1
|
Seale B, Slotabec L, Nguyen JD, Wang H, Patterson C, Filho F, Rouhi N, Adenawoola MI, Li J. Sestrin2 serves as a scaffold protein to maintain cardiac energy and metabolic homeostasis during pathological stress. FASEB J 2024; 38:e70106. [PMID: 39404019 DOI: 10.1096/fj.202401404r] [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: 06/19/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024]
Abstract
Cardiovascular diseases (CVDs) are a leading cause of morbidity and mortality worldwide. Metabolic imbalances and pathological stress often contribute to increased mortality. Sestrin2 (Sesn2) is a stress-inducible protein crucial in maintaining cardiac energy and metabolic homeostasis under pathological conditions. Sesn2 is upregulated in response to various stressors, including oxidative stress, hypoxia, and energy depletion, and mediates multiple cellular pathways to enhance antioxidant defenses, promote autophagy, and inhibit inflammation. This review explores the mechanisms through which Sesn2 regulates these pathways, focusing on the AMPK-mTORC1, Sesn2-Nrf2, and HIF1α-Sesn2 pathways, among others. We can identify the potential therapeutic targets for treating CVDs and related metabolic disorders by comprehending these complex mechanisms. Sesn2's unique ability to respond thoroughly to metabolic challenges, oxidative stress, and inflammation makes it a promising prospect for enhancing cardiac health and resilience against pathological stress.
Collapse
Affiliation(s)
- Blaise Seale
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lily Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Research, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Jennie D Nguyen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Cory Patterson
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Fernanda Filho
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael I Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Research, G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
2
|
Huang Y, Cui Y, Huang J, Xinyuan H, Zihang W, Luo T, Li J. Proanthocyanidins protects 3-NPA-induced ovarian function decline by activating SESTRIN2-NRF2-mediated oxidative stress in mice. Sci Rep 2024; 14:25643. [PMID: 39465303 PMCID: PMC11514188 DOI: 10.1038/s41598-024-76743-w] [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: 04/22/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
Abnormal apoptosis of ovarian cells caused by oxidative stress is an important cause of premature ovarian failure (POF). Previous studies revealed that proanthocyanidins (PCs) are powerful natural antioxidants that can safely prevent oxidative damage in humans. However, the protective effect and mechanism of PCs on ovarian function during the course of POF remain unknown. In this study, female mice were injected with 3-nitropropionic acid (3-NPA) to establish an ovarian oxidative stress model; at the same time, the mice were treated with PC via gavage. Thereafter, the expression of various apoptosis genes, hormones, and related molecules was assessed. Compared with those in the control group, the ovarian index, follicle count at all levels, expression of MVH, PCNA and BCL2, and estradiol (E2) and progesterone (P) levels were significantly lower in the POF group, but significant recovery was observed in terms of MVH and PCNA expression and E2 and P levels in the POF + PCs group. The apoptosis marker genes BAX and ROS were significantly increased in the POF group but were notably restored in the POF + PCs group. In addition, the expression of Sestrin2, an antiapoptotic protein, was significantly increased in the PCs treatment group, as were the upstream and downstream regulatory factors NRF2 and SOD2, and the indices of the Sestrin2 overexpression group were similar to those of the PCs treatment group. In summary, these findings suggest that PCs have potential as innovative therapeutic agents for preventing and treating POF by activating the protective SESTRIN2-NRF2 pathway against oxidative stress.
Collapse
Affiliation(s)
- Yupei Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Yanfan Cui
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Jian Huang
- Clinical Medicine Center, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Huang Xinyuan
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Wang Zihang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Tao Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
- Institute of Biomedical Innovation, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Jia Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China.
| |
Collapse
|
3
|
Rouholamini FS, Aminaei M, Aminizadeh S. The effect of eight weeks of endurance training and MitoQ supplementation on antioxidant capacity and the expression of sestrin-2 and AMPK in cardiac tissue of aged rats. Exp Gerontol 2024; 196:112572. [PMID: 39233194 DOI: 10.1016/j.exger.2024.112572] [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: 05/12/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/06/2024]
Abstract
PURPOSE The present study aimed to investigate the effects of endurance training (ET) in combination with MitoQ supplementation on antioxidant indices and the expression of sesterin-2 (SESN2) as an anti-aging factor and AMPK as an energy sensor in aged male Wistar rats. METHODS Twenty-eight aged Wistar rats (410 ± 15 g, 22 ± 1.5 months old) were randomly divided into four groups (n = 7): Control, ET (eight weeks endurance training on the treadmill), MitoQ (250 μ/L in drinking water), and ET + MitoQ. We measured the protein and gene expression of SESN2 and AMPK in the heart tissue by western blotting and real-time PCR, respectively. In addition, antioxidant indices, superoxide dismutase (SOD), and glutathione peroxidase (GPx) activity, and oxidant malondialdehyde (MDA) concentration in the cardiac tissue and serum were measured. RESULTS SESN2 and AMPK protein expression significantly increased in the MitoQ group compared to the control group (P = 0.002, P = 0.0003). MDA content in tissue and serum remained unchanged in all groups (P > 0.05). MitoQ supplementation significantly increased SOD and GPx enzyme activity in serum and cardiac tissue (P = 0.001). CONCLUSION Overall, ET and MitoQ alone and in combination have anti-aging effects and improve the expression of AMPK and SESN2. Additionally, ET and MitoQ lead to improved antioxidant capacity in aged rats by ameliorating the activity of antioxidant enzymes.
Collapse
Affiliation(s)
- Fatemeh Sadat Rouholamini
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mohsen Aminaei
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Soheil Aminizadeh
- Physiology Research Center, Institute of Neuropharmacology, Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
4
|
Haidurov A, Budanov AV. Locked in Structure: Sestrin and GATOR-A Billion-Year Marriage. Cells 2024; 13:1587. [PMID: 39329768 PMCID: PMC11429811 DOI: 10.3390/cells13181587] [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: 07/11/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Sestrins are a conserved family of stress-responsive proteins that play a crucial role in cellular metabolism, stress response, and ageing. Vertebrates have three Sestrin genes (SESN1, SESN2, and SESN3), while invertebrates encode only one. Initially identified as antioxidant proteins that regulate cell viability, Sestrins are now recognised as crucial inhibitors of the mechanistic target of rapamycin complex 1 kinase (mTORC1), a central regulator of anabolism, cell growth, and autophagy. Sestrins suppress mTORC1 through an inhibitory interaction with the GATOR2 protein complex, which, in concert with GATOR1, signals to inhibit the lysosomal docking of mTORC1. A leucine-binding pocket (LBP) is found in most vertebrate Sestrins, and when bound with leucine, Sestrins do not bind GATOR2, prompting mTORC1 activation. This review examines the evolutionary conservation of Sestrins and their functional motifs, focusing on their origins and development. We highlight that the most conserved regions of Sestrins are those involved in GATOR2 binding, and while analogues of Sestrins exist in prokaryotes, the unique feature of eukaryotic Sestrins is their structural presentation of GATOR2-binding motifs.
Collapse
Affiliation(s)
- Alexander Haidurov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, D02 R590 Dublin, Ireland
| | - Andrei V. Budanov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, D02 R590 Dublin, Ireland
| |
Collapse
|
5
|
Wang Y, Zhao Y, Gong W, Hou Y, Ren J, Duan C, Zhang H, Nie X, Li J. Aspirin exposure coupled with hypoxia interferes energy metabolism, antioxidant and autophagic processes and causes liver injury in estuarine goby Mugilogobius chulae. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135071. [PMID: 38996678 DOI: 10.1016/j.jhazmat.2024.135071] [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: 03/21/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024]
Abstract
Toxicity assessments of pollutants often overlook the impact of environmental factors like hypoxia, which can alter chemical toxicity with unexpected consequences. In this study, Mugilogobius chulae, an estuarine fish, was used to investigate the effects of hypoxia (H), aspirin (ASA), and their combination (H_ASA) exposure over 24, 72, and 168 h. We employed RNA-seq analysis, expression of key gene expression profiling, enzymatic activity assays, and histopathological and ultrastructural examinations of liver tissue to explore the effects and mechanisms of ASA-coupled hypoxia exposure in fish. Results showed that glycolysis was inhibited, and lipolysis was enhanced in ASA/H_ASA groups. The PPAR signaling pathway was activated, increasing fatty acid β-oxidation and lipophagy to mitigate energy crisis. Both ASA and H_ASA exposures induced p53 expression and inhibited the TOR pathway to combat environmental stress. However, a greater energy demand and heightened sensitivity to ASA were observed in H_ASA compared to ASA exposure. Disruptions in energy and detoxification pathways led to increased stress responses, including enhanced antioxidant activities, autophagy, and apoptotic events, as observed in organelle structures. Overall, sub-chronic H_ASA exposure caused liver injury in M. chulae by affecting energy metabolism, antioxidant regulation, and autophagy processes. This study highlights the influence of hypoxia on ASA toxicity in fish, providing valuable insights for ecological risk assessment of NSAIDs.
Collapse
Affiliation(s)
- Yimeng Wang
- Department of Ecology, Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou 510663, China
| | - Yufei Zhao
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Weibo Gong
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Yingshi Hou
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Jinzhi Ren
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Chunni Duan
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Huiyu Zhang
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Xiangping Nie
- Department of Ecology, Jinan University, Guangzhou 510632, China; Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, Jinan University, Guangzhou 510632, China.
| | - Jianjun Li
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou 510663, China
| |
Collapse
|
6
|
Sun D, Cui H, Rong L, Ma T, Li X, Ye Z, Li Z. Sestrin2 Protects Human Lens Epithelial Cells (HLECs) Against Apoptosis in Cataracts Formation: Interaction Between Endoplasmic Reticulum (ER) Stress and Oxidative Stress (OS) is Involved. Curr Eye Res 2024; 49:949-960. [PMID: 38780907 DOI: 10.1080/02713683.2024.2352058] [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: 12/16/2023] [Revised: 04/07/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE To explore the correlation of endoplasmic reticulum stress (ERS) and oxidative stress (OS), and the protective effect of Sestrin2 (SESN2) on human lens epithelial cells (HLECs). METHODS Tunicamycin (TM) was used to induce ERS in HLECs. 4-Phenylbutyric acid (4-PBA) was used to inhibit ERS. Eupatilin applied to HLECs as SESN2 agonist. SESN2 expression was knocked down via si-RNA in HLECs. The morphological changes of HLECs were observed by microscope. ER-tracker to evaluate ERS, ROS production assay to measure ROS, flow cytometry to calculate cell apoptosis rate. Immunofluorescence to observe Nrf2 translocation, and effects of TM or EUP on SESN2. Western blot and qPCR were used to evaluate the expression of GRP78, PERK, ATF4, CHOP, Nrf2, and SESN2 expression in HLECs with different treatment groups. RESULTS ERS can elevate the expression of ROS and Nrf2 to induce OS. Upregulation of SESN2 was observed in ERS-mediate OS. Overexpression of SESN2 can reduce the overexpression of ERS-related protein GRP78, PERK, ATF4, proapoptotic protein CHOP, OS-related protein Nrf2, as well as ROS, and alleviate ERS injury at the same time. Whereas knockdown of SESN2 can upregulate the expression of GRP78, PERK, ATF4, CHOP, Nrf2, ROS, and deteriorate ERS damage. CONCLUSIONS ERS can induce OS, they form a vicious cycle to induce apoptosis in HLECs, which may contribute to cataract formation. SESN2 could protect HLECs against the apoptosis by regulating the vicious cycle between ERS and OS.
Collapse
Affiliation(s)
- Di Sun
- Chinese PLA Medical School, Beijing, China
- Senior Department of Ophthalmology, 3rd Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Hui Cui
- Chinese PLA Medical School, Beijing, China
- Senior Department of Ophthalmology, 3rd Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Liyuan Rong
- Senior Department of Ophthalmology, 3rd Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Tianju Ma
- Senior Department of Ophthalmology, 3rd Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Xuanlong Li
- Chinese PLA Medical School, Beijing, China
- Senior Department of Ophthalmology, 3rd Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Zi Ye
- Senior Department of Ophthalmology, 3rd Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Zhaohui Li
- Senior Department of Ophthalmology, 3rd Medical Center of Chinese, PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Sun Y, Wu Y, Jing R, Yang K, Wang X, Zhao X, Fede C, Stecco C. Interventions of sestrin proteins: Insights to clinical therapy. Heliyon 2024; 10:e34590. [PMID: 39130415 PMCID: PMC11315081 DOI: 10.1016/j.heliyon.2024.e34590] [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: 01/25/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Sestrin proteins, conserved family proteins which mainly induced by ROS, DNA damage, inflammation, and other injuries. Growing evidences proved sestrin proteins exert protective functions in cardiovascular diseases, chronic degenerative osteoarthritis, musculoskeletal diseases, aging and others, sestrin proteins exhibit an anti-inflammatory response, improving metabolism and other valuable character. However, there is no comprehensive and detailed summary and literature research on the intervention methods of sestrin proteins at present. As the advance of research during last several years, exercise training and other interventions are considered to be the potential methods to up-regulate expression level of protein. In view of the physiological function of this protein, a review of the main studies on regulating the expression level of this protein can provide a novel approach for the clinical treatment and scientific research. In present study, all related researches about interventions and potential mechanisms were reviewed and the mainstream methodologies were described.
Collapse
Affiliation(s)
- Yunfeng Sun
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical Univeristy, China
- Padova Neuroscience Center, University of Padova, Italy
| | - Yawei Wu
- Affiliated Traditional Chinese Medicine Hospital of Xinjiang Medical University, Xinjiang Medical University, China
| | - Ronghua Jing
- Affiliated Traditional Chinese Medicine Hospital of Xinjiang Medical University, Xinjiang Medical University, China
| | - Keping Yang
- Yangtze University, Jingzhou Central Hospital, China
| | - Xiaoya Wang
- Yangtze University, Jingzhou Central Hospital, China
| | - Xiaoxiao Zhao
- Padova Neuroscience Center, University of Padova, Italy
| | - Caterina Fede
- Department of Neuroscience, University of Padova, 35122, Padova, Italy
| | - Carla Stecco
- Department of Neuroscience, University of Padova, 35122, Padova, Italy
| |
Collapse
|
8
|
Lesmana R, Tandean S, Christoper A, Suwantika AA, Wathoni N, Abdulah R, Fearnley J, Bankova V, Zulhendri F. Propolis as an autophagy modulator in relation to its roles in redox balance and inflammation regulation. Biomed Pharmacother 2024; 175:116745. [PMID: 38761422 DOI: 10.1016/j.biopha.2024.116745] [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: 03/21/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/20/2024] Open
Abstract
Autophagy is a degradation process that is evolutionarily conserved and is essential in maintaining cellular and physiological homeostasis through lysosomal removal and elimination of damaged peptides, proteins and cellular organelles. The dysregulation of autophagy is implicated in various diseases and disorders, including cancers, infection-related, and metabolic syndrome-related diseases. Propolis has been demonstrated in various studies including many human clinical trials to have antimicrobial, antioxidant, anti-inflammatory, immune-modulator, neuro-protective, and anti-cancer. Nevertheless, the autophagy modulation properties of propolis have not been extensively studied and explored. The role of propolis and its bioactive compounds in modulating cellular autophagy is possibly due to their dual role in redox balance and inflammation. The present review attempts to discuss the activities of propolis as an autophagy modulator in biological models in relation to various diseases/disorders which has implications in the development of propolis-based nutraceuticals, functional foods, and complementary therapies.
Collapse
Affiliation(s)
- R Lesmana
- Physiology Division, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Indonesia; Biological Activity Division, Central Laboratory, Universitas Padjadjaran, Indonesia.
| | - S Tandean
- Department of Neurosurgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Sumatera Utara 20222, Indonesia.
| | - A Christoper
- Postgraduate Program of Medical Science, Faculty of Medicine, Universitas Padjadjaran, Bandung 45363, Indonesia.
| | - A A Suwantika
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia.
| | - N Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; Research Center of Biopolymers for Drug and Cosmetic Delivery, Bandung 45363, Indonesia.
| | - R Abdulah
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung 45363, Indonesia; Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia.
| | - J Fearnley
- Apiceutical Research Centre, Unit 3b Enterprise Way, Whitby, North Yorkshire YO18 7NA, UK.
| | - V Bankova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev str., bl. 9, Sofia 1113, Bulgaria.
| | - F Zulhendri
- Kebun Efi, Kabanjahe, North Sumatra 22171, Indonesia; Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Indonesia.
| |
Collapse
|
9
|
Mbiandjeu SCT, Siciliano A, Mattè A, Federti E, Perduca M, Melisi D, Andolfo I, Amoresano A, Iolascon A, Valenti MT, Turrini F, Bovi M, Pisani A, Recchiuti A, Mattoscio D, Riccardi V, Dalle Carbonare L, Brugnara C, Mohandas N, De Franceschi L. Nrf2 Plays a Key Role in Erythropoiesis during Aging. Antioxidants (Basel) 2024; 13:454. [PMID: 38671902 PMCID: PMC11047311 DOI: 10.3390/antiox13040454] [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: 03/15/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Aging is characterized by increased oxidation and reduced efficiency of cytoprotective mechanisms. Nuclear factor erythroid-2-related factor (Nrf2) is a key transcription factor, controlling the expression of multiple antioxidant proteins. Here, we show that Nrf2-/- mice displayed an age-dependent anemia, due to the combined contributions of reduced red cell lifespan and ineffective erythropoiesis, suggesting a role of Nrf2 in erythroid biology during aging. Mechanistically, we found that the expression of antioxidants during aging is mediated by activation of Nrf2 function by peroxiredoxin-2. The absence of Nrf2 resulted in persistent oxidation and overactivation of adaptive systems such as the unfolded protein response (UPR) system and autophagy in Nrf2-/- mouse erythroblasts. As Nrf2 is involved in the expression of autophagy-related proteins such as autophagy-related protein (Atg) 4-5 and p62, we found impairment of late phase of autophagy in Nrf2-/- mouse erythroblasts. The overactivation of the UPR system and impaired autophagy drove apoptosis of Nrf2-/- mouse erythroblasts via caspase-3 activation. As a proof of concept for the role of oxidation, we treated Nrf2-/- mice with astaxanthin, an antioxidant, in the form of poly (lactic-co-glycolic acid) (PLGA)-loaded nanoparticles (ATS-NPs) to improve its bioavailability. ATS-NPs ameliorated the age-dependent anemia and decreased ineffective erythropoiesis in Nrf2-/- mice. In summary, we propose that Nrf2 plays a key role in limiting age-related oxidation, ensuring erythroid maturation and growth during aging.
Collapse
Affiliation(s)
| | - Angela Siciliano
- Dipartimento Ingegneria per la Medicina di Innovazione—DIMI, University of Verona, 37134 Verona, Italy; (A.S.); (E.F.); (V.R.); (L.D.C.)
- Department of Medicine, AOUI Verona, 37134 Verona, Italy
| | - Alessandro Mattè
- Department of Medicine, University of Verona, 37134 Verona, Italy; (S.C.T.M.); (A.M.); (D.M.)
| | - Enrica Federti
- Dipartimento Ingegneria per la Medicina di Innovazione—DIMI, University of Verona, 37134 Verona, Italy; (A.S.); (E.F.); (V.R.); (L.D.C.)
- Department of Medicine, AOUI Verona, 37134 Verona, Italy
| | - Massimiliano Perduca
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (M.P.); (M.B.); (A.P.)
| | - Davide Melisi
- Department of Medicine, University of Verona, 37134 Verona, Italy; (S.C.T.M.); (A.M.); (D.M.)
| | - Immacolata Andolfo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (I.A.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80131 Naples, Italy
| | - Angela Amoresano
- Department of Chimical Sciences, University Federico II, 80138 Naples, Italy;
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (I.A.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80131 Naples, Italy
| | | | | | - Michele Bovi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (M.P.); (M.B.); (A.P.)
| | - Arianna Pisani
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (M.P.); (M.B.); (A.P.)
| | - Antonio Recchiuti
- Department of Medical, Oral, and Biotechnology Science, “G. d’Annunzio” University Chieti–Pescara, 66013 Chieti, Italy; (A.R.); (D.M.)
| | - Domenico Mattoscio
- Department of Medical, Oral, and Biotechnology Science, “G. d’Annunzio” University Chieti–Pescara, 66013 Chieti, Italy; (A.R.); (D.M.)
| | - Veronica Riccardi
- Dipartimento Ingegneria per la Medicina di Innovazione—DIMI, University of Verona, 37134 Verona, Italy; (A.S.); (E.F.); (V.R.); (L.D.C.)
| | - Luca Dalle Carbonare
- Dipartimento Ingegneria per la Medicina di Innovazione—DIMI, University of Verona, 37134 Verona, Italy; (A.S.); (E.F.); (V.R.); (L.D.C.)
- Department of Medicine, AOUI Verona, 37134 Verona, Italy
| | - Carlo Brugnara
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA 02114, USA;
- Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Narla Mohandas
- New York Blood Center Enterprises, New York, NY 10065, USA;
| | - Lucia De Franceschi
- Dipartimento Ingegneria per la Medicina di Innovazione—DIMI, University of Verona, 37134 Verona, Italy; (A.S.); (E.F.); (V.R.); (L.D.C.)
- Department of Medicine, AOUI Verona, 37134 Verona, Italy
| |
Collapse
|
10
|
Zhang LL, Ding K, Liao SS, Zhang YG, Liao HY, Chen R, Meng QT. Sestrin2 reduces ferroptosis via the Keap1/Nrf2 signaling pathway after intestinal ischemia-reperfusion. Free Radic Biol Med 2024; 214:115-128. [PMID: 38331008 DOI: 10.1016/j.freeradbiomed.2024.02.003] [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: 01/01/2024] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Sestrins are metabolic regulators that respond to stress by reducing the levels of reactive oxygen species (ROS) and inhibiting the activity of target of rapamycin complex 1 (mTORC1). Previous research has demonstrated that Sestrin2 mitigates ischemia-reperfusion (IR) injury in the heart, liver, and kidneys. However, its specific role in intestinal ischemia-reperfusion (IIR) injury remains unclear. To elucidate the role of Sestrin2 in IIR injury, we conducted an experimental study using a C57BL/6J mouse model of IIR. We noticed an increase in the levels of Sestrin2 expression and indicators associated with ferroptosis. Our study revealed that manipulating Sestrin2 expression in Caco-2 cells through overexpression or knockdown resulted in a corresponding decrease or increase, respectively, in ferroptosis levels. Furthermore, our investigation revealed that Sestrin2 alleviated ferroptosis caused by IIR injury through the activation of the Keap1/Nrf2 signal pathway. This finding highlights the potential of Sestrin2 as a therapeutic target for alleviating IIR injury. These findings indicated that the modulation of Sestrin2 could be a promising strategy for managing prolonged IIR injury.
Collapse
Affiliation(s)
- Le-le Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shi-Shi Liao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi-Guo Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hui-Yang Liao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China; Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing-Tao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China; Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
11
|
Kim K, Kim JH, Kim I, Seong S, Koh JT, Kim N. Sestrin2 inhibits RANKL-induced osteoclastogenesis through AMPK activation and ROS inhibition. Free Radic Biol Med 2024; 211:77-88. [PMID: 38101586 DOI: 10.1016/j.freeradbiomed.2023.12.009] [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: 09/12/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023]
Abstract
Sestrins are stress-responsive proteins with antioxidant properties. They participate in cellular redox balance and protect against oxidative damage. This study investigated the effects of Sestrin2 (Sesn2) on osteoclast differentiation and function. Overexpressing Sesn2 in osteoclast precursor cells significantly inhibited receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclastogenesis. This was assessed as reduced expression of various osteoclast markers, including c-Fos, nuclear factor of activated T cells 1 (NFATc1), osteoclast-associated receptor, tartrate-resistant acid phosphatase, and cathepsin K. Conversely, downregulation of Sesn2 produced the opposite effect. Mechanistically, Sesn2 overexpression enhanced AMPK activation and the nuclear translocation of nuclear factor erythroid-derived factor 2-related factor 2 (Nrf2), promoting antioxidant enzymes. Moreover, azithromycin (Azm) induced Sesn2 expression, which suppressed RANKL-induced osteoclast differentiation. Specifically, Azm treatment reduced RANKL-induced production of reactive oxygen species in osteoclasts. Furthermore, intraperitoneal administration of Azm ameliorated RANKL-induced bone loss by reducing osteoclast activity in mice. Taken together, our results suggested that Azm-induced Sesn2 act as a negative regulator of RANKL-induced osteoclast differentiation through the AMPK/NFATc1 signaling pathway. Concisely, targeting Sesn2 can be a potential pharmacological intervention in osteoporosis.
Collapse
Affiliation(s)
- Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jeong-Tae Koh
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea; Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
12
|
Fang H, Shi X, Wan J, Zhong X. Role of sestrins in metabolic and aging-related diseases. Biogerontology 2024; 25:9-22. [PMID: 37516672 DOI: 10.1007/s10522-023-10053-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/14/2023] [Indexed: 07/31/2023]
Abstract
Sestrins are a type of highly conserved stress-inducing protein that has antioxidant and mTORC1 inhibitory functions. Metabolic dysfunction and aging are the main risk factors for development of human diseases, such as diabetes, neurodegenerative diseases, and cancer. Sestrins have important roles in regulating glucose and lipid metabolism, anti-tumor functions, and aging by inhibiting the reactive oxygen species and mechanistic target of rapamycin complex 1 pathways. In this review, the structure and biological functions of sestrins are summarized, and how sestrins are activated and contribute to regulation of the downstream signal pathways of metabolic and aging-related diseases are discussed in detail with the goal of providing new ideas and therapeutic targets for the treatment of related diseases.
Collapse
Affiliation(s)
- Huan Fang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, China
| | - Xiaomin Shi
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, China.
| | - Xiaolin Zhong
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, China.
| |
Collapse
|
13
|
Wang X, Tian Z, He L, Meng H, Zhu J, Li Y, Wang J, Hua X, Huang H, Huang C. DNMT3a-mediated upregulation of the stress inducible protein sestrin-2 contributes to malignant transformation of human bronchial epithelial cells following nickel exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 271:115954. [PMID: 38232523 DOI: 10.1016/j.ecoenv.2024.115954] [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: 08/01/2023] [Revised: 12/07/2023] [Accepted: 01/06/2024] [Indexed: 01/19/2024]
Abstract
BACKGROUND Nickel is a confirmed human lung carcinogen. Nonetheless, the molecular mechanisms driving its carcinogenic impact on lung tissue remain poorly defined. In this study, we assessed SESN2 expression and the signaling pathways responsible for cellular transformation in human bronchial epithelial cells (HBECs) as a result of nickel exposure. METHODS We employed the Western blotting to determine the induction of SESN2 by nickel. To clarify the signaling pathways leading to cellular transformation following nickel exposure, we applied techniques such as gene knockdown, methylation-specific PCR, and chromatin immunoprecipitation. RESULT Exposure to nickel results in the upregulation of SESN2 and the initiation of autophagy in human bronchial epithelial cells (HBECs). This leads to degradation of HUR protein and consequently downregulation of USP28 mRNA, PP2AC protein, β-catenin protein, and diminished VHL transcription, culminating in the accumulation of hypoxia-inducible factor-1α (HIF-1α) and the malignant transformation of these cells. Mechanistic studies revealed that the increased expression of SESN2 is attributed to the demethylation of the SESN2 promoter induced by nickel, a process facilitated by decreased DNA methyl-transferase 3 A (DNMT3a) expression, while The downregulation of VHL transcription is linked to the suppression of the PP2A-C/GSK3β/β-Catenin/C-Myc pathway. Additionally, we discovered that SESN2-mediated autophagy triggers the degradation of HUR protein, which subsequently reduces the stability of USP28 mRNA and inhibits the PP2A-C/GSK3β/β-Catenin pathway and c-Myc transcription in HBECs post nickel exposure. CONCLUSION Our results reveal that nickel exposure leads to the downregulation of DNMT3a, resulting in the hypomethylation of the SESN2 promoter and its protein induction. This triggers autophagy-dependent suppression of the HUR/USP28/PP2A/β-Catenin/c-Myc pathway, subsequently leading to reduced VHL transcription, accumulation of HIF-1α protein, and the malignant transformation of human bronchial epithelial cells (HBECs). Our research offers novel insights into the molecular mechanisms that underlie the lung carcinogenic effects of nickel exposure. Specifically, nickel induces aberrant DNA methylation in the SESN2 promoter region through the decrease of DNMT3a levels, which ultimately leads to HIF-1α protein accumulation and the malignant transformation of HBECs. Specifically, nickel initiates DNA-methylation of the SESN2 promoter region by decreasing DNMT3a, ultimately resulting in HIF-1α protein accumulation and malignant transformation of HBECs. This study highlights DNMT3a as a potential prognostic biomarker or therapeutic target to improve clinical outcomes in lung cancer patients.
Collapse
Affiliation(s)
- Xinxing Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory, Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zhongxian Tian
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory, Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lijiong He
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory, Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hao Meng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory, Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Junlan Zhu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory, Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yang Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory, Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jingjing Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory, Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaohui Hua
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory, Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Haishan Huang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory, Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chuanshu Huang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory, Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
14
|
Yang G, Zhang Q, Dong C, Hou G, Li J, Jiang X, Xin Y. Nrf2 prevents diabetic cardiomyopathy via antioxidant effect and normalization of glucose and lipid metabolism in the heart. J Cell Physiol 2024; 239:e31149. [PMID: 38308838 DOI: 10.1002/jcp.31149] [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: 07/03/2023] [Revised: 10/11/2023] [Accepted: 10/25/2023] [Indexed: 02/05/2024]
Abstract
Metabolic disorders and oxidative stress are the main causes of diabetic cardiomyopathy. Activation of nuclear factor erythroid 2-related factor 2 (Nrf2) exerts a powerful antioxidant effect and prevents the progression of diabetic cardiomyopathy. However, the mechanism of its cardiac protection and direct action on cardiomyocytes are not well understood. Here, we investigated in a cardiomyocyte-restricted Nrf2 transgenic mice (Nrf2-TG) the direct effect of Nrf2 on cardiomyocytes in DCM and its mechanism. In this study, cardiomyocyte-restricted Nrf2 transgenic mice (Nrf2-TG) were used to directly observe whether cardiomyocyte-specific overexpression of Nrf2 can prevent diabetic cardiomyopathy and correct glucose and lipid metabolism disorders in the heart. Compared to wild-type mice, Nrf2-TG mice showed resistance to diabetic cardiomyopathy in a streptozotocin-induced type 1 diabetes mouse model. This was primarily manifested as improved echocardiography results as well as reduced myocardial fibrosis, cardiac inflammation, and oxidative stress. These results showed that Nrf2 can directly act on cardiomyocytes to exert a cardioprotective role. Mechanistically, the cardioprotective effects of Nrf2 depend on its antioxidation activity, partially through improving glucose and lipid metabolism by directly targeting lipid metabolic pathway of AMPK/Sirt1/PGC-1α activation via upstream genes of sestrin2 and LKB1, and indirectly enabling AKT/GSK-3β/HK-Ⅱ activity via AMPK mediated p70S6K inhibition.
Collapse
Affiliation(s)
- Ge Yang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Qihe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Chao Dong
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Guowen Hou
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Jinjie Li
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
15
|
Cortés-Espinar AJ, Ibarz-Blanch N, Soliz-Rueda JR, Calvo E, Bravo FI, Mulero M, Ávila-Román J. Abrupt Photoperiod Changes Differentially Modulate Hepatic Antioxidant Response in Healthy and Obese Rats: Effects of Grape Seed Proanthocyanidin Extract (GSPE). Int J Mol Sci 2023; 24:17057. [PMID: 38069379 PMCID: PMC10707189 DOI: 10.3390/ijms242317057] [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: 11/02/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Disruptions of the light/dark cycle and unhealthy diets can promote misalignment of biological rhythms and metabolic alterations, ultimately leading to an oxidative stress condition. Grape seed proanthocyanidin extract (GSPE), which possesses antioxidant properties, has demonstrated its beneficial effects in metabolic-associated diseases and its potential role in modulating circadian disruptions. Therefore, this study aimed to assess the impact of GSPE administration on the liver oxidant system of healthy and diet-induced obese rats undergoing a sudden photoperiod shift. To this end, forty-eight photoperiod-sensitive Fischer 344/IcoCrl rats were fed either a standard (STD) or a cafeteria diet (CAF) for 6 weeks. A week before euthanizing, rats were abruptly transferred from a standard photoperiod of 12 h of light/day (L12) to either a short (6 h light/day, L6) or a long photoperiod (18 h light/day, L18) while receiving a daily oral dose of vehicle (VH) or GSPE (25 mg/kg). Alterations in body weight gain, serum and liver biochemical parameters, antioxidant gene and protein expression, and antioxidant metabolites were observed. Interestingly, GSPE partially ameliorated these effects by reducing the oxidative stress status in L6 through an increase in GPx1 expression and in hepatic antioxidant metabolites and in L18 by increasing the NRF2/KEAP1/ARE pathway, thereby showing potential in the treatment of circadian-related disorders by increasing the hepatic antioxidant response in a photoperiod-dependent manner.
Collapse
Affiliation(s)
- Antonio J. Cortés-Espinar
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.J.C.-E.); (N.I.-B.); (J.R.S.-R.); (E.C.); (F.I.B.)
- Nutrigenomics Research Group, Institut d’Investigació Sanitària Pere Virgili, 43007 Tarragona, Spain
| | - Néstor Ibarz-Blanch
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.J.C.-E.); (N.I.-B.); (J.R.S.-R.); (E.C.); (F.I.B.)
- Nutrigenomics Research Group, Institut d’Investigació Sanitària Pere Virgili, 43007 Tarragona, Spain
| | - Jorge R. Soliz-Rueda
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.J.C.-E.); (N.I.-B.); (J.R.S.-R.); (E.C.); (F.I.B.)
- Nutrigenomics Research Group, Institut d’Investigació Sanitària Pere Virgili, 43007 Tarragona, Spain
| | - Enrique Calvo
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.J.C.-E.); (N.I.-B.); (J.R.S.-R.); (E.C.); (F.I.B.)
- Nutrigenomics Research Group, Institut d’Investigació Sanitària Pere Virgili, 43007 Tarragona, Spain
| | - Francisca Isabel Bravo
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.J.C.-E.); (N.I.-B.); (J.R.S.-R.); (E.C.); (F.I.B.)
- Nutrigenomics Research Group, Institut d’Investigació Sanitària Pere Virgili, 43007 Tarragona, Spain
| | - Miquel Mulero
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.J.C.-E.); (N.I.-B.); (J.R.S.-R.); (E.C.); (F.I.B.)
- Nutrigenomics Research Group, Institut d’Investigació Sanitària Pere Virgili, 43007 Tarragona, Spain
| | - Javier Ávila-Román
- Molecular and Applied Pharmacology Group (FARMOLAP), Department of Pharmacology, Universidad de Sevilla, 41012 Sevilla, Spain
| |
Collapse
|
16
|
Krishna S, Echevarria KG, Reed CH, Eo H, Wintzinger M, Quattrocelli M, Valentine RJ, Selsby JT. A fat- and sucrose-enriched diet causes metabolic alterations in mdx mice. Am J Physiol Regul Integr Comp Physiol 2023; 325:R692-R711. [PMID: 37811713 PMCID: PMC11178302 DOI: 10.1152/ajpregu.00246.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 08/18/2023] [Accepted: 09/10/2023] [Indexed: 10/10/2023]
Abstract
Duchenne muscular dystrophy (DMD), a progressive muscle disease caused by the absence of functional dystrophin protein, is associated with multiple cellular, physiological, and metabolic dysfunctions. As an added complication to the primary insult, obesity/insulin resistance (O/IR) is frequently reported in patients with DMD; however, how IR impacts disease severity is unknown. We hypothesized a high-fat, high-sucrose diet (HFHSD) would induce O/IR, exacerbate disease severity, and cause metabolic alterations in dystrophic mice. To test this hypothesis, we treated 7-wk-old mdx (disease model) and C57 mice with a control diet (CD) or an HFHSD for 15 wk. The HFHSD induced insulin resistance, glucose intolerance, and hyperglycemia in C57 and mdx mice. Of note, mdx mice on CD were also insulin resistant. In addition, visceral adipose tissue weights were increased with HFHSD in C57 and mdx mice though differed by genotype. Serum creatine kinase activity and histopathological analyses using Masson's trichrome staining in the diaphragm indicated muscle damage was driven by dystrophin deficiency but was not augmented by diet. In addition, markers of inflammatory signaling, mitochondrial abundance, and autophagy were impacted by disease but not diet. Despite this, in addition to disease signatures in CD-fed mice, metabolomic and lipidomic analyses demonstrated a HFHSD caused some common changes in C57 and mdx mice and some unique signatures of O/IR within the context of dystrophin deficiency. In total, these data revealed that in mdx mice, 15 wk of HFHSD did not overtly exacerbate muscle injury but further impaired the metabolic status of dystrophic muscle.
Collapse
Affiliation(s)
- Swathy Krishna
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | | | - Carter H Reed
- Department of Kinesiology, Iowa State University, Ames, Iowa, United States
| | - Hyeyoon Eo
- Department of Kinesiology, Iowa State University, Ames, Iowa, United States
| | - Michelle Wintzinger
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Rudy J Valentine
- Department of Kinesiology, Iowa State University, Ames, Iowa, United States
| | - Joshua T Selsby
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| |
Collapse
|
17
|
Dai G, Li M, Xu H, Quan N. Status of Research on Sestrin2 and Prospects for its Application in Therapeutic Strategies Targeting Myocardial Aging. Curr Probl Cardiol 2023; 48:101910. [PMID: 37422038 DOI: 10.1016/j.cpcardiol.2023.101910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/10/2023]
Abstract
Cardiac aging is accompanied by changes in the heart at the cellular and molecular levels, leading to alterations in cardiac structure and function. Given today's increasingly aging population, the decline in cardiac function caused by cardiac aging has a significant impact on quality of life. Antiaging therapies to slow the aging process and attenuate changes in cardiac structure and function have become an important research topic. Treatment with drugs, including metformin, spermidine, rapamycin, resveratrol, astaxanthin, Huolisu oral liquid, and sulforaphane, has been demonstrated be effective in delaying cardiac aging by stimulating autophagy, delaying ventricular remodeling, and reducing oxidative stress and the inflammatory response. Furthermore, caloric restriction has been shown to play an important role in delaying aging of the heart. Many studies in cardiac aging and cardiac aging-related models have demonstrated that Sestrin2 has antioxidant and anti-inflammatory effects, stimulates autophagy, delays aging, regulates mitochondrial function, and inhibits myocardial remodeling by regulation of relevant signaling pathways. Therefore, Sestrin2 is likely to become an important target for antimyocardial aging therapy.
Collapse
Affiliation(s)
- Gaoying Dai
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Meina Li
- Department of Infection Control, The First Hospital of Jilin University, Changchun, China
| | - He Xu
- Department of Integrative Medicine, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Nanhu Quan
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
18
|
Li K, Geng Y, Lin B, Xi Z. Molecular mechanisms underlying mitochondrial damage, endoplasmic reticulum stress, and oxidative stress induced by environmental pollutants. Toxicol Res (Camb) 2023; 12:1014-1023. [PMID: 38145103 PMCID: PMC10734609 DOI: 10.1093/toxres/tfad094] [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: 04/28/2023] [Revised: 08/09/2023] [Accepted: 09/15/2023] [Indexed: 12/26/2023] Open
Abstract
Mitochondria and endoplasmic reticulum (ER) are essential organelles playing pivotal roles in the regulation of cellular metabolism, energy production, and protein synthesis. In addition, these organelles are important targets susceptible to external stimuli, such as environmental pollutants. Exposure to environmental pollutants can cause the mitochondrial damage, endoplasmic reticulum stress (ERS), and oxidative stress, leading to cellular dysfunction and death. Therefore, understanding the toxic effects and molecular mechanisms of environmental pollution underlying these processes is crucial for developing effective strategies to mitigate the adverse effects of environmental pollutants on human health. In the present study, we summarized and reviewed the toxic effects and molecular mechanisms of mitochondrial damage, ERS, and oxidative stress caused by exposure to environmental pollutants as well as interactions inducing the cell apoptosis and the roles in exposure to environmental pollutants.
Collapse
Affiliation(s)
- Kang Li
- Department of Health Toxicology, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yanpei Geng
- Department of Health Toxicology, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Bencheng Lin
- Department of Health Toxicology, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zhuge Xi
- Department of Health Toxicology, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| |
Collapse
|
19
|
Kırça M, Yeşilkaya A. Angiotensin II reduces glyoxalase 1 activity and expression in vascular smooth muscle cells: Implications for diabetic vascular complications. Cell Biochem Funct 2023; 41:1430-1441. [PMID: 37915258 DOI: 10.1002/cbf.3879] [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: 06/24/2023] [Revised: 10/13/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
Angiotensin II (Ang II), a key mediator of vascular diseases, is linked to methylglyoxal (MGO) formation, a by-product of glucose metabolism implicated in vascular complications. The glyoxalase system, consisting of glyoxalase 1 (Glo1) and reduced glutathione (GSH), is responsible for detoxifying MGO. This study investigated the effect of Ang II on Glo1 activity and expression in vascular smooth muscle cells (VSMCs). Primary VSMCs were isolated from rat aortas and exposed to Ang II under standard or high glucose conditions. We examined Glo1 activity, expression, intracellular GSH, and methylglyoxal-derived hydroimidazolone 1 (MG-H1) levels. We also analyzed the expressions of nuclear factor-κB (NF-κB) p65 and nuclear factor erythroid 2-related factor 2 (Nrf2) as potential regulators of Glo1 expression. The results demonstrated that Ang II reduced Glo1 activity, expression, and GSH levels while increasing MG-H1 levels in VSMCs. Telmisartan and irbesartan, AT1R blockers, restored Glo1 activity, expression, and GSH levels and alleviated MG-H1 levels. Treatment with AT1R blockers or inhibitors targeting signaling pathways involved in Ang II-induced responses mitigated these effects. High glucose exacerbated the reduction in Glo1 activity and expression. In conclusion, this study provides evidence that Ang II reduces Glo1 activity and expression in VSMCs, which may contribute to developing vascular complications in diabetes. AT1R blockers and inhibitors targeting specific signaling pathways show potential in restoring Glo1 function and mitigating MGO-associated damage. These findings highlight the complex interactions between RAS, MGO, and vascular diseases, highlighting potential therapeutic targets for diabetic vascular complications.
Collapse
Affiliation(s)
- Mustafa Kırça
- Department of Medical Biochemistry, Faculty of Medicine, Kütahya Health Sciences University, Kütahya, Turkey
| | - Akın Yeşilkaya
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
20
|
Ivarsson J, Ferrara F, Vallese A, Guiotto A, Colella S, Pecorelli A, Valacchi G. Comparison of Pollutant Effects on Cutaneous Inflammasomes Activation. Int J Mol Sci 2023; 24:16674. [PMID: 38068996 PMCID: PMC10706824 DOI: 10.3390/ijms242316674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
The skin is the outermost layer of the body and, therefore, is exposed to a variety of stressors, such as environmental pollutants, known to cause oxinflammatory reactions involved in the exacerbation of several skin conditions. Today, inflammasomes are recognized as important modulators of the cutaneous inflammatory status in response to air pollutants and ultraviolet (UV) light exposure. In this study, human skin explants were exposed to the best-recognized air pollutants, such as microplastics (MP), cigarette smoke (CS), diesel engine exhaust (DEE), ozone (O3), and UV, for 1 or 4 days, to explore how each pollutant can differently modulate markers of cutaneous oxinflammation. Exposure to environmental pollutants caused an altered oxidative stress response, accompanied by increased DNA damage and signs of premature skin aging. The effect of specific pollutants being able to exert different inflammasomes pathways (NLRP1, NLRP3, NLRP6, and NLRC4) was also investigated in terms of scaffold formation and cell pyroptosis. Among all environmental pollutants, O3, MP, and UV represented the main pollutants affecting cutaneous redox homeostasis; of note, the NLRP1 and NLRP6 inflammasomes were the main ones modulated by these outdoor stressors, suggesting their role as possible molecular targets in preventing skin disorders and the inflammaging events associated with environmental pollutant exposure.
Collapse
Affiliation(s)
- John Ivarsson
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA;
| | - Francesca Ferrara
- Department of Chemical, Pharmaceuticals and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Andrea Vallese
- Department of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA; (A.V.); (A.G.); (A.P.)
- Department of Environmental Sciences and Prevention, University of Ferrara, 44121 Ferrara, Italy
| | - Anna Guiotto
- Department of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA; (A.V.); (A.G.); (A.P.)
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Sante Colella
- Department of Biotechnology, Chemistry and Pharmaceutical Sciences, University of Siena, 53100 Siena, Italy;
| | - Alessandra Pecorelli
- Department of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA; (A.V.); (A.G.); (A.P.)
- Department of Environmental Sciences and Prevention, University of Ferrara, 44121 Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA; (A.V.); (A.G.); (A.P.)
- Department of Environmental Sciences and Prevention, University of Ferrara, 44121 Ferrara, Italy
- Department of Food and Nutrition, Kyung Hee University, Seoul 26723, Republic of Korea
| |
Collapse
|
21
|
Kim JM, Jung IA, Kim JM, Choi MH, Yang JH. Anti-Inflammatory Effect of Cinnamomum japonicum Siebold's Leaf through the Inhibition of p38/JNK/AP-1 Signaling. Pharmaceuticals (Basel) 2023; 16:1402. [PMID: 37895873 PMCID: PMC10610235 DOI: 10.3390/ph16101402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/23/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Cinnamomum japonicum Siebold (CJ) branch bark, commonly known as Japanese cinnamon, has been used for various culinary and medicinal applications for many centuries. Although the efficacy of CJ branch bark's anti-inflammatory and antioxidant activity for the treatment of various diseases has been confirmed, the efficacy of CJ leaves (CJLs) has not been examined. We therefore investigated whether CJL3, an ethyl acetate extract of a 70% ethanol CJL extract, exerts anti-inflammatory effects on lipopolysaccharide (LPS)-activated Kupffer cells, specialized macrophages found in the liver. Liver inflammation can activate Kupffer cells, inducing the release of pro-inflammatory molecules that contribute to tissue damage. We found that CJL3 has high 2,2-diphenyl-1-picrylhydrazyl and 2,2-azino-bis (3-ethylbenzthiazoline-6-sulfonic acid) radical-scavenging activity. Among the CJL extracts, CJL3 exhibited the greatest polyphenol content, with protocatechuic acid and 4-hydroxybenzoic acid being the most abundant. In addition, we verified that CJL3, which has strong antioxidant properties, ameliorates LPS-induced pro-inflammatory responses by inhibiting p38/JNK/AP-1 signaling. CJL3 therefore has potential for treating liver disease, including hepatitis.
Collapse
Affiliation(s)
- Ji Min Kim
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea; (J.M.K.); (I.A.J.); (J.M.K.)
| | - In A Jung
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea; (J.M.K.); (I.A.J.); (J.M.K.)
| | - Jae Min Kim
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea; (J.M.K.); (I.A.J.); (J.M.K.)
| | - Moon-Hee Choi
- Department of Biochemical Engineering, College of Engineering, Chosun University, Gwangju 61452, Republic of Korea
- Sumsumbio Co., Ltd., Jangseong-gun 57248, Republic of Korea
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea; (J.M.K.); (I.A.J.); (J.M.K.)
| |
Collapse
|
22
|
Agaoglu MO, Agaoglu Z, Yucel KY, Ozturk FH, Caglar T. Evaluation of maternal serum sestrin-2 levels in intrauterine growth restriction. Ir J Med Sci 2023; 192:2259-2264. [PMID: 36877413 DOI: 10.1007/s11845-023-03329-2] [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: 01/19/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Sestrin-2 (SESN2) is a antioxidant protein that can be activated by a number of conditions, including DNA damage and hypoxia. AIMS Our objective was to evaluate maternal serum SESN2 levels in patients with intrauterine growth restriction (IUGR) and its association with adverse perinatal outcomes. METHODS This prospective study included a total of 87 pregnant women admitted to our tertiary care center between 2018 August and 2019 July. The study group consisted of a total of 44 patients who had been diagnosed with IUGR. Forty-three low-risk and gestational age-matched pregnant women were taken as control group. Demographic data, maternal serum SESN2 levels, and maternal-neonatal outcomes were evaluated. SESN2 levels were analyzed by the enzyme-linked immunosorbent assay (ELISA) method and compared between groups. RESULTS Maternal serum SESN2 levels were significantly higher in the IUGR group compared to control group (22.38 ng/ml vs. 13.0 ng/ml, p < 0.001). In correlation analysis, a negative significant correlation was found between SESN2 levels and gestational week at delivery (r = - 0.387, p < 0.001). The ideal cut-off value for detecting IUGR was 9.5 ng/ml, and the area under the curve was 0.719 (95%CI: 0.610-0.827). Birth interval, gestational week at birth, birth weight, and 1-5-min Apgar scores were lower in the IUGR group (p < 0.001). CONCLUSIONS Maternal serum SESN2 levels are elevated in IUGR and are associated with adverse neonatal outcome. Considering that SESN2 is involved in pathogenesis, it can be used as a new marker for the evaluation of IUGR.
Collapse
Affiliation(s)
- Merve Ozturk Agaoglu
- Department of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey.
| | - Zahid Agaoglu
- Department of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Kadriye Yakut Yucel
- Department of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Filiz Halıcı Ozturk
- Department of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Turhan Caglar
- Department of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
23
|
Ning B, Hang S, Zhang W, Mao C, Li D. An update on the bridging factors connecting autophagy and Nrf2 antioxidant pathway. Front Cell Dev Biol 2023; 11:1232241. [PMID: 37621776 PMCID: PMC10445655 DOI: 10.3389/fcell.2023.1232241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Macroautophagy/autophagy is a lysosome-dependent catabolic pathway for the degradation of intracellular proteins and organelles. Autophagy dysfunction is related to many diseases, including lysosomal storage diseases, cancer, neurodegenerative diseases, cardiomyopathy, and chronic metabolic diseases, in which increased reactive oxygen species (ROS) levels are also observed. ROS can randomly oxidize proteins, lipids, and DNA, causing oxidative stress and damage. Cells have developed various antioxidant pathways to reduce excessive ROS and maintain redox homeostasis. Treatment targeting only one aspect of diseases with autophagy dysfunction and oxidative stress shows very limited effects. Herein, identifying the bridging factors that can regulate both autophagy and antioxidant pathways is beneficial for dual-target therapies. This review intends to provide insights into the current identified bridging factors that connect autophagy and Nrf2 antioxidant pathway, as well as their tight interconnection with each other. These factors could be potential dual-purpose targets for the treatment of diseases implicated in both autophagy dysfunction and oxidative stress.
Collapse
Affiliation(s)
- Baike Ning
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Shuqi Hang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wenhe Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Caiwen Mao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Dan Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
24
|
Kim JM, Cho SS, Kang S, Moon C, Yang JH, Ki SH. Castanopsis sieboldii Extract Alleviates Acute Liver Injury by Antagonizing Inflammasome-Mediated Pyroptosis. Int J Mol Sci 2023; 24:11982. [PMID: 37569359 PMCID: PMC10419291 DOI: 10.3390/ijms241511982] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Castanopsis sieboldii (CS), a subtropical species, was reported to have antioxidant and antibacterial effects. However, the anti-inflammatory effects of CS have not been studied. This study aimed to investigate whether the 70% ethanol extract of the CS leaf (CSL3) inhibited lipopolysaccharide (LPS)-induced inflammatory responses and LPS and ATP-induced pyroptosis in macrophages. CSL3 treatment inhibited NO release and iNOS expression in LPS-stimulated cells. CSL3 antagonized NF-κB and AP-1 activation, which was due to MAPK (p38, ERK, and JNK) inhibition. CSL3 successfully decreased NLRP3 inflammasome activation and increased IL-1β expression. CSL3 treatment diminished LPS and ATP-induced pore formation in GSDMD. The in vivo effect of CSL3 on acute liver injury was evaluated in a CCl4-treated mouse model. CCl4 treatment increased the activity of serum alanine aminotransferase and aspartate aminotransferase, which decreased by CSL3. In addition, CCl4-induced an increase in TNF-α, and IL-6 levels decreased by CSL3 treatment. Furthermore, we verified that the CCl4-induced inflammasome and pyroptosis-related gene expression in liver tissue and release of IL-1β into serum were suppressed by CSL3 treatment. Our results suggest that CSL3 protects against acute liver injury by inhibiting inflammasome formation and pyroptosis.
Collapse
Affiliation(s)
- Jae Min Kim
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea; (J.M.K.); (S.S.C.)
| | - Sam Seok Cho
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea; (J.M.K.); (S.S.C.)
| | - Sohi Kang
- College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea; (S.K.); (C.M.)
| | - Changjong Moon
- College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea; (S.K.); (C.M.)
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea; (J.M.K.); (S.S.C.)
| |
Collapse
|
25
|
Didik S, Wang H, James AS, Slotabec L, Li J. Sestrin2 as a Potential Target in Hypertension. Diagnostics (Basel) 2023; 13:2374. [PMID: 37510117 PMCID: PMC10378131 DOI: 10.3390/diagnostics13142374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/24/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Hypertension is a highly complex, intricate condition affecting millions of individuals across the globe. Nearly half of adults in the United States are diagnosed with hypertension, with incident rates projected to rise over the next decade. Hypertension is a precursor to many cardiovascular diseases including atherosclerosis, stroke, myocardial infarction, heart failure, and peripheral artery disease. This review describes the major processes contributing to the development of hypertension and how Sestrin2 (Sesn2), an antioxidative protein, could be a potential target in the treatment of hypertension. In hypertension, increased reactive oxygen species (ROS) production is a critical component in the etiology of the condition. The increased ROS in hypertension is derived from a variety of sources, all of which are covered in depth in this review. Increased ROS is generated from mitochondrial stress, endoplasmic reticulum (ER) stress, NADPH oxidase (NOX) overactivity, and the uncoupling of endothelial nitric oxidase synthase (eNOS). Sesn2, a highly conserved, stress-inducible protein, has the structural and functional characteristics to be a potential therapeutic target to alleviate the progression of hypertension. The structure, function, genetics, and characteristics of Sesn2 are presented in the review. The Nrf2/Sesn2, Sesn2/AMPK/mTOR, and Sesn2/Angiotensin II signaling pathways are described in detail in this review. Sesn2 can be utilized in a multitude of ways as a therapeutic modality in hypertension. This review explores potential Sesn2 inducers and activators and how Sesn2 can be incorporated into gene therapy for the treatment of hypertension.
Collapse
Affiliation(s)
- Steven Didik
- Department of Surgery, University of South Florida, Tampa, FL 33612, USA
- James A. Haley Veterans' Hospital, Tampa, FL 33612, USA
| | - Hao Wang
- Department of Surgery, University of South Florida, Tampa, FL 33612, USA
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | - Lily Slotabec
- Department of Surgery, University of South Florida, Tampa, FL 33612, USA
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
26
|
Kozak J, Jonak K. Association between the antioxidant properties of SESN proteins and anti-cancer therapies. Amino Acids 2023:10.1007/s00726-023-03281-6. [PMID: 37284849 PMCID: PMC10372130 DOI: 10.1007/s00726-023-03281-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/17/2023] [Indexed: 06/08/2023]
Abstract
Since the beginning of SESN protein development, they have attracted highly progressive attention due to their regulatory role in multiple signalling pathways. Through their antioxidant activity and autophagy regulation implication, they can function as powerful antioxidants to reduce oxidative stress in cells. SESN proteins received special attention in the field of regulation of reactive oxygen species level in the cell and its interplay with signalling pathways determining energy and nutrient homeostasis. Since perturbations in these pathways are implicated in cancer onset and development, SESNs might constitute potential novel therapeutic targets of broad interest. In this review, we discuss the impact of SESN proteins on anti-cancer therapy based on naturally occurring compounds and conventionally used drugs that influence oxidative stress and autophagy-induced cellular signalling pathways. The significant changes in reactive oxygen species level and nutrient status in cancer cells generate subsequent biological effect through the regulation of SESN-dependent pathways. Thus, SESN may serve as the key molecule for regulating anti-cancer drugs' induced cellular response.
Collapse
Affiliation(s)
- Joanna Kozak
- Chair of Fundamental Sciences, Department of Human Anatomy, Medical University of Lublin, Kazimierza Jaczewskiego 4, 20-090, Lublin, Poland.
| | - Katarzyna Jonak
- Department of Foreign Languages, Interfaculty Centre for Didactics, Medical University of Lublin, 20-081, Lublin, Poland
| |
Collapse
|
27
|
Wang X, Yang J, Li H, Mu H, Zeng L, Cai S, Su P, Li H, Zhang L, Xiang W. miR-484 mediates oxidative stress-induced ovarian dysfunction and promotes granulosa cell apoptosis via SESN2 downregulation. Redox Biol 2023; 62:102684. [PMID: 36963287 PMCID: PMC10060268 DOI: 10.1016/j.redox.2023.102684] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023] Open
Abstract
Ovarian dysfunction is a common cause of female infertility, which is associated with genetic, autoimmune and environmental factors. Granulosa cells (GCs) constitute the largest cell population of ovarian follicles. Changes in GCs, including oxidative stress (OS) and excessive reactive oxygen species (ROS), are involved in regulating ovary function. miR-484 is highly expressed in 3-NP-induced oxidative stress models of ovaries and GCs. miR-484 overexpression aggravated GCs dysfunction and thereby intensified ovarian oxidative stress injury in mice. Moreover, bioinformatic analyses, luciferase assays and pull-down assays indicated that LINC00958 acted as a competing endogenous RNA (ceRNA) for miR-484 and formed a signaling axis with Sestrin2(SESN2) under oxidative stress conditions, which in turn regulated mitochondrial functions and mitochondrial-related apoptosis in GCs. Additionally, the inhibition of miR-484 alleviated GCs dysfunction under ovarian oxidative stress condition. Our present study revealed the role of miR-484 in oxidative stress of ovaries and GCs and the function of LINC00958/miR-484/SESN2 axis in mitochondrial function and mitochondria-related apoptosis.
Collapse
Affiliation(s)
- Xiaofei Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Jiahao Yang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Huiying Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Hongbei Mu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Ling Zeng
- Medical Genetics Center, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Siying Cai
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Ping Su
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China; Wuhan Huake Reproductive Hospital, 128 Sanyang Road, Wuhan, 430013, China
| | - Huaibiao Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| | - Ling Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China; Wuhan Huake Reproductive Hospital, 128 Sanyang Road, Wuhan, 430013, China.
| | - Wenpei Xiang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China; Wuhan Huake Reproductive Hospital, 128 Sanyang Road, Wuhan, 430013, China.
| |
Collapse
|
28
|
Wang K, Shen K, Han F, Bai X, Fang Z, Jia Y, Zhang J, Li Y, Cai W, Wang X, Luo L, Guo K, Wang H, Yang X, Wang H, Hu D. Activation of Sestrin2 accelerates deep second-degree burn wound healing through PI3K/AKT pathway. Arch Biochem Biophys 2023; 743:109645. [PMID: 37225009 DOI: 10.1016/j.abb.2023.109645] [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: 02/25/2023] [Revised: 05/07/2023] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Deep second-degree burns heal slowly, and promoting the healing process is a focus of clinical research. Sestrin2 is a stress-inducible protein with antioxidant and metabolic regulatory effects. However, its role during acute dermal and epidermal re-epithelialization in deep second-degree burns is unknown. In this study, we aimed to explore the role and molecular mechanism of sestrin2 in deep second-degree burns as a potential treatment target for burn wounds. To explore the effects of sestrin2 on burn wound healing, we established a deep second-degree burn mouse model. Then we detected the expression of sestrin2 by western blot and immunohistochemistry after obtaining the wound margin of full-thickness burned skin. The effects of sestrin2 on burn wound healing were explored in vivo and in vitro through interfering sestrin2 expression using siRNAs or the small molecule agonist of sestrin2, eupatilin. We also investigated the molecular mechanism of sestrin2 in promoting burn wound healing by western blot and CCK-8 assay. Our in vivo and in vitro deep second-degree burn wound healing model demonstrated that sestrin2 was promptly induced at murine skin wound edges. The small molecule agonist of sestrin2 accelerated the proliferation and migration of keratinocytes, as well as burn wound healing. Conversely, the healing of burn wounds was delayed in sestrin2-deficient mice and was accompanied by the secretion of inflammatory cytokines as well as the suppression of keratinocyte proliferation and migration. Mechanistically, sestrin2 promoted the phosphorylation of the PI3K/AKT pathway, and inhibition of PI3K/AKT pathway abrogated the promoting role of sestrin2 in keratinocyte proliferation and migration. Therefore, sestrin2 plays a critical role in activation of the PI3K/AKT pathway to promote keratinocyte proliferation and migration, as well as re-epithelialization in the process of deep second-degree burn wound repair.
Collapse
Affiliation(s)
- Kejia Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Kuo Shen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Fu Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Zhuoqun Fang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Yanhui Jia
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Jian Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Yan Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Weixia Cai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xujie Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Kai Guo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Hongtao Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xuekang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
29
|
Sestrin2 as a Protective Shield against Cardiovascular Disease. Int J Mol Sci 2023; 24:ijms24054880. [PMID: 36902310 PMCID: PMC10003517 DOI: 10.3390/ijms24054880] [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: 12/31/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 03/06/2023] Open
Abstract
A timely and adequate response to stress is inherently present in each cell and is important for maintaining the proper functioning of the cell in changing intracellular and extracellular environments. Disruptions in the functioning or coordination of defense mechanisms against cellular stress can reduce the tolerance of cells to stress and lead to the development of various pathologies. Aging also reduces the effectiveness of these defense mechanisms and results in the accumulation of cellular lesions leading to senescence or death of the cells. Endothelial cells and cardiomyocytes are particularly exposed to changing environments. Pathologies related to metabolism and dynamics of caloric intake, hemodynamics, and oxygenation, such as diabetes, hypertension, and atherosclerosis, can overwhelm endothelial cells and cardiomyocytes with cellular stress to produce cardiovascular disease. The ability to cope with stress depends on the expression of endogenous stress-inducible molecules. Sestrin2 (SESN2) is an evolutionary conserved stress-inducible cytoprotective protein whose expression is increased in response to and defend against different types of cellular stress. SESN2 fights back the stress by increasing the supply of antioxidants, temporarily holding the stressful anabolic reactions, and increasing autophagy while maintaining the growth factor and insulin signaling. If the stress and the damage are beyond repair, SESN2 can serve as a safety valve to signal apoptosis. The expression of SESN2 decreases with age and its levels are associated with cardiovascular disease and many age-related pathologies. Maintaining sufficient levels or activity of SESN2 can in principle prevent the cardiovascular system from aging and disease.
Collapse
|
30
|
Lu C, Jiang Y, Xu W, Bao X. Sestrin2: multifaceted functions, molecular basis, and its implications in liver diseases. Cell Death Dis 2023; 14:160. [PMID: 36841824 PMCID: PMC9968343 DOI: 10.1038/s41419-023-05669-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/26/2023]
Abstract
Sestrin2 (SESN2), a highly conserved stress-responsive protein, can be triggered by various noxious stimuli, such as hypoxia, DNA damage, oxidative stress, endoplasmic reticulum (ER) stress, and inflammation. Multiple transcription factors regulate SESN2 expression, including hypoxia-inducible factor 1 (HIF-1), p53, nuclear factor E2-related factor 2 (Nrf2), activating transcription factor 4 (ATF4), ATF6, etc. Upon induction, SESN2 generally leads to activation of adenosine monophosphate-activated protein kinase (AMPK) and inhibition of mechanistic target of rapamycin complex 1 (mTORC1). To maintain cellular homeostasis, SESN2 and its downstream molecules directly scavenge reactive oxygen species or indirectly influence the expression patterns of key genes associated with redox, macroautophagy, mitophagy, ER stress, apoptosis, protein synthesis, and inflammation. In liver diseases including acute liver injury, fatty liver diseases, hepatic fibrosis, and hepatocellular carcinoma (HCC), SESN2 is abnormally expressed and correlated with disease progression. In NAFLD, SESN2 helps with postponing disease progression through balancing glycolipid metabolism and macroautophagy (lipophagy), and rectifying oxidative damage and ER stress. During hepatic fibrosis, SESN2 represses HSCs activation and intrahepatic inflammation, hindering the occurrence and progress of fibrogenesis. However, the role of SESN2 in HCC is controversial due to its paradoxical pro-autophagic and anti-apoptotic effects. In conclusion, this review summarizes the biological functions of SESN2 in hypoxia, genotoxic stress, oxidative stress, ER stress, and inflammation, and specifically emphasizes the pathophysiological significance of SESN2 in liver diseases, aiming to providing a comprehensive understanding for SESN2 as a potential therapeutic target in liver diseases.
Collapse
Affiliation(s)
- Chunfeng Lu
- grid.260483.b0000 0000 9530 8833School of Pharmacy, Nantong University, 226001 Nantong, Jiangsu China
| | - Yiming Jiang
- grid.260483.b0000 0000 9530 8833School of Pharmacy, Nantong University, 226001 Nantong, Jiangsu China
| | - Wenxuan Xu
- School of Life Science and Technology, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China.
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, 226001, Nantong, Jiangsu, China.
| |
Collapse
|
31
|
Cinnamomum japonicum Siebold Branch Extracts Attenuate NO and ROS Production via the Inhibition of p38 and JNK Phosphorylation. Molecules 2023; 28:molecules28041974. [PMID: 36838961 PMCID: PMC9960860 DOI: 10.3390/molecules28041974] [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: 12/30/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Cinnamomum japonicum (CJ) is widely distributed in Asian countries like Korea, China, and Japan. Modern pharmacological studies have demonstrated that it exhibits various biological activities, including antioxidant and anti-inflammatory effects. However, most studies have confirmed the efficacy of its water extract but not that of its other extracts. Therefore, in this study, Cinnamomum japonicum Siebold branches (CJB: 70% EtOH extract) were separated using hexane, chloroform, ethyl acetate (CJB3), butanol, and water. Then, their antioxidative activities and phenolic contents were measured. Results revealed that the antioxidant activities and phenolic contents of CJB3 were higher than those of the other extracts. Further, the inhibitory and anti-inflammatory effect of CJB3 on lipopolysaccharide (LPS)-induced reactive oxygen species (ROS) production and LPS-activated macrophages, respectively, was determined. CJB3 suppressed oxidative stress in LPS-activated cells and dose-dependently decreased LPS-stimulated ROS production. CJB3 reduced oxidative stress and reversed the glutathione decrease in LPS-activated RAW264.7 cells. The inhibitory and reducing effect of CJB3 on LPS-induced nitric oxide (NO) production and inducible NO synthase protein and messenger RNA levels, respectively, was investigated. CJB3 inhibited LPS-induced cytokine production and p38 and c-Jun N-terminal kinase (JNK) phosphorylation but not extracellular signal-regulated kinase phosphorylation. Overall, the study results suggest that CJB3 may exert its anti-inflammatory effects via the suppression of p38, JNK, and c-Jun activation.
Collapse
|
32
|
Ala M. Sestrin2 Signaling Pathway Regulates Podocyte Biology and Protects against Diabetic Nephropathy. J Diabetes Res 2023; 2023:8776878. [PMID: 36818747 PMCID: PMC9937769 DOI: 10.1155/2023/8776878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/22/2022] [Accepted: 02/04/2023] [Indexed: 02/12/2023] Open
Abstract
Sestrin2 regulates cell homeostasis and is an upstream signaling molecule for several signaling pathways. Sestrin2 leads to AMP-activated protein kinase- (AMPK-) and GTPase-activating protein activity toward Rags (GATOR) 1-mediated inhibition of mammalian target of rapamycin complex 1 (mTORC1), thereby enhancing autophagy. Sestrin2 also improves mitochondrial biogenesis via AMPK/Sirt1/peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) signaling pathway. Blockade of ribosomal protein synthesis and augmentation of autophagy by Sestrin2 can prevent misfolded protein accumulation and attenuate endoplasmic reticulum (ER) stress. In addition, Sestrin2 enhances P62-mediated autophagic degradation of Keap1 to release nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 release by Sestrin2 vigorously potentiates antioxidant defense in diabetic nephropathy. Impaired autophagy and mitochondrial biogenesis, severe oxidative stress, and ER stress are all deeply involved in the development and progression of diabetic nephropathy. It has been shown that Sestrin2 expression is lower in the kidney of animals and patients with diabetic nephropathy. Sestrin2 knockdown aggravated diabetic nephropathy in animal models. In contrast, upregulation of Sestrin2 enhanced autophagy, mitophagy, and mitochondrial biogenesis and suppressed oxidative stress, ER stress, and apoptosis in diabetic nephropathy. Consistently, overexpression of Sestrin2 ameliorated podocyte injury, mesangial proliferation, proteinuria, and renal fibrosis in animal models of diabetic nephropathy. By suppressing transforming growth factor beta (TGF-β)/Smad and Yes-associated protein (YAP)/transcription enhancer factor 1 (TEF1) signaling pathways in experimental models, Sestrin2 hindered epithelial-mesenchymal transition and extracellular matrix accumulation in diabetic kidneys. Moreover, modulation of the downstream molecules of Sestrin2, for instance, augmentation of AMPK or Nrf2 signaling and inhibition of mTORC1, has been protective in diabetic nephropathy. Regarding the beneficial effects of Sestrin2 on diabetic nephropathy and its interaction with several signaling molecules, it is worth targeting Sestrin2 in diabetic nephropathy.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
33
|
Li Y, Li S, Wu L, Wu T, Li M, Du D, Chen Y, Wang C, Li X, Zhang S, Zhao Z, Zheng L, Chen M, Li M, Li T, Shi X, Qiao Y. Sestrin 2 Deficiency Exacerbates Noise-Induced Cochlear Injury Through Inhibiting ULK1/Parkin-Mediated Mitophagy. Antioxid Redox Signal 2023; 38:115-136. [PMID: 35708118 PMCID: PMC9885551 DOI: 10.1089/ars.2021.0283] [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: 01/09/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 02/03/2023]
Abstract
Aims: Noise damage to auditory hair cells is associated with oxidative stress and mitochondrial dysfunction. This study aimed to investigate the possible effect of sestrin 2 (SESN2), an endogenous antioxidant protein, on noise-induced hearing loss (NIHL) and the underlying mechanisms. Results: We identified SESN2 as a protective factor against oxidative stress in NIHL through activation of Parkin-mediated mitophagy. Consistently, SESN2 expression was increased and mitophagy was induced during the early stage after a temporary threshold shift due to noise exposure or hydrogen peroxide(H2O2) stimulation; conversely, SESN2 deficiency blocked mitophagy and exacerbated acoustic trauma. Mechanistically, SESN2 interacted with Unc-51-like protein kinase 1(ULK1), promoting ULK1 protein-level stabilization by interfering with its proteasomal degradation. This stabilization is essential for mitophagy initiation, since restoring ULK1 expression in SESN2-silenced cells rescued mitophagy defects. Innovation and Conclusion: Our results provide novel insights regarding SESN2 as a therapeutic target against noise-induced cochlear injury, possibly through improved mitophagy. Antioxid. Redox Signal. 38, 115-136.
Collapse
Affiliation(s)
- Yalan Li
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Shengsheng Li
- Department of Neurosurgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, P.R. China
| | - Liyuan Wu
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Tingting Wu
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Mengxiao Li
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Deliang Du
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Yalin Chen
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Caiji Wang
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Xuanyi Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, P.R. China
| | - Shili Zhang
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Zeqi Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, P.R. China
| | - Liting Zheng
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Mengbing Chen
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Menghua Li
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Ting Li
- School of Life Sciences, Xuzhou Medical University, Xuzhou, P.R. China
| | - Xi Shi
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Yuehua Qiao
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, P.R. China
| |
Collapse
|
34
|
Wang Z, Jin S, Xia T, Liu Y, Zhou Y, Liu X, Pan R, Liao Y, Yan M, Chang Q. Nelumbinis Stamen Ameliorates Chronic Restraint Stress-Induced Muscle Dysfunction and Fatigue in Mice by Decreasing Serum Corticosterone Levels and Activating Sestrin2. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:16188-16200. [PMID: 36529943 DOI: 10.1021/acs.jafc.2c06318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Nelumbo nucifera Gaertn. is an important aquatic vegetable, and its dried stamen (Nelumbinis stamen, NS) is a valuable nutraceutical usually used as a herbal tea. Here, we used ultrahigh-performance liquid chromatography (UPLC)-quadrupole time-of-flight mass spectrometry and high-performance liquid chromatography (HPLC) to chemically profile NS and quantify their main constituent flavonoids, respectively. In total, 44 components were identified, including organic acids, flavonoids, monoterpene glycosides, and fatty acids. Experimental mice were induced with fatigue by exposure to chronic restraint stress (CRS) for 8 h daily for 15 days and then treated with an aqueous extract of NS (0.5 and 1 g/kg) via gavage. NS significantly mitigated CRS-induced skeletal muscle dysfunction and fatigue in mice possibly by lowering serum corticosterone levels and restoring Sestrin2 expression in the gastrocnemius to regulate metabolism, preserve mitochondrial homeostasis, and promote antioxidant capacity. These results demonstrate that NS can be used as a nutraceutical or supplement for controlling stress-induced muscle dysfunction and fatigue.
Collapse
Affiliation(s)
- Zhi Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Suwei Jin
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Tianji Xia
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yongguang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yunfeng Zhou
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Xinmin Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Ruile Pan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yonghong Liao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Mingzhu Yan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| |
Collapse
|
35
|
Cho SS, Yang JH, Lee JH, Baek JS, Ku SK, Cho IJ, Kim KM, Ki SH. Ferroptosis contribute to hepatic stellate cell activation and liver fibrogenesis. Free Radic Biol Med 2022; 193:620-637. [PMID: 36370962 DOI: 10.1016/j.freeradbiomed.2022.11.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022]
Abstract
Ferroptosis is a widely known regulator of cell death in connection with the redox state as a consequence of the depletion of glutathione or accumulation of lipid peroxidation. Hepatic stellate cells (HSCs) play a pivotal role in the progression of hepatic fibrosis by increasing the production and secretion of the extracellular matrix. However, the role of ferroptosis in HSC activation and liver fibrogenesis has not been clearly elucidated. The ferroptosis inducer RAS-selective lethal 3 (RSL3) or erastin treatment in HSCs caused cell death. This effect was suppressed only after exposure to ferroptosis inhibitors. We observed induction of ferroptosis by RSL3 treatment in HSCs supported by decreased glutathione peroxidase 4, glutathione deficiency, reactive oxygen species generation, or lipid peroxidation. Interestingly, RSL3 treatment upregulated the expression of plasminogen activator inhibitor-1, a representative fibrogenic marker of HSCs. In addition, treatment with ferroptosis-inducing compounds increased c-JUN phosphorylation and activator protein 1 luciferase activity but did not alter Smad phosphorylation and Smad-binding element luciferase activity. Chronic administration of iron dextran to mice causes ferroptosis of liver in vivo. The expression of fibrosis markers, such as alpha-smooth muscle actin and plasminogen activator inhibitor-1, was increased in the livers of mice with iron accumulation. Hepatic injury accompanying liver fibrosis was observed based on the levels of alanine aminotransferase, aspartate aminotransferase, and hematoxylin and eosin staining. Furthermore, we found that both isolated primary hepatocyte and HSCs undergo ferroptosis. Consistently, cirrhotic liver tissue of patients indicated glutathione peroxidase 4 downregulation in fibrotic region. In conclusion, our results suggest that ferroptosis contribute to HSC activation and the progression of hepatic fibrosis.
Collapse
Affiliation(s)
- Sam Seok Cho
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ji Hyun Lee
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Jin Sol Baek
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeongsangbuk-do, 38610, Republic of Korea
| | - Il Je Cho
- College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeongsangbuk-do, 38610, Republic of Korea
| | - Kyu Min Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea; Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju, 61452, Republic of Korea.
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
36
|
Li H, Wu M, Guo C, Zhai R, Chen J. Tanshinone IIA Regulates Keap1/Nrf2 Signal Pathway by Activating Sestrin2 to Restrain Pulmonary Fibrosis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:2125-2151. [PMID: 36309810 DOI: 10.1142/s0192415x22500914] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tanshinone IIA (Tan-IIA) is a major component extracted from the traditional herbal medicine Danshen, which has shown antipulmonary fibrosis by suppress reactive oxygen species-mediated activation of myofibroblast. However, the exact mechanism of Tan-IIA against pulmonary fibrosis (PF) remains unclear. This work aimed to explore the underlying mechanism of the protective effects of Tan-IIA on PF. By using high-throughput RNA-Seq analysis, we have compared the genome-wide gene expression profiles and pathway enrichment of Tan-IIA-treated NIH-3T3 cells with or without transforming growth factor beta 1 (TGF-[Formula: see text]1) induction. In normal NIH-3T3 cells, Tan-IIA treatment up-regulated 181 differential expression genes (DEGs) and down-regulated 137 DEGs. In TGF-[Formula: see text]1-induced NIH-3T3 cells, Tan-IIA treatment up-regulated 709 DEGs and down-regulated 1075 DEGs, and these DEGs were enriched in extracellular matrix organization, collagen fibril organization, cell adhesion, ECM-receptor interaction, PI3K-Akt signaling pathway and P53 signaling pathway. Moreover, there were 207 co-expressed DEGs between Tan-IIA treatment vs. the Control and TGF-[Formula: see text]1 plus Tan-IIA treatment vs. TGF-[Formula: see text]1 alone treatment, some of which were related to anti-oxidative stress. In both normal and TGF-[Formula: see text]1-induced NIH-3T3 cells, protein-protein interaction network analysis indicated that Tan-IIA can regulate the expression of several common anti-oxidant genes including Heme oxygenase 1 (Ho-1, also known as Homx1), Sestrin2 (Sesn2), GCL modifier subunit (Gclm), GCL catalytic subunit (Gclc) and Sequestosome-1 (Sqstm1). Quantitative Real-time polymerase chain reaction analysis confirmed some DEGs specifically expressing on Tan-IIA treated cells, which provided new candidates for further functional studies of Tan-IIA. In both in vitro and in vivo PF models, the protein expression of Sesn2 was significantly enhanced by Tan-IIA treatment. Overexpression and knockdown experiments showed that Sesn2 is required for Tan-IIA against TGF-[Formula: see text]1-induced myofibroblast activation by reinforcing nuclear factor-erythroid 2-related factor 2 (Nrf2)-mediated anti-oxidant response via downregulation of kelch-like ECH-associated protein 1 (Keap1). These results suggest Tan-IIA inhibits myofibroblast activation by activating Sesn2-Nrf2 signaling pathway, and provide a new insight into the essential role of Sesn2 in PF.
Collapse
Affiliation(s)
- Hongxia Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Nanjing, Jiangsu 210009, P. R. China.,Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P. R. China
| | - Mingyu Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Nanjing, Jiangsu 210009, P. R. China.,Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P. R. China
| | - Congying Guo
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Nanjing, Jiangsu 210009, P. R. China.,Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P. R. China
| | - Rao Zhai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Nanjing, Jiangsu 210009, P. R. China.,Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P. R. China
| | - Jun Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Lane, Nanjing, Jiangsu 210009, P. R. China.,Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, P. R. China
| |
Collapse
|
37
|
Tamari S, Menju T, Toyazaki T, Miyamoto H, Chiba N, Noguchi M, Ishikawa H, Miyata R, Kayawake H, Tanaka S, Yamada Y, Yutaka Y, Nakajima D, Ohsumi A, Hamaji M, Date H. Nrf2/p‑Fyn/ABCB1 axis accompanied by p‑Fyn nuclear accumulation plays pivotal roles in vinorelbine resistance in non‑small cell lung cancer. Oncol Rep 2022; 48:171. [PMID: 35959810 DOI: 10.3892/or.2022.8386] [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: 04/12/2022] [Accepted: 07/19/2022] [Indexed: 11/05/2022] Open
Abstract
Adjuvant cisplatin‑vinorelbine is a standard therapy for stage II/III lung cancer. However, a poor survival rate of patients with lung cancer is attributed to vinorelbine resistance arising from ATP‑binding cassette (ABC) sub‑family B member 1 (ABCB1) and phosphorylated Fyn (p‑Fyn) overexpression. However, the underlying mechanisms remain unclear. NF‑E2‑related factor 2 (Nrf2) regulates the ABC family and activates the nuclear transport of Fyn. The present study evaluated the roles of the Nrf2/p‑Fyn/ABCB1 axis in vinorelbine‑resistant (VR) cells and clinical samples. To establish VR cells, H1299 cells were exposed to vinorelbine, and the intracellular reactive oxygen species (ROS) level in the H1299 cells was determined using a DCFH‑DA assay. The total and subcellular expression of Nrf2, ABCB1 and p‑Fyn in VR cells was evaluated. Immunofluorescence was used to detect the subcellular localization of p‑Fyn in VR cells. A cell viability assay was used to examine whether the sensitivity of VR cells to vinorelbine is dependent on Nrf2 activity. Immunohistochemistry was performed on 104 tissue samples from patients with lung cancer who underwent surgery followed by cisplatin‑vinorelbine treatment. The results revealed that persistent exposure to vinorelbine induced intracellular ROS formation in H1299 cells. p‑Fyn was localized in the nucleus, and ABCB1 and Nrf2 were overexpressed in VR cells. ABCB1 expression was dependent on Nrf2 downstream activation. The decreased expression of Nrf2 restored the sensitivity of VR cells to vinorelbine. In the surgical samples, Nrf2 and ABCB1 were associated with disease‑free survival, and p‑Fyn was associated with overall survival (P<0.05). On the whole, the present study demonstrates that Nrf2 upregulates ABCB1 and, accompanied by the nuclear accumulation of p‑Fyn, induces vinorelbine resistance. These findings may facilitate the development of drug resistance prevention strategies or new drug targets against non‑small cell lung cancer.
Collapse
Affiliation(s)
- Shigeyuki Tamari
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Toshiya Toyazaki
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Hideaki Miyamoto
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Naohisa Chiba
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Misa Noguchi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Hiroaki Ishikawa
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Ryo Miyata
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Hidenao Kayawake
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Satona Tanaka
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Yoshito Yamada
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Yojiro Yutaka
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Masatsugu Hamaji
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606‑8507, Japan
| |
Collapse
|
38
|
Serum Sestrin2 Was Lower in Septic Shock Patients with Cardiomyopathy. DISEASE MARKERS 2022; 2022:1390373. [PMID: 36092963 PMCID: PMC9458382 DOI: 10.1155/2022/1390373] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022]
Abstract
Background To determine the clinical significance of variations in serum sestrin2 protein levels in the development of septic cardiomyopathy in septic shock patients. Methods The serum sestrin2 concentrations of each sample were determined using ELISA in a total of 67 control persons and 188 patients with septic shock. Furthermore, using transthoracic echocardiography, septic shock patients were split into two groups based on whether or not cardiomyopathy had developed, and the differences in each index between the two groups were analyzed. We looked at the relationship between serum sestrin2 levels, norepinephrine dosage, and NTproBNP levels. The influencing variables for the prediction of septic cardiomyopathy linked with the development of septic cardiomyopathy and clinical prognosis in septic cardiomyopathy were determined using multivariate binary logistic regression. Results Assessment of left ventricular systolic function by measurement of LVEF revealed that 61/188 (32.4%) of the 188 patients with septic shock included in the research satisfied the diagnostic criteria for septic cardiomyopathy. (1) Sestrin2 protein levels showed a significant difference between septic shock and healthy controls (p < 0.01). (2) Compared to the group without septic cardiomyopathy, the group with combined septic cardiomyopathy had lower serum sestrin2 protein levels (p < 0.05), lower systolic blood pressure (p < 0.05), and higher plasma NTproBNP levels (p < 0.01) and used greater norepinephrine dosages (p < 0.01). The levels of serum sestrin2 protein revealed a little negative relationship with NTproBNP and norepinephrine dose. However, a binary logistic regression analysis revealed that none of these factors was an independent predictor of septic shock. (3) Age, lactate level, SOFA score, positive bacteremia, and sestrin2 protein were shown to be substantial discrepancies in clinical outcomes in patients with septic cardiomyopathy, becoming variables that impact clinical outcomes. Positive bacteremia (p = 0.031, OR = 5.084), SOFA score (p = 0.021, OR = 1.304), and sestrin2 protein (p = 0.039, OR = 0.897) were revealed to have independent influences in predicting clinical mortality outcome in septic cardiomyopathy using multivariate binary logistic regression. Conclusion High serum sestrin2 levels clearly distinguish septic shock patients from healthy controls, whereas low serum sestrin2 levels are related with cardiac dysfunction to some extent but are not an independent influence factor for septic cardiomyopathy. Low serum sestrin2 levels were shown to be useful in predicting clinical outcome in patients with septic cardiomyopathy.
Collapse
|
39
|
Krause-Hauch M, Fedorova J, Zoungrana LI, Wang H, Fatmi MK, Li Z, Iglesias M, Slotabec L, Li J. Targeting on Nrf2/Sesn2 Signaling to Rescue Cardiac Dysfunction during High-Fat Diet-Induced Obesity. Cells 2022; 11:cells11162614. [PMID: 36010689 PMCID: PMC9406590 DOI: 10.3390/cells11162614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/14/2022] Open
Abstract
Obesity is of concern to the population because it is known to cause inflammation and oxidative stress throughout the body, leading to patient predisposition for health conditions such as diabetes, hypertension, and some cancers. However, some proteins that are activated in times of oxidative stress may provide cytoprotective properties. In this study, we aim to gain further understanding of the interconnection between Nrf2 and Sesn2 during obesity-related stress and how this relationship can play a role in cardio-protection. Cardiomyocyte-specific Sesn2 knockout (cSesn2-/-) and Sesn2 overexpressed (tTa-tet-Sesn2) mice and their wildtype littermates (Sesn2flox/flox and tet-Sesn2, respectively) were assigned to either a normal chow (NC) or a high-fat (HF) diet to induce obesity. After 16 weeks of dietary intervention, heart function was evaluated via echocardiography and cardiac tissue was collected for analysis. Immunoblotting, histology, and ROS staining were completed. Human heart samples were obtained via the LifeLink Foundation and were also subjected to analysis. Overall, these results indicated that the overexpression of Sesn2 appears to have cardio-protective effects on the obese heart through the reduction of ROS and fibrosis present in the tissues and in cardiac function. These results were consistent for both mouse and human heart samples. In human samples, there was an increase in Sesn2 and Nrf2 expression in the obese patients' LV tissue. However, there was no observable pattern of Sesn2/Nrf2 expression in mouse LV tissue samples. Further investigation into the link between the Sesn2/Nrf2 pathway and obesity-related oxidative stress is needed.
Collapse
Affiliation(s)
- Meredith Krause-Hauch
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A. Haley Veterans’ Hospital, Tampa, FL 33612, USA
| | - Julia Fedorova
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Linda Ines Zoungrana
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Hao Wang
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Mohammad Kasim Fatmi
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Zehui Li
- Department of Medical Engineering, College of Engineering and Morsani College of Medicine, Tampa, FL 33612, USA
| | - Migdalia Iglesias
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Lily Slotabec
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Ji Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A. Haley Veterans’ Hospital, Tampa, FL 33612, USA
- Correspondence: ; Tel.: +1-813-974-4917
| |
Collapse
|
40
|
Oxidative Stress-Induced Protein of SESTRIN2 in Cardioprotection Effect. DISEASE MARKERS 2022; 2022:7439878. [PMID: 35937943 PMCID: PMC9355779 DOI: 10.1155/2022/7439878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/12/2022] [Accepted: 07/16/2022] [Indexed: 12/02/2022]
Abstract
Because of the rich mitochondria and high energy metabolic requirements, excessive oxidative stress generated by ROS is a key pathogenic mechanism in heart disease. SESTRIN2, the well-known antioxidant protein, plays a vital role in diminishing the production and accumulation of ROS, thus sparing cells from oxidative damage. From this new perspective, we first examine SESTRIN2 structure-function relationships; then, we describe how SESTRIN2 expression is regulated under oxidative stress conditions, emphasizing SESTRIN2's antioxidant mechanism via multiple signal transductions; and finally, we discuss SESTRIN2's role in a variety of oxidative stress-related cardiac diseases, including age-related heart disease, diabetic cardiomyopathy, ischemia-reperfusion myocardial injury, septic cardiomyopathy, and chronic cardiac insufficiency. The goal of this review is to identify the SESTRIN2 protein as a potential biomarker and new therapy target for oxidative stress-related cardiac diseases.
Collapse
|
41
|
Yu TJ, Shiau JP, Tang JY, Yen CH, Hou MF, Cheng YB, Shu CW, Chang HW. Physapruin A Induces Reactive Oxygen Species to Trigger Cytoprotective Autophagy of Breast Cancer Cells. Antioxidants (Basel) 2022; 11:antiox11071352. [PMID: 35883843 PMCID: PMC9311569 DOI: 10.3390/antiox11071352] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023] Open
Abstract
Physalis peruviana-derived physapruin A (PHA) is a potent compound that selectively generates reactive oxygen species (ROS) and induces cancer cell death. Autophagy, a cellular self-clearance pathway, can be induced by ROS and plays a dual role in cancer cell death. However, the role of autophagy in PHA-treated cancer cells is not understood. Our study initially showed that autophagy inhibitors such as bafilomycin A1 enhanced the cytotoxic effects of PHA in breast cancer cell lines, including MCF7 and MDA-MB-231. PHA treatment decreased the p62 protein level and increased LC3-II flux. PHA increased the fluorescence intensity of DAPGreen and DALGreen, which are used to reflect the formation of autophagosome/autolysosome and autolysosome, respectively. ROS scavenger N-acetylcysteine (NAC) decreased PHA-elevated autophagy activity, implying that PHA-induced ROS may be required for autophagy induction in breast cancer cells. Moreover, the autophagy inhibitor increased ROS levels and enhanced PHA-elevated ROS levels, while NAC scavenges the produced ROS resulting from PHA and autophagy inhibitor. In addition, the autophagy inhibitor elevated the PHA-induced proportion of annexin V/7-aminoactinmycin D and cleavage of caspase-3/8/9 and poly (ADP-ribose) polymerase. In contrast, NAC and apoptosis inhibitor Z-VAD-FMK blocked the proportion of annexin V/7-aminoactinmycin D and the activation of caspases. Taken together, PHA induced ROS to promote autophagy, which might play an antioxidant and anti-apoptotic role in breast cancer cells.
Collapse
Affiliation(s)
- Tzu-Jung Yu
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (T.-J.Y.); (C.-H.Y.)
| | - Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (J.-P.S.); (M.-F.H.)
- Department of Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 81267, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (T.-J.Y.); (C.-H.Y.)
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (J.-P.S.); (M.-F.H.)
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yuan-Bin Cheng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
| | - Chih-Wen Shu
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Correspondence: (C.-W.S.); (H.-W.C.); Tel.: +886-7-525-2000 (ext. 5828) (C.-W.S.); +886-7-312-1101 (ext. 2691) (H.-W.C.)
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (C.-W.S.); (H.-W.C.); Tel.: +886-7-525-2000 (ext. 5828) (C.-W.S.); +886-7-312-1101 (ext. 2691) (H.-W.C.)
| |
Collapse
|
42
|
Sawa R, Ohnishi A, Ohno M, Nagata M, Wake I, Okimura Y. Specific amino acids regulate Sestrin2 mRNA and protein levels in an ATF4-dependent manner in C2C12 myocytes. Biochim Biophys Acta Gen Subj 2022; 1866:130174. [PMID: 35597502 DOI: 10.1016/j.bbagen.2022.130174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/29/2022] [Accepted: 05/16/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Sestrin2 is a conserved protein in several species, and its expression is upregulated in cells under various environmental stresses. Sestrin2 content is involved in the function of mechanistic target of rapamycin complex 1 (mTORC1) in mouse embryonic fibroblasts and C2C12 cells. METHODS C2C12 cells were treated with amino acid-free DMEM (AF-DMEM) for 5 h. The effects of the addition of specific amino acids to AF-DMEM on Sestrin2 mRNA and protein expression were examined using RT-qPCR and western blotting, respectively. The mechanism by which amino acids regulate Sestrin2 mRNA expression was examined using blocking and siRNA experiments. RESULTS AF-DMEM increased the mRNA and protein levels of both Sestrin2 and activating transcription factor 4 (ATF4). The addition of a specific amino acid changed Sestrin2 mRNA and protein levels. The response pattern of Sestrin2 to specific amino acids was similar to that of ATF4. ATF4 siRNA reduced Sestrin2 mRNA levels. AF-DMEM increased eukaryotic initiation factor 2α (eIF2α) phosphorylation as early as 10 min after the treatment; however, ATF4 and Sestrin2 were increased 300 min after the treatment. Nuclear factor erythroid 2-related factor 2 and pancreatic and duodenal homeobox 1 siRNA did not affect ATF4 and Sestrin2 mRNA expression. CONCLUSIONS Specific Amino acids regulate Sestrin2 levels in an ATF4-dependent manner in C2C12 cells. GENERAL SIGNIFICANCE The results of the present study indicate that amino acids regulate levels of Sestrin2, which might cause phenotypic alterations, including mTORC1 activity, in C2C12 cells.
Collapse
Affiliation(s)
- Ran Sawa
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan
| | - Ayaka Ohnishi
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan
| | - Maya Ohno
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan
| | - Maika Nagata
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan
| | - Ikumi Wake
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan
| | - Yasuhiko Okimura
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan.
| |
Collapse
|
43
|
Abstract
AbstractSestrin2 is a conserved antioxidant, metabolism regulator, and downstream of P53. Sestrin2 can suppress oxidative stress and inflammation, thereby preventing the development and progression of cancer. However, Sestrin2 attenuates severe oxidative stress by activating nuclear factor erythroid 2-related factor 2 (Nrf2), thereby enhancing cancer cells survival and chemoresistance. Sestrin2 inhibits endoplasmic reticulum stress and activates autophagy and apoptosis in cancer cells. Attenuation of endoplasmic reticulum stress and augmentation of autophagy hinders cancer development but can either expedite or impede cancer progression under specific conditions. Furthermore, Sestrin2 can vigorously inhibit oncogenic signaling pathways through downregulation of mammalian target of rapamycin complex 1 (mTORC1) and hypoxia-inducible factor 1-alpha (HIF-1α). Conversely, Sestrin2 decreases the cytotoxic activity of T cells and natural killer cells which helps tumor cells immune evasion. Sestrin2 can enhance tumor cells viability in stress conditions such as glucose or glutamine deficiency. Cancer cells can also upregulate Sestrin2 during chemotherapy or radiotherapy to attenuate severe oxidative stress and ER stress, augment autophagy and resist the treatment. Recent studies unveiled that Sestrin2 is involved in the development and progression of several types of human cancer. The effect of Sestrin2 may differ depending on the type of tumor, for instance, several studies revealed that Sestrin2 protects against colorectal cancer, whereas results are controversial regarding lung cancer. Furthermore, Sestrin2 expression correlates with metastasis and survival in several types of human cancer such as colorectal cancer, lung cancer, and hepatocellular carcinoma. Targeted therapy for Sestrin2 or regulation of its expression by new techniques such as non-coding RNAs delivery and vector systems may improve cancer chemotherapy and overcome chemoresistance, metastasis and immune evasion that should be investigated by future trials.
Collapse
|
44
|
Jiménez A, Lu D, Kalocsay M, Berberich MJ, Balbi P, Jambhekar A, Lahav G. Time‐series transcriptomics and proteomics reveal alternative modes to decode p53 oscillations. Mol Syst Biol 2022; 18:e10588. [PMID: 35285572 PMCID: PMC8919251 DOI: 10.15252/msb.202110588] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 12/21/2022] Open
Affiliation(s)
- Alba Jiménez
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
| | - Dan Lu
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
| | - Marian Kalocsay
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
- Laboratory of Systems Pharmacology Blavatnik Institute at Harvard Medical School Boston MA USA
| | - Matthew J Berberich
- Laboratory of Systems Pharmacology Blavatnik Institute at Harvard Medical School Boston MA USA
- Center for Protein Degradation Dana‐Farber Cancer Institute Boston MA USA
| | - Petra Balbi
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
| | - Ashwini Jambhekar
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
- Ludwig Center at Harvard Medical School Boston MA USA
| | - Galit Lahav
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
- Ludwig Center at Harvard Medical School Boston MA USA
| |
Collapse
|
45
|
Rahul, Siddique YH. Neurodegenerative Diseases and Flavonoids: Special Reference to Kaempferol. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 20:327-342. [PMID: 33511932 DOI: 10.2174/1871527320666210129122033] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/21/2020] [Accepted: 09/13/2020] [Indexed: 12/20/2022]
Abstract
Neurodegenerative diseases like Alzheimer's disease, Parkinson's disease, Huntington's disease, Multiple Sclerosis and Ischemic stroke have become a major health problem worldwide. Pre-clinical studies have demonstrated the beneficial effects of flavonoids on neurodegenerative diseases and suggest them to be used as therapeutic agents. Kaempferol is found in many plants such as tea, beans, broccoli, strawberries, and neuroprotective effects against the development of many neurodegenerative diseases such as Parkinson, Alzheimer's disease and Huntington's disease. The present study summarizes the neuroprotective effects of kaempferol in various models of neurodegenerative diseases. Kaempferol delays the initiation as well as the progression of neurodegenerative disorders by acting as a scavenger of free radicals and preserving the activity of various antioxidant enzymes. Kaempferol can cross the Blood-Brain Barrier (BBB), and therefore results in an enhanced protective effect. The multi-target property of kaempferol makes it a potential dietary supplement in preventing and treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Rahul
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| | - Yasir H Siddique
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, Uttar Pradesh, India
| |
Collapse
|
46
|
Chang KC, Liu PF, Chang CH, Lin YC, Chen YJ, Shu CW. The interplay of autophagy and oxidative stress in the pathogenesis and therapy of retinal degenerative diseases. Cell Biosci 2022; 12:1. [PMID: 34980273 PMCID: PMC8725349 DOI: 10.1186/s13578-021-00736-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/19/2021] [Indexed: 12/27/2022] Open
Abstract
Oxidative stress is mainly caused by intracellular reactive oxygen species (ROS) production, which is highly associated with normal physiological homeostasis and the pathogenesis of diseases, particularly ocular diseases. Autophagy is a self-clearance pathway that removes oxidized cellular components and regulates cellular ROS levels. ROS can modulate autophagy activity through transcriptional and posttranslational mechanisms. Autophagy further triggers transcription factor activation and degrades impaired organelles and proteins to eliminate excessive ROS in cells. Thus, autophagy may play an antioxidant role in protecting ocular cells from oxidative stress. Nevertheless, excessive autophagy may cause autophagic cell death. In this review, we summarize the mechanisms of interaction between ROS and autophagy and their roles in the pathogenesis of several ocular diseases, including glaucoma, age-related macular degeneration (AMD), diabetic retinopathy (DR), and optic nerve atrophy, which are major causes of blindness. The autophagy modulators used to treat ocular diseases are further discussed. The findings of the studies reviewed here might shed light on the development and use of autophagy modulators for the future treatment of ocular diseases.
Collapse
Affiliation(s)
- Kun-Che Chang
- Department of Ophthalmology and Neurobiology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Science, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Hsuan Chang
- Institute of BioPharmaceutical Sciences, National Sun Yat-Sen University, No. 70, Lianhai Rd., Gushan Dist., Kaohsiung, 80424, Taiwan
| | - Ying-Cheng Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yen-Ju Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chih-Wen Shu
- Institute of BioPharmaceutical Sciences, National Sun Yat-Sen University, No. 70, Lianhai Rd., Gushan Dist., Kaohsiung, 80424, Taiwan.
| |
Collapse
|
47
|
Chen Y, Huang T, Yu Z, Yu Q, Wang Y, Hu J, Shi J, Yang G. The functions and roles of sestrins in regulating human diseases. Cell Mol Biol Lett 2022; 27:2. [PMID: 34979914 PMCID: PMC8721191 DOI: 10.1186/s11658-021-00302-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Sestrins (Sesns), highly conserved stress-inducible metabolic proteins, are known to protect organisms against various noxious stimuli including DNA damage, oxidative stress, starvation, endoplasmic reticulum (ER) stress, and hypoxia. Sesns regulate metabolism mainly through activation of the key energy sensor AMP-dependent protein kinase (AMPK) and inhibition of mammalian target of rapamycin complex 1 (mTORC1). Sesns also play pivotal roles in autophagy activation and apoptosis inhibition in normal cells, while conversely promoting apoptosis in cancer cells. The functions of Sesns in diseases such as metabolic disorders, neurodegenerative diseases, cardiovascular diseases, and cancer have been broadly investigated in the past decades. However, there is a limited number of reviews that have summarized the functions of Sesns in the pathophysiological processes of human diseases, especially musculoskeletal system diseases. One aim of this review is to discuss the biological functions of Sesns in the pathophysiological process and phenotype of diseases. More significantly, we include some new evidence about the musculoskeletal system. Another purpose is to explore whether Sesns could be potential biomarkers or targets in the future diagnostic and therapeutic process.
Collapse
Affiliation(s)
- Yitong Chen
- Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Tingben Huang
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Zhou Yu
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Qiong Yu
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Ying Wang
- Department of Oral Medicine, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Ji'an Hu
- Department of Oral Pathology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China.
| | - Jiejun Shi
- Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China.
| | - Guoli Yang
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
48
|
Mao EW, Cheng XB, Li WC, Kan CX, Huang N, Wang HS, Hou NN, Sun XD. Association between serum Sestrin2 level and diabetic peripheral neuropathy in type 2 diabetic patients. World J Clin Cases 2021; 9:11156-11164. [PMID: 35071546 PMCID: PMC8717510 DOI: 10.12998/wjcc.v9.i36.11156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/06/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diabetic peripheral neuropathy (DPN) is a chronic and serious microvascular complication of diabetes linked to redox imbalance. Sestrin2, a novel inducible stress protein, participates in glucose metabolic regulation and redox homeostasis. However, the association between serum Sestrin2 and DPN is unknown.
AIM To explore the association between serum Sestrin2 and DPN in patients with type 2 diabetes mellitus (T2DM).
METHODS A total of 96 T2DM patients and 39 healthy volunteers, matched by age and sex, participated in this cross-sectional study. Clinical features and metabolic indices were identified. Serum Sestrin2 was measured by ELISA. The association between Sestrin2 and DPN was studied. Correlation and logistic regression analyses were used to evaluate the associations of different metabolic indices with Sestrin2 and DPN.
RESULTS The 96 patients with T2DM were divided into DPN (n = 47) and patients without DPN (n = 49). Serum Sestrin2 was significantly lower in healthy volunteers than in all T2DM patients combined [9.10 (5.41-13.53) ng/mL vs 12.75 (7.44-23.80) ng/mL, P < 0.01]. T2DM patients without DPN also had significantly higher levels of Sestrin2 than healthy volunteers [14.58 (7.93-26.62) ng/mL vs 9.10 (5.41-13.53) ng/mL, P < 0.01]. However, T2DM patients with DPN had lower circulating Sestrin2 levels compared to T2DM patients without DPN [9.86 (6.72-21.71) ng/mL vs 14.58 (7.93-26.62) ng/mL, respectively, P < 0.01]. Bivariate correlation analysis revealed that serum Sestrin2 was positively correlated with body mass index (r = 0.672, P = 0.000), hemoglobin A1c (HbA1c) (r = 0.292, P = 0.000), serum creatinine (r = 0.206, P = 0.016), triglycerides (r = 0.731, P = 0.000), and fasting glucose (r = 0.202, P = 0.040), and negatively associated with estimated glomerular filtration rate (r = -0.230, P = 0.007). After adjustment for sex, age, HbA1c, and diabetes duration, multiple regression analysis revealed that Sestrin2 was independently correlated with body mass index and triglyceride levels (P = 0.000). Logistic regression analyses indicated that Sestrin2, diabetes duration, and high-density lipoprotein were strongly associated with DPN (odds ratio = 0.855, 1.411, and 0.041, respectively).
CONCLUSION Our results show Sestrin2 is decreased in T2DM patients with DNP. As lower Sestrin2 is independently associated with DPN, Sestrin2 may contribute to progression of DPN in T2DM patients.
Collapse
Affiliation(s)
- En-Wen Mao
- Department of Endocrinology and Metabolism, Clinical Research Center, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Xue-Bing Cheng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Wen-Chao Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Cheng-Xia Kan
- Department of Endocrinology and Metabolism, Clinical Research Center, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Na Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Hong-Sheng Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Ning-Ning Hou
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| | - Xiao-Dong Sun
- Department of Endocrinology and Metabolism, Clinical Research Center, The Affiliated Hospital of Weifang Medical University, Weifang 261031, Shandong Province, China
| |
Collapse
|
49
|
Cho SS, Lee JH, Kim KM, Park EY, Ku SK, Cho IJ, Yang JH, Ki SH. REDD1 attenuates hepatic stellate cell activation and liver fibrosis via inhibiting of TGF-β/Smad signaling pathway. Free Radic Biol Med 2021; 176:246-256. [PMID: 34614448 DOI: 10.1016/j.freeradbiomed.2021.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 12/20/2022]
Abstract
Liver fibrosis is caused by repetitive hepatic injury. Regulated in development and DNA damage response 1 (REDD1) gene is induced by various stresses and has been studied in cell proliferation and survival. However, the role of REDD1 in hepatic stellate cell activation and hepatic fibrogenesis has not yet been investigated. In the current study, we examined the effect of REDD1 on hepatic fibrogenesis and the underlying molecular mechanism. REDD1 protein was upregulated in the activated primary hepatic stellate cells and transforming growth factor-β (TGF-β)-treated LX-2 cells. REDD1 mRNA levels were also elevated by TGF-β treatment. TGF-β signaling is primarily transduced via the activation of the Smad transcription factor. However, TGF-β-mediated REDD1 induction was not Smad-dependent. Thus, we investigated the transcription factors that influence the REDD1 expression by TGF-β. We found that c-JUN, a component of AP-1, upregulated the REDD1 expression that was specifically suppressed by p38 inhibitor. In silico analysis of the REDD1 promoter region showed putative AP-1-binding sites; additionally, its deletion mutants demonstrated that the AP-1-binding site between -716 and -587 bp within the REDD1 promoter is critical for TGF-β-mediated REDD1 induction. Moreover, REDD1 overexpression markedly inhibited TGF-β-induced plasminogen activator inhibitor-1 (PAI-1) expression and Smad phosphorylation. REDD1 adenovirus infection inhibited CCl4-induced hepatic injury in mice, which was demonstrated by reduced ALT/AST levels and collagen accumulation. In addition, we observed that REDD1 inhibited CCl4-induced fibrogenic gene induction and restored GSH and malondialdehyde levels. Our findings implied that REDD1 has the potential to inhibit HSC activation and protect against liver fibrosis.
Collapse
Affiliation(s)
- Sam Seok Cho
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Ji Hyun Lee
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Kyu Min Kim
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea; Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju, 61452, Republic of Korea
| | - Eun Young Park
- College of Pharmacy, Mokpo National University, Muan-gun, Jeollanam-do, 58554, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeongsangbuk-do, 38610, Republic of Korea
| | - Il Je Cho
- College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeongsangbuk-do, 38610, Republic of Korea
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do, 58245, Republic of Korea.
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
50
|
Lee S, Pham DV, Park PH. Sestrin2 induction contributes to anti-inflammatory responses and cell survival by globular adiponectin in macrophages. Arch Pharm Res 2021; 45:38-50. [PMID: 34797495 DOI: 10.1007/s12272-021-01364-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022]
Abstract
Adiponectin, an adipose tissue-derived hormone, exhibits a modulatory effect on cell death/survival and possesses potent anti-inflammatory properties. However, the underlying molecular mechanisms remain elusive. Sestrin2, a stress-inducible metabolic protein, has shown cytoprotective and inflammation-modulatory effects under stressful conditions. In this study, we examined the role of sestrin2 signaling in the modulation of cell survival and inflammatory responses by globular adiponectin (gAcrp) in macrophages. We observed that gAcrp induced a significant increase in sestrin2 expression in both RAW 264.7 murine macrophages and primary murine macrophages. Notably, gAcrp treatment markedly increased expression of hypoxia inducible factor-1 α (HIF-1α) and gene silencing of HIF-1α blocked sestrin2 induction by gAcrp. In addition, pretreatment with a pharmacological inhibitor of ERK or PI3K abrogated both sestrin2 and HIF-1α expression by gAcrp, indicating that ERK/PI3K-mediated HIF-1α signaling pathway plays a critical role in sestrin2 induction by gAcrp. Furthermore, sestrin2 induction is implicated in autophagy activation, and knockdown of sestrin2 prevented enhanced cell viability by gAcrp. Moreover, gene silencing of sestrin2 caused restoration of gAcrp-induced expression of anti-inflammatory genes in a gene-selective manner. Taken together, these results indicate that sestrin2 induction critically contributes to cell survival and anti-inflammatory responses by gAcrp in macrophages.
Collapse
Affiliation(s)
- Sumin Lee
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Duc-Vinh Pham
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea. .,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|