1
|
Cui J, Shen Y, Song Z, Fan D, Hu B. Mechanism by which Rab5 promotes regeneration and functional recovery of zebrafish Mauthner axons. Neural Regen Res 2025; 20:1816-1824. [PMID: 39104118 PMCID: PMC11688562 DOI: 10.4103/nrr.nrr-d-23-00529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/02/2023] [Accepted: 01/26/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202506000-00031/figure1/v/2024-08-05T133530Z/r/image-tiff Rab5 is a GTPase protein that is involved in intracellular membrane trafficking. It functions by binding to various effector proteins and regulating cellular responses, including the formation of transport vesicles and their fusion with the cellular membrane. Rab5 has been reported to play an important role in the development of the zebrafish embryo; however, its role in axonal regeneration in the central nervous system remains unclear. In this study, we established a zebrafish Mauthner cell model of axonal injury using single-cell electroporation and two-photon axotomy techniques. We found that overexpression of Rab5 in single Mauthner cells promoted marked axonal regeneration and increased the number of intra-axonal transport vesicles. In contrast, treatment of zebrafish larvae with the Rab kinase inhibitor CID-1067700 markedly inhibited axonal regeneration in Mauthner cells. We also found that Rab5 activated phosphatidylinositol 3-kinase (PI3K) during axonal repair of Mauthner cells and promoted the recovery of zebrafish locomotor function. Additionally, rapamycin, an inhibitor of the mechanistic target of rapamycin downstream of PI3K, markedly hindered axonal regeneration. These findings suggest that Rab5 promotes the axonal regeneration of injured zebrafish Mauthner cells by activating the PI3K signaling pathway.
Collapse
Affiliation(s)
- Jiantao Cui
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Yueru Shen
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Zheng Song
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Dinggang Fan
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Bing Hu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| |
Collapse
|
2
|
Zheng C, Xia W, Zhang J. Rock inhibitors in Alzheimer's disease. FRONTIERS IN AGING 2025; 6:1547883. [PMID: 40182055 PMCID: PMC11965611 DOI: 10.3389/fragi.2025.1547883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025]
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disease and cause of dementia. AD pathology primarily involves the formation of amyloid β (Aβ) plaques and neurofibrillary tangles containing hyperphosphorylated tau (p-tau). While Aβ targeted treatments have shown clinical promise, other aspects of AD pathology such as microgliosis, astrocytosis, synaptic loss, and hypometabolism may be viable targets for treatment. Among notable novel therapeutic approaches, the Ras homolog (Rho)-associated kinases (ROCKs) are being investigated as targets for AD treatment, based on the observations that ROCK1/2 levels are elevated in AD, and activation or inhibition of ROCKs changes dendritic/synaptic structures, protein aggregate accumulation, inflammation, and gliosis. This review will highlight key findings on the effects of ROCK inhibition in Aβ and ptau pathologies, as well as its effects on neuroinflammation, synaptic density, and potentially metabolism and bioenergetics.
Collapse
Affiliation(s)
- Chao Zheng
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Departments of Psychiatry, Chemistry, Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Weiming Xia
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biological Sciences, University of Massachusetts Kennedy College of Science, Lowell, MA, United States
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
3
|
Lv R, Liu B, Jiang Z, Zhou R, Liu X, Lu T, Bao Y, Huang C, Zou G, Zhang Z, Lu L, Yin Q. Intermittent fasting and neurodegenerative diseases: Molecular mechanisms and therapeutic potential. Metabolism 2025; 164:156104. [PMID: 39674569 DOI: 10.1016/j.metabol.2024.156104] [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: 08/13/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Neurodegenerative disorders are straining public health worldwide. During neurodegenerative disease progression, aberrant neuronal network activity, bioenergetic impairment, adaptive neural plasticity impairment, dysregulation of neuronal Ca2+ homeostasis, oxidative stress, and immune inflammation manifest as characteristic pathological changes in the cellular milieu of the brain. There is no drug for the treatment of neurodegenerative disorders, and therefore, strategies/treatments for the prevention or treatment of neurodegenerative disorders are urgently needed. Intermittent fasting (IF) is characterized as an eating pattern that alternates between periods of fasting and eating, requiring fasting durations that vary depending on the specific protocol implemented. During IF, depletion of liver glycogen stores leads to the production of ketone bodies from fatty acids derived from adipocytes, thereby inducing an altered metabolic state accompanied by cellular and molecular adaptive responses within neural networks in the brain. At the cellular level, adaptive responses can promote the generation of synapses and neurons. At the molecular level, IF triggers the activation of associated transcription factors, thereby eliciting the expression of protective proteins. Consequently, this regulatory process governs central and peripheral metabolism, oxidative stress, inflammation, mitochondrial function, autophagy, and the gut microbiota, all of which contribute to the amelioration of neurodegenerative disorders. Emerging evidence suggests that weight regulation significantly contributes to the neuroprotective effects of IF. By alleviating obesity-related factors such as blood-brain barrier dysfunction, neuroinflammation, and β-amyloid accumulation, IF enhances metabolic flexibility and insulin sensitivity, further supporting its potential in mitigating neurodegenerative disorders. The present review summarizes animal and human studies investigating the role and underlying mechanisms of IF in physiology and pathology, with an emphasis on its therapeutic potential. Furthermore, we provide an overview of the cellular and molecular mechanisms involved in regulating brain energy metabolism through IF, highlighting its potential applications in neurodegenerative disorders. Ultimately, our findings offer novel insights into the preventive and therapeutic applications of IF for neurodegenerative disorders.
Collapse
Affiliation(s)
- Renjun Lv
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Bin Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan 250014, China
| | - Ziying Jiang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Runfa Zhou
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehlstr. 13-17, Mannheim 68167, Germany
| | - Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Chunxia Huang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Guichang Zou
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Zongyong Zhang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China; National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871 Beijing, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
4
|
Azam M, Jastrzebska B. Mechanisms of Rhodopsin-Related Inherited Retinal Degeneration and Pharmacological Treatment Strategies. Cells 2025; 14:49. [PMID: 39791750 PMCID: PMC11720364 DOI: 10.3390/cells14010049] [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: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
Retinitis pigmentosa (RP) is a hereditary disease characterized by progressive vision loss ultimately leading to blindness. This condition is initiated by mutations in genes expressed in retinal cells, resulting in the degeneration of rod photoreceptors, which is subsequently followed by the loss of cone photoreceptors. Mutations in various genes expressed in the retina are associated with RP. Among them, mutations in the rhodopsin gene (RHO) are the most common cause of this condition. Due to the involvement of numerous genes and multiple mutations in a single gene, RP is a highly heterogeneous disease making the development of effective treatments particularly challenging. The progression of this disease involves complex cellular responses to restore cellular homeostasis, including the unfolded protein response (UPR) signaling, autophagy, and various cell death pathways. These mechanisms, however, often fail to prevent photoreceptor cell degradation and instead contribute to cell death under certain conditions. Current research focuses on the pharmacological modulation of the components of these pathways and the direct stabilization of mutated receptors as potential treatment strategies. Despite these efforts, the intricate interplay between these mechanisms and the diverse causative mutations involved has hindered the development of effective treatments. Advancing our understanding of the interactions between photoreceptor cell death mechanisms and the specific genetic mutations driving RP is critical to accelerate the discovery and development of therapeutic strategies for this currently incurable disease.
Collapse
Affiliation(s)
- Maria Azam
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Beata Jastrzebska
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
- Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| |
Collapse
|
5
|
Tian RZ, Zhuang DL, Vong CT, He X, Ouyang Q, Liang JH, Guo YP, Wang YH, Zhao S, Yuan H, Ide Nasser M, Li G, Zhu P. Role of Autophagy in Myocardial Remodeling After Myocardial Infarction. J Cardiovasc Pharmacol 2025; 85:1-11. [PMID: 39454200 DOI: 10.1097/fjc.0000000000001646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/08/2024] [Indexed: 10/27/2024]
Abstract
ABSTRACT Autophagy is the process of reusing the body's senescent and damaged cell components, which can be regarded as the cellular circulatory system. There are 3 distinct forms of autophagy: macroautophagy, microautophagy, and chaperone-mediated autophagy. In the heart, autophagy is regulated mainly through mitophagy because of the metabolic changes of cardiomyocytes caused by ischemia and hypoxia. Myocardial remodeling is characterized by gradual heart enlargement, cardiac dysfunction, and extraordinary molecular changes. Cardiac remodeling after myocardial infarction is almost inevitable, which is the leading cause of heart failure. Autophagy has a protective effect on myocardial remodeling improvement. Autophagy can minimize cardiac remodeling by preventing misfolded protein accumulation and oxidative stress. This review summarizes the nestest molecular mechanisms of autophagy and myocardial remodeling, the protective effects, and the new target of autophagy medicine in cardiac remodeling. The future development and challenges of autophagy in heart disease are also summarized.
Collapse
Affiliation(s)
- Run-Ze Tian
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Dong-Lin Zhuang
- Department of Structural Heart Disease, National Center for Cardiovascular Disease, China & Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chi Teng Vong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
- Macau Centre for Research and Development in Chinese Medicine, University of Macau, Macau, China
| | - Xuyu He
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qing Ouyang
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jing-Hua Liang
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Yan-Ping Guo
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yu-Hong Wang
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Shuang Zhao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China; and
| | - Haiyun Yuan
- Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Moussa Ide Nasser
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ge Li
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ping Zhu
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Da Graça J, Delevoye C, Morel E. Morphodynamical adaptation of the endolysosomal system to stress. FEBS J 2025; 292:248-260. [PMID: 38706230 PMCID: PMC11734881 DOI: 10.1111/febs.17154] [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: 11/27/2023] [Revised: 03/28/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024]
Abstract
In eukaryotes, the spatiotemporal control of endolysosomal organelles is central to the maintenance of homeostasis. By providing an interface between the cytoplasm and external environment, the endolysosomal system is placed at the forefront of the response to a wide range of stresses faced by cells. Endosomes are equipped with a dedicated set of membrane-associated proteins that ensure endosomal functions as well as crosstalk with the secretory or the autophagy pathways. Morphodynamical processes operate through local spatialization of subdomains, enabling specific remodeling and membrane contact capabilities. Consequently, the plasticity of endolysosomal organelles can be considered a robust and flexible tool exploited by cells to cope with homeostatic deviations. In this review, we provide insights into how the cellular responses to various stresses (osmotic, UV, nutrient deprivation, or pathogen infections) rely on the adaptation of the endolysosomal system morphodynamics.
Collapse
Affiliation(s)
- Juliane Da Graça
- Université Paris Cité, INSERM UMR‐S1151, CNRS UMR‐S8253, Institut Necker Enfants MaladesFrance
| | - Cédric Delevoye
- Université Paris Cité, INSERM UMR‐S1151, CNRS UMR‐S8253, Institut Necker Enfants MaladesFrance
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane CompartmentsParisFrance
| | - Etienne Morel
- Université Paris Cité, INSERM UMR‐S1151, CNRS UMR‐S8253, Institut Necker Enfants MaladesFrance
| |
Collapse
|
7
|
Villadangos L, Serrador JM. Subcellular Localization Guides eNOS Function. Int J Mol Sci 2024; 25:13402. [PMID: 39769167 PMCID: PMC11678294 DOI: 10.3390/ijms252413402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Nitric oxide synthases (NOS) are enzymes responsible for the cellular production of nitric oxide (NO), a highly reactive signaling molecule involved in important physiological and pathological processes. Given its remarkable capacity to diffuse across membranes, NO cannot be stored inside cells and thus requires multiple controlling mechanisms to regulate its biological functions. In particular, the regulation of endothelial nitric oxide synthase (eNOS) activity has been shown to be crucial in vascular homeostasis, primarily affecting cardiovascular disease and other pathophysiological processes of importance for human health. Among other factors, the subcellular localization of eNOS plays an important role in regulating its enzymatic activity and the bioavailability of NO. The aim of this review is to summarize pioneering studies and more recent publications, unveiling some of the factors that influence the subcellular compartmentalization of eNOS and discussing their functional implications in health and disease.
Collapse
Affiliation(s)
| | - Juan M. Serrador
- Interactions with the Environment Program, Immune System Development and Function Unit, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas (CSIC)—Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| |
Collapse
|
8
|
Georgoulis I, Giantsis IA, Michaelidis B, Feidantsis K. Heat Hardening Ameliorates Apoptotic and Inflammatory Effects Through Increased Autophagy in Mussels. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:1271-1286. [PMID: 39240443 DOI: 10.1007/s10126-024-10371-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
The severity, frequency, and duration of extreme events, in the context of global warming, have placed many marine ecosystems at high risk. Therefore, the application of methods that can mediate the impacts of global warming on marine organisms seems to be an emerging necessity in the near term. In this context, enhancing the thermal resilience of marine organisms may be crucial for their sustainability. It has been shown that the repeated time-limited exposure of an organism to an environmental stimulus modifies its response mode, thus enhancing resilience and allowing adaptation of the physiological and developmental phenotype to environmental stress. In the present study, we investigated the "stress memory" effect caused by heat hardening on Mytilus galloprovincialis cellular pathways to identify the underlying biochemical mechanisms that enhance mussel thermal tolerance. Heat hardening resulted in increased ETS activity and ATP production and increased autophagic performance at all elevated temperatures (24 °C, 26 °C, and 28 °C). Furthermore, at these increased temperatures, apoptosis and inflammation remain at significantly lower levels in pregnant individuals than in nonhardened individuals. Autophagy, as a negative regulator of apoptosis, may lead to decreased damage to surrounding cells, which in turn alleviates inflammatory effects. In conclusion, the exposure of mussels to heat hardening seems to provide a physiological response that enhances heat tolerance and increases cell survival through increased energy production and reduced cell death and inflammatory responses. The latter can be utilized for the management and conservation of aquatic species of economic value or endangered status.
Collapse
Affiliation(s)
- Ioannis Georgoulis
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| | - Ioannis A Giantsis
- Department of Animal Science, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Basile Michaelidis
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Konstantinos Feidantsis
- Department of Fisheries & Aquaculture, School of Agricultural Sciences, University of Patras, 26504, Mesolonghi, Greece.
| |
Collapse
|
9
|
Nam HY, Park SH, Lee GH, Kim EY, Lee S, Chang HW, Chang EJ, Choi KC, Kim SW. TIGAR coordinates senescence-associated secretory phenotype via lysosome repositioning and α-tubulin deacetylation. Exp Mol Med 2024; 56:2726-2738. [PMID: 39633033 DOI: 10.1038/s12276-024-01362-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/06/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024] Open
Abstract
TP53-induced glycolysis and apoptosis regulator (TIGAR) regulates redox homeostasis and provides the intermediates necessary for cell growth by reducing the glycolytic rate. During cellular senescence, cells undergo metabolic rewiring towards the glycolytic pathway, along with the development of the senescence-associated secretory phenotype (SASP), also known as the secretome. We observed that TIGAR expression increased during replicative senescence following the in vitro expansion of human mesenchymal stromal cells (MSCs) and that TIGAR knockout (KO) decreased SASP factors and triggered premature senescence with decelerated progression. Additionally, TIGAR KO impaired flexible lysosomal movement to the perinuclear region and decreased the autophagic flux of MSCs. Research on the mechanism of lysosomal movement revealed that, while native senescent MSCs presented low levels of Ac-α-tubulin (lysine 40) and increased sirtuin 2 (SIRT2) activity compared with those in growing cells, TIGAR KO-MSCs maintained Ac-α-tubulin levels and exhibited decreased SIRT2 activity despite being in a senescent state. The overexpression of SIRT2 reduced Ac-α-tubulin as a protein target of SIRT2 and induced the positioning of lysosomes at the perinuclear region, restoring the cytokine secretion of TIGAR KO-MSCs. Furthermore, TIGAR expression was positively correlated with SIRT2 activity, indicating that TIGAR affects SIRT2 activity partly by modulating the NAD+ level. Thus, our study demonstrated that TIGAR provides a foundation that translates the regulation of energy metabolism into lysosome positioning, affecting the secretome for senescence development. Considering the functional value of the cell-secretome in aging-related diseases, these findings suggest the feasibility of TIGAR for the regulation of secretory phenotypes.
Collapse
Affiliation(s)
- Hae Yun Nam
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| | - Seung-Ho Park
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Geun-Hee Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Eun-Young Kim
- Department Hematology and Medical Oncology, Whinship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - SangEun Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Hyo Won Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Kyung-Chul Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| |
Collapse
|
10
|
Eo H, Kim S, Jung UJ, Kim SR. Alpha-Synuclein and Microglia in Parkinson's Disease: From Pathogenesis to Therapeutic Prospects. J Clin Med 2024; 13:7243. [PMID: 39685702 DOI: 10.3390/jcm13237243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/14/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by both motor symptoms and non-motor features. A hallmark of PD is the misfolding and accumulation of alpha-synuclein (α-syn), which triggers neuroinflammation and drives neurodegeneration. Microglia, brain cells that play a central role in neuroinflammatory responses and help clear various unnecessary molecules within the brain, thus maintaining the brain's internal environment, respond to α-syn through mechanisms involving inflammation, propagation, and clearance. This review delves into the complex interplay between α-syn and microglia, elucidating how these interactions drive PD pathogenesis. Furthermore, we discuss emerging therapeutic strategies targeting the α-syn-microglia axis, with a focus on modulating microglial functions to mitigate neuroinflammation, enhance clearance, and prevent α-syn propagation, emphasizing their potential to slow PD progression.
Collapse
Affiliation(s)
- Hyemi Eo
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Sehwan Kim
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
11
|
Chavas C, Sapanidou VG, Feidantsis K, Lavrentiadou SN, Mavrogianni D, Zarogoulidou I, Fletouris DJ, Tsantarliotou MP. Treatment with Pterostilbene Ameliorates the Antioxidant Status of Bovine Spermatozoa and Modulates Cell Death Pathways. Antioxidants (Basel) 2024; 13:1437. [PMID: 39765765 PMCID: PMC11672693 DOI: 10.3390/antiox13121437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Reactive Oxygen Species (ROS) play an important role in sperm physiology. They are required in processes such as capacitation and fertilization. However, the exposure of spermatozoa to ROS generated from internal or external sources may create a potentially detrimental redox imbalance. Antioxidant supplementation in semen is now a rather common approach to protect spermatozoa from oxidative stress (OS) during their handling and/or cryopreservation. Supplementation with pterostilbene, a potent antioxidant, protects spermatozoa from OS and ameliorates their post-thawing characteristics and viability. In the present study, we used freezing/thawing as a model of natural ROS overproduction and investigated the molecular mechanisms modulated by pterostilbene. Specifically, bovine frozen/thawed spermatozoa were incubated with 10 or 25 μM pterostilbene for 60 min. Results have shown that in a dose-independent manner, pterostilbene decreased lipid peroxidation and increased intracellular GSH levels. Moreover, pterostilbene ameliorated energy production, as ATP and AMP/ATP levels were restored, and increased autophagy levels through AMP-activated protein kinase (AMPK) activation, which finally resulted in the inhibition of apoptotic cell death in bovine spermatozoa when exposed to OS. This study sheds light on spermatozoa redox state, the crosstalk between apoptotic and autophagic pathways, and its role in determining the beneficial or detrimental effect of ROS in spermatozoa.
Collapse
Affiliation(s)
- Christos Chavas
- Laboratory of Animal Physiology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (C.C.); (V.G.S.); (S.N.L.); (I.Z.)
| | - Vasiliki G. Sapanidou
- Laboratory of Animal Physiology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (C.C.); (V.G.S.); (S.N.L.); (I.Z.)
| | - Konstantinos Feidantsis
- Department of Fisheries & Aquaculture, School of Agricultural Sciences, University of Patras, GR-26504 Mesolonghi, Greece
| | - Sophia N. Lavrentiadou
- Laboratory of Animal Physiology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (C.C.); (V.G.S.); (S.N.L.); (I.Z.)
| | - Despoina Mavrogianni
- First Department of Obstetrics and Gynecology, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, GR-11528 Athens, Greece;
| | - Ioanna Zarogoulidou
- Laboratory of Animal Physiology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (C.C.); (V.G.S.); (S.N.L.); (I.Z.)
| | - Dimitrios J. Fletouris
- Department of Hygiene and Technology of Animal Origin Products, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece;
| | - Maria P. Tsantarliotou
- Laboratory of Animal Physiology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (C.C.); (V.G.S.); (S.N.L.); (I.Z.)
| |
Collapse
|
12
|
Syed RU, Alshammari MD, Banu H, Khojali WMA, Jafar M, Nagaraju P, Alshammari A. Targeting the autophagy-miRNA axis in prostate cancer: toward novel diagnostic and therapeutic strategies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7421-7437. [PMID: 38761210 DOI: 10.1007/s00210-024-03153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
Since prostate cancer is one of the leading causes of cancer-related death, a better understanding of the molecular pathways guiding its development is imperative. A key factor in prostate cancer is autophagy, a cellular mechanism that affects both cell survival and death. Autophagy is essential in maintaining cellular homeostasis. Autophagy is a physiological mechanism wherein redundant or malfunctioning cellular constituents are broken down and recycled. It is essential for preserving cellular homeostasis and is implicated in several physiological and pathological conditions, including cancer. Autophagy has been linked to metastasis, tumor development, and treatment resistance in prostate cancer. The deregulation of miRNAs related to autophagy appears to be a crucial element in the etiology of prostate cancer. These miRNAs influence the destiny of cancer cells by finely regulating autophagic mechanisms. Numerous investigations have emphasized the dual function of specific miRNAs in prostate cancer, which alter autophagy-related pathways to function as either tumor suppressors or oncogenes. Notably, miRNAs have been linked to the control of autophagy and the proliferation, apoptosis, and migration of prostate cancer cells. To create customized therapy approaches, it is imperative to comprehend the dynamic interplay between autophagy and miRNAs in prostate cancer. The identification of key miRNAs provides potential diagnostic and prognostic markers. Unraveling the complex network of lncRNAs, like PCA3, also expands the repertoire of molecular targets for therapeutic interventions. This review explores the intricate interplay between autophagy and miRNAs in prostate cancer, focusing on their regulatory roles in cellular processes ranging from survival to programmed cell death.
Collapse
Affiliation(s)
- Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Hail, 81442, Hail, Saudi Arabia.
| | - Maali D Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, 81442, Hail, Saudi Arabia
| | - Humera Banu
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Hail, Hail, Kingdom of Saudi Arabia
| | - Weam M A Khojali
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, 81442, Hail, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Omdurman Islamic University, Omdurman, 14415, Sudan
| | - Mohammed Jafar
- Department of Pharmaceutics, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam, 34212, Saudi Arabia.
| | - Potnuri Nagaraju
- Department of Pharmaceutics, Mandesh Institute of Pharmaceutical Science and Research Center, Mhaswad, Maharashtra, India
| | - Alia Alshammari
- Department of Pharmaceutics, College of Pharmacy, University of Hail, 81442, Hail, Saudi Arabia
| |
Collapse
|
13
|
Wu J, Chen Y, Zou H, Xu K, Hou J, Wang M, Tian S, Gao M, Ren Q, Sun C, Lu S, Wang Q, Shu Y, Wang S, Wang X. 6-Phosphogluconate dehydrogenase promotes glycolysis and fatty acid synthesis by inhibiting the AMPK pathway in lung adenocarcinoma cells. Cancer Lett 2024; 601:217177. [PMID: 39179096 DOI: 10.1016/j.canlet.2024.217177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/27/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024]
Abstract
Abnormal metabolism has emerged as a prominent hallmark of cancer and plays a pivotal role in carcinogenesis and progression of lung adenocarcinoma (LUAD). In this study, single-cell sequencing revealed that the metabolic enzyme 6-phosphogluconate dehydrogenase (PGD), which is a critical regulator of the pentose phosphate pathway (PPP), is significantly upregulated in the malignant epithelial cell subpopulation during malignant progression. However, the precise functional significance of PGD in LUAD and its underlying mechanisms remain elusive. Through the integration of TCGA database analysis and LUAD tissue microarray data, it was found that PGD expression was significantly upregulated in LUAD and closely correlated with a poor prognosis in LUAD patients. Moreover, in vitro and in vivo analyses demonstrated that PGD knockout and inhibition of its activity mitigated the proliferation, migration, and invasion of LUAD cells. Mechanistically, immunoprecipitation-mass spectrometry (IP-MS) revealed for the first time that IQGAP1 is a robust novel interacting protein of PGD. PGD decreased p-AMPK levels by competitively interacting with the IQ domain of the known AMPKα binding partner IQGAP1, which promoted glycolysis and fatty acid synthesis in LUAD cells. Furthermore, we demonstrated that the combination of Physcion (a PGD-specific inhibitor) and metformin (an AMPK agonist) could inhibit tumor growth more effectively both in vivo and in vitro. Collectively, these findings suggest that PGD is a potential prognostic biomarker and therapeutic target for LUAD.
Collapse
Affiliation(s)
- Jun Wu
- Medical College, Yangzhou University, Yangzhou, China; Department of Thoracic Surgery, Northern Jiangsu People's Hospital, Yangzhou, China; Yangzhou Key Laboratory of Thoracic and Cardiac Surgery, Yangzhou, China
| | - Yong Chen
- Department of Thoracic Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hui Zou
- The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou, China; Department of Thoracic Surgery, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Kaiyue Xu
- Department of Radiation Oncology, Suzhou Municipal Hospital, Suzhou, China
| | - Jiaqi Hou
- First College of Clinical Medicine, Dalian Medical University, Dalian, China
| | - Mengmeng Wang
- First College of Clinical Medicine, Dalian Medical University, Dalian, China
| | - Shuyu Tian
- First College of Clinical Medicine, Dalian Medical University, Dalian, China
| | - Mingjun Gao
- First College of Clinical Medicine, Dalian Medical University, Dalian, China
| | - Qinglin Ren
- First College of Clinical Medicine, Dalian Medical University, Dalian, China
| | - Chao Sun
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Shichun Lu
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Provincial Innovation Institute for Pharmaceutical Basic Research, Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China
| | - Yusheng Shu
- The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou, China; Department of Thoracic Surgery, Northern Jiangsu People's Hospital, Yangzhou, China; Yangzhou Key Laboratory of Thoracic and Cardiac Surgery, Yangzhou, China.
| | - Shouyu Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Provincial Innovation Institute for Pharmaceutical Basic Research, Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China.
| | - Xiaolin Wang
- Medical College, Yangzhou University, Yangzhou, China; Department of Thoracic Surgery, Northern Jiangsu People's Hospital, Yangzhou, China; Yangzhou Key Laboratory of Thoracic and Cardiac Surgery, Yangzhou, China.
| |
Collapse
|
14
|
Sowers AL, Gohain S, Edmondson EF, Choudhuri R, Krishna MC, Cook JA, Mitchell JB. Rapamycin Reduces Carcinogenesis and Enhances Survival in Mice when Administered after Nonlethal Total-Body Irradiation. Radiat Res 2024; 202:639-648. [PMID: 39142650 PMCID: PMC11556393 DOI: 10.1667/rade-24-00111.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/26/2024] [Indexed: 08/16/2024]
Abstract
The rationale of this study stems from the concern of a radiation-induced accident or terrorist-mediated nuclear attack resulting in large populations of people exposed to nonlethal radiation doses or after a course of definitive radiation therapy which could substantially increase the risk for cancer induction after exposure. Currently, there are no safe and effective interventions to reduce this increased cancer risk to humans. We have tested the hypothesis that the mTOR inhibitor, rapamycin, administered in the diet of mice would reduce or delay radiation-induced cancer when given after radiation exposure. A total-body irradiation (TBI) of 3 Gy was administered to female C3H/Hen mice. Immediately after TBI, along with untreated control groups, animals were placed on chow containing different concentrations of encapsulated rapamycin (14, 40, 140 mg/kg chow). Animals remained on the respective control or rapamycin diets and were followed for their entire lifespan (total of 795 mice). The endpoint for the study was tumor formation (not to exceed 1 cm) or until the animal reached a humane endpoint at which time the animal was euthanized and evaluated for the presence of tumors (pathology evaluated on all animals). Kaplan-Meier survival curves revealed that all three concentrations of rapamycin afforded a significant survival advantage by delaying the time at which tumors appeared and reduction of the incidence of certain tumor types such as hepatocellular carcinomas. The survival advantage was dependent on the rapamycin concentration used. Further, there was a survival advantage when delaying the rapamycin chow by 1 month after TBI. Rapamycin is FDA-approved for human use and could be considered for use in individuals exposed to nonlethal TBI from a nuclear accident or attack or after significant therapeutic doses for cancer treatment.
Collapse
Affiliation(s)
- Anastasia L. Sowers
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Sangeeta Gohain
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Elijah F. Edmondson
- Molecular Histopathology Laboratory, NCI-Frederick Cancer Research Center, Frederick, Maryland 21702
| | - Rajani Choudhuri
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Murali C. Krishna
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - John A. Cook
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - James B. Mitchell
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
15
|
Zhang F, Guo J, Yu S, Zheng Y, Duan M, Zhao L, Wang Y, Yang Z, Jiang X. Cellular senescence and metabolic reprogramming: Unraveling the intricate crosstalk in the immunosuppressive tumor microenvironment. Cancer Commun (Lond) 2024; 44:929-966. [PMID: 38997794 PMCID: PMC11492308 DOI: 10.1002/cac2.12591] [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: 11/26/2023] [Revised: 06/23/2024] [Accepted: 07/07/2024] [Indexed: 07/14/2024] Open
Abstract
The intrinsic oncogenic mechanisms and properties of the tumor microenvironment (TME) have been extensively investigated. Primary features of the TME include metabolic reprogramming, hypoxia, chronic inflammation, and tumor immunosuppression. Previous studies suggest that senescence-associated secretory phenotypes that mediate intercellular information exchange play a role in the dynamic evolution of the TME. Specifically, hypoxic adaptation, metabolic dysregulation, and phenotypic shifts in immune cells regulated by cellular senescence synergistically contribute to the development of an immunosuppressive microenvironment and chronic inflammation, thereby promoting the progression of tumor events. This review provides a comprehensive summary of the processes by which cellular senescence regulates the dynamic evolution of the tumor-adapted TME, with focus on the complex mechanisms underlying the relationship between senescence and changes in the biological functions of tumor cells. The available findings suggest that components of the TME collectively contribute to the progression of tumor events. The potential applications and challenges of targeted cellular senescence-based and combination therapies in clinical settings are further discussed within the context of advancing cellular senescence-related research.
Collapse
Affiliation(s)
- Fusheng Zhang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
- Department of Hepatobiliary and Pancreatic SurgeryPeking University First HospitalBeijingP. R. China
| | - Junchen Guo
- Department of RadiologyThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Shengmiao Yu
- Outpatient DepartmentThe Fourth Affiliated HospitalChina Medical UniversityShenyangLiaoningP. R. China
| | - Youwei Zheng
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Meiqi Duan
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Liang Zhao
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Yihan Wang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Zhi Yang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Xiaofeng Jiang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| |
Collapse
|
16
|
Galli F, Bartolini D, Ronco C. Oxidative stress, defective proteostasis and immunometabolic complications in critically ill patients. Eur J Clin Invest 2024; 54:e14229. [PMID: 38676423 DOI: 10.1111/eci.14229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/31/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Oxidative stress (OS) develops in critically ill patients as a metabolic consequence of the immunoinflammatory and degenerative processes of the tissues. These induce increased and/or dysregulated fluxes of reactive species enhancing their pro-oxidant activity and toxicity. At the same time, OS sustains its own inflammatory and immunometabolic pathogenesis, leading to a pervasive and vitious cycle of events that contribute to defective immunity, organ dysfunction and poor prognosis. Protein damage is a key player of these OS effects; it generates increased levels of protein oxidation products and misfolded proteins in both the cellular and extracellular environment, and contributes to forms DAMPs and other proteinaceous material to be removed by endocytosis and proteostasis processes of different cell types, as endothelial cells, tissue resident monocytes-macrophages and peripheral immune cells. An excess of OS and protein damage in critical illness can overwhelm such cellular processes ultimately interfering with systemic proteostasis, and consequently with innate immunity and cell death pathways of the tissues thus sustaining organ dysfunction mechanisms. Extracorporeal therapies based on biocompatible/bioactive membranes and new adsorption techniques may hold some potential in reducing the impact of OS on the defective proteostasis of patients with critical illness. These can help neutralizing reactive and toxic species, also removing solutes in a wide spectrum of molecular weights thus improving proteostasis and its immunometabolic corelates. Pharmacological therapy is also moving steps forward which could help to enhance the efficacy of extracorporeal treatments. This narrative review article explores the aspects behind the origin and pathogenic role of OS in intensive care and critically ill patients, with a focus on protein damage as a cause of impaired systemic proteostasis and immune dysfunction in critical illness.
Collapse
Affiliation(s)
- Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Claudio Ronco
- Department of Medicine, International Renal Research Institute of Vicenza, University of Padova, San Bortolo Hospital Vicenza, Vicenza, Italy
| |
Collapse
|
17
|
Feidantsis K, Panteli N, Bousdras T, Gai F, Gasco L, Antonopoulou E. Dietary Tenebrio molitor larvae meal effects on cellular stress responses, antioxidant status and intermediate metabolism of Oncorhynchus mykiss. J Anim Physiol Anim Nutr (Berl) 2024; 108:1270-1285. [PMID: 38657021 DOI: 10.1111/jpn.13970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024]
Abstract
In the context of evaluating the impact of environmentally friendly and sustainably produced alternative protein sources in fish feed, the present study's aim was to examine the overall physiological stress response in one of the main fish species of European freshwater aquaculture, Oncorhynchus mykiss (rainbow trout), following the partial substitution of fish meal (FM) with a Tenebrio molitor (TM) (yellow mealworm) full-fat meal. In total, 222 rainbow trout individuals (115.2 ± 14.2 g) were allocated randomly into six tanks, three per dietary treatment, and were fed a formulated diet containing 60% yellow mealworm (TM60) compared to a control diet without insect meal (TM0). Both diets contained equal amounts of crude protein, dry matter and, lipid content, while the FM in TM60 was 100 g kg-1 corresponding to the one seventh of the TM0. Heat shock response (HSR), MAPK signalling, cell death pathways (apoptosis and autophagy), antioxidant defence mechanisms, and intermediate metabolism were evaluated. In general, HSR and MAPK signalling were activated in response to the inclusion of T. molitor. Moreover, triggering of apoptotic and autophagic processes and the onset of antioxidant defence mechanisms underlined the existence of physiological stress. Despite the apparent dietary-induced stress, rainbow trout in the present study exhibited no mortality and no significant effects regarding growth performance parameters. Specifically, TM60 dietary inclusion resulted in no changes in final body weight, weight gain, and specific growth rate. However, feed intake depicted a statistically significant decrease in TM60 fish compared to TM0 individuals. Nevertheless, nutrient stress should be considered a limiting factor regarding the utilization of T. molitor in O. mykiss diet due to the associated risks for health and welfare.
Collapse
Affiliation(s)
- Konstantinos Feidantsis
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Department of Fisheries & Aquaculture, University of Patras, Mesolonghi, Greece
| | - Nikolas Panteli
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Thomas Bousdras
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Francesco Gai
- Institute of Sciences of Food Production, National Research Council, Grugliasco, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Laura Gasco
- Department of Agricultural, Forest and Food Sciences, University of Turin, Grugliasco, Italy
| | - Efthimia Antonopoulou
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
18
|
Cai X, Liu Y, Li H, Que Y, Xiao M, Wang Y, Wang X, Li D. XPO1 inhibition displays anti-leukemia efficacy against DNMT3A-mutant acute myeloid leukemia via downregulating glutathione pathway. Ann Hematol 2024; 103:2311-2322. [PMID: 38519605 DOI: 10.1007/s00277-024-05706-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/10/2024] [Indexed: 03/25/2024]
Abstract
Acute myeloid leukemia (AML) patients with DNA methyltransferase 3A (DNMT3A) mutation display poor prognosis, and targeted therapy is not available currently. Our previous study identified increased expression of Exportin1 (XPO1) in DNMT3AR882H AML patients. Therefore, we further investigated the therapeutic effect of XPO1 inhibition on DNMT3AR882H AML. Three types of DNMT3AR882H AML cell lines were generated, and XPO1 was significantly upregulated in all DNMT3AR882H cells compared with the wild-type (WT) cells. The XPO1 inhibitor selinexor displayed higher potential in the inhibition of proliferation, promotion of apoptosis, and blockage of the cell cycle in DNMT3AR882H cells than WT cells. Selinexor also significantly inhibited the proliferation of subcutaneous tumors in DNMT3AR882H AML model mice. Primary cells with DNMT3A mutations were more sensitive to selinexor in chemotherapy-naive AML patients. RNA sequencing of selinexor treated AML cells revealed that the majority of metabolic pathways were downregulated after selinexor treatment, with the most significant change in the glutathione metabolic pathway. Glutathione inhibitor L-Buthionine-(S, R)-sulfoximine (BSO) significantly enhanced the apoptosis-inducing effect of selinexor in DNMT3AWT/DNMT3AR882H AML cells. In conclusion, our work reveals that selinexor displays anti-leukemia efficacy against DNMT3AR882H AML via downregulating glutathione pathway. Combination of selinexor and BSO provides novel therapeutic strategy for AML treatment.
Collapse
Affiliation(s)
- Xiaoya Cai
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Liu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huimin Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yimei Que
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiong Wang
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Dengju Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
19
|
Nag S, Mitra O, Maturi B, Kaur SP, Saini A, Nama M, Roy S, Samanta S, Chacko L, Dutta R, Sayana SB, Subramaniyan V, Bhatti JS, Kandimalla R. Autophagy and mitophagy as potential therapeutic targets in diabetic heart condition: Harnessing the power of nanotheranostics. Asian J Pharm Sci 2024; 19:100927. [PMID: 38948399 PMCID: PMC11214300 DOI: 10.1016/j.ajps.2024.100927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 03/29/2024] [Accepted: 04/13/2024] [Indexed: 07/02/2024] Open
Abstract
Autophagy and mitophagy pose unresolved challenges in understanding the pathology of diabetic heart condition (DHC), which encompasses a complex range of cardiovascular issues linked to diabetes and associated cardiomyopathies. Despite significant progress in reducing mortality rates from cardiovascular diseases (CVDs), heart failure remains a major cause of increased morbidity among diabetic patients. These cellular processes are essential for maintaining cellular balance and removing damaged or dysfunctional components, and their involvement in the development of diabetic heart disease makes them attractive targets for diagnosis and treatment. While a variety of conventional diagnostic and therapeutic strategies are available, DHC continues to present a significant challenge. Point-of-care diagnostics, supported by nanobiosensing techniques, offer a promising alternative for these complex scenarios. Although conventional medications have been widely used in DHC patients, they raise several concerns regarding various physiological aspects. Modern medicine places great emphasis on the application of nanotechnology to target autophagy and mitophagy in DHC, offering a promising approach to deliver drugs beyond the limitations of traditional therapies. This article aims to explore the potential connections between autophagy, mitophagy and DHC, while also discussing the promise of nanotechnology-based theranostic interventions that specifically target these molecular pathways.
Collapse
Affiliation(s)
- Sagnik Nag
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Oishi Mitra
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Bhanu Maturi
- Department of Internal Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Simran Preet Kaur
- Department of Microbiology, University of Delhi (South Campus), Benito Juarez Road, New Delhi 110021, India
| | - Ankita Saini
- Department of Microbiology, University of Delhi (South Campus), Benito Juarez Road, New Delhi 110021, India
| | - Muskan Nama
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Soumik Roy
- Department of Biotechnology, Indian Institute of Technology, Hyderabad (IIT-H), Sangareddy, Telangana 502284, India
| | - Souvik Samanta
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Leena Chacko
- BioAnalytical Lab, Meso Scale Discovery, 1601 Research Blvd, Rockville, MD, USA
| | - Rohan Dutta
- Department of Bio-Sciences, School of Bio-Sciences & Technology (SBST), Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Suresh Babu Sayana
- Department of Pharmacology, Government Medical College, Suryapet, Telangana, India
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, India
| |
Collapse
|
20
|
Han YZ, Du BX, Zhu XY, Wang YZY, Zheng HJ, Liu WJ. Lipid metabolism disorder in diabetic kidney disease. Front Endocrinol (Lausanne) 2024; 15:1336402. [PMID: 38742197 PMCID: PMC11089115 DOI: 10.3389/fendo.2024.1336402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetic kidney disease (DKD), a significant complication associated with diabetes mellitus, presents limited treatment options. The progression of DKD is marked by substantial lipid disturbances, including alterations in triglycerides, cholesterol, sphingolipids, phospholipids, lipid droplets, and bile acids (BAs). Altered lipid metabolism serves as a crucial pathogenic mechanism in DKD, potentially intertwined with cellular ferroptosis, lipophagy, lipid metabolism reprogramming, and immune modulation of gut microbiota (thus impacting the liver-kidney axis). The elucidation of these mechanisms opens new potential therapeutic pathways for DKD management. This research explores the link between lipid metabolism disruptions and DKD onset.
Collapse
Affiliation(s)
- Yi-Zhen Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Bo-Xuan Du
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xing-Yu Zhu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang-Zhi-Yuan Wang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Hui-Juan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei-Jing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
21
|
Qiao Q, Wang X, Su Z, Han C, Zhao K, Qi K, Xie Z, Huang X, Zhang S. PuNDH9, a subunit of ETC Complex I regulates plant defense by interacting with PuPR1. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2024; 341:112009. [PMID: 38316345 DOI: 10.1016/j.plantsci.2024.112009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/08/2024] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
NAD+ and NADH play critical roles in energy metabolism, cell death, and gene expression. The NADH-ubiquinone oxidoreductase complex (Complex I) has been long known as a key enzyme in NAD+ and NADH metabolism. In the present study, we found and analyzed a new subunit of Complex I (NDH9), which was isolated from Pyrus ussuriensis combined with RT-PCR. Following infection with A. alternata, RT-qPCR analysis demonstrated an increase in the expression of PuNDH9. Genetic manipulation of PuNDH9 levels suggested that PuNDH9 plays key roles in NADH/NAD+ homeostasis, defense enzyme activities, ROS generation, cell death, gene expression, energy metabolism, and mitochondrial functions during the pear- A. alternata interaction. Furthermore, Y2H, GST-pull down, and a split-luciferase complementation imaging assays revealed that PuNDH9 interacts with PuPR1. We discover that PuNDH9 and PuPR1 synergistically activate defense enzyme activities, ROS accumulation, cell death, and plant defenses. Collectively, our findings reveal that PuNDH9 is likely important for plant defenses.
Collapse
Affiliation(s)
- Qinghai Qiao
- College of Life Science, Nanjing Agricultural University, Nanjing 210095, China; State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing 210095, China
| | - Xin Wang
- College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China; State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhiyuan Su
- College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China; State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing 210095, China
| | - Chenyang Han
- College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China; State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing 210095, China
| | - Keke Zhao
- College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China; State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing 210095, China
| | - Kaijie Qi
- College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China; State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhihua Xie
- College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China; State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaosan Huang
- College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China; State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing 210095, China.
| | - Shaoling Zhang
- College of Life Science, Nanjing Agricultural University, Nanjing 210095, China; College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China; State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
22
|
Bischof L, Schweitzer F, Heinisch JJ. Functional Conservation of the Small GTPase Rho5/Rac1-A Tale of Yeast and Men. Cells 2024; 13:472. [PMID: 38534316 PMCID: PMC10969153 DOI: 10.3390/cells13060472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Small GTPases are molecular switches that participate in many essential cellular processes. Amongst them, human Rac1 was first described for its role in regulating actin cytoskeleton dynamics and cell migration, with a close relation to carcinogenesis. More recently, the role of Rac1 in regulating the production of reactive oxygen species (ROS), both as a subunit of NADPH oxidase complexes and through its association with mitochondrial functions, has drawn attention. Malfunctions in this context affect cellular plasticity and apoptosis, related to neurodegenerative diseases and diabetes. Some of these features of Rac1 are conserved in its yeast homologue Rho5. Here, we review the structural and functional similarities and differences between these two evolutionary distant proteins and propose yeast as a useful model and a device for high-throughput screens for specific drugs.
Collapse
Affiliation(s)
| | | | - Jürgen J. Heinisch
- AG Genetik, Fachbereich Biologie/Chemie, University of Osnabrück, Barbarastrasse 11, D-49076 Osnabrück, Germany; (L.B.); (F.S.)
| |
Collapse
|
23
|
Xu L, Cao Y, Xu Y, Li R, Xu X. Redox-Responsive Polymeric Nanoparticle for Nucleic Acid Delivery and Cancer Therapy: Progress, Opportunities, and Challenges. Macromol Biosci 2024; 24:e2300238. [PMID: 37573033 DOI: 10.1002/mabi.202300238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/25/2023] [Indexed: 08/14/2023]
Abstract
Cancer development and progression of cancer are closely associated with the activation of oncogenes and loss of tumor suppressor genes. Nucleic acid drugs (e.g., siRNA, mRNA, and DNA) are widely used for cancer therapy due to their specific ability to regulate the expression of any cancer-associated genes. However, nucleic acid drugs are negatively charged biomacromolecules that are susceptible to serum nucleases and cannot cross cell membrane. Therefore, specific delivery tools are required to facilitate the intracellular delivery of nucleic acid drugs. In the past few decades, a variety of nanoparticles (NPs) are designed and developed for nucleic acid delivery and cancer therapy. In particular, the polymeric NPs in response to the abnormal redox status in cancer cells have garnered much more attention as their potential in redox-triggered nanostructure dissociation and rapid intracellular release of nucleic acid drugs. In this review, the important genes or signaling pathways regulating the abnormal redox status in cancer cells are briefly introduced and the recent development of redox-responsive NPs for nucleic acid delivery and cancer therapy is systemically summarized. The future development of NPs-mediated nucleic acid delivery and their challenges in clinical translation are also discussed.
Collapse
Affiliation(s)
- Lei Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Yuan Cao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Ya Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Rong Li
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| |
Collapse
|
24
|
Zhong X, Deng Y, Yang H, Du X, Liu P, Du Y. Role of autophagy in skin photoaging: A narrative review. Medicine (Baltimore) 2024; 103:e37178. [PMID: 38394552 PMCID: PMC11309671 DOI: 10.1097/md.0000000000037178] [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: 10/30/2023] [Accepted: 01/17/2024] [Indexed: 02/25/2024] Open
Abstract
As the largest organ of the human body, the skin serves as the primary barrier against external damage. The continuous increase in human activities and environmental pollution has resulted in the ongoing depletion of the ozone layer. Excessive exposure to ultraviolet (UV) radiation enhances the impact of external factors on the skin, leading to photoaging. Photoaging causes physical and psychological damage to the human body. The prevention and management of photoaging have attracted increased attention in recent years. Despite significant progress in understanding and mitigating UV-induced photoaging, the precise mechanisms through which autophagy contributes to the prevention of photoaging remain unclear. Given the important role of autophagy in repairing UV-induced DNA damage and scavenging oxidized lipids, autophagy is considered a novel strategy for preventing the occurrence of photoaging and other UV light-induced skin diseases. This review aims to elucidate the biochemical and clinical features of photoaging, the relationship of skin photoaging and chronological aging, the mechanisms underlying skin photoaging and autophagy, and the role of autophagy in skin photoaging.
Collapse
Affiliation(s)
- Xiaojiao Zhong
- Medical Cosmetic Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ying Deng
- Medical Cosmetic Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hongqiu Yang
- Medical Cosmetic Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xiaoshuang Du
- Medical Cosmetic Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ping Liu
- Medical Cosmetic Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Yu Du
- Medical Cosmetic Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
25
|
Zhou Y, Yan J, Qiao L, Zeng J, Cao F, Sheng X, Qi X, Long C, Liu B, Wang X, Yao H, Xiao L. Bone Marrow Mesenchymal Stem Cell-Derived Exosomes Ameliorate Aging-Induced BTB Impairment in Porcine Testes by Activating Autophagy and Inhibiting ROS/NLRP3 Inflammasomes via the AMPK/mTOR Signaling Pathway. Antioxidants (Basel) 2024; 13:183. [PMID: 38397781 PMCID: PMC10886345 DOI: 10.3390/antiox13020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
As a pivotal player in spermatogenesis, the blood-testis barrier (BTB) made from junction apparatus coexisting in Sertoli cells (SCs) is impaired with an increase in age and ultimately induces spermatogenic dysfunction or even infertility. It has been corroborated that bone marrow mesenchymal stem cell (BMSC) transplantation can efficiently repair and regenerate the testicular function. As vital mediators of cell-to-cell communication, MSC-derived exosomes (Exos) can directly serve as therapeutic agents for tissue repair and regeneration. However, the therapeutic value of BMSC-Exos in aging-induced BTB damage remains to be confirmed. In this study, we explored that the old porcine testes had defective autophagy, which aggravated BTB disruption in SCs. BMSC-Exos could decrease ROS production and NLRP3 inflammasome activation but enhanced autophagy and tight junction (TJ) function in D-gal-triggered aging porcine SCs and mouse model testes, according to in vitro and in vivo experiments. Furthermore, rapamycin, NAC, MCC950, and IL-1Ra restored the TJ function in D-gal-stimulated aging porcine SCs, while BMSC-Exos' stimulatory effect on TJ function was inhibited by chloroquine. Moreover, the treatment with BMSC-Exos enhanced autophagy in D-gal-induced aging porcine SCs by means of the AMPK/mTOR signal transduction pathway. These findings uncovered through the present study that BMSC-Exos can enhance the BTB function in aging testes by improving autophagy via the AMPK/mTOR signaling pathway, thereby suppressing ROS production and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Yi Zhou
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| | - Jiale Yan
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| | - Limin Qiao
- Department of Animal Husbandry and Veterinary, Beijing Vocational College of Agriculture, Beijing 102445, China;
| | - Jiaqin Zeng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| | - Fuyu Cao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| | - Xihui Sheng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| | - Xiaolong Qi
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| | - Cheng Long
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| | - Bingying Liu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| | - Xiangguo Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| | - Hua Yao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| | - Longfei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (Y.Z.); (J.Y.); (J.Z.); (F.C.); (X.S.); (X.Q.); (C.L.); (B.L.); (X.W.)
| |
Collapse
|
26
|
Ahamed A, Hosea R, Wu S, Kasim V. The Emerging Roles of the Metabolic Regulator G6PD in Human Cancers. Int J Mol Sci 2023; 24:17238. [PMID: 38139067 PMCID: PMC10743588 DOI: 10.3390/ijms242417238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Metabolic reprogramming, especially reprogrammed glucose metabolism, is a well-known cancer hallmark related to various characteristics of tumor cells, including proliferation, survival, metastasis, and drug resistance. Glucose-6-phosphate dehydrogenase (G6PD) is the first and rate-limiting enzyme of the pentose phosphate pathway (PPP), a branch of glycolysis, that converts glucose-6-phosphate (G6P) into 6-phosphogluconolactone (6PGL). Furthermore, PPP produces ribose-5-phosphate (R5P), which provides sugar-phosphate backbones for nucleotide synthesis as well as nicotinamide adenine dinucleotide phosphate (NADPH), an important cellular reductant. Several studies have shown enhanced G6PD expression and PPP flux in various tumor cells, as well as their correlation with tumor progression through cancer hallmark regulation, especially reprogramming cellular metabolism, sustaining proliferative signaling, resisting cell death, and activating invasion and metastasis. Inhibiting G6PD could suppress tumor cell proliferation, promote cell death, reverse chemoresistance, and inhibit metastasis, suggesting the potential of G6PD as a target for anti-tumor therapeutic strategies. Indeed, while challenges-including side effects-still remain, small-molecule G6PD inhibitors showing potential anti-tumor effect either when used alone or in combination with other anti-tumor drugs have been developed. This review provides an overview of the structural significance of G6PD, its role in and regulation of tumor development and progression, and the strategies explored in relation to G6PD-targeted therapy.
Collapse
Affiliation(s)
- Alfar Ahamed
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Rendy Hosea
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China
| |
Collapse
|
27
|
Teng H, Yang B, Su Y, Chen J, Cui L, Sun R, Zhao J, Liu Q, Qin A. Aminooxyacetic acid hemihydrochloride leads to decreased intracellular ATP levels and altered cell cycle of prostate cancer cells by suppressing energy metabolism. Biomed Pharmacother 2023; 167:115605. [PMID: 37801901 DOI: 10.1016/j.biopha.2023.115605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/08/2023] Open
Abstract
The second most common cancer among men is prostate cancer, which is also the fifth leading reason for male cancer deaths worldwide. Bone metastases are the main factor affecting the prognosis of prostate cancer. Consequently, antitumor and anti-prostate cancer-induced bone destruction medicines are urgently needed. We previously discovered that aminooxyacetic acid hemihydrochloride (AOAA) suppressed bone resorption and osteoclast growth by decreasing adenosine triphosphate (ATP) production and limiting oxidative phosphorylation (OXPHOS). Here, we evaluated the impacts of AOAA on prostate cancer RM-1 cells in vitro. It's found that AOAA significantly inhibited cell proliferation, migration, and invasiveness, decreased ATP levels, increased ROS, halted the cell cycle phase, and triggered apoptosis. AOAA also decreased mitochondrial membrane potential and the ability to uptake glucose, suggesting that the antitumor effects of AOAA were expressed through the inhibition of OXPHOS and glycolysis. Furthermore, we assessed the effects of AOAA in vivo using a prostate cancer-induced bone osteolysis mice model. AOAA also delayed tumor growth and bone destruction in vivo. On the whole, our findings imply that AOAA may potentially have therapeutic effects on prostate cancer and prostate cancer-induced osteolysis.
Collapse
Affiliation(s)
- Hailong Teng
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Biao Yang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuangang Su
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Junchun Chen
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Lei Cui
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Ran Sun
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinmin Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China.
| | - An Qin
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China; Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
28
|
Liu S, Hu Q, Xie Z, Chen S, Li Y, Quan N, Huang K, Li R, Fang L. An endoplasmic reticulum stress-related signature could robustly predict prognosis and closely associate with response to immunotherapy in pancreatic ductal adenocarcinoma. J Cancer Res Clin Oncol 2023; 149:15589-15608. [PMID: 37653101 PMCID: PMC10620278 DOI: 10.1007/s00432-023-05312-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant tumors. Endoplasmic reticulum stress (ERS) plays an essential role in PDAC progression. Here, we aim to identify the ERS-related genes in PDAC and build reliable risk models for diagnosis, prognosis and immunotherapy response of PDAC patients as well as investigate the potential mechanism. METHODS We obtained PDAC cohorts with transcriptional profiles and clinical data from the ArrayExpress, The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. Univariate Cox regression, LASSO regression and multivariate Cox regression analyses were used to construct an ERS-related prognostic signature. The CIBERSORT and ssGSEA algorithms were applied to explore the correlation between the prognostic signature and immune cell infiltration and immune-related pathways. The GDSC database and TIDE algorithm were used to predict responses to chemotherapy and immunotherapy, identifying potential drugs for treating patients with PDAC. RESULTS We established and validated an ERS-related prognostic signature comprising eight genes (HMOX1, TGFB1, JSRP1, GAPDH, CAV1, CHRNE, CD74 and ERN2). Patients with higher risk scores displayed worse outcomes than those with lower risk scores. PDAC patients in low-risk groups might benefit from immunotherapy. Dasatinib and lapatinib might have potential therapeutic implications in high-risk PDAC patients. CONCLUSION We established and validated an ERS-related prognostic signature comprising eight genes to predict the overall survival outcome of PDAC patients, which closely correlating with the response to immunotherapy and sensitivity to anti-tumor drugs, as well as could be beneficial for formulating clinical strategies and administering individualized treatments.
Collapse
Affiliation(s)
- Shuguang Liu
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-Sun University, Shenzhen, 518033, China.
| | - Qianying Hu
- Medical Research Center, The Eighth Affiliated Hospital, Sun Yat-Sun University, Shenzhen, 518033, China
| | - Zishan Xie
- Department of Breast Surgery, The Eighth Affiliated Hospital, Sun Yat-Sun University, Shenzhen, 518033, China
| | - Shaojing Chen
- Medical Research Center, The Eighth Affiliated Hospital, Sun Yat-Sun University, Shenzhen, 518033, China
| | - Yixuan Li
- Medical Research Center, The Eighth Affiliated Hospital, Sun Yat-Sun University, Shenzhen, 518033, China
| | - Nali Quan
- Clinical Laboratory, The Eighth Affiliated Hospital, Sun Yat-Sun University, Shenzhen, 518033, China
| | - Kaimeng Huang
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA.
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Riqing Li
- Shenzhen Agricultural Technology Promotion Center, Shenzhen, 518005, China.
| | - Lishan Fang
- Medical Research Center, The Eighth Affiliated Hospital, Sun Yat-Sun University, Shenzhen, 518033, China.
| |
Collapse
|
29
|
Zhu M, Liang H, Zhang Z, Jiang H, Pu J, Hang X, Zhou Q, Xiang J, He X. Distinct mononuclear diploid cardiac subpopulation with minimal cell-cell communications persists in embryonic and adult mammalian heart. Front Med 2023; 17:939-956. [PMID: 37294383 DOI: 10.1007/s11684-023-0987-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 01/31/2023] [Indexed: 06/10/2023]
Abstract
A small proportion of mononuclear diploid cardiomyocytes (MNDCMs), with regeneration potential, could persist in adult mammalian heart. However, the heterogeneity of MNDCMs and changes during development remains to be illuminated. To this end, 12 645 cardiac cells were generated from embryonic day 17.5 and postnatal days 2 and 8 mice by single-cell RNA sequencing. Three cardiac developmental paths were identified: two switching to cardiomyocytes (CM) maturation with close CM-fibroblast (FB) communications and one maintaining MNDCM status with least CM-FB communications. Proliferative MNDCMs having interactions with macrophages and non-proliferative MNDCMs (non-pMNDCMs) with minimal cell-cell communications were identified in the third path. The non-pMNDCMs possessed distinct properties: the lowest mitochondrial metabolisms, the highest glycolysis, and high expression of Myl4 and Tnni1. Single-nucleus RNA sequencing and immunohistochemical staining further proved that the Myl4+Tnni1+ MNDCMs persisted in embryonic and adult hearts. These MNDCMs were mapped to the heart by integrating the spatial and single-cell transcriptomic data. In conclusion, a novel non-pMNDCM subpopulation with minimal cell-cell communications was unveiled, highlighting the importance of microenvironment contribution to CM fate during maturation. These findings could improve the understanding of MNDCM heterogeneity and cardiac development, thus providing new clues for approaches to effective cardiac regeneration.
Collapse
Affiliation(s)
- Miaomiao Zhu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, `, Wuhan, 430030, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huamin Liang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, `, Wuhan, 430030, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhe Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, `, Wuhan, 430030, China
| | - Hao Jiang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, `, Wuhan, 430030, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingwen Pu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, `, Wuhan, 430030, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoyi Hang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, `, Wuhan, 430030, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian Zhou
- Department of Physiology, School of Basic Medicine, Tongji Medical College, `, Wuhan, 430030, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiacheng Xiang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, `, Wuhan, 430030, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ximiao He
- Department of Physiology, School of Basic Medicine, Tongji Medical College, `, Wuhan, 430030, China.
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
30
|
Javali PS, Sekar M, Kumar A, Thirumurugan K. Dynamics of redox signaling in aging via autophagy, inflammation, and senescence. Biogerontology 2023; 24:663-678. [PMID: 37195483 DOI: 10.1007/s10522-023-10040-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 05/18/2023]
Abstract
Review paper attempts to explain the dynamic aspects of redox signaling in aging through autophagy, inflammation, and senescence. It begins with ROS source in the cell, then states redox signaling in autophagy, and regulation of autophagy in aging. Next, we discuss inflammation and redox signaling with various pathways involved: NOX pathway, ROS production via TNF-α, IL-1β, xanthine oxidase pathway, COX pathway, and myeloperoxidase pathway. Also, we emphasize oxidative damage as an aging marker and the contribution of pathophysiological factors to aging. In senescence-associated secretory phenotypes, we link ROS with senescence, aging disorders. Relevant crosstalk between autophagy, inflammation, and senescence using a balanced ROS level might reduce age-related disorders. Transducing the context-dependent signal communication among these three processes at high spatiotemporal resolution demands other tools like multi-omics aging biomarkers, artificial intelligence, machine learning, and deep learning. The bewildering advancement of technology in the above areas might progress age-related disorders diagnostics with precision and accuracy.
Collapse
Affiliation(s)
- Prashanth S Javali
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Mouliganesh Sekar
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Ashish Kumar
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Kavitha Thirumurugan
- #412J, Structural Biology Lab, Pearl Research Park, School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
31
|
Peng XX, Zhang H, Zhang R, Li ZH, Yang ZS, Zhang J, Gao S, Zhang JL. Gallium Triggers Ferroptosis through a Synergistic Mechanism. Angew Chem Int Ed Engl 2023; 62:e202307838. [PMID: 37452698 DOI: 10.1002/anie.202307838] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/18/2023]
Abstract
The gallium ion (Ga3+ ) has long been believed to disrupt ferric homeostasis in the body by competing with iron cofactors in metalloproteins, ultimately leading to cell death. This study revealed that through an indirect pathway, gallium can trigger ferroptosis, a type of non-apoptotic cell death regulated by iron. This is exemplified by the gallium complex of the salen ligand (Ga-1); we found that Ga-1 acts as an effective anion transporter that can affect the pH gradient and change membrane permeability, leading to mitochondrial dysfunction and the release of ferrous iron from the electron transfer chain (ETC). In addition, Ga-1 also targeted protein disulfide isomerases (PDIs) located in the endoplasmic reticulum (ER) membrane, preventing the repair of the antioxidant glutathione (GSH) system and thus enforcing ferroptosis. Finally, a combination treatment of Ga-1 and dietary polyunsaturated fatty acids (PUFAs), which enhances lipid peroxidation during ferroptosis, showed a synergistic therapeutic effect both in vitro and in vivo. This study provided us with a strategy to synergistically induce Ferroptosis in tumor cells, thereby enhancing the anti-neoplastic effect.
Collapse
Affiliation(s)
- Xin-Xin Peng
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Hang Zhang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Ruijing Zhang
- Spin-X Institute, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
| | - Ze-Hao Li
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zi-Shu Yang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Jing Zhang
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Song Gao
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
- Spin-X Institute, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, 515031, P. R. China
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Jun-Long Zhang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, 515031, P. R. China
| |
Collapse
|
32
|
Zhang L, Chen L, Qi M, Yu F, Ni X, Hong H, Xu H, Xu S. Glyphosate induces autophagy in hepatic L8824 cell line through NO-mediated activation of RAS/RAF/MEK/ERK signaling pathway and energy metabolism disorders. FISH & SHELLFISH IMMUNOLOGY 2023; 137:108772. [PMID: 37100311 DOI: 10.1016/j.fsi.2023.108772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/22/2023]
Abstract
Glyphosate is an herbicide commonly used worldwide, and its substantial use causes widespread pollution with runoff. However, research on glyphosate toxicity has mostly remained at the embryonic level and existing studies are limited. In the present study, we investigated whether glyphosate can induce autophagy in hepatic L8824 cells by regulating energy metabolism and rat sarcoma (RAS)/rapidly accelerated fibrosarcoma (RAF)/mitogen-activated extracellular signal-regulated kinase (MEK)/extracellular regulated protein kinases (ERK) signaling by activating nitric oxide (NO). First, we selected 0, 50, 200, and 500 μg/mL as the challenge doses, according to the inhibitory concentration of 50% (IC50) of glyphosate. The results showed that glyphosate exposure increased the enzyme activity of inducible nitric oxide synthase (iNOS), which in turn increased the NO content. The activity and expression of enzymes related to energy metabolism, such as hexokinase (HK)1, HK2, phosphofructokinase (PFK), phosphokinase (PK), succinate dehydrogenase (SDH), and nicotinamide adenine dinucleotide with hydrogen (NADH), were inhibited, and the RAS/RAF/MEK/ERK signaling pathway was activated. This led to the negative expression of mammalian target of rapamycin (mTOR) and P62 in hepatic L8824 cells and the activation of the autophagy marker genes microtubule-associated proteins light chain 3 (LC3) and Beclin1 to induce autophagy. The above results were dependent on glyphosate concentration. To verify whether autophagy can be excited by the RAS/RAF/MEK/ERK signaling pathway, we treated L8824 cells with the ERK inhibitor U0126 and found that the autophagy gene LC3 was reduced due to the inhibition of ERK, thus demonstrating the reliability of the results. In conclusion, our results demonstrate that glyphosate can induce autophagy in hepatic L8824 cells by activating NO, thus regulating energy metabolism and the RAS/RAF/MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Linlin Zhang
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Lu Chen
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Meng Qi
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Fuchang Yu
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Xiaotong Ni
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Haozheng Hong
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China
| | - Haotian Xu
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China.
| | - Shiwen Xu
- College of Animal Science and Technology, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China; Key Laboratory of Tarim Animal Husbandry Technology Corps, Tarim University, Alar, Xinjiang Uygur Autonomous Region, 843300, PR China.
| |
Collapse
|
33
|
Zhang X, Wang S, Jin Y, Wang J, Wang R, Yang X, Zhang S, Yan T, Jia Y. Wei-Tong-Xin ameliorated cisplatin-induced mitophagy and apoptosis in gastric antral mucosa by activating the Nrf2/HO-1 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116253. [PMID: 36806345 DOI: 10.1016/j.jep.2023.116253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/24/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wei-Tong-Xin (WTX) originated from the famous ancient Chinese formula "Wan Ying Yuan", recorded in the ancient Chinese medicine book "Zhong Zang Jing" by Hua Tuo. As "Jun" drugs, Dahuang and Muxiang have the effects of clearing heat and expelling fire, reducing food retention, regulating Qi and relieving pain. As "Chen" drug, Qianniuzi has the effect of assisting "Jun" drugs. Zhuyazao and Gancao, as "Zuo-Shi" drugs, can reduce toxicity and modulate the medicinal properties of other herbs. AIM OF THE STUDY The present study aimed to investigate the effect and mechanism of WTX on the oxidative stress of gastric antrum mucosa in mice with cisplatin (CIS)-induced dyspepsia. MATERIALS AND. METHODS A variety of experimental methods, including western blot, qRT-PCR, immunofluorescence and immunohistochemistry were performed in vivo and in vitro. RESULTS In vivo, WTX restored the number and function of interstitial cells of Cajal (ICCs), accompanied by the inhibition of lipid peroxidation. Moreover, WTX inhibited the activation of Parkin-dependent mitophagy and apoptosis. In vitro, WTX activated the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway and inactivated mitophagy in GES-1 cells. To explore the role of Nrf2 in WTX's improvement of CIS-induced cell damage, Nrf2 inhibitor ML385 was used in cell experiments. We found that ML385 counteracted the regulation of WTX on mitophagy and apoptosis. Finally, N-acetylcysteine (NAC), a reactive oxygen species (ROS) scavenger, was applied in our experiments, and the results suggested that WTX suppressed the CIS-induced apoptosis via mitochondrial pathway. CONCLUSIONS The above results, for the first time, indicated that WTX inhibited mitophagy and apoptosis of gastric antral mucosal cells induced by CIS through the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Xiaoying Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Shiyu Wang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Yanjun Jin
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Jinyu Wang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Ruixuan Wang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Xihan Yang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Shuanglin Zhang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China.
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, China.
| |
Collapse
|
34
|
Lattos A, Feidantsis K, Giantsis IA, Theodorou JA, Michaelidis B. Seasonality in Synergism with Multi-Pathogen Presence Leads to Mass Mortalities of the Highly Endangered Pinna nobilis in Greek Coastlines: A Pathophysiological Approach. Microorganisms 2023; 11:1117. [PMID: 37317091 DOI: 10.3390/microorganisms11051117] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/22/2023] [Accepted: 04/23/2023] [Indexed: 06/16/2023] Open
Abstract
Mortalities of Pinna nobilis populations set at risk the survival of the species from many Mediterranean coastline habitats. In many cases, both Haplosporidium pinnae and Mycobacterium spp. are implicated in mass mortalities of P. nobilis populations, leading the species into extinction. In the context of the importance of these pathogens' role in P. nobilis mortalities, the present study investigated two Greek populations of the species hosting different microbial loads (one only H. pinnae and the second both pathogens) by the means of pathophysiological markers. More specifically, the populations from Kalloni Gulf (Lesvos Island) and from Maliakos Gulf (Fthiotis), seasonally sampled, were chosen based on the host pathogens in order to investigate physiological and immunological biomarkers to assess those pathogens' roles. In order to determine if the haplosporidian parasite possesses a major role in the mortalities or if both pathogens are involved in these phenomena, a variety of biomarkers, including apoptosis, autophagy, inflammation and heat shock response were applied. The results indicated a decreased physiological performance of individuals hosting both pathogens in comparison with those hosting only H. pinnae. Our findings provide evidence for the synergistic role of those pathogens in the mortality events, which is also enhanced by the influence of seasonality.
Collapse
Affiliation(s)
- Athanasios Lattos
- Laboratory of Animal Physiology, Department of Zoology, Faculty of Science, School of Biology, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Konstantinos Feidantsis
- Laboratory of Animal Physiology, Department of Zoology, Faculty of Science, School of Biology, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Ioannis A Giantsis
- Department of Animal Science, Faculty of Agricultural Sciences, University of Western Macedonia, GR-53100 Florina, Greece
| | - John A Theodorou
- Department of Fisheries & Aquaculture, University of Patras, GR-23200 Mesolonghi, Greece
| | - Basile Michaelidis
- Laboratory of Animal Physiology, Department of Zoology, Faculty of Science, School of Biology, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| |
Collapse
|
35
|
Makri V, Feidantsis K, Porlou D, Ntokou A, Georgoulis I, Giantsis IA, Anestis A, Michaelidis B. Red porgy's (Pagrus pagrus) cellular physiology and antioxidant defense in response to seasonality. J Therm Biol 2023; 113:103527. [PMID: 37055131 DOI: 10.1016/j.jtherbio.2023.103527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Physiological stress patterns of marine organisms in their natural habitats are considerably complex in space and time. These patterns can eventually contribute in the shaping of fish' thermal limits under natural conditions. In the view of the knowledge gap regarding red porgy's thermal physiology, in combination with the characterization of the Mediterranean Sea as a climate change ''hotspot'', the aim of the present study was to investigate this species biochemical responses to constantly changing field conditions. To achieve this goal, Heat Shock Response (HSR), MAPKs pathway, autophagy, apoptosis, lipid peroxidation and antioxidant defense were estimated and exhibited a seasonal pattern. In general, all the examined biochemical indicators expressed high levels parallel to the increasing seawater temperature in spring, although several bio-indicators have shown increased levels when fish were cold-acclimatized. Similar to other sparids, the observed patterns of physiological responses in red porgy may support the concept of eurythermy.
Collapse
|
36
|
Guo J, Zhou M, Zhao M, Li S, Fang Z, Li A, Zhang M. TIGAR deficiency induces caspase-1-dependent trophoblasts pyroptosis through NLRP3-ASC inflammasome. Front Immunol 2023; 14:1114620. [PMID: 37122710 PMCID: PMC10140348 DOI: 10.3389/fimmu.2023.1114620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/06/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Gestational diabetes mellitus (GDM), a common complication of pregnancy, is risky for both mother and fetus. Previous studies about TP53-induced glycolysis and apoptosis regulator (TIGAR) focused on the occurrence and development of cancer, cardiovascular disease, and neurological disease, however, it is still unclear whether TIGAR plays a regulatory role in gestational diabetes mellitus (GDM). Methods Utilizing HG exposure, we explored the role of TIGAR in oxidative stress limitation, excessive inflammatory toxicity defense, and pyroptosis prevention. Results TIGAR was up-regulated in vivo and in vitro under HG condition, and loss of TIGAR increased ROS in trophoblast cells which drove a phenotypic switch and hindered the capacity of migration, invasion, and tube formation. This switch depended on the increased activation of NLRP3-ASC-caspase-1 signaling, which caused a distinctive characteristic of pyroptosis, and these findings could finally be reverted by antioxidant treatment (NAC) and receptor block (MCC950). Collectively, trophoblast pyroptosis is an upstream event of TIGAR deficiency-induced inflammation, which is promoted by ROS accumulation through NLRP3-ASC inflammasome. Conclusion Taken together, our results uncovered that, as the upstream event of TIGAR deficiency-induced inflammation, pyroptosis is stimulated by ROS accumulation through NLRP3-ASC inflammasome.
Collapse
Affiliation(s)
| | | | | | | | | | - Anna Li
- *Correspondence: Anna Li, ; Meihua Zhang,
| | | |
Collapse
|
37
|
Intracellular galectin-3 is a lipopolysaccharide sensor that promotes glycolysis through mTORC1 activation. Nat Commun 2022; 13:7578. [PMID: 36481721 PMCID: PMC9732310 DOI: 10.1038/s41467-022-35334-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
How the carbohydrate binding protein galectin-3 might act as a diabetogenic and tumorogenic factor remains to be investigated. Here we report that intracellular galectin-3 interacts with Rag GTPases and Ragulator on lysosomes. We show that galectin-3 senses lipopolysaccharide (LPS) to facilitate the interaction of Rag GTPases and Ragulator, leading to the activation of mTORC1. We find that the lipopolysaccharide/galectin-3-Rag GTPases/Ragulator-mTORC1 axis regulates a cohort of genes including GLUT1, and HK2, and PKM2 that are critically involved in glucose uptake and glycolysis. Indeed, galectin-3 deficiency severely compromises LPS-promoted glycolysis. Importantly, the expression of HK2 is significantly reduced in diabetes patients. In multiple types of cancer including hepatocellular carcinoma (HCC), galectin-3 is highly expressed, and its level of expression is positively correlated with that of HK2 and PKM2 and negatively correlated with the prognosis of HCC patients. Our study unravels that galectin-3 is a sensor of LPS, an important modulator of the mTORC1 signaling, and a critical regulator of glucose metabolism.
Collapse
|
38
|
Therapeutic Stimulation of Glycolytic ATP Production for Treating ROS-Mediated Cellular Senescence. Metabolites 2022; 12:metabo12121160. [PMID: 36557198 PMCID: PMC9781421 DOI: 10.3390/metabo12121160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Cellular senescence is conditioned through two interrelated processes, i.e., a reduction in adenosine triphosphate (ATP) and the enhancement of reactive oxygen species (ROS) production levels in mitochondria. ATP shortages primarily influence the energy-intensive synthesis of large biomolecules, such as deoxyribonucleic acid (DNA). In addition, as compared to small biomolecules, large biomolecules are more prone to ROS-mediated damaging effects. Based on the available evidence, we suggest that the stimulation of anaerobic glycolytic ROS-independent ATP production could restrain cellular senescence. Consistent with this notion, non-drug related intermittent hypoxia (IH)-based therapy could be effectively applied in sports medicine, as well as for supporting the physical activity of elderly patients and prophylactics of various age-related disorders. Moreover, drug therapy aiming to achieve the partial blockade of respiratory chain and downstream compensatory glycolysis enhancement could prove to be useful for treating cardiovascular, neurological and hormonal diseases. We maintain that non-drug/drug-related therapeutic interventions applied in combination over the entire lifespan could significantly rejuvenate and prolong a high quality of life for individuals.
Collapse
|
39
|
Huang B, Lang X, Li X. The role of TIGAR in nervous system diseases. Front Aging Neurosci 2022; 14:1023161. [DOI: 10.3389/fnagi.2022.1023161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/05/2022] [Indexed: 11/10/2022] Open
Abstract
TP53-induced glycolysis and apoptosis regulator (TIGAR) mainly regulates pentose phosphate pathway by inhibiting glycolysis, so as to synthesize ribose required by DNA, promote DNA damage repair and cell proliferation, maintain cell homeostasis and avoid body injury. Its physiological functions include anti-oxidative stress, reducing inflammation, maintaining mitochondrial function, inhibiting apoptosis, reducing autophagy etc. This paper reviews the research of TIGAR in neurological diseases, including stroke, Parkinson’s disease (PD), Alzheimer’s disease (AD), seizures and brain tumors, aiming to provide reference for the development of new therapeutic targets.
Collapse
|
40
|
Bousdras T, Feidantsis K, Panteli N, Chatzifotis S, Piccolo G, Gasco L, Gai F, Antonopoulou E. Dietary Tenebrio molitor Larvae Meal Inclusion Exerts Tissue-Specific Effects on Cellular, Metabolic, and Antioxidant Status in European Sea Bass ( Dicentrarchus labrax) and Gilthead Seabream ( Sparus aurata). AQUACULTURE NUTRITION 2022; 2022:9858983. [PMID: 36860434 PMCID: PMC9973136 DOI: 10.1155/2022/9858983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 09/30/2022] [Indexed: 06/01/2023]
Abstract
The present study addresses the effects of dietary Tenebrio molitor (TM) larvae meal inclusion on cytoprotective, cell death pathways, antioxidant defence, and intermediate metabolism in the heart, muscle, and digestive tract of gilthead seabream (Sparus aurata) and European sea bass (Dicentrarchus labrax). Three experimental diets were formulated to contain 0%, 25%, or 50% inclusion TM levels. Heat Shock Proteins (HSPs) induction was apparent in both species' muscle at 50% inclusion. Conversely, p44/42 Mitogen-Activated Protein Kinase (MAPK) activation was increased (p < 0.05) in both species' muscle and digestive tract at 25% inclusion. Regarding the apoptotic machinery, TM inclusion exerted no influence on gilthead seabream, while suppression through autophagy may have occurred in the muscle. However, significant apoptosis (p < 0.05) was evident in European sea bass muscle and digestive tract. Both fish species' heart seemed to additionally rely on lipids compared to muscle and digestive tract. In contrast to gilthead seabream, European sea bass exhibited increased (p < 0.05) antioxidant activity at 50% TM inclusion. The present findings highlight the dietary derived induction of cellular responses in a species- and tissue-specific manner, whereas European sea bass appears to be more susceptible to TM inclusion.
Collapse
Affiliation(s)
- Thomas Bousdras
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Konstantinos Feidantsis
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Nikolas Panteli
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Stavros Chatzifotis
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research, Gournes Pediados, P.O. Box 2214, GR-71003, Heraklion, Crete, Greece
| | - Giovanni Piccolo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy
| | - Laura Gasco
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, Italy
| | - Francesco Gai
- Institute of Sciences of Food Production, National Research Council, Largo Paolo Braccini 2, 10095 Grugliasco, Italy
| | - Efthimia Antonopoulou
- Laboratory of Animal Physiology, Department of Zoology, School of Biology, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| |
Collapse
|
41
|
Yan F, Zhao Q, Li Y, Zheng Z, Kong X, Shu C, Liu Y, Shi Y. The role of oxidative stress in ovarian aging: a review. J Ovarian Res 2022; 15:100. [PMID: 36050696 PMCID: PMC9434839 DOI: 10.1186/s13048-022-01032-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 08/21/2022] [Indexed: 11/29/2022] Open
Abstract
Ovarian aging refers to the process by which ovarian function declines until eventual failure. The pathogenesis of ovarian aging is complex and diverse; oxidative stress (OS) is considered to be a key factor. This review focuses on the fact that OS status accelerates the ovarian aging process by promoting apoptosis, inflammation, mitochondrial damage, telomere shortening and biomacromolecular damage. Current evidence suggests that aging, smoking, high-sugar diets, pressure, superovulation, chemotherapeutic agents and industrial pollutants can be factors that accelerate ovarian aging by exacerbating OS status. In addition, we review the role of nuclear factor E2-related factor 2 (Nrf2), Sirtuin (Sirt), mitogen-activated protein kinase (MAPK), protein kinase B (AKT), Forkhead box O (FoxO) and Klotho signaling pathways during the process of ovarian aging. We also explore the role of antioxidant therapies such as melatonin, vitamins, stem cell therapies, antioxidant monomers and Traditional Chinese Medicine (TCM), and investigate the roles of these supplements with respect to the reduction of OS and the improvement of ovarian function. This review provides a rationale for antioxidant therapy to improve ovarian aging.
Collapse
Affiliation(s)
- Fei Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Qi Zhao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Ying Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Zhibo Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Xinliang Kong
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Chang Shu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Yanfeng Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China.
| | - Yun Shi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People's Republic of China.
| |
Collapse
|
42
|
Wang Y, Zhang Z, Sun W, Zhang J, Xu Q, Zhou X, Mao L. Ferroptosis in colorectal cancer: Potential mechanisms and effective therapeutic targets. Biomed Pharmacother 2022; 153:113524. [PMID: 36076606 DOI: 10.1016/j.biopha.2022.113524] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 01/17/2023] Open
|
43
|
Neuroprotective effect of geraniol on neurological disorders: a review article. Mol Biol Rep 2022; 49:10865-10874. [PMID: 35900613 DOI: 10.1007/s11033-022-07755-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/29/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Neurological disorders are structural, biochemical, and electrical abnormalities that affect the peripheral and central nervous systems. Paralysis, muscle weakness, tremors, spasms, and partial or complete loss of sensation are some symptoms of these disorders. Neurorehabilitation is the main treatment for neurological disorders. Treatments can improve the quality of life of patients. Neuroprotective substances of natural origin are used for the treatments of these disorders. METHODS AND RESULTS Online databases, such as Google Scholar, PubMed, ScienceDirect, and Scopus were searched to evaluate articles from 1981-2021 using the Mesh words of geraniol (GER), neurological disorders, epilepsy, spinal cord injury (SCI), Parkinson's diseases (PD), and depression. A total of 87 studies were included in this review. GER with antioxidant, anti-inflammatory, and neuroprotective effects can improve the symptoms and reduce the progression of neurological diseases. GER exhibits neuroprotective effects by binding to GABA and glycine receptors as well as by inhibiting the activation of nuclear factor kappa B (NF-κB) pathway and regulating the expression of nucleotide-binding oligomerization of NLRP3 inflammasome. In this study, the effect of GER was investigated on neurological disorders, such as epilepsy, SCI, PD, and depression. CONCLUSION Although the medicinal uses of GER have been reported, more clinical and experimental studies are needed to investigate the effect of using traditional medicine on improving lifethreatening diseases and the quality of life of patients.
Collapse
|
44
|
Drehmer E, Navarro-Moreno MÁ, Carrera-Juliá S, Moreno ML. A comparative study between olive oil and corn oil on oxidative metabolism. Food Funct 2022; 13:7157-7167. [PMID: 35699154 DOI: 10.1039/d2fo00919f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fats are an important part of diet, but not all lipids have the same structure and chemical properties. Unsaturated fatty acids have one or more double bonds in their structure and can be monounsaturated or polyunsaturated, respectively. Most vegetable oils, such as olive oil and corn oil, contain significant amounts of these fatty acids. The presence of double bonds in the molecule of a fatty acid constitutes vulnerable sites for oxidation reactions generating lipid peroxides, potentially toxic compounds that can cause cellular damage. In response to this oxidative damage, aerobic organisms have intracellular enzymatic antioxidant defense mechanisms. The aim of the present investigation was to study comparatively the effects of control liquid diets, of a defined composition, containing olive oil or corn oil as a lipid source respectively of monounsaturated and polyunsaturated fatty acids, on the oxidative metabolism of rats. Rats were divided into three groups which received a control animal feed diet (A.F.), olive oil liquid diet (O.O) and corn oil liquid diet (C.O) for 30 days. It was observed that the activity of the antioxidant enzymes superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx), increased in the liver and white fat tissue of rats fed with olive oil when compared to the corn oil group. However, in brown fat tissue and blood cells, the enzyme activities showed a tendency to decrease in the olive oil group. In addition, the effect of olive oil and corn oil on several glucose metabolism parameters (pyruvate, lactate, LDH, acetoacetate and beta-hydroxybutyrate) showed that corn oil impairs to a greater extent the cellular metabolism. All these results helped in concluding that some body tissues are more adversely affected than others by the administration of corn oil or olive oil, and their antioxidant defenses and cellular metabolism respond differently too.
Collapse
Affiliation(s)
- Eraci Drehmer
- Department of Health Sciences, Universidad Católica de Valencia "San Vicente Mártir", Valencia, Spain
| | | | - Sandra Carrera-Juliá
- Department of Nutrition and Dietetics, Universidad Católica de Valencia "San Vicente Mártir", Valencia, Spain
| | - Mari Luz Moreno
- Department of Human Physiology and Anatomy, Universidad Católica de Valencia "San Vicente Mártir", C/Ramiro de Maeztu, 14., 46900 Torrente, Valencia, Spain.
| |
Collapse
|
45
|
Daniels RC, Tiba MH, Cummings B, Yap YR, Ansari S, McCracken B, Sun Y, Jennaro T, Ward KR, Stringer KA. Redox Potential Correlates with Changes in Metabolite Concentrations Attributable to Pathways Active in Oxidative Stress Response in Swine Traumatic Shock. Shock 2022; 57:282-290. [PMID: 35670453 PMCID: PMC10314677 DOI: 10.1097/shk.0000000000001944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Oxidation-reduction (redox) reactions, and the redox potential (RP) that must be maintained for proper cell function, lie at the heart of physiologic processes in critical illness. Imbalance in RP reflects systemic oxidative stress, and whole blood RP measures have been shown to correlate with oxygen debt level over time in swine traumatic shock. We hypothesize that RP measures reflect changing concentrations of metabolites involved in oxidative stress. To test this hypothesis, we compared blood and urine RP with concentrations of multiple metabolites in a swine traumatic shock model to identify meaningful RP-metabolite relationships. METHODS Seven swine were subjected to traumatic shock. Mixed venous (MV) RP, urine RP, and concurrent MV and urine metabolite concentrations were assessed at baseline, max O 2 Debt (80 mL/kg), end resuscitation, and 2 h post-resuscitation. RP was measured at collection via open circuit potential using nanoporous gold electrodes with Ag/AgCl reference and a ParstatMC potentiostat. Metabolite concentrations were measured by quantitative 1 H-NMR spectroscopy. MV and urine RP were compared with time-matched metabolites across all swine. LASSO regression with leave-one-out cross validation was used to determine meaningful RP/metabolite relationships. Metabolites had to maintain magnitude and direction of coefficients across 6 or more swine to be considered as having a meaningful relationship. KEGG IDs of these metabolites were uploaded into Metscape for pathway identification and evaluation for physiologic function. RESULTS Meaningful metabolite relationships (and mean coefficients across cross-validation folds) with MV RP included: choline (-6.27), ATP (-4.39), glycine (5.93), ADP (1.84), glucose (15.96), formate (-13.09), pyruvate (6.18), and taurine (-7.18). Relationships with urine RP were: betaine (4.81), urea (4.14), glycine (-2.97), taurine (10.32), 3-hydroxyisobutyrate (-7.67), N-phenylacetylglycine, PAG (-14.52), hippurate (12.89), and formate (-5.89). These meaningful metabolites were found to scavenge extracellular peroxide (pyruvate), inhibit ROS and activate cellular antioxidant defense (taurine), act as indicators of antioxidant mobilization against oxidative stress (glycine + PAG), and reflect renal hydroxyl radical trapping (hippurate), among other activities. CONCLUSIONS Real-time RP measures demonstrate significant relationships with metabolites attributable to metabolic pathways involved in systemic responses to oxidative stress, as well as those involved in these processes. These data support RP measures as a feasible, biologically relevant marker of oxidative stress. As a direct measure of redox state, RP may be a useful biomarker and clinical tool in guiding diagnosis and therapy in states of increased oxidative stress and may offer value as a marker for organ injury in these states as well.
Collapse
Affiliation(s)
- Rodney C. Daniels
- Pediatric Critical Care Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI
- Michigan Center for Integrative Research in Critical Care (MCIRCC), University of Michigan, Ann Arbor, MI
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI
| | - M. Hakam Tiba
- Michigan Center for Integrative Research in Critical Care (MCIRCC), University of Michigan, Ann Arbor, MI
- Department of Emergency Medicine, University of Michigan, Ann Arbor, MI
| | - Brandon Cummings
- Michigan Center for Integrative Research in Critical Care (MCIRCC), University of Michigan, Ann Arbor, MI
| | - Yan Rou Yap
- Pediatric Critical Care Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI
- Michigan Center for Integrative Research in Critical Care (MCIRCC), University of Michigan, Ann Arbor, MI
| | - Sardar Ansari
- Michigan Center for Integrative Research in Critical Care (MCIRCC), University of Michigan, Ann Arbor, MI
| | - Brendan McCracken
- Michigan Center for Integrative Research in Critical Care (MCIRCC), University of Michigan, Ann Arbor, MI
- Department of Emergency Medicine, University of Michigan, Ann Arbor, MI
| | - Yihan Sun
- NMR Metabolomics Laboratory, Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI
- Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Teddy Jennaro
- NMR Metabolomics Laboratory, Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI
- Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Kevin R. Ward
- Michigan Center for Integrative Research in Critical Care (MCIRCC), University of Michigan, Ann Arbor, MI
- Department of Emergency Medicine, University of Michigan, Ann Arbor, MI
| | - Kathleen A. Stringer
- Michigan Center for Integrative Research in Critical Care (MCIRCC), University of Michigan, Ann Arbor, MI
- NMR Metabolomics Laboratory, Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI
- Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor, MI
| |
Collapse
|
46
|
Chen J, Tang YX, Kang JX, Xu YR, Elsherbeni AIA, Gharib HBA, Li JL. Astragalus polysaccharide alleviates transport stress-induced heart injury in newly hatched chicks via ERS-UPR-Autophagy dependent pathway. Poult Sci 2022; 101:102030. [PMID: 35905545 PMCID: PMC9334333 DOI: 10.1016/j.psj.2022.102030] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/12/2022] [Accepted: 06/20/2022] [Indexed: 02/08/2023] Open
Abstract
Transport stress (TS) not only affects animal welfare but also eventually leads to higher morbidity and mortality. Moreover, TS could induce heart injury in animals, but the possible mechanism has yet to be fully explored. Astragalus polysaccharide (APS) is a main active component of Radix Astragali, which has an extensive anti-stress effect. However, the effect of APS on TS-induced heart injury has not yet been elucidated. In this study, a chick model of simulated TS was used. 240 newly hatched chicks were arranged into 4 groups: Control (Con), Transport group (T), Transport + water group (TW), and Transport + APS group (TA). Before transport, the chicks of the TW and TA groups were treated with deionized water and APS (0.25 mg/mL, 100 µL) by oral drops respectively. The histopathological analysis of myocardial tissue was assessed by hematoxylin and eosin staining. qRT-PCR and Western Blotting assays were employed to measure the expression of genes and proteins. Semiquantitative PCR was performed for the X box-binding protein-1 (XBP-1) mRNA splicing assay. The results indicated that APS significantly reduced TS-induced myocardial histopathological changes. Meanwhile, TS induced endoplasmic reticulum stress (ERS), evidenced by an activation of the unfolded protein response (UPR) signaling pathway and up-regulation of ERS-markers (P < 0.05). Moreover, TS markedly triggered autophagy induction by activating AMP-activated protein kinase (AMPK), reflected by augmented LC3-II/LC3-I, AMPK phosphorylation and autophagy-related genes (ATGs) expression (P < 0.05). Importantly, our study manifested that treatment of APS could reduce TS-induced ERS and AMPK-activated autophagy, accordingly alleviating heart injury of transported chicks. In summary, these findings indicate that TS induces heart injury in chicks via an ERS-UPR-autophagy-dependent pathway, and APS as an effective therapeutic method to alleviate it.
Collapse
Affiliation(s)
- Jian Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yi-Xi Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jian-Xun Kang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Ya-Ru Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | | | | | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
47
|
Yuan Y, Liang B, Liu XL, Liu WJ, Huang BH, Yang SB, Gao YZ, Meng JS, Li MJ, Ye T, Wang CZ, Hu XK, Xing DM. Targeting NAD+: is it a common strategy to delay heart aging? Cell Death Dis 2022; 8:230. [PMID: 35474295 PMCID: PMC9042931 DOI: 10.1038/s41420-022-01031-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/17/2022]
Abstract
Heart aging is the main susceptible factor to coronary heart disease and significantly increases the risk of heart failure, especially when the aging heart is suffering from ischemia-reperfusion injury. Numerous studies with NAD+ supplementations have suggested its use in anti-aging treatment. However, systematic reviews regarding the overall role of NAD+ in cardiac aging are scarce. The relationship between NAD+ signaling and heart aging has yet to be clarified. This review comprehensively summarizes the current studies on the role of NAD+ signaling in delaying heart aging from the following aspects: the influence of NAD+ supplementations on the aging heart; the relationship and cross-talks between NAD+ signaling and other cardiac aging-related signaling pathways; Importantly, the therapeutic potential of targeting NAD+ in delaying heart aging will be discussed. In brief, NAD+ plays a vital role in delaying heart aging. However, the abnormalities such as altered glucose and lipid metabolism, oxidative stress, and calcium overload could also interfere with NAD+ function in the heart. Therefore, the specific physiopathology of the aging heart should be considered before applying NAD+ supplementations. We believe that this article will help augment our understanding of heart aging mechanisms. In the meantime, it provides invaluable insights into possible therapeutic strategies for preventing age-related heart diseases in clinical settings.
Collapse
Affiliation(s)
- Yang Yuan
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Bing Liang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Xin-Lin Liu
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Wen-Jing Liu
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Bing-Huan Huang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Shan-Bo Yang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Yuan-Zhen Gao
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Jing-Sen Meng
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Meng-Jiao Li
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Ting Ye
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Chuan-Zhi Wang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Xiao-Kun Hu
- Interventional Medicine Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dong-Ming Xing
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China. .,School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
48
|
Mooli RGR, Mukhi D, Ramakrishnan SK. Oxidative Stress and Redox Signaling in the Pathophysiology of Liver Diseases. Compr Physiol 2022; 12:3167-3192. [PMID: 35578969 PMCID: PMC10074426 DOI: 10.1002/cphy.c200021] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The increased production of derivatives of molecular oxygen and nitrogen in the form of reactive oxygen species (ROS) and reactive nitrogen species (RNS) lead to molecular damage called oxidative stress. Under normal physiological conditions, the ROS generation is tightly regulated in different cells and cellular compartments. Any disturbance in the balance between the cellular generation of ROS and antioxidant balance leads to oxidative stress. In this article, we discuss the sources of ROS (endogenous and exogenous) and antioxidant mechanisms. We also focus on the pathophysiological significance of oxidative stress in various cell types of the liver. Oxidative stress is implicated in the development and progression of various liver diseases. We narrate the master regulators of ROS-mediated signaling and their contribution to liver diseases. Nonalcoholic fatty liver diseases (NAFLD) are influenced by a "multiple parallel-hit model" in which oxidative stress plays a central role. We highlight the recent findings on the role of oxidative stress in the spectrum of NAFLD, including fibrosis and liver cancer. Finally, we provide a brief overview of oxidative stress biomarkers and their therapeutic applications in various liver-related disorders. Overall, the article sheds light on the significance of oxidative stress in the pathophysiology of the liver. © 2022 American Physiological Society. Compr Physiol 12:3167-3192, 2022.
Collapse
Affiliation(s)
- Raja Gopal Reddy Mooli
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dhanunjay Mukhi
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sadeesh K Ramakrishnan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
49
|
Teng X, Brown J, Morel L. Redox Homeostasis Involvement in the Pharmacological Effects of Metformin in Systemic Lupus Erythematosus. Antioxid Redox Signal 2022; 36:462-479. [PMID: 34619975 PMCID: PMC8982129 DOI: 10.1089/ars.2021.0070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 09/13/2021] [Accepted: 10/05/2021] [Indexed: 12/21/2022]
Abstract
Significance: Metformin has been proposed as a treatment for systemic lupus erythematosus (SLE). The primary target of metformin, the electron transport chain complex I in the mitochondria, is associated with redox homeostasis in immune cells, which plays a critical role in the pathogenesis of autoimmune diseases. This review addresses the evidence and knowledge gaps on whether a beneficial effect of metformin in lupus may be due to a restoration of a balanced redox state. Recent Advances: Clinical trials in SLE patients with mild-to-moderate disease activity and preclinical studies in mice have provided encouraging results for metformin. The mechanism by which this therapeutic effect was achieved is largely unknown. Metformin regulates redox homeostasis in a context-specific manner. Multiple cell types contribute to SLE, with evidence of increased mitochondrial oxidative stress in T cells and neutrophils. Critical Issues: The major knowledge gaps are whether the efficacy of metformin is linked to a restored redox homeostasis in the immune system, and if it does, in which cell types it occurs? We also need to know which patients may have a better response to metformin, and whether it corresponds to a specific mechanism? Finally, the identification of biomarkers to predict treatment outcomes would be of great value. Future Directions: Mechanistic studies must address the context-dependent pharmacological effects of metformin. Multiple cell types as well as a complex disease etiology should be considered. These studies must integrate the rapid advances made in understanding how metabolic programs direct the effector functions of immune cells. Antioxid. Redox Signal. 36, 462-479.
Collapse
Affiliation(s)
- Xiangyu Teng
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Josephine Brown
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Laurence Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
50
|
Han FX, Zhang R, Yang XX, Ma SB, Hu SJ, Li B. Neuroprotective effect of aldose reductase knockout in a mouse model of spinal cord injury involves NF-κB pathway. Exp Brain Res 2022; 240:853-859. [PMID: 35066597 DOI: 10.1007/s00221-021-06223-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/13/2021] [Indexed: 11/27/2022]
Abstract
The inflammatory response following spinal cord injury (SCI) involves the activation of resident microglia and the infiltration of macrophages. Activated microglia/macrophages have either detrimental or beneficial effects on neural regeneration based on their functional polarized M1/M2 subsets. Aldose reductase (AR) has recently been shown to be a key component of the innate immune response. However, the mechanisms involved in AR and innate immune response remain unclear. In this study, wild-type (WT) or AR-deficiency (KO) mice were subjected to SCI by a spinal crush injury model. AR KO mice showed better locomotor recovery and smaller injury lesion areas after spinal cord crushing compared with WT mice. Here, we first demonstrated that AR deficiency repressed the expression level of inducible nitric oxide synthase (iNOS) induced by lipopolysaccharide (LPS) in vitro via the activation of autophagy. AR deficiency caused 4-hydroxy-2-(E)-nonenal (4-HNE) accumulation in LPS-induced macrophages. We also found that exogenous addition of low concentrations of 4-HNE in LPS-induced macrophages had the effect of promoting further activation of NF-κB pathway, whereas high concentrations of 4-HNE had inhibitory effects. Together, these results indicated that autophagy as a mechanism underlying AR and 4-HNE in LPS-induced macrophages.
Collapse
Affiliation(s)
- Fu-Xin Han
- Department of Neurosurgery, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, 710004, P. R. China
| | - Rui Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Xin-Xing Yang
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Shan-Bo Ma
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China
| | - Shi-Jie Hu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China.
| | - Bing Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, P. R. China.
| |
Collapse
|