedded in cellular signaling networks implicated in aging and diseases such as cancer, diabetes, and neurodegenerative disorders.
Collapse
|
40
|
Walker CL, Pomatto LCD, Tripathi DN, Davies KJA. Redox Regulation of Homeostasis and Proteostasis in Peroxisomes. Physiol Rev 2018; 98:89-115. [PMID: 29167332 PMCID: PMC6335096 DOI: 10.1152/physrev.00033.2016] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 06/19/2017] [Accepted: 06/21/2017] [Indexed: 02/08/2023] Open
Abstract
Peroxisomes are highly dynamic intracellular organelles involved in a variety of metabolic functions essential for the metabolism of long-chain fatty acids, d-amino acids, and many polyamines. A byproduct of peroxisomal metabolism is the generation, and subsequent detoxification, of reactive oxygen and nitrogen species, particularly hydrogen peroxide (H2O2). Because of its relatively low reactivity (as a mild oxidant), H2O2 has a comparatively long intracellular half-life and a high diffusion rate, all of which makes H2O2 an efficient signaling molecule. Peroxisomes also have intricate connections to mitochondria, and both organelles appear to play important roles in regulating redox signaling pathways. Peroxisomal proteins are also subject to oxidative modification and inactivation by the reactive oxygen and nitrogen species they generate, but the peroxisomal LonP2 protease can selectively remove such oxidatively damaged proteins, thus prolonging the useful lifespan of the organelle. Peroxisomal homeostasis must adapt to the metabolic state of the cell, by a combination of peroxisome proliferation, the removal of excess or badly damaged organelles by autophagy (pexophagy), as well as by processes of peroxisome inheritance and motility. More recently the tumor suppressors ataxia telangiectasia mutate (ATM) and tuberous sclerosis complex (TSC), which regulate mTORC1 signaling, have been found to regulate pexophagy in response to variable levels of certain reactive oxygen and nitrogen species. It is now clear that any significant loss of peroxisome homeostasis can have devastating physiological consequences. Peroxisome dysregulation has been implicated in several metabolic diseases, and increasing evidence highlights the important role of diminished peroxisomal functions in aging processes.
Collapse
Affiliation(s)
- Cheryl L Walker
- Center for Precision Environmental Health and Departments of Molecular & Cellular Biology and Medicine, Baylor College of Medicine, Houston, Texas; and Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center and Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, The University of Southern California, Los Angeles, California
| | - Laura C D Pomatto
- Center for Precision Environmental Health and Departments of Molecular & Cellular Biology and Medicine, Baylor College of Medicine, Houston, Texas; and Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center and Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, The University of Southern California, Los Angeles, California
| | - Durga Nand Tripathi
- Center for Precision Environmental Health and Departments of Molecular & Cellular Biology and Medicine, Baylor College of Medicine, Houston, Texas; and Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center and Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, The University of Southern California, Los Angeles, California
| | - Kelvin J A Davies
- Center for Precision Environmental Health and Departments of Molecular & Cellular Biology and Medicine, Baylor College of Medicine, Houston, Texas; and Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center and Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, The University of Southern California, Los Angeles, California
| |
Collapse
|
41
|
Reductive Stress in Inflammation-Associated Diseases and the Pro-Oxidant Effect of Antioxidant Agents. Int J Mol Sci 2017; 18:ijms18102098. [PMID: 28981461 PMCID: PMC5666780 DOI: 10.3390/ijms18102098] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/16/2017] [Accepted: 09/30/2017] [Indexed: 12/18/2022] Open
Abstract
Abstract: Reductive stress (RS) is the counterpart oxidative stress (OS), and can occur in response to conditions that shift the redox balance of important biological redox couples, such as the NAD⁺/NADH, NADP⁺/NADPH, and GSH/GSSG, to a more reducing state. Overexpression of antioxidant enzymatic systems leads to excess reducing equivalents that can deplete reactive oxidative species, driving the cells to RS. A feedback regulation is established in which chronic RS induces OS, which in turn, stimulates again RS. Excess reducing equivalents may regulate cellular signaling pathways, modify transcriptional activity, induce alterations in the formation of disulfide bonds in proteins, reduce mitochondrial function, decrease cellular metabolism, and thus, contribute to the development of some diseases in which NF-κB, a redox-sensitive transcription factor, participates. Here, we described the diseases in which an inflammatory condition is associated to RS, and where delayed folding, disordered transport, failed oxidation, and aggregation are found. Some of these diseases are aggregation protein cardiomyopathy, hypertrophic cardiomyopathy, muscular dystrophy, pulmonary hypertension, rheumatoid arthritis, Alzheimer's disease, and metabolic syndrome, among others. Moreover, chronic consumption of antioxidant supplements, such as vitamins and/or flavonoids, may have pro-oxidant effects that may alter the redox cellular equilibrium and contribute to RS, even diminishing life expectancy.
Collapse
|
42
|
Osuagwuh UI, Palomo MJ. Effect of semen washing on thawed ram spermatozoa subjected to a four hour post-thawing thermal evaluation test. Small Rumin Res 2017. [DOI: 10.1016/j.smallrumres.2017.09.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
43
|
Tanaka LY, Laurindo FRM. Vascular remodeling: A redox-modulated mechanism of vessel caliber regulation. Free Radic Biol Med 2017; 109:11-21. [PMID: 28109889 DOI: 10.1016/j.freeradbiomed.2017.01.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 01/13/2017] [Accepted: 01/15/2017] [Indexed: 11/17/2022]
Abstract
Vascular remodeling, i.e. whole-vessel structural reshaping, determines lumen caliber in (patho)physiology. Here we review mechanisms underlying vessel remodeling, with emphasis in redox regulation. First, we discuss confusing terminology and focus on strictu sensu remodeling. Second, we propose a mechanobiological remodeling paradigm based on the concept of tensional homeostasis as a setpoint regulator. We first focus on shear-mediated models as prototypes of remodeling closely dominated by highly redox-sensitive endothelial function. More detailed discussions focus on mechanosensors, integrins, extracellular matrix, cytoskeleton and inflammatory pathways as potential of mechanisms potentially coupling tensional homeostasis to redox regulation. Further discussion of remodeling associated with atherosclerosis and injury repair highlights important aspects of redox vascular responses. While neointima formation has not shown consistent responsiveness to antioxidants, vessel remodeling has been more clearly responsive, indicating that despite the multilevel redox signaling pathways, there is a coordinated response of the whole vessel. Among mechanisms that may orchestrate redox pathways, we discuss roles of superoxide dismutase activity and extracellular protein disulfide isomerase. We then discuss redox modulation of aneurysms, a special case of expansive remodeling. We propose that the redox modulation of vascular remodeling may reflect (1) remodeling pathophysiology is dominated by a particularly redox-sensitive cell type, e.g., endothelial cells (2) redox pathways are temporospatially coordinated at an organ level across distinct cellular and acellular structures or (3) the tensional homeostasis setpoint is closely connected to redox signaling. The mechanobiological/redox model discussed here can be a basis for improved understanding of remodeling and helps clarifying mechanisms underlying prevalent hard-to-treat diseases.
Collapse
Affiliation(s)
- Leonardo Y Tanaka
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, Av. Enéas Carvalho Aguiar, 44, Annex II, 9th Floor, São Paulo CEP 05403-000, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, Av. Enéas Carvalho Aguiar, 44, Annex II, 9th Floor, São Paulo CEP 05403-000, Brazil.
| |
Collapse
|
44
|
Handy DE, Loscalzo J. Responses to reductive stress in the cardiovascular system. Free Radic Biol Med 2017; 109:114-124. [PMID: 27940350 PMCID: PMC5462861 DOI: 10.1016/j.freeradbiomed.2016.12.006] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 11/29/2016] [Accepted: 12/03/2016] [Indexed: 12/13/2022]
Abstract
There is a growing appreciation that reductive stress represents a disturbance in the redox state that is harmful to biological systems. On a cellular level, the presence of increased reducing equivalents and the lack of beneficial fluxes of reactive oxygen species can prevent growth factor-mediated signaling, promote mitochondrial dysfunction, increase apoptosis, and decrease cell survival. In this review, we highlight the importance of redox balance in maintaining cardiovascular homeostasis and consider the tenuous balance between oxidative and reductive stress. We explain the role of reductive stress in models of protein aggregation-induced cardiomyopathies, such as those caused by mutations in αB-crystallin. In addition, we discuss the role of NADPH oxidases in models of heart failure and ischemia-reperfusion to illustrate how oxidants may mediate the adaptive responses to injury. NADPH oxidase 4, a hydrogen peroxide generator, also has a major role in promoting vascular homeostasis through its regulation of vascular tone, angiogenic responses, and effects on atherogenesis. In contrast, the lack of antioxidant enzymes that reduce hydrogen peroxide, such as glutathione peroxidase 1, promotes vascular remodeling and is deleterious to endothelial function. Thus, we consider the role of oxidants as necessary signals to promote adaptive responses, such as the activation of Nrf2 and eNOS, and the stabilization of Hif1. In addition, we discuss the adaptive metabolic reprogramming in hypoxia that lead to a reductive state, and the subsequent cellular redistribution of reducing equivalents from NADH to other metabolites. Finally, we discuss the paradoxical ability of excess reducing equivalents to stimulate oxidative stress and promote injury.
Collapse
Affiliation(s)
- Diane E Handy
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, USA.
| |
Collapse
|
45
|
Sun MA, Wang Y, Zhang Q, Xia Y, Ge W, Guo D. Prediction of reversible disulfide based on features from local structural signatures. BMC Genomics 2017; 18:279. [PMID: 28376774 PMCID: PMC5379614 DOI: 10.1186/s12864-017-3668-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 03/28/2017] [Indexed: 11/12/2022] Open
Abstract
Background Disulfide bonds are traditionally considered to play only structural roles. In recent years, increasing evidence suggests that the disulfide proteome is made up of structural disulfides and reversible disulfides. Unlike structural disulfides, reversible disulfides are usually of important functional roles and may serve as redox switches. Interestingly, only specific disulfide bonds are reversible while others are not. However, whether reversible disulfides can be predicted based on structural information remains largely unknown. Methods In this study, two datasets with both types of disulfides were compiled using independent approaches. By comparison of various features extracted from the local structural signatures, we identified several features that differ significantly between reversible and structural disulfides, including disulfide bond length, along with the number, amino acid composition, secondary structure and physical-chemical properties of surrounding amino acids. A SVM-based classifier was developed for predicting reversible disulfides. Results By 10-fold cross-validation, the model achieved accuracy of 0.750, sensitivity of 0.352, specificity of 0.953, MCC of 0.405 and AUC of 0.751 using the RevSS_PDB dataset. The robustness was further validated by using RevSS_RedoxDB as independent testing dataset. This model was applied to proteins with known structures in the PDB database. The results show that one third of the predicted reversible disulfide containing proteins are well-known redox enzymes, while the remaining are non-enzyme proteins. Given that reversible disulfides are frequently reported from functionally important non-enzyme proteins such as transcription factors, the predictions may provide valuable candidates of novel reversible disulfides for further experimental investigation. Conclusions This study provides the first comparative analysis between the reversible and the structural disulfides. Distinct features remarkably different between these two groups of disulfides were identified, and a SVM-based classifier for predicting reversible disulfides was developed accordingly. A web server named RevssPred can be accessed freely from: http://biocomputer.bio.cuhk.edu.hk/RevssPred. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3668-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ming-An Sun
- School of Life Sciences and the State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, SAR, China
| | - Yejun Wang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Shenzhen University Health Science Center, Nanhai Ave 3688, Shenzhen, 518060, China
| | - Qing Zhang
- School of Life Sciences and the State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, SAR, China
| | - Yiji Xia
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong, SAR, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Dianjing Guo
- School of Life Sciences and the State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, SAR, China.
| |
Collapse
|
46
|
Kaneko C, Ogura J, Sasaki S, Okamoto K, Kobayashi M, Kuwayama K, Narumi K, Iseki K. Fructose suppresses uric acid excretion to the intestinal lumen as a result of the induction of oxidative stress by NADPH oxidase activation. Biochim Biophys Acta Gen Subj 2016; 1861:559-566. [PMID: 27913188 DOI: 10.1016/j.bbagen.2016.11.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/08/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND A high intake of fructose increases the risk for hyperuricemia. It has been reported that long-term fructose consumption suppressed renal uric acid excretion and increased serum uric acid level. However, the effect of single administration of fructose on excretion of uric acid has not been clarified. METHODS We used male Wistar rats, which were orally administered fructose (5g/kg). Those rats were used in each experiment at 12h after administration. RESULTS Single administration of fructose suppressed the function of ileal uric acid excretion and had no effect on the function of renal uric acid excretion. Breast cancer resistance protein (BCRP) predominantly contributes to intestinal excretion of uric acid as an active homodimer. Single administration of fructose decreased BCRP homodimer level in the ileum. Moreover, diphenyleneiodonium (DPI), an inhibitor of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox), recovered the suppression of the function of ileal uric acid excretion and the Bcrp homodimer level in the ileum of rats that received single administration of fructose. CONCLUSIONS Single administration of fructose decreases in BCRP homodimer level, resulting in the suppression the function of ileal uric acid excretion. The suppression of the function of ileal uric acid excretion by single administration of fructose is caused by the activation of Nox. The results of our study provide a new insight into the mechanism of fructose-induced hyperuricemia.
Collapse
Affiliation(s)
- Chihiro Kaneko
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Jiro Ogura
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Shunichi Sasaki
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Keisuke Okamoto
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Masaki Kobayashi
- Department of Pharmacy, Hokkaido University Hospital, Kita-14-jo, Nishi-5-chome, Kita-ku, Sapporo 060-8648, Japan.
| | - Kaori Kuwayama
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Katsuya Narumi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Ken Iseki
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan; Department of Pharmacy, Hokkaido University Hospital, Kita-14-jo, Nishi-5-chome, Kita-ku, Sapporo 060-8648, Japan.
| |
Collapse
|
47
|
Wang Y, Zhong Y, Wang Q, Yang XF, Li Z, Li H. Ratiometric Fluorescent Probe for Vicinal Dithiol-Containing Proteins in Living Cells Designed via Modulating the Intramolecular Charge Transfer–Twisted Intramolecular Charge Transfer Conversion Process. Anal Chem 2016; 88:10237-10244. [DOI: 10.1021/acs.analchem.6b02923] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Yuanyuan Wang
- Key
Laboratory of Synthetic and Natural Functional Molecule Chemistry
of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi’an, Shaanxi 710127, P. R. China
| | - Yaogang Zhong
- College
of Life Sciences, Northwest University, Xi’an, Shaanxi 710069, P. R. China
| | - Qin Wang
- Key
Laboratory of Synthetic and Natural Functional Molecule Chemistry
of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi’an, Shaanxi 710127, P. R. China
| | - Xiao-Feng Yang
- Key
Laboratory of Synthetic and Natural Functional Molecule Chemistry
of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi’an, Shaanxi 710127, P. R. China
| | - Zheng Li
- College
of Life Sciences, Northwest University, Xi’an, Shaanxi 710069, P. R. China
| | - Hua Li
- Key
Laboratory of Synthetic and Natural Functional Molecule Chemistry
of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi’an, Shaanxi 710127, P. R. China
- College
of Chemistry and Chemical Engineering, Xi’an Shiyou University, Xi’an, Shaanxi 710065, P. R. China
| |
Collapse
|
48
|
Zhou T, Frabutt DA, Moremen KW, Zheng YH. ERManI (Endoplasmic Reticulum Class I α-Mannosidase) Is Required for HIV-1 Envelope Glycoprotein Degradation via Endoplasmic Reticulum-associated Protein Degradation Pathway. J Biol Chem 2015. [PMID: 26205822 DOI: 10.1074/jbc.m115.675207] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previously, we reported that the mitochondrial translocator protein (TSPO) induces HIV-1 envelope (Env) degradation via the endoplasmic reticulum (ER)-associated protein degradation (ERAD) pathway, but the mechanism was not clear. Here we investigated how the four ER-associated glycoside hydrolase family 47 (GH47) α-mannosidases, ERManI, and ER-degradation enhancing α-mannosidase-like (EDEM) proteins 1, 2, and 3, are involved in the Env degradation process. Ectopic expression of these four α-mannosidases uncovers that only ERManI inhibits HIV-1 Env expression in a dose-dependent manner. In addition, genetic knock-out of the ERManI gene MAN1B1 using CRISPR/Cas9 technology disrupts the TSPO-mediated Env degradation. Biochemical studies show that HIV-1 Env interacts with ERManI, and between the ERManI cytoplasmic, transmembrane, lumenal stem, and lumenal catalytic domains, the catalytic domain plays a critical role in the Env-ERManI interaction. In addition, functional studies show that inactivation of the catalytic sites by site-directed mutagenesis disrupts the ERManI activity. These studies identify ERManI as a critical GH47 α-mannosidase in the ER-associated protein degradation pathway that initiates the Env degradation and suggests that its catalytic domain and enzymatic activity play an important role in this process.
Collapse
Affiliation(s)
- Tao Zhou
- From the Harbin Veterinary Research Institute, CAAS-Michigan State University Joint Laboratory of Innate Immunity, State Key Laboratory of Veterinary Biotechnology, Chinese Academy of Agricultural Sciences, Harbin, 150001, China, BEACON Center for the Study of Evolution in Action and Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824
| | - Dylan A Frabutt
- BEACON Center for the Study of Evolution in Action and Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602, and
| | - Yong-Hui Zheng
- From the Harbin Veterinary Research Institute, CAAS-Michigan State University Joint Laboratory of Innate Immunity, State Key Laboratory of Veterinary Biotechnology, Chinese Academy of Agricultural Sciences, Harbin, 150001, China, BEACON Center for the Study of Evolution in Action and Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
49
|
Ogura J, Kuwayama K, Sasaki S, Kaneko C, Koizumi T, Yabe K, Tsujimoto T, Takeno R, Takaya A, Kobayashi M, Yamaguchi H, Iseki K. Reactive oxygen species derived from xanthine oxidase interrupt dimerization of breast cancer resistance protein, resulting in suppression of uric acid excretion to the intestinal lumen. Biochem Pharmacol 2015; 97:89-98. [PMID: 26119820 DOI: 10.1016/j.bcp.2015.06.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/22/2015] [Indexed: 02/04/2023]
Abstract
The prevalence of hyperuricemia/gout increases with aging. However, the effect of aging on function for excretion of uric acid to out of the body has not been clarified. We found that ileal uric acid clearance in middle-aged rats (11-12 months) was decreased compared with that in young rats (2 months). In middle-aged rats, xanthine oxidase (XO) activity in the ileum was significantly higher than that in young rats. Inosine-induced reactive oxygen species (ROS), which are derived from XO, also decreased ileal uric acid clearance. ROS derived from XO decreased the active homodimer level of breast cancer resistance protein (BCRP), which is a uric acid efflux transporter, in the ileum. Pre-administration of allopurinol recovered the BCRP homodimer level, resulting in the recovering ileal uric acid clearance. Moreover, we investigated the effects of ROS derived from XO on BCRP homodimer level directly in Caco-2 cells using hypoxanthine. Treatment with hypoxanthine decreased BCRP homodimer level. Treatment with hypoxanthine induced mitochondrial dysfunction, suggesting that the decreasing BCRP homodimer level might be caused by mitochondrial dysfunction. In conclusion, ROS derived from XO decrease BCRP homodimer level, resulting in suppression of function for uric acid excretion to the ileal lumen. ROS derived from XO may cause the suppression of function of the ileum for the excretion of uric acid with aging. The results of our study provide a new insight into the causes of increasing hyperuricemia/gout prevalence with aging.
Collapse
Affiliation(s)
- Jiro Ogura
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Kaori Kuwayama
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Shunichi Sasaki
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Chihiro Kaneko
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Takahiro Koizumi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Keisuke Yabe
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Takashi Tsujimoto
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Reiko Takeno
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Atsushi Takaya
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Hiroaki Yamaguchi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Ken Iseki
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
50
|
Liemburg-Apers DC, Willems PHGM, Koopman WJH, Grefte S. Interactions between mitochondrial reactive oxygen species and cellular glucose metabolism. Arch Toxicol 2015; 89:1209-26. [PMID: 26047665 PMCID: PMC4508370 DOI: 10.1007/s00204-015-1520-y] [Citation(s) in RCA: 269] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 04/27/2015] [Indexed: 12/20/2022]
Abstract
Mitochondrial reactive oxygen species (ROS) production and detoxification are tightly balanced. Shifting this balance enables ROS to activate intracellular signaling and/or induce cellular damage and cell death. Increased mitochondrial ROS production is observed in a number of pathological conditions characterized by mitochondrial dysfunction. One important hallmark of these diseases is enhanced glycolytic activity and low or impaired oxidative phosphorylation. This suggests that ROS is involved in glycolysis (dys)regulation and vice versa. Here we focus on the bidirectional link between ROS and the regulation of glucose metabolism. To this end, we provide a basic introduction into mitochondrial energy metabolism, ROS generation and redox homeostasis. Next, we discuss the interactions between cellular glucose metabolism and ROS. ROS-stimulated cellular glucose uptake can stimulate both ROS production and scavenging. When glucose-stimulated ROS production, leading to further glucose uptake, is not adequately counterbalanced by (glucose-stimulated) ROS scavenging systems, a toxic cycle is triggered, ultimately leading to cell death. Here we inventoried the various cellular regulatory mechanisms and negative feedback loops that prevent this cycle from occurring. It is concluded that more insight in these processes is required to understand why they are (un)able to prevent excessive ROS production during various pathological conditions in humans.
Collapse
Affiliation(s)
- Dania C. Liemburg-Apers
- />Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Peter H. G. M. Willems
- />Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Werner J. H. Koopman
- />Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Sander Grefte
- />Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (RUMC), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
- />Department of Human and Animal Physiology, Wageningen University, P.O. Box 338, 6700 AH Wageningen, The Netherlands
| |
Collapse
|