1
|
Dhapola R, Kumari S, Sharma P, Vellingiri B, HariKrishnaReddy D. Advancements in autophagy perturbations in Alzheimer's disease: Molecular aspects and therapeutics. Brain Res 2025; 1851:149494. [PMID: 39922409 DOI: 10.1016/j.brainres.2025.149494] [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: 09/23/2024] [Revised: 12/11/2024] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Emerging evidences suggest that autophagy, a key cellular process responsible for degrading and recycling damaged organelles and proteins, plays a crucial role in maintaining neuronal health. Dysfunctional autophagy has been linked to the pathogenesis of Alzheimer's disease (AD), contributing to the accumulation of misfolded proteins and cellular debris. Molecular mechanisms underlying autophagy dysfunction in AD involve amyloid-beta (Aβ) and tau accumulation, neuroinflammation, mitochondrial dysfunction, oxidative stress and endoplasmic reticulum stress. Disrupted signaling pathways such as TRIB3, Nmnat and BAG3 that regulate key processes like autophagosome initiation, lysosome function, and protein homeostasis also play a crucial role in the pathogenesis. Restoration of autophagy by modulating these molecular and signaling pathways may be an effective therapeutic strategy for AD. Studies have found few drugs targeting autophagy dysregulation in AD. These drugs include metformin that has been found to modulate the expression of TRIB3 for autophagy regulation. Another drug, resveratrol has been reported to augment the activity of Nmnat thus, increases autophagy flux. BACE1 and mTOR inhibitors like arctigenin, nilvadipine and dapagliflozin were also found to restore autophagy. This study elaborates recent advances in signaling and molecular pathways and discusses current and emerging therapeutic interventions targeting autophagy dysfunction in AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab 151401 Bathinda, Punjab, India.
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India.
| |
Collapse
|
2
|
Sun FW, Tian Y. Intermittent Fasting Improves Sevoflurane-Induced Cognitive Dysfunction in Rats Through SIRT1-Mediated Autophagy. Neurochem Res 2025; 50:81. [PMID: 39831923 DOI: 10.1007/s11064-025-04335-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/15/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Perioperative neurocognitive disorders (PND) is a common complication affecting the central nervous system, commonly induced by anesthesia and surgical procedures. PND has garnered considerable attention in recent years, not only due to its high morbidity but also its negative impact on patient prognosis, such as increased rates of dementia and mortality. Sevoflurane, a common volatile anesthetic in clinical practice, is increasingly linked to being a potential risk factor for PND with prolonged inhalation, yet effective prevention and treatment methods remain elusive. Autophagy, a crucial regulatory process for maintaining organism function, has been shown to play a key role in sevoflurane-induced cognitive dysfunction. In recent years, intermittent fasting (IF), a unique dietary pattern, has gained significant recognition. IF has been shown in multiple studies to offer neuroprotective advantages in different central nervous system conditions. disorders. This study aims to explore the potential neuroprotective effects of intermittent fasting preconditioning through the autophagic pathway in sevoflurane-induced cognitive impairment in rats and its underlying mechanisms.
Collapse
Affiliation(s)
- Feng-Wei Sun
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yue Tian
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
刘 本, 王 业, 任 海, 欧 丽, 邓 轩, 黄 梦, 吴 鑫, 龚 权. [3-Methyladenine alleviates early renal injury in diabetic mice by inhibiting AKT signaling]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1236-1242. [PMID: 39051069 PMCID: PMC11270656 DOI: 10.12122/j.issn.1673-4254.2024.07.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE To explore the mechanism of 3-methyladenine (3-MA) for alleviating early diabetic renal injury. METHODS Mouse models of streptozotocin (STZ) -induced diabetes mellitus were randomized into model group and 3-MA treatment group for daily treatments with normal saline and 10 mg/kg 3-MA by gavage for 6 weeks, respectively. Body weight and fasting blood glucose of the mice were recorded every week. After the treatments, the kidneys of the mice were collected for measurement kidney/body weight ratio, examination of glomerular size with PAS staining, and detection of α-SMA and PCNA expressions using Western blotting and immunohistochemistry. SV40 MES 13 cells cultured in normal glucose (5.6 mmol/L) and high glucose (30 mmol/L) were treated with 24.4 mmol/L mannitol and 5 mmol/L 3-MA for 24 h, respectively, and the changes in cell viability and PCNA expression were examined using CCK8 assay and Western blotting. Bioinformatics analysis of the intersecting gene targets of diabetic kidney disease (DKD) and 3-MA was performed, and the results were verified by Western blotting both in vivo and in vitro. RESULTS In the diabetic mice, treatment with 3-MA produced a short-term hypoglycemic effect, reduced the kidney/body weight ratio and glomerular hypertrophy, and decreased the expressions of α‑SMA and PCNA in the renal cortex. In the in vitro study, 3-MA significantly lowered the viability and reduced PCNA expression in SV40 MES 13 cells exposed to high glucose. The results of bioinformatic analysis identified AKT1 as the key gene in the therapeutic mechanism of 3-MA for DKD. Western blotting confirmed that 3-MA inhibited the phosphorylation of AKT and S6 in both the renal cortex of diabetic mice and high glucose-treated SV40 MES 13 cells. CONCLUSION 3-MA suppresses mesangial cell proliferation and alleviates early diabetic renal injury in mice possibly by inhibiting AKT signaling.
Collapse
|
4
|
Yin XM, Song YY, Jiang WY, Zhang HT, Chen JW, Murao K, Han MX, Sun WP, Zhang GX. Mitochondrial K ATP channel-mediated autophagy contributes to angiotensin II-induced vascular dysfunction in mice. Nutr Metab Cardiovasc Dis 2024; 34:1571-1580. [PMID: 38418351 DOI: 10.1016/j.numecd.2024.01.019] [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: 02/23/2023] [Revised: 10/28/2023] [Accepted: 01/15/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND AND AIM The present study aimed to investigate whether the mitochondrial KATP channel contributes to angiotensin II (Ang II)-induced vascular dysfunction, the development of hypertension, and atherosclerosis. METHODS AND RESULTS ApoE (-/-) mice fed a high-fat diet were chronically infused with Ang II for eight weeks and concomitantly treated with losartan (ARB), apocynin, or 5-hydroxy decanoate (5-HD), or 3-methyladenine (3-MA). Systolic blood pressure was measured, and pathological changes of aortic or liver tissue were observed. Nitric oxide (NO), superoxide dismutase 2 (SOD2) levels and vasorelaxation rate were measured, and protein and mRNA expressions were examined by western blot and RT-PCR. Ang II-induced development of hypertension was suppressed not only by ARB, and apocynin but also by 5-HD or 3-MA. Ang II infusion decreased aortic NO production and relaxation, as well as SOD2 activity in liver, which were improved by all treatments. In addition, Ang II-induced activation of autophagy was suppressed by 5-HD in aortic tissue, furthermore, Ang II increases the atherosclerotic index in plasma and exacerbates the development of atherosclerosis by increases of fat deposition in the aorta and liver. Lipid metabolism-related mRNA expressions (LXR-α, LDLR, SRBI, Acca, and FASN) were changed by Ang II. Similarly, not only ARB, and apocynin, but also 5-HD and 3-MA suppressed Ang II-induced these changes. CONCLUSIONS Our present findings evidence that mitochondrial KATP channel-mediated autophagy contributes to Ang II-induced vascular dysfunction, development of hypertension, and atherosclerosis.
Collapse
Affiliation(s)
- Xue-Min Yin
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Yi-Yi Song
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Wen-Yi Jiang
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Hao-Tian Zhang
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Jing-Wei Chen
- Department of Internal Medicine, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 18 Yang-Su Road, Suzhou 215003, PR China
| | - Koji Murao
- Department of Endocrine and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Meng-Xiao Han
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China.
| | - Wan-Ping Sun
- Laboratory of Molecular Diagnostics, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China.
| | - Guo-Xing Zhang
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China; Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China.
| |
Collapse
|
5
|
Riitano G, Recalchi S, Capozzi A, Manganelli V, Misasi R, Garofalo T, Sorice M, Longo A. The Role of Autophagy as a Trigger of Post-Translational Modifications of Proteins and Extracellular Vesicles in the Pathogenesis of Rheumatoid Arthritis. Int J Mol Sci 2023; 24:12764. [PMID: 37628944 PMCID: PMC10454292 DOI: 10.3390/ijms241612764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease, characterized by persistent joint inflammation, leading to cartilage and bone destruction. Autoantibody production is directed to post-translational modified (PTM) proteins, i.e., citrullinated or carbamylated. Autophagy may be the common feature in several types of stress (smoking, joint injury, and infections) and may be involved in post-translational modifications (PTMs) in proteins and the generation of citrullinated and carbamylated peptides recognized by the immune system in RA patients, with a consequent breakage of tolerance. Interestingly, autophagy actively provides information to neighboring cells via a process called secretory autophagy. Secretory autophagy combines the autophagy machinery with the secretion of cellular content via extracellular vesicles (EVs). A role for exosomes in RA pathogenesis has been recently demonstrated. Exosomes are involved in intercellular communications, and upregulated proteins and RNAs may contribute to the development of inflammatory arthritis and the progression of RA. In RA, most of the exosomes are produced by leukocytes and synoviocytes, which are loaded with PTM proteins, mainly citrullinated proteins, inflammatory molecules, and enzymes that are implicated in RA pathogenesis. Microvesicles derived from cell plasma membrane may also be loaded with PTM proteins, playing a role in the immunopathogenesis of RA. An analysis of changes in EV profiles, including PTM proteins, could be a useful tool for the prevention of inflammation in RA patients and help in the discovery of personalized medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maurizio Sorice
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (G.R.); (S.R.); (A.C.); (V.M.); (R.M.); (T.G.); (A.L.)
| | | |
Collapse
|
6
|
Wear D, Bhagirath E, Balachandar A, Vegh C, Pandey S. Autophagy Inhibition via Hydroxychloroquine or 3-Methyladenine Enhances Chemotherapy-Induced Apoptosis in Neuro-Blastoma and Glioblastoma. Int J Mol Sci 2023; 24:12052. [PMID: 37569432 PMCID: PMC10418453 DOI: 10.3390/ijms241512052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Neuroblastoma is the most common tumour in children under 1 year old, accounting for 12-15% of childhood cancer deaths. Although current treatments are relatively efficacious against this cancer, associated adverse effects could be detrimental to growth and development. In contrast, glioblastoma accounts for 52% of brain tumours and has an extremely poor prognosis. Current chemotherapeutics include temozolomide, which has numerous negative side-effects and a low-effective rate. Previous studies have shown the manipulation of autophagy to be a promising method for targeting cancers, including glioblastoma. We sought to determine the effects of autophagic alterations in combination with current chemotherapies in both neuroblastoma and glioblastoma. Supplementing cisplatin or temozolomide with autophagy activator rapamycin stabilized cancer cell mitochondria, despite having little effect on apoptosis or oxidative stress. Autophagy inhibition via 3-methyladenine or hydroxychloroquine alongside standard chemotherapies enhanced apoptosis and oxidative stress, with 3-methyladenine also disrupting mitochondrial health. Importantly, combining hydroxychloroquine with 0.5 µM cisplatin or 50 µg/mL temozolomide was as or more effective than 2 µM cisplatin or 100 µg/mL temozolomide alone. Analyzing these interesting results, a combined treatment of autophagy inhibitor with a standard chemotherapeutic agent could help to improve patient prognosis and reduce chemotherapy doses and their associated side-effects.
Collapse
Affiliation(s)
- Darcy Wear
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada; (D.W.); (E.B.); (A.B.); (C.V.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5R 0A3, Canada
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Eesha Bhagirath
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada; (D.W.); (E.B.); (A.B.); (C.V.)
- Public Health, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Arpana Balachandar
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada; (D.W.); (E.B.); (A.B.); (C.V.)
- Department of Medicine, University of Toronto, Toronto, ON M5R 0A3, Canada
| | - Caleb Vegh
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada; (D.W.); (E.B.); (A.B.); (C.V.)
| | - Siyaram Pandey
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada; (D.W.); (E.B.); (A.B.); (C.V.)
| |
Collapse
|
7
|
Pizzimenti C, Fiorentino V, Franchina M, Martini M, Giuffrè G, Lentini M, Silvestris N, Di Pietro M, Fadda G, Tuccari G, Ieni A. Autophagic-Related Proteins in Brain Gliomas: Role, Mechanisms, and Targeting Agents. Cancers (Basel) 2023; 15:cancers15092622. [PMID: 37174088 PMCID: PMC10177137 DOI: 10.3390/cancers15092622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
The present review focuses on the phenomenon of autophagy, a catabolic cellular process, which allows for the recycling of damaged organelles, macromolecules, and misfolded proteins. The different steps able to activate autophagy start with the formation of the autophagosome, mainly controlled by the action of several autophagy-related proteins. It is remarkable that autophagy may exert a double role as a tumour promoter and a tumour suppressor. Herein, we analyse the molecular mechanisms as well as the regulatory pathways of autophagy, mainly addressing their involvement in human astrocytic neoplasms. Moreover, the relationships between autophagy, the tumour immune microenvironment, and glioma stem cells are discussed. Finally, an excursus concerning autophagy-targeting agents is included in the present review in order to obtain additional information for the better treatment and management of therapy-resistant patients.
Collapse
Affiliation(s)
- Cristina Pizzimenti
- Translational Molecular Medicine and Surgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy
| | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Mariausilia Franchina
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Maurizio Martini
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Giuseppe Giuffrè
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Maria Lentini
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Nicola Silvestris
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Oncology Section, University of Messina, 98125 Messina, Italy
| | - Martina Di Pietro
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Oncology Section, University of Messina, 98125 Messina, Italy
| | - Guido Fadda
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Giovanni Tuccari
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| |
Collapse
|
8
|
Liu H, He H, Tian Y, Cui J, Wang S, Wang H. Cyclophilin A accelerates SiO 2-induced macrophage foaming. Cell Signal 2023; 103:110562. [PMID: 36535629 DOI: 10.1016/j.cellsig.2022.110562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Silicosis is a common occupational disease characterized by lung inflammation, fibrosis and pulmonary dysfunction caused by long-term inhalation of free SiO2. Cell foaming and the change of CyPA have been observed in SiO2-induced macrophages, but the specific mechanism of CyPA in SiO2-induced foam cells remains poorly understood. The purpose of this study is to explore the mechanism of CyPA in SiO2-induced macrophage foaming and its effect on silicosis. We found that overexpression of CyPA promoted the macrophage foaming and the expression of COL I and α-SMA, while silencing CyPA inhibites the macrophage foaming and the expression of COL I and α-SMA. After blocking the expression of CD36 on the basis of overexpression CyPA, we found it inhibites the macrophage foaming. In conclusion, CyPA can affect the foaming of macrophages and may participate in silicosis fibrosis.
Collapse
Affiliation(s)
- Heliang Liu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China; Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Hailan He
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Ying Tian
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Jie Cui
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Shuang Wang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Hongli Wang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China.
| |
Collapse
|
9
|
Xu Z, Li X, Ding Z, Zhang Y, Peng Z, Yang X, Cao W, Du R. LRPPRC inhibits autophagy and promotes foam cell formation in atherosclerosis. FEBS J 2022; 289:7545-7560. [PMID: 35792704 DOI: 10.1111/febs.16567] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 05/13/2022] [Accepted: 06/27/2022] [Indexed: 01/14/2023]
Abstract
Lipid-laden macrophages are considered as the main source of foam cells in atherosclerosis; however, the mechanism for macrophage foam cell formation remains unknown. Here, we explore the mechanism behind foam cell formation to potentially identify a novel treatment for atherosclerosis. Our data demonstrated that leucine-rich pentatricopeptide repeat-containing protein (LRPPRC) increased in the atherosclerotic plaques of LDLR-/- mice fed with a Western diet. LRPPRC was also upregulated in mice peritoneal macrophages and RAW 264.7 cells treated with oxidative low density lipoprotein, whereas knockdown of LRPPRC by transfecting with small interfering (Si)-LRPPRC in RAW 264.7 cells decreased foam cell formation. Furthermore, Si-LRPPRC promoted autophagy and increased the expression of cholesterol efflux protein ATP-binding cassette transporter A1 in RAW 264.7 cells. Moreover, intervention with MHY1485 in RAW 264.7 cells revealed that autophagy was inhibited by LRPPRC via the Akt-mechanistic target of rapamycin pathway. Taken together, we confirm for the first time that LRPPRC is increased within the atherosclerotic plaques of mice and enhances the process of foam cell formation. The knockdown of LRPPRC inhibited foam cell formation by activating macrophage autophagy. Our findings indicate that the regulation of macrophage LRPPRC expression may be a novel strategy for ameliorating atherosclerosis.
Collapse
Affiliation(s)
- Zhou Xu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xinran Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Zhiquan Ding
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yuyang Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Zhiwei Peng
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xin Yang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Wangsen Cao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Ronghui Du
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China.,State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
10
|
Wu W, Hendrix A, Nair S, Cui T. Nrf2-Mediated Dichotomy in the Vascular System: Mechanistic and Therapeutic Perspective. Cells 2022; 11:cells11193042. [PMID: 36231004 PMCID: PMC9563590 DOI: 10.3390/cells11193042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor-erythroid 2-related factor 2 (Nrf2), a transcription factor, controls the expression of more than 1000 genes that can be clustered into different categories with distinct functions ranging from redox balance and metabolism to protein quality control in the cell. The biological consequence of Nrf2 activation can be either protective or detrimental in a context-dependent manner. In the cardiovascular system, most studies have focused on the protective properties of Nrf2, mainly as a key transcription factor of antioxidant defense. However, emerging evidence revealed an unexpected role of Nrf2 in mediating cardiovascular maladaptive remodeling and dysfunction in certain disease settings. Herein we review the role of Nrf2 in cardiovascular diseases with a focus on vascular disease. We discuss the negative effect of Nrf2 on the vasculature as well as the potential underlying mechanisms. We also discuss the clinical relevance of targeting Nrf2 pathways for the treatment of cardiovascular and other diseases.
Collapse
Affiliation(s)
- Weiwei Wu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Andrew Hendrix
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Sharad Nair
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
- Columbia VA Health System, Wm. Jennings Bryan Dorn VA Medical Center, Columbia, SC 29209, USA
| | - Taixing Cui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209, USA
- Columbia VA Health System, Wm. Jennings Bryan Dorn VA Medical Center, Columbia, SC 29209, USA
- Correspondence: ; Tel.: +1-803-216-3804
| |
Collapse
|
11
|
Sears SM, Feng JL, Orwick A, Vega AA, Krueger AM, Shah PP, Doll MA, Beverly LJ, Siskind LJ. Pharmacological inhibitors of autophagy have opposite effects in acute and chronic cisplatin-induced kidney injury. Am J Physiol Renal Physiol 2022; 323:F288-F298. [PMID: 35796459 PMCID: PMC9394729 DOI: 10.1152/ajprenal.00097.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 12/25/2022] Open
Abstract
The nephrotoxicity of cisplatin remains a major hurdle in the field of oncology. Thirty percent of patients treated with cisplatin develop acute kidney injury, and all patients are at risk for long-term impacts on kidney function. There are currently no Federal Drug Administration-approved agents to prevent or treat cisplatin-induced kidney injury. The dosing regimen used in preclinical models of nephrotoxicity may impact the success of therapeutic candidates in clinical trials. Here, we demonstrated that pharmacological inhibitors of autophagy have opposite effects when used as interventions in two different models of cisplatin-induced kidney injury. Eight-week-old male C57BL/6 mice were treated with either one dose of 20 mg/kg cisplatin or weekly doses of 9 mg/kg cisplatin for 4 wk or until body weight loss exceeded 30%. Concurrently, mice were administered multiple doses of 60 mg/kg chloroquine or 15 mg/kg 3-methyladenine attempting to globally inhibit autophagy. Mice that received a single high dose of cisplatin had worsened kidney function, inflammation, and cell death with the addition of chloroquine. 3-Methlyadenine did not impact the development of acute kidney injury in this model. In contrast, mice that received repeated low doses of cisplatin showed improved kidney function, reduced inflammation, and reduced fibrosis when treated with either chloroquine or 3-methyladenine. This study highlights how therapeutic candidates can have drastically different effects on the development of cisplatin-induced kidney injury depending on the dosing model used. This emphasizes the importance of choosing the appropriate model of injury for preclinical studies.NEW & NOTEWORTHY This study examined how inhibition of autophagy has opposite effects on the development of acute and chronic kidney injury. Autophagy inhibition exacerbated the development of acute kidney injury following a single high dose of cisplatin but prevented the development of injury and fibrosis following repeated low doses of cisplatin.
Collapse
Affiliation(s)
- Sophia M Sears
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Joanna L Feng
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Andrew Orwick
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Alexis A Vega
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky
| | - Austin M Krueger
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Parag P Shah
- Department of Medicine, University of Louisville, Louisville, Kentucky
- Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Mark A Doll
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Levi J Beverly
- Department of Medicine, University of Louisville, Louisville, Kentucky
- Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
- Brown Cancer Center, University of Louisville, Louisville, Kentucky
| |
Collapse
|
12
|
Kwon Y, Haam CE, Byeon S, Choi SK, Lee YH. Effects of 3-methyladenine, an autophagy inhibitor, on the elevated blood pressure and arterial dysfunction of angiotensin II-induced hypertensive mice. Biomed Pharmacother 2022; 154:113588. [PMID: 35994821 DOI: 10.1016/j.biopha.2022.113588] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/19/2022] Open
Abstract
Autophagy is an intracellular degradation system that disassembles cytoplasmic components through autophagosomes fused with lysosomes. Recently, it has been reported that autophagy is associated with cardiovascular diseases, including pulmonary hypertension, atherosclerosis, and myocardial ischemia. However, the involvement of autophagy in hypertension is not well understood. In the present study, we hypothesized that excessive autophagy contributes to the dysfunction of mesenteric arteries in angiotensin II (Ang II)-induced hypertensive mice. Treatment of an autophagy inhibitor, 3-methyladenine (3-MA), reduced the elevated blood pressure and wall thickness, and improved endothelium-dependent relaxation in mesenteric arteries of Ang II-treated mice. The expression levels of autophagy markers, beclin1 and LC3 II, were significantly increased by Ang II infusion, which was reduced by treatment of 3-MA. Furthermore, treatment of 3-MA induced vasodilation in the mesenteric resistance arteries pre-contracted with U46619 or phenylephrine, which was dependent on endothelium. Interestingly, nitric oxide production and phosphorylated endothelial nitric oxide synthase (p-eNOS) at S1177 in the mesenteric arteries of Ang II-treated mice were increased by treatment with 3-MA. In HUVECs, p-eNOS was reduced by Ang II, which was increased by treatment of 3-MA. 3-MA had direct vasodilatory effect on the pre-contracted mesenteric arteries. In cultured vascular smooth muscle cells (VSMCs), Ang II induced increase in beclin1 and LC3 II and decrease in p62, which was reversed by treatment of 3-MA. These results suggest that autophagy inhibition exerts beneficial effects on the dysfunction of mesenteric arteries in hypertension.
Collapse
Affiliation(s)
- Youngin Kwon
- Department of Physiology, College of Medicine, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University, 50 Yonseiro, Seodaemun-gu, Seoul 03722, the Republic of the Korea
| | - Chae Eun Haam
- Department of Physiology, College of Medicine, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University, 50 Yonseiro, Seodaemun-gu, Seoul 03722, the Republic of the Korea
| | - Seonhee Byeon
- Department of Physiology, College of Medicine, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University, 50 Yonseiro, Seodaemun-gu, Seoul 03722, the Republic of the Korea
| | - Soo-Kyoung Choi
- Department of Physiology, College of Medicine, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University, 50 Yonseiro, Seodaemun-gu, Seoul 03722, the Republic of the Korea.
| | - Young-Ho Lee
- Department of Physiology, College of Medicine, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University, 50 Yonseiro, Seodaemun-gu, Seoul 03722, the Republic of the Korea.
| |
Collapse
|
13
|
López A, Fuentes E, Yusà V, Ibáñez M, Coscollà C. Identification of Unknown Substances in Ambient Air (PM10), Profiles and Differences between Rural, Urban and Industrial Areas. TOXICS 2022; 10:toxics10050220. [PMID: 35622634 PMCID: PMC9145881 DOI: 10.3390/toxics10050220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 12/10/2022]
Abstract
A fast and automated strategy has been developed for identifying unknown substances in the atmosphere (concretely, in the particulate matter, PM10) using LC-HRMS (MS3). A total of 15 samples were collected in three different areas (rural, urban and industrial). A sampling flow rate of 30 m3 h−1 was applied for 24 h, sampling a total volume of around 720 m3. A total of 49 compounds were tentatively identified using very restrictive criteria regarding exact mass, retention time, isotopic profile and both MS2 and MS3 spectra. Pesticides, pharmaceutical active compounds, drugs, plasticizers and metabolites were the most identified compounds. To verify whether the developed methodology was suitable, 11 substances were checked with their analytical standards and all of them were confirmed. Different profiles for industrial, rural and urban areas were examined. The Principal Component Analysis (PCA) model allowed us to separate the obtained data of the three assessed area. When the profiles obtained in the three evaluated areas were compared using a Volcano plot (the rural area was taken as reference), 11 compounds were confirmed as being discriminant: three of them (3-hydroxy-2-methylpyridine, 3-methyladenine and nicotine) were more likely to be found in industrial sites; ten compounds (3-hydroxy-2-methylpyridine, 3-methyladenine, azoxystrobin, cocaine, cotinine, ethoprophos, imidacloprid, metalaxyl-M, nicotine and pyrimethanil) were more probable in the case of urban sites; finally, triisopropanolamine was more likely to be detected in rural locations.
Collapse
Affiliation(s)
- Antonio López
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region, FISABIO-Public Health, 21, Avenida Catalunya, 46020 Valencia, Spain; (A.L.); (E.F.); (V.Y.)
| | - Esther Fuentes
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region, FISABIO-Public Health, 21, Avenida Catalunya, 46020 Valencia, Spain; (A.L.); (E.F.); (V.Y.)
- Environmental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water, University Jaume I, S/N, Avenida Sos Baynat, 12071 Castelló de la Plana, Spain;
| | - Vicent Yusà
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region, FISABIO-Public Health, 21, Avenida Catalunya, 46020 Valencia, Spain; (A.L.); (E.F.); (V.Y.)
- Public Health Laboratory of Valencia, 21, Avenida Catalunya, 46020 Valencia, Spain
- Analytical Chemistry Department, University of Valencia, Edifici Jeroni Muñoz, Dr. Moliner 50, 46100 Burjassot, Spain
| | - María Ibáñez
- Environmental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water, University Jaume I, S/N, Avenida Sos Baynat, 12071 Castelló de la Plana, Spain;
| | - Clara Coscollà
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region, FISABIO-Public Health, 21, Avenida Catalunya, 46020 Valencia, Spain; (A.L.); (E.F.); (V.Y.)
- Correspondence: ; Tel.: +34-96-192-6333
| |
Collapse
|
14
|
Dasgupta A, Arneson-Wissink PC, Schmitt RE, Cho DS, Ducharme AM, Hogenson TL, Krueger EW, Bamlet WR, Zhang L, Razidlo GL, Fernandez-Zapico ME, Doles JD. Anticachectic regulator analysis reveals Perp-dependent antitumorigenic properties of 3-methyladenine in pancreatic cancer. JCI Insight 2022; 7:153842. [PMID: 34874916 PMCID: PMC8855816 DOI: 10.1172/jci.insight.153842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/01/2021] [Indexed: 12/14/2022] Open
Abstract
Approximately 80% of pancreatic cancer patients suffer from cachexia, and one-third die due to cachexia-related complications such as respiratory failure and cardiac arrest. Although there has been considerable research into cachexia mechanisms and interventions, there are, to date, no FDA-approved therapies. A major contributing factor for the lack of therapy options could be the failure of animal models to accurately recapitulate the human condition. In this study, we generated an aged model of pancreatic cancer cachexia to compare cachexia progression in young versus aged tumor-bearing mice. Comparative skeletal muscle transcriptome analyses identified 3-methyladenine (3-MA) as a candidate antiwasting compound. In vitro analyses confirmed antiwasting capacity, while in vivo analysis revealed potent antitumor effects. Transcriptome analyses of 3-MA-treated tumor cells implicated Perp as a 3-MA target gene. We subsequently (a) observed significantly higher expression of Perp in cancer cell lines compared with control cells, (b) noted a survival disadvantage associated with elevated Perp, and (c) found that 3-MA-associated Perp reduction inhibited tumor cell growth. Finally, we have provided in vivo evidence that survival benefits conferred by 3-MA administration are independent of its effect on tumor progression. Taken together, we report a mechanism linking 3-MA to Perp inhibition, and we further implicate Perp as a tumor-promoting factor in pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Tara L. Hogenson
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology
| | - Eugene W. Krueger
- Department of Biochemistry and Molecular Biology,,Division of Gastroenterology and Hepatology
| | | | - Lizhi Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gina L. Razidlo
- Department of Biochemistry and Molecular Biology,,Division of Gastroenterology and Hepatology
| | | | | |
Collapse
|
15
|
Mueller AL, Payandeh Z, Mohammadkhani N, Mubarak SMH, Zakeri A, Alagheband Bahrami A, Brockmueller A, Shakibaei M. Recent Advances in Understanding the Pathogenesis of Rheumatoid Arthritis: New Treatment Strategies. Cells 2021; 10:cells10113017. [PMID: 34831240 PMCID: PMC8616543 DOI: 10.3390/cells10113017] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/03/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is considered a chronic systemic, multi-factorial, inflammatory, and progressive autoimmune disease affecting many people worldwide. While patients show very individual courses of disease, with RA focusing on the musculoskeletal system, joints are often severely affected, leading to local inflammation, cartilage destruction, and bone erosion. To prevent joint damage and physical disability as one of many symptoms of RA, early diagnosis is critical. Auto-antibodies play a pivotal clinical role in patients with systemic RA. As biomarkers, they could help to make a more efficient diagnosis, prognosis, and treatment decision. Besides auto-antibodies, several other factors are involved in the progression of RA, such as epigenetic alterations, post-translational modifications, glycosylation, autophagy, and T-cells. Understanding the interplay between these factors would contribute to a deeper insight into the causes, mechanisms, progression, and treatment of the disease. In this review, the latest RA research findings are discussed to better understand the pathogenesis, and finally, treatment strategies for RA therapy are presented, including both conventional approaches and new methods that have been developed in recent years or are currently under investigation.
Collapse
Affiliation(s)
- Anna-Lena Mueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; (A.-L.M.); (A.B.)
| | - Zahra Payandeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166-15731, Iran;
| | - Niloufar Mohammadkhani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran;
- Children’s Medical Center, Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Shaden M. H. Mubarak
- Department of Clinical Laboratory Science, Faculty of Pharmacy, University of Kufa, Najaf 1967365271, Iraq;
| | - Alireza Zakeri
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran 1678815811, Iran;
| | - Armina Alagheband Bahrami
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran;
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; (A.-L.M.); (A.B.)
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany; (A.-L.M.); (A.B.)
- Correspondence: ; Tel.: +49-89-2180-72624
| |
Collapse
|
16
|
Dai S, Liu F, Ren M, Qin Z, Rout N, Yang XF, Wang H, Tomlinson S, Qin X. Complement Inhibition Targeted to Injury Specific Neoepitopes Attenuates Atherogenesis in Mice. Front Cardiovasc Med 2021; 8:731315. [PMID: 34651027 PMCID: PMC8505745 DOI: 10.3389/fcvm.2021.731315] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 08/30/2021] [Indexed: 12/22/2022] Open
Abstract
Rationale: Previous studies have indicated an important role for complement in atherosclerosis, a lipid-driven chronic inflammatory disease associated to oxidative stress in the vessel wall. However, it remains unclear how complement is activated in the process of atherogenesis. An accepted general model for complement activation in the context of ischemia reperfusion injury is that ischemia induces the exposure of neoepitopes that are recognized by natural self-reactive IgM antibodies, and that in turn activate complement. Objective: We investigated whether a similar phenomenon may be involved in the pathogenesis of atherosclerosis, and whether interfering with this activation event, together with inhibition of subsequent amplification of the cascade at the C3 activation step, can provide protection against atherogenesis. Methods and Results: We utilized C2scFv-Crry, a novel construct consisting of a single chain antibody (scFv) linked to Crry, a complement inhibitor that functions at C3 activation. The scFv moiety was derived from C2 IgM mAb that specifically recognizes phospholipid neoepitopes known to be expressed after ischemia. C2scFv-Crry targeted to the atherosclerotic plaque of Apoe -/- mice, demonstrating expression of the C2 neoepitope. C2scFv-Crry administered twice per week significantly attenuated atherosclerotic plaque in the aorta and aortic root of Apoe -/- mice fed with a high-fat diet (HFD) for either 2 or 4 months, and treatment reduced C3 deposition and membrane attack complex formation as compared to vehicle treated mice. C2scFv-Crry also inhibited the uptake of oxidized low-density-lipoprotein (oxLDL) by peritoneal macrophages, which has been shown to play a role in pathogenesis, and C2scFv-Crry-treated mice had decreased lipid content in the lesion with reduced oxLDL levels in serum compared to vehicle-treated mice. Furthermore, C2scFv-Crry reduced the deposition of endogenous total IgM in the plaque, although it did not alter serum IgM levels, further indicating a role for natural IgM in initiating complement activation. Conclusion: Neoepitope targeted complement inhibitors represent a novel therapeutic approach for atherosclerosis.
Collapse
Affiliation(s)
- Shen Dai
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Neuroscience, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Fengming Liu
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Neuroscience, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Mi Ren
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Zhongnan Qin
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Namita Rout
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Xiao-Feng Yang
- Center for Metabolic Disease Research and Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Hong Wang
- Center for Metabolic Disease Research and Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Xuebin Qin
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Neuroscience, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
17
|
Synthesis and Characterization of Sr-Doped ZnSe Nanoparticles for Catalytic and Biological Activities. WATER 2021. [DOI: 10.3390/w13162189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of cost-effective and ecofriendly approaches toward water purification and antibacterial activity is a hot research topic in this era. Purposely, strontium-doped zinc selenide (Sr-doped ZnSe) nanoparticles, with different molar ratios of Sr2+ cations (0.01, 0.05, and 0.1), were prepared via the co-precipitation method, in which sodium borohydride (NaBH4) and 2-mercaptoethanol were employed as reducing and stabilizing agents, respectively. The ZnSe cubic structure expanded by Sr2+ cations was indicated by X-ray diffraction (XRD) analysis. The absorption of the chemical compounds on the surface was observed via Fourier transform infrared (FT-IR) spectroscopy. The optical orientation was measured by ultraviolet–visible diffused reflectance spectroscopy (UV-DRS) analysis. The surface area, morphology, and elemental purity were analyzed using field emission scanning electron microscopy (FE-SEM), high-resolution transmission electron microscopy (HR-TEM), and energy-dispersive spectroscopy (EDS) analyses. The oxidation state and valency of the synthesized nanoparticles were analyzed using X-ray photoelectron spectroscopy (XPS). Sr-doped ZnSe nanoparticles were investigated for photocatalytic degradation of methyl orange (MO), and their antibacterial potential was investigated against different bacterial strains. The antibacterial activity examined against Staphylococcus aureus and Escherichia coli implied the excellent biological activity of the nanoparticles. Moreover, the Sr-doped ZnSe nanoparticles were evaluated by the successful degradation of methyl orange under visible light irradiation. Therefore, Sr-doped ZnSe nanoparticles have tremendous potential in biological and water remediation fields.
Collapse
|
18
|
Florance I, Ramasubbu S, Mukherjee A, Chandrasekaran N. Polystyrene nanoplastics dysregulate lipid metabolism in murine macrophages in vitro. Toxicology 2021; 458:152850. [PMID: 34217793 DOI: 10.1016/j.tox.2021.152850] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/20/2021] [Accepted: 06/30/2021] [Indexed: 10/21/2022]
Abstract
Micro and nanoplastics are one of the major emerging environmental contaminants. Their impact on human health is less explored. There are several in vitro studies on their cellular uptake and accumulation, where micro and nanoplastics were mostly reported to be non-cytotoxic. The effects caused by the direct contact of nanoplastics with the immune system, especially at the cellular level is less known. Here we report that RAW 264.7 macrophages undergo differentiation into lipid laden foam cells when exposed to polystyrene nanoplastics (50 μg/mL). We found that exposure of RAW 264.7 macrophages to sulfate-modified polystyrene nanoplastics results in the accumulation of lipid droplets in the cytoplasm leading to foam cell formation. Exposure to high concentration of polystyrene nanoplastics (100 and 200 μg/mL) results in increased reactive oxygen species and impair lysosomes in macrophages. The exposure of BV2 microglial cells to polystyrene nanoplastics (50 μg/mL) induces lipid accumulation. In addition, our results indicate the role of polystyrene nanoplastics in altering the lipid metabolism in murine macrophages in vitro. In the present study we reported that polystyrene nanoplastics stabilized with anionic surfactants can be potent stimuli for lipotoxicity and foam cell formation leading to the pathogenesis of atherosclerosis posing major threat for animal and human health.
Collapse
Affiliation(s)
- Ida Florance
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India
| | - Seenivasan Ramasubbu
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India
| | - Amitava Mukherjee
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India
| | - Natarajan Chandrasekaran
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India.
| |
Collapse
|
19
|
Batara DCR, Choi MC, Shin HU, Kim H, Kim SH. Friend or Foe: Paradoxical Roles of Autophagy in Gliomagenesis. Cells 2021; 10:1411. [PMID: 34204169 PMCID: PMC8227518 DOI: 10.3390/cells10061411] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/30/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive type of primary brain tumor in adults, with a poor median survival of approximately 15 months after diagnosis. Despite several decades of intensive research on its cancer biology, treatment for GBM remains a challenge. Autophagy, a fundamental homeostatic mechanism, is responsible for degrading and recycling damaged or defective cellular components. It plays a paradoxical role in GBM by either promoting or suppressing tumor growth depending on the cellular context. A thorough understanding of autophagy's pleiotropic roles is needed to develop potential therapeutic strategies for GBM. In this paper, we discussed molecular mechanisms and biphasic functions of autophagy in gliomagenesis. We also provided a summary of treatments for GBM, emphasizing the importance of autophagy as a promising molecular target for treating GBM.
Collapse
Affiliation(s)
- Don Carlo Ramos Batara
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (D.C.R.B.); (H.-U.S.)
| | - Moon-Chang Choi
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea;
| | - Hyeon-Uk Shin
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (D.C.R.B.); (H.-U.S.)
| | - Hyunggee Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea;
| | - Sung-Hak Kim
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (D.C.R.B.); (H.-U.S.)
| |
Collapse
|
20
|
Tadokoro K, Yamashita T, Shang J, Ohta Y, Nomura E, Morihara R, Omote Y, Takemoto M, Abe K. Switching the Proteolytic System from the Ubiquitin-Proteasome System to Autophagy in the Spinal Cord of an Amyotrophic Lateral Sclerosis Mouse Model. Neuroscience 2021; 466:47-57. [PMID: 33974963 DOI: 10.1016/j.neuroscience.2021.04.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/22/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022]
Abstract
The degradation of damaged proteins takes place via two major proteolytic pathways: the ubiquitin-proteasome system (UPS) and autophagy. However, since it is unclear how these two proteolytic pathways contribute to the pathogenesis of amyotrophic lateral sclerosis (ALS), we investigated the switching mechanism from UPS to autophagy by pharmacologically modifying these pathways by treating the spinal cords of female ALS mouse model bearing G93A human SOD1 (G93A mice) with MG132 or 3-methyladenine (3MA). G93A mice exhibited a progressive increase in the amount of ubiquitin and p62 aggregates, BAG3 expression, and LC3-II/LC3-I ratio in both astroglia and motor neurons. Treatment with MG132 or 3MA significantly increased the clinical hanging wire score and exacerbated α-motor neuron loss at 18 weeks in G93A mice, and increased the amount of ubiquitin, p62 aggregates, and BAG3 expression. This study's results demonstrate that the molecular switch from UPS to autophagy occurred not only in motor neurons but also in astroglia at the end stage (18 weeks) when the autophagic flux was impaired in G93A mice. This finding suggests that the defense system was disrupted against aggregate-prone protein production in ALS.
Collapse
Affiliation(s)
- Koh Tadokoro
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-Ku, Okayama 700-8558, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-Ku, Okayama 700-8558, Japan
| | - Jingwei Shang
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-Ku, Okayama 700-8558, Japan
| | - Yasuyuki Ohta
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-Ku, Okayama 700-8558, Japan
| | - Emi Nomura
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-Ku, Okayama 700-8558, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-Ku, Okayama 700-8558, Japan
| | - Yoshio Omote
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-Ku, Okayama 700-8558, Japan
| | - Mami Takemoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-Ku, Okayama 700-8558, Japan
| | - Koji Abe
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-Ku, Okayama 700-8558, Japan.
| |
Collapse
|
21
|
Bastonini E, Kovacs D, Raffa S, Delle Macchie M, Pacifico A, Iacovelli P, Torrisi MR, Picardo M. A protective role for autophagy in vitiligo. Cell Death Dis 2021; 12:318. [PMID: 33767135 PMCID: PMC7994839 DOI: 10.1038/s41419-021-03592-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022]
Abstract
A growing number of studies supports the existence of a dynamic interplay between energetic metabolism and autophagy, whose induction represents an adaptive response against several stress conditions. Autophagy is an evolutionarily conserved and a highly orchestrated catabolic recycling process that guarantees cellular homeostasis. To date, the exact role of autophagy in vitiligo pathogenesis is still not clear. Here, we provide the first evidence that autophagy occurs in melanocytes and fibroblasts from non-lesional skin of vitiligo patients, as a result of metabolic surveillance response. More precisely, this study is the first to reveal that induction of autophagy exerts a protective role against the intrinsic metabolic stress and attempts to antagonize degenerative processes in normal appearing vitiligo skin, where melanocytes and fibroblasts are already prone to premature senescence.
Collapse
Affiliation(s)
- Emanuela Bastonini
- Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Rome, Italy.
| | - Daniela Kovacs
- Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Salvatore Raffa
- Ultrastructural Pathology Lab., Medical Genetics and Advanced Cellular Diagnostics Unit, Sant'Andrea University Hospital & Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marina Delle Macchie
- Ultrastructural Pathology Lab., Medical Genetics and Advanced Cellular Diagnostics Unit, Sant'Andrea University Hospital & Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessia Pacifico
- Clinical Dermatology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Paolo Iacovelli
- Clinical Dermatology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Maria Rosaria Torrisi
- Ultrastructural Pathology Lab., Medical Genetics and Advanced Cellular Diagnostics Unit, Sant'Andrea University Hospital & Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mauro Picardo
- Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Rome, Italy.
| |
Collapse
|
22
|
Fang S, Wan X, Zou X, Sun S, Hao X, Liang C, Zhang Z, Zhang F, Sun B, Li H, Yu B. Arsenic trioxide induces macrophage autophagy and atheroprotection by regulating ROS-dependent TFEB nuclear translocation and AKT/mTOR pathway. Cell Death Dis 2021; 12:88. [PMID: 33462182 PMCID: PMC7814005 DOI: 10.1038/s41419-020-03357-1] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 01/30/2023]
Abstract
Inducing autophagy and inhibiting apoptosis may provide a therapeutic treatment for atherosclerosis (AS). For the treatment of progressive AS, arsenic trioxide (ATO) has been used to coat vascular stents. However, the effect of ATO on autophagy of macrophages is still unknown. Therefore, the aims of this study were to characterize the effects and the mechanism of actions of ATO on autophagy in macrophages. Our results showed that ATO-induced activation of autophagy was an earlier event than ATO-induced inhibition of the expression of apoptosis markers in macrophages and foam cells. Nuclear transcription factor EB (TFEB) prevents atherosclerosis by activating macrophage autophagy and promoting lysosomal biogenesis. Here, we report that ATO triggered the nuclear translocation of TFEB, which in turn promoted autophagy and autophagosome-lysosome fusion. Both the latter events were prevented by TFEB knockdown. Moreover, ATO decreased the p-AKT and p-mTOR in the PI3K/AKT/mTOR signaling pathway, thus inducing autophagy. Correspondingly, treatment with the autophagy inhibitor 3-methyladenine (3-MA) abolished the autophagy-inducing effects of ATO. Meanwhile, PI3K inhibitor (LY294002) and mTOR inhibitor (rapamycin) cooperated with ATO to induce autophagy. Furthermore, reactive oxygen species (ROS) were generated in macrophages after treatment with ATO. The ROS scavenger N-acetyl-1-cysteine (NAC) abolished ATO-induced nuclear translocation of TFEB, as well as changes in key molecules of the AKT/mTOR signaling pathway and downstream autophagy. More importantly, ATO promoted autophagy in the aorta of ApoE-/- mice and reduced atherosclerotic lesions in early AS, which were reversed by 3-MA treatment. In summary, our data indicated that ATO promoted ROS induction, which resulted in nuclear translocation of TFEB and inhibition of the PI3K/AKT/mTOR pathway. These actions ultimately promoted macrophage autophagy and reduced atherosclerotic lesions at early stages. These findings may provide a new perspective for the clinical treatment of early-stage atherosclerosis and should be further studied.
Collapse
Affiliation(s)
- Shaohong Fang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Xin Wan
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoyi Zou
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Song Sun
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinran Hao
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Chenchen Liang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Zhenming Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Fangni Zhang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Bo Sun
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hulun Li
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.
| |
Collapse
|
23
|
Yuan W, He X, Morin D, Barrière G, Liu X, Li J, Zhu Y. Autophagy Induction Contributes to the Neuroprotective Impact of Intermittent Fasting on the Acutely Injured Spinal Cord. J Neurotrauma 2020; 38:373-384. [PMID: 33076741 DOI: 10.1089/neu.2020.7166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) is one of the leading causes of neurological disability and death. So far, there is no satisfactory treatment for SCI, because of its complex and ill-defined pathophysiology. Recently, autophagy has been implicated as protective in acute SCI rat models. Here, we investigated the therapeutic value of a dietary intervention, namely, intermittent fasting (IF), on neuronal survival after acute SCI in rats, and its underlying mechanism related to autophagy regulation. We found remarkable improvement in both behavioral performance and neuronal survival at the injured segment of the spinal cord of animals previously subjected to IF. Western blotting revealed a marked decrease in apoptosis-related markers such as cleaved caspase 3 levels and the bax/bcl-2 ratio in the IF group, which suggested an inhibition of the intrinsic apoptosis pathway. In addition, the expression of the autophagy markers LC3-II and beclin 1 was also increased in the IF group compared with ad libitum fed animals. In parallel, IF decreased the levels of the substrate protein of autophagy, p62, indicative of an upregulation of the autophagic processes. Treatment with 3-methyladenine (3-MA), a selective inhibitor of autophagy, reversed the downregulated apoptosis-related markers by IF. Finally, IF could activate the adenosine monophosphate (AMP)-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway and enhance lysosome function by upregulating transcription factor (TF)EB expression. Altogether, the present findings suggest that IF exerts a neuroprotective effect after acute SCI via the upregulation of autophagy, and further points to dietary interventions as a promising combinatorial treatment for SCI.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Université de Bordeaux, Bordeaux, France
| | - Xin He
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Didier Morin
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Université de Bordeaux, Bordeaux, France
| | - Grégory Barrière
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Université de Bordeaux, Bordeaux, France
| | - Xuan Liu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
- Department of Orthopedics, Affiliated Hospital of Chengdu University, Chengdu, China
| | - Jiatong Li
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| | - Yue Zhu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
24
|
Webb LG, Veloz J, Pintado-Silva J, Zhu T, Rangel MV, Mutetwa T, Zhang L, Bernal-Rubio D, Figueroa D, Carrau L, Fenutria R, Potla U, Reid SP, Yount JS, Stapleford KA, Aguirre S, Fernandez-Sesma A. Chikungunya virus antagonizes cGAS-STING mediated type-I interferon responses by degrading cGAS. PLoS Pathog 2020; 16:e1008999. [PMID: 33057424 PMCID: PMC7591055 DOI: 10.1371/journal.ppat.1008999] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 10/27/2020] [Accepted: 09/21/2020] [Indexed: 12/24/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus known to cause epidemics resulting in predominantly symptomatic infections, which in rare cases cause long term debilitating arthritis and arthralgia. Significant progress has been made in understanding the roles of canonical RNA sensing pathways in the host recognition of CHIKV; however, less is known regarding antagonism of CHIKV by cytosolic DNA sensing pathways like that of cyclic GMP-AMP synthase (cGAS) and Stimulator of Interferon Genes (STING). With the use of cGAS or STING null cells we demonstrate that the pathway restricts CHIKV replication in fibroblasts and immune cells. We show that DNA accumulates in the cytoplasm of infected cells and that CHIKV blocks DNA dependent IFN-β transcription. This antagonism of DNA sensing is via an early autophagy-mediated degradation of cGAS and expression of the CHIKV capsid protein is sufficient to induce cGAS degradation. Furthermore, we identify an interaction of CHIKV nsP1 with STING and map the interaction to 23 residues in the cytosolic loop of the adaptor protein. This interaction stabilizes the viral protein and increases the level of palmitoylated nsP1 in cells. Together, this work supports previous publications highlighting the relevance of the cGAS-STING pathway in the early detection of (+)ssRNA viruses and provides direct evidence that CHIKV interacts with and antagonizes cGAS-STING signaling.
Collapse
Affiliation(s)
- L. G. Webb
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - J. Veloz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - J. Pintado-Silva
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - T. Zhu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - M. V. Rangel
- Department of Microbiology, New York University School of Medicine, New York, NY, United States of America
| | - T. Mutetwa
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - L. Zhang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States of America
| | - D. Bernal-Rubio
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - D. Figueroa
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - L. Carrau
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - R. Fenutria
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - U. Potla
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - St. P. Reid
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - J. S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States of America
| | - K. A. Stapleford
- Department of Microbiology, New York University School of Medicine, New York, NY, United States of America
| | - S. Aguirre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - A. Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- The Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| |
Collapse
|
25
|
Pyrroloquinoline Quinone Inhibits Rotenone-Induced Microglia Inflammation by Enhancing Autophagy. Molecules 2020; 25:molecules25194359. [PMID: 32977419 PMCID: PMC7582530 DOI: 10.3390/molecules25194359] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation is a feature common to neurodegenerative diseases, such as Parkinson’s disease (PD), which might be responsive to therapeutic intervention. Rotenone has been widely used to establish PD models by inducing mitochondrial dysfunction and inflammation. Our previous studies have reported that pyrroloquinoline quinone (PQQ), a naturally occurring redox cofactor, could prevent mitochondrial dysfunction in rotenone induced PD models by regulating mitochondrial functions. In the present study, we aimed to investigate the effect of PQQ on neuroinflammation and the mechanism involved. BV2 microglia cells were pre-treated with PQQ followed by rotenone incubation. The data showed that PQQ did not affect the cell viability of BV2 cells treated with rotenone, while the conditioned medium (CM) of BV2 cells pre-treated with PQQ significantly increased cell viability of SH-SY5Y cells. In rotenone-treated BV2 cells, PQQ dose-dependently decreased lactate dehydrogenase (LDH) release and suppressed the up-regulation of pro-inflammation factors, such as interleukin-1β (IL-1β), IL-6 and tumor necrosis factor-α (TNF-α) in the cultured media, as well as nitric oxide (NO) release induced by rotenone. PQQ pretreatment also increased the ratio of LC3-II/LC3-I and expression of Atg5 in BV2 cells stimulated with rotenone. Additionally, the autophagosome observed by transmission electron microscopy (TEM) and co-localization of mitochondria with lysosomes indicated that mitophagy was induced by PQQ in rotenone-injured BV2 cells, and the PINK1/parkin mediated mitophagy pathway was regulated by PQQ. Further, autophagy inhibitor, 3-methyladenine (3-MA), partially abolished the neuroprotective effect of PQQ and attenuated the inhibition of inflammation with PQQ pretreatment. Taken together, our data extend our understanding of the neuroprotective effect of PQQ against rotenone-induced injury and provide evidence that autophagy enhancement might be a novel therapeutic strategy for PD treatment.
Collapse
|
26
|
Abstract
Purpose of Review To summarize recent insights into long non-coding RNAs (lncRNAs) involved in atherosclerosis. Because atherosclerosis is the main underlying pathology of cardiovascular diseases (CVD), the world’s deadliest disease, finding novel therapeutic strategies is of high interest. Recent Findings LncRNAs can bind to proteins, DNA, and RNA regulating disease initiation and plaque growth as well as plaque stability in different cell types such as endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and macrophages. A number of lncRNAs have been implicated in cholesterol homeostasis and foam cell formation such as LASER, LeXis, and CHROME. Among others, MANTIS, lncRNA-CCL2, and MALAT1 were shown to be involved in vascular inflammation. Further regulations include, but are not limited to, DNA damage response in ECs, phenotypic switch of VSMCs, and various cell death mechanisms. Interestingly, some lncRNAs are closely correlated with response to statin treatment, such as NEXN-AS1 or LASER. Additionally, some lncRNAs may serve as CVD biomarkers. Summary LncRNAs are a potential novel therapeutic target to treat CVD, but research of lncRNA in atherosclerosis is still in its infancy. With increasing knowledge of the complex and diverse regulations of lncRNAs in the heterogeneous environment of atherosclerotic plaques, lncRNAs hold promise for their clinical translation in the near future.
Collapse
Affiliation(s)
- Tatjana Josefs
- Department of Physiology, Amsterdam Cardiovascular Science, VU University, Amsterdam UMC, Postbus 7057, 1007 MB, Amsterdam, The Netherlands
| | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Science, VU University, Amsterdam UMC, Postbus 7057, 1007 MB, Amsterdam, The Netherlands. .,Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany. .,German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany.
| |
Collapse
|
27
|
Karami J, Masoumi M, Khorramdelazad H, Bashiri H, Darvishi P, Sereshki HA, Shekarabi M, Sahebkar A. Role of autophagy in the pathogenesis of rheumatoid arthritis: Latest evidence and therapeutic approaches. Life Sci 2020; 254:117734. [PMID: 32380080 DOI: 10.1016/j.lfs.2020.117734] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/17/2020] [Accepted: 04/25/2020] [Indexed: 12/19/2022]
Abstract
Autophagy is considered as an important intracellular mechanism that degrades cytoplasmic components to furnish additional energy. It has cytoprotective effects through the degradation of intracellular pathogens, damaged organelles, and protein aggregates. On the other hand, there are reports of an association between autophagy and autoimmune diseases. Indeed, it has been evident that autophagy is dysregulated in various autoimmune diseases including rheumatoid arthritis (RA). Autophagy is implicated in the maturation survival and proliferation of various immune and non-immune cells, which play pivotal roles in RA pathogenesis. Additionally, autophagy seems to be involved in citrullination and presentation of citrullinated peptides to T lymphocyte cells. Presentation of citrullinated peptides through MHC compartments to the T cells leads to immune response and chronic inflammation. Evidence suggests that autophagy could be implicated in apoptosis resistance of RA fibroblast-like synoviocyte (RA FLS), osteoclastogenesis, and finally severe bone and cartilage destruction. Since autophagy could be an important phenomenon in RA pathogenesis, we summarized the roles of autophagy in citrullination, osteoclastogenesis, RA FLS cells survival, apoptosis resistance of cells, lymphocyte homeostasis and its clinical outcomes in RA disease.
Collapse
Affiliation(s)
- Jafar Karami
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Masoumi
- Clinical Research Development Center, Shahid Beheshti Hospital, Qom University of Medical Sciences, Qom, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hamidreza Bashiri
- Department of Rheumatology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Parisa Darvishi
- Department of Immunology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Hale Abdoli Sereshki
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shekarabi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
28
|
Bo Q, Shen M, Xiao M, Liang J, Zhai Y, Zhu H, Jiang M, Wang F, Luo X, Sun X. 3-Methyladenine Alleviates Experimental Subretinal Fibrosis by Inhibiting Macrophages and M2 Polarization Through the PI3K/Akt Pathway. J Ocul Pharmacol Ther 2020; 36:618-628. [PMID: 32552228 DOI: 10.1089/jop.2019.0112] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose: To explore the effects of 3-methyladenine (3-MA), a selective inhibitor of phosphatidylinositol-3-kinase (PI3K), on experimental subretinal fibrosis (SRF) in mice. Methods: The SRF mouse model was established by 532 nm laser photocoagulation at each fundus of mice on day 0. 3-MA was administered every 2 days from day 0 to 35. Immunofluorescence of choroidal flat mounts was performed to evaluate the size of SRF area, local macrophages, and polarization, respectively. Besides, Western blot analysis was carried out to assess the expression levels of macrophage polarization-related genes, Arg-1, Ym-1, and transforming growth factor-β2 (TGF-β2). Co-culture and migration experiments were used to demonstrate the inhibitory effect of 3-MA on fibroblasts. The gene knockout and Western blot analysis were used to explore the signal pathways related to macrophage polarization. Results: Compared with the control group, the 3-MA-treated group showed significantly less size of SRF area. 3-MA treatment reduced both circulating and local macrophages, and counteracted M2 polarization. Moreover, 3-MA inhibited fibroblast recruitment. Mechanistically, we proved that 3-MA inhibits macrophage M2 polarization by suppressing PI3K/Akt signal pathway rather than the PI3K-autophagy-related signal pathway. Conclusions: 3-MA exerts antifibrotic effects on experimental SRF by targeting circulating and local macrophages and M2 polarization, through PI3K/Akt signal pathway. These results support the potential use of 3-MA as a new therapeutic modality for SRF associated with neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Qiyu Bo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengxi Shen
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meichun Xiao
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Liang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Yuanqi Zhai
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Hong Zhu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Mei Jiang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Fenghua Wang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Xueting Luo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
29
|
Yang Y, Li X, Wang T, Guo Q, Xi T, Zheng L. Emerging agents that target signaling pathways in cancer stem cells. J Hematol Oncol 2020; 13:60. [PMID: 32456660 PMCID: PMC7249421 DOI: 10.1186/s13045-020-00901-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/18/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer stem cells (CSCs) contribute to the initiation, recurrence, and metastasis of cancer; however, there are still no drugs targeting CSCs in clinical application. There are several signaling pathways playing critical roles in CSC progression, such as the Wnt, Hedgehog, Notch, Hippo, and autophagy signaling pathways. Additionally, targeting the ferroptosis signaling pathway was recently shown to specifically kill CSCs. Therefore, targeting these pathways may suppress CSC progression. The structure of small-molecule drugs shows a good spatial dispersion, and its chemical properties determine its good druggability and pharmacokinetic properties. These characteristics make small-molecule drugs show a great advantage in drug development, which is increasingly popular in the market. Thus, in this review, we will summarize the current researches on the small-molecule compounds suppressing CSC progression, including inhibitors of Wnt, Notch, Hedgehog, and autophagy pathways, and activators of Hippo and ferroptosis pathways. These small-molecule compounds emphasize CSC importance in tumor progression and propose a new strategy to treat cancer in clinic via targeting CSCs.
Collapse
Affiliation(s)
- Yue Yang
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Ting Wang
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Qianqian Guo
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450003, People's Republic of China
| | - Tao Xi
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
30
|
Wang X, Wu Y. Protective effects of autophagy inhibitor 3-methyladenine on ischemia-reperfusion-induced retinal injury. Int Ophthalmol 2020; 40:1095-1101. [PMID: 31916058 DOI: 10.1007/s10792-019-01272-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/30/2019] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To investigate the protective effects of autophagy inhibitor 3-methyladenine (3-MA) in a rat model of ischemic-reperfusion injury (IRI). METHODS Forty Sprague-Dawley male rats (weight 220-250 g) were randomly divided into four groups: a control group (NC, n = 10), a Sham surgery group (n = 10), an IRI group (n = 10), and a 3-MA-treated IRI group [10 μL 3-MA (10 mmol/L) was injected in vitreous after the injury, n = 10]. The retinal IRI was induced by elevating the intraocular pressure to 110 mmHg for 60 min. Hematoxylin and eosin (HE) staining was used to calculate the number of retinal ganglion cells (RGCs). The level of microtubule-associated protein 1A/1B light chain 3 (LC3), Beclin-1, and Caspase-3 in the retina was detected using the immunofluorescence staining method. The LC3, Beclin-1, B-cell lymphoma/leukemia-2 (Bcl-2), and Caspase-3 protein levels were examined by Western blotting. RESULTS The number of RGCs in IRI group was significantly lower than that in NC group (P < 0.05), demonstrated by HE staining. Western blotting results indicated that the protein expression of LC3 and Beclin-1 in the IRI group was significantly elevated compared with those in the NC group (P < 0.05). However, with 3-MA treatment, the number of RCGs in 3-MA-treated IRI group was elevated and protein levels of LC3, Beclin-1 were down-regulated, compared with those in the IRI group (P < 0.05). Further immunohistochemistry staining and Western blot showed that 3-MA-treated IRI group presented down-regulated Caspase-3 and up-regulated Bcl-2 protein expression with comparison of IRI group (P < 0.05). CONCLUSIONS Retina IRI-caused RGCs loss involved activated autophagy pathway and apoptosis, which could be prevented by autophagy inhibitor 3-MA. Autophagy inhibitor 3-MA may act as a potent therapeutic tool in attenuating retina IRI.
Collapse
Affiliation(s)
- Xiaorui Wang
- Ophthalmology Department, Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Yuyu Wu
- Ophthalmology Department, Second Affiliated Hospital, Fujian Medical University, Quanzhou, China.
| |
Collapse
|
31
|
Zhang X, Ramírez CM, Aryal B, Madrigal-Matute J, Liu X, Diaz A, Torrecilla-Parra M, Suárez Y, Cuervo AM, Sessa WC, Fernández-Hernando C. Cav-1 (Caveolin-1) Deficiency Increases Autophagy in the Endothelium and Attenuates Vascular Inflammation and Atherosclerosis. Arterioscler Thromb Vasc Biol 2020; 40:1510-1522. [PMID: 32349535 PMCID: PMC7253189 DOI: 10.1161/atvbaha.120.314291] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Endothelial Cav-1 (caveolin-1) expression plays a relevant role during atherogenesis by controlling NO production, vascular inflammation, LDL (low-density lipoprotein) transcytosis, and extracellular matrix remodeling. Additional studies have identified cholesterol-rich membrane domains as important regulators of autophagy by recruiting ATGs (autophagy-related proteins) to the plasma membrane. Here, we investigate how the expression of Cav-1 in the aortic endothelium influences autophagy and whether enhanced autophagy contributes to the atheroprotective phenotype observed in Cav-1–deficient mice.
Collapse
Affiliation(s)
- Xinbo Zhang
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (X.Z., C.M.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Cristina M Ramírez
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (X.Z., C.M.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,IMDEA Research Institute of Food and Health Sciences, Madrid, Spain (C.M.R., M.T.-P.)
| | - Binod Aryal
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (X.Z., C.M.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Julio Madrigal-Matute
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (J.M.-M., A.D., A.M.C.)
| | - Xinran Liu
- Department of Cell Biology (X.L.), Yale University School of Medicine, New Haven, CT
| | - Antonio Diaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (J.M.-M., A.D., A.M.C.)
| | | | - Yajaira Suárez
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (X.Z., C.M.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| | - Ana M Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (J.M.-M., A.D., A.M.C.)
| | - William C Sessa
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Department of Pharmacology (W.C.S.), Yale University School of Medicine, New Haven, CT
| | - Carlos Fernández-Hernando
- From the Vascular Biology and Therapeutics Program (X.Z., C.M.R., B.A., Y.S., W.C.S., C.F.-H.), Yale University School of Medicine, New Haven, CT.,Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology (X.Z., C.M.R., B.A., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT
| |
Collapse
|
32
|
Zhang P, Li Y, Fu Y, Huang L, Liu B, Zhang L, Shao XM, Xiao D. Inhibition of Autophagy Signaling via 3-methyladenine Rescued Nicotine-Mediated Cardiac Pathological Effects and Heart Dysfunctions. Int J Biol Sci 2020; 16:1349-1362. [PMID: 32210724 PMCID: PMC7085229 DOI: 10.7150/ijbs.41275] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/31/2020] [Indexed: 12/15/2022] Open
Abstract
Rationale: Cigarette smoking is a well-established risk factor for myocardial infarction and sudden cardiac death. The deleterious effects are mainly due to nicotine, but the mechanisms involved and theranostics remain unclear. Thus, we tested the hypothesis that nicotine exposure increases the heart sensitivity to ischemia/reperfusion injury and dysfunction, which can be rescued by autophagy inhibitor. Methods: Nicotine or saline was administered to adult rats via subcutaneous osmotic minipumps in the absence or presence of an autophagy inhibitor, 3-methyladenine (3-MA). After 30 days of nicotine treatment, the rats underwent the cardiac ischemia/reperfusion (I/R) procedure and echocardiography analysis, and the heart tissues were isolated for molecular biological studies. Results: Nicotine exposure increased I/R-induced cardiac injury and cardiac dysfunction as compared to the control. The levels of autophagy-related proteins including LC3 II, P62, Beclin1, and Atg5 were upregulated in the reperfused hearts isolated from nicotine-treated group. In addition, nicotine enhanced cardiac and plasma ROS production, and increased the phosphorylation of GSK3β (ser9) in the left ventricle tissues. Treatment with 3-MA abolished nicotine-mediated increase in the levels of autophagy-related proteins and phosphorylation of GSK3β, but had no effect on ROS production. Of importance, 3-MA ameliorated the augmented I/R-induced cardiac injury and dysfunction in the nicotine-treated group as compared to the control. Conclusion: Our results demonstrate that nicotine exposure enhances autophagy signaling pathway, resulting in development of ischemic-sensitive phenotype of heart. It suggests a potentially novel therapeutic strategy of autophagy inhibition for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Peng Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA.,Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Li
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Yingjie Fu
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Lei Huang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Bailin Liu
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Xuesi M Shao
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California, USA
| | - Daliao Xiao
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
33
|
Sun G, Li Y, Ji Z. Up-regulation of MIAT aggravates the atherosclerotic damage in atherosclerosis mice through the activation of PI3K/Akt signaling pathway. Drug Deliv 2019; 26:641-649. [PMID: 31237148 PMCID: PMC6598488 DOI: 10.1080/10717544.2019.1628116] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study is performed to elucidate the role of long non-coding RNA myocardial infarction associated transcript (lncRNA MIAT) in vulnerable plaque formation in rats with atherosclerosis (AS) through the regulation of the PI3K/Akt signaling pathway. The mice model of AS was established, and the successful modeled AS mice were treated with overexpressed MIAT and silenced MIAT. The levels of blood lipids, atherosclerotic plaques (AP) formation, the lipid content, collagen content, apoptosis of aortic cells, angiogenesis as well as the expression of inflammatory factors, such as tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) were determined through a series of experiments. MIAT was found to be upregulated in AS. Additionally, MIAT up-regulated the levels of blood lipids, promoted AP formation, increased the lipid content and decreased the collagen content of AP, promoted the apoptosis of aortic cells in AS mice by activating the PI3K/Akt signaling pathway. Meanwhile, MIAT was determined to promote angiogenesis as well as the expression of inflammatory factors (IL-1β, IL-6, and TNF-α) in AS mice through the activation of the PI3K/Akt signaling pathway. Furthermore, MIAT activated the PI3K/Akt signaling pathway to participate in AS progression. Our study suggests that upregulation of MIAT can aggravate AS injury in AS mice via the activation of the PI3K/Akt signaling pathway, which could provide a novel target for the treatment of AS.
Collapse
Affiliation(s)
- Guoqiang Sun
- a Department of Cardiology , The First Hospital of Jilin University , Changchun , PR China
| | - Yubo Li
- b Department of Pediatrics , Medical College , Changchun , PR China
| | - Zhiyong Ji
- c Department of Critical Care Medicine , The First Hospital of Jilin University , Changchun , PR China
| |
Collapse
|
34
|
Wang B, Dong G, Zhang Q, Yan F, Li Z, Li C, Zhang H, Ma Q, Dai J, Si C, Xiong H. The inhibitor of autophagy SBI-0206965 aggravates atherosclerosis through decreasing myeloid-derived suppressor cells. Exp Ther Med 2019; 19:1370-1378. [PMID: 32010311 PMCID: PMC6966176 DOI: 10.3892/etm.2019.8317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis (AS) is currently the leading cause of mortality worldwide, with the development of new strategies to prevent the formation and rupture of atherosclerotic plaques being a paramount area of research. Amounting evidence suggests autophagy has an important role in the pathogenesis of AS and may be a potential therapeutic target. In this study, the effect of SBI-0206965(6965), a novel inhibitor of autophagy, was tested on the development of AS in apolipoprotein E deficient (ApoE−/−) mice. Systemic application of 6965 was found to aggravate AS, with increased plaque size and decreased plaque stability in comparison with the control. Of note, it was observed that 6965 decreased the proportion of myeloid-derived suppressor cells (MDSCs). Further investigation demonstrated MDSCs markedly alleviated AS in ApoE−/− mice; while 6965 reduced the viability and promoted apoptosis of MDSCs in vitro. This is the first study describing an association between autophagy and MDSCs in AS models, providing a novel mechanism to potentially target in the management of this condition.
Collapse
Affiliation(s)
- Bo Wang
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China.,Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Guanjun Dong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Qingqiing Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Fenglian Yan
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Zhihua Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Chunxia Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Qun Ma
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Jun Dai
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Chuanping Si
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Huabao Xiong
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
35
|
Fu W, Chen M, Ou L, Li T, Chang X, Huang R, Zhang J, Zhang Z. Xiaoyaosan prevents atherosclerotic vulnerable plaque formation through heat shock protein/glucocorticoid receptor axis-mediated mechanism. Am J Transl Res 2019; 11:5531-5545. [PMID: 31632527 PMCID: PMC6789251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/31/2019] [Indexed: 06/10/2023]
Abstract
Atherosclerosis is a metabolic and chronic inflammatory disease caused by deposition of lipoproteins in arteries. However, the diagnostic drug and the mechanism for this vascular disease are less studied. In the present study, atherosclerosis model was developed using apolipoprotein E-deficient mice that was treated with long-term high-fat food and chronic stresses. Xiaoyaosan (XYS) and glucocorticoid receptor (GR) antagonist RU 38486 were orally administrated to the mice. The change of aortic root vessels was observed by histological analysis. The results indicate that high-fat food coupled with chronic stress induced atherosclerosis in mice model, with plaque formation in the entire aortas foam cells and macrophage infiltration in aortic tissues. However, XYS granules inhibited the development of atherosclerotic lesion, with down-regulation of GC, TC, TG, HDL-C, ox-LDL, LDL-C, IFN-γ, IL-6, IL-1β, and TNF-α measured by ELISA method; XYS inhibited the expressions of GR, CD36, HSP27/60/90, and induced ABCA1 in atherosclerotic mice, which was measured by qPCR and Western blot, which showed similar effect as positive control RU 38486 did. The interaction between HSP90-GR complexes and CD36 was validated in atherosclerotic mice. Our results inferred that the HSP/GR complex-mediated CD36 axis was involved in the regulation of atherosclerosis development in mice verified by Co-IP assay, EMSA, and Chip-PCR. These findings not only provide the potential therapeutic value of Xiaoyaosan for atherosclerotic vulnerable plaque but also brought forth a novel strategy for preventing the formation and treatment of atherosclerotic vulnerable plaques through the elucidated mechanism of XYS on vulnerable plaque.
Collapse
Affiliation(s)
- Wenjun Fu
- Centre for Integrative Medicine, School of Basic Medical Science, Guangzhou University of Chinese MedicineGuangzhou 510006, Guangdong, China
| | - Mingtai Chen
- Department of Cardiovascular, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Lijun Ou
- Department of Cardiovascular, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Tao Li
- Department of Cardiovascular, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Xiao Chang
- Department of Intensive Care Unit, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Ruolan Huang
- Department of Intensive Care Unit, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| | - Jian Zhang
- Heart Failure Center, Fuwai Hospital Chinese Academy of Medical SciencesBeijing 100037, China
| | - Zhong Zhang
- Department of Cardiovascular, Shenzhen Traditional Chinese Medicine Hospital, The Forth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen 518033, Guangdong, China
| |
Collapse
|
36
|
Guo FX, Wu Q, Li P, Zheng L, Ye S, Dai XY, Kang CM, Lu JB, Xu BM, Xu YJ, Xiao L, Lu ZF, Bai HL, Hu YW, Wang Q. The role of the LncRNA-FA2H-2-MLKL pathway in atherosclerosis by regulation of autophagy flux and inflammation through mTOR-dependent signaling. Cell Death Differ 2019; 26:1670-1687. [PMID: 30683918 PMCID: PMC6748100 DOI: 10.1038/s41418-018-0235-z] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 10/28/2018] [Accepted: 10/30/2018] [Indexed: 01/13/2023] Open
Abstract
Atherosclerosis is a progressive, chronic inflammation in arterial walls. Long noncoding RNAs (lncRNAs) participate in inflammation, but the exact mechanism in atherosclerosis is unclear. Our microarray analyses revealed that the levels of lncRNA-FA2H-2 were significantly decreased by oxidized low-density lipoprotein (OX-LDL). Bioinformatics analyses indicated that mixed lineage kinase domain-like protein (MLKL) might be regulated by lncRNA-FA2H-2. In vitro experiments showed that lncRNA-FA2H-2 interacted with the promoter of the MLKL gene, downregulated MLKL expression, and the binding sites between -750 and 471 were necessary for lncRNA-FA2H-2 responsiveness to MLKL. Silencing lncRNA-FA2H-2 and overexpression of MLKL could activate inflammation and inhibited autophagy flux. Both lncRNA-FA2H-2 knockdown and overexpression of MLKL could significantly aggravate inflammatory responses induced by OX-LDL. We found that the 3-methyladenine (3-MA) and Atg7-shRNA enhanced inflammatory responses induced by knockdown of lncRNA-FA2H-2 and overexpression of MLKL. We demonstrated that the effects of MLKL on autophagy might be associated with a mechanistic target of rapamycin (mTOR)-dependent signaling pathways. In vivo experiments with apoE knockout mice fed a western diet demonstrated that LncRNA-FA2H-2 knockdown decreased microtubule-associated expression of microtubule-associated protein 1 light chain 3 II and lysosome-associated membrane protein 1, but increased expression of sequestosome 1 (p62), MLKL, vascular cell adhesion molecule-1, monocyte chemoattractant protein-1, and interleukin-6 in atherosclerotic lesions. Our findings indicated that the lncRNA-FA2H-2-MLKL pathway is essential for regulation of autophagy and inflammation, and suggested that lncRNA-FA2H-2 and MLKL could act as potential therapeutic targets to ameliorate atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Feng-Xia Guo
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qian Wu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Pan Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shu Ye
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Xiao-Yan Dai
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Chun-Min Kang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing-Bo Lu
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bang-Ming Xu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuan-Jun Xu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Xiao
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhi-Feng Lu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Huan-Lan Bai
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
37
|
Yang G, Zeng X, Li J, Leung CK, Zhang D, Hong S, He Y, Huang J, Li L, Li Z. Protective effect of gastrodin against methamphetamine-induced autophagy in human dopaminergic neuroblastoma SH-SY5Y cells via the AKT/mTOR signaling pathway. Neurosci Lett 2019; 707:134287. [PMID: 31128157 DOI: 10.1016/j.neulet.2019.134287] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 02/06/2023]
Abstract
Methamphetamine (METH) has been shown to induce neuropathological dysfunction and irreversible brain cell damage. Prior studies indicated the involvement of autophagy in METH-induced neurotoxicity. However, the underlying mechanism by which autophagy contributes to METH-induced neurotoxicity remains elusive. Gastrodin, a primary bioactive constituent of Gastrodia elata-an orchid used in traditional Chinese medicine-is used widely to treat stroke, dementia, and headache. This study investigates whether METH induces autophagy in the human dopaminergic neuroblastoma cell line SH-SY5Y, then examines the neuroprotective effects of gastrodin against autophagy in METH-treated SH-SY5Y cells. The effects of METH on the protein expressions of autophagy-related genes (LC3B and Beclin-1) were evaluated with and without gastrodin. The presence of autophagosomes in the METH-induced treatment with and without gastrodin is revealed through transmission electron microscopy. Pharmacological intervention was employed to study the role of the AKT/mTOR signaling pathway in the gastrodin-mediated neuroprotection against METH-induced autophagy. The present results indicate that METH exposure elevates the protein expression levels of LC3B and Beclin-1 in a dose- and time-dependent manner. Gastrodin is observed to block the METH-induced upregulation of LC3B and Beclin-1 protein expression significantly. Gastrodin is found to exhibit an anti-autophagic effect on the inhibition of the METH-induced Beclin-1 protein expression, partly via the AKT/mTOR These findings may aid the development of a gastrodin-based therapeutic strategy for treating METH-induced neurotoxicity.
Collapse
Affiliation(s)
- Genmeng Yang
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Xiaofeng Zeng
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Juan Li
- School of Basic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Chi-Kwan Leung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; CUHK-SDU Joint Laboratory of Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Dongxian Zhang
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Shijun Hong
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yongwang He
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Jian Huang
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Lihua Li
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan Province, China.
| | - Zhen Li
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|
38
|
Lee W, Kim SH. Autophagy at synapses in neurodegenerative diseases. Arch Pharm Res 2019; 42:407-415. [DOI: 10.1007/s12272-019-01148-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/21/2019] [Indexed: 12/31/2022]
|
39
|
Shih JH, Chiu CH, Ma KH, Huang YS, Shiue CY, Yeh TY, Kao LT, Lin YY, Li IH. Autophagy inhibition plays a protective role against 3, 4-methylenedioxymethamphetamine (MDMA)-induced loss of serotonin transporters and depressive-like behaviors in rats. Pharmacol Res 2019; 142:283-293. [PMID: 30826457 DOI: 10.1016/j.phrs.2019.02.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/27/2018] [Accepted: 02/24/2019] [Indexed: 02/06/2023]
Abstract
The 3,4-methylenedioxymethamphetamine (MDMA) is a popular recreational drug, which ultimately leads to serotonergic (5-HT) neurotoxicity and psychiatric disorders. Previous in vitro studies have consistently demonstrated that MDMA provokes autophagic activation, as well as damage of 5-HT axons and nerve fibers. So far, whether autophagy, a well-conserved cellular process that is critical for cell fate, also participates in MDMA-induced neurotoxicity in vivo remains elusive. Here, we first examined time-course of autophagy-related changes during repeated administration of MDMA (10 mg/kg s.c. twice daily for 4 consecutive days) using immunofluorescent staining for tryptophan hydroxylase and microtubule-associated protein 1 light chain 3 beta in rats. We also evaluated the protective effects of 3-methyadanine (3-MA, an autophagy inhibitor, 15 mg/kg i.p.) against MDMA-induced acute and long-term reductions in serotonin transporters (SERT) density in various brain regions using immunohistochemical staining and positron emission tomography (PET) imaging respectively. Plasma corticosterone measurements and forced swim tests were performed to evaluate the depressive performance. The staining results showed that repeated administration of MDMA increased expression of autophagosome and caused reduction in SERT densities of striatum and frontal cortex, which was ameliorated in the presence of 3-MA. PET imaging data also revealed that 3-MA could ameliorate MDMA-induced long-term decreased SERT availability in various brain regions of rats. Furthermore, immobility time of forced swim tests and plasma corticosterone levels were less in the group of MDMA co-injected with 3-MA compared with that of MDMA group. Together, these findings suggest that autophagy inhibition may confer protection against neurobiological and behavioral changes induced by MDMA.
Collapse
Affiliation(s)
- Jui-Hu Shih
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Chuang-Hsin Chiu
- Department of Nuclear Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Hsing Ma
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yuahn-Sieh Huang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Chyng-Yann Shiue
- Department of Nuclear Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ting-Yin Yeh
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Li-Ting Kao
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan; Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
| | - Yang-Yi Lin
- Department of Pharmacy, Chi Mei Medical Center, Tainan, Taiwan
| | - I-Hsun Li
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
40
|
Biglycan evokes autophagy in macrophages via a novel CD44/Toll-like receptor 4 signaling axis in ischemia/reperfusion injury. Kidney Int 2019; 95:540-562. [PMID: 30712922 DOI: 10.1016/j.kint.2018.10.037] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/09/2018] [Accepted: 10/24/2018] [Indexed: 01/14/2023]
Abstract
Biglycan, a small leucine-rich proteoglycan, acts as a danger signal and is classically thought to promote macrophage recruitment via Toll-like receptors (TLR) 2 and 4. We have recently shown that biglycan signaling through TLR 2/4 and the CD14 co-receptor regulates inflammation, suggesting that TLR co-receptors may determine whether biglycan-TLR signaling is pro- or anti-inflammatory. Here, we sought to identify other co-receptors and characterize their impact on biglycan-TLR signaling. We found a marked increase in the number of autophagic macrophages in mice stably overexpressing soluble biglycan. In vitro, stimulation of murine macrophages with biglycan triggered autophagosome formation and enhanced the flux of autophagy markers. Soluble biglycan also promoted autophagy in human peripheral blood macrophages. Using macrophages from mice lacking TLR2 and/or TLR4, CD14, or CD44, we demonstrated that the pro-autophagy signal required TLR4 interaction with CD44, a receptor involved in adhesion, migration, lymphocyte activation, and angiogenesis. In vivo, transient overexpression of circulating biglycan at the onset of renal ischemia/reperfusion injury (IRI) enhanced M1 macrophage recruitment into the kidneys of Cd44+/+ and Cd44-/- mice but not Cd14-/- mice. The biglycan-CD44 interaction increased M1 autophagy and the number of renal M2 macrophages and reduced tubular damage following IRI. Thus, CD44 is a novel signaling co-receptor for biglycan, an interaction that is required for TLR4-CD44-dependent pro-autophagic activity in macrophages. Interfering with the interaction between biglycan and specific TLR co-receptors could represent a promising therapeutic intervention to curtail kidney inflammation and damage.
Collapse
|
41
|
Vomero M, Barbati C, Colasanti T, Perricone C, Novelli L, Ceccarelli F, Spinelli FR, Di Franco M, Conti F, Valesini G, Alessandri C. Autophagy and Rheumatoid Arthritis: Current Knowledges and Future Perspectives. Front Immunol 2018; 9:1577. [PMID: 30072986 PMCID: PMC6058034 DOI: 10.3389/fimmu.2018.01577] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/26/2018] [Indexed: 01/07/2023] Open
Abstract
Autophagy is a degradation mechanism by which cells recycle cytoplasmic components to generate energy. By influencing lymphocyte development, survival, and proliferation, autophagy regulates the immune responses against self and non-self antigens. Deregulation of autophagic pathway has recently been implicated in the pathogenesis of several autoimmune diseases, including rheumatoid arthritis (RA). Indeed, autophagy seems to be involved in the generation of citrullinated peptides, and also in apoptosis resistance in RA. In this review, we summarize the current knowledge on the role of autophagy in RA and discuss the possibility of a clinical application of autophagy modulation in this disease.
Collapse
|
42
|
Hsiao YT, Kuo CL, Chueh FS, Liu KC, Bau DT, Chung JG. Curcuminoids Induce Reactive Oxygen Species and Autophagy to Enhance Apoptosis in Human Oral Cancer Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:1145-1168. [PMID: 29976081 DOI: 10.1142/s0192415x1850060x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Numerous studies support the use of herbal medicine or natural products for chemotherapy in human cancers. Reports have associated curcumin (CUR), dimethoxy curcumin (DMC) and bisdemethoxycurcumin (BDMC) with numerous biological activities including anticancer activities, but no available information have shown that these induced apoptotic cell death and autophagy in human oral cancer cells. In the present study, we investigated the effect of CUR, DMC and BDMC on the cell viability, apoptotic cell death, reactive oxygen species (ROS), Ca[Formula: see text], mitochondria membrane potential (MMP) and caspase activities using flow cytometry assay and autophagy by monodansylcadaverine (MDC) and acridine orange (AO) staining in human oral cancer SAS cells. Results indicated that CUR, DMC and BDMC decreased total viable cell number through the induction of cell autophagy and apoptosis in SAS cells. Cells were pretreated with N-acetyl-cysteine (NAC), 3-methyladenine (3MA), rapamycin and carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoro-methylketone (Z-VAD-fmk) and then were treated with CUR, DMC and BDMC that led to increased total viable cell number when compared to CUR, DMC and BDMC treatments only. Results indicated induced apoptotic cell death through ROS, mitochondria-dependent pathway and induction of cell autophagy. Based on those observations, we suggest that CUR, DMC and BDMC could be used as a potential anticancer agent in human oral cancer.
Collapse
Affiliation(s)
- Yung-Ting Hsiao
- * Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chao-Lin Kuo
- † Department of Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Fu-Shin Chueh
- ¶ Department of Food Nutrition and Health Biotechnology, Asia University, Wufeng, Taichung, Taiwan
| | - Kuo-Ching Liu
- ‡ Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Da-Tian Bau
- § Graduate Institute of Biomedical and Sciences, China Medical University, Taichung, Taiwan.,** Terry Fox Cancer Research Laboratory, China Medical University Hospital, Taichung, Taiwan
| | - Jing-Gung Chung
- * Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.,∥ Department of Biotechnology, Asia University, Wufeng, Taichung, Taiwan
| |
Collapse
|
43
|
Li YL, Qi RQ, Yang Y, Wang HX, Jiang HH, Li ZX, Xiao BH, Zheng S, Hong YX, Li JH, Chen HD, Gao XH. Screening and identification of differentially expressed serum proteins in patients with vitiligo using two‑dimensional gel electrophoresis coupled with mass spectrometry. Mol Med Rep 2017; 17:2651-2659. [PMID: 29207142 DOI: 10.3892/mmr.2017.8159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 11/22/2017] [Indexed: 11/06/2022] Open
Abstract
In the clinic, vitiligo is characterized by two stages: Stable and progressive. The pathogenesis of vitiligo is still not clear. Here, we identified serum markers of vitiligo by screening for differentially expressed proteins in patients with vitiligo compared to healthy individuals. Serum samples were collected from patients with vitiligo (n=10 for both the stable and progressive stages) and healthy individuals (n=10). Two‑dimensional gel electrophoresis followed by matrix‑assisted laser desorption/ionization time‑of‑flight mass spectrometry and western blotting were used to validate the differential expression of the proteins in the serum (n=20 each, at both stages for patients and healthy individuals). A total of 48 differentially expressed proteins were identified by gel image analysis. There were 28 differentially expressed proteins in patients with progressive vitiligo (PV) and 13 differentially expressed proteins in patients with stable vitiligo (SV) compared with that in healthy individuals. Additionally, 7 differentially expressed proteins were identified in patients with PV compared with those in patients with SV. The western blotting results showed that Peroxiredoxin‑6, apolipoprotein L1, apolipoprotein E and mannose‑binding protein were differentially expressed in patients with different stages of vitiligo. Our results showed that change serum levels of several proteins might be useful as biomarkers or in understanding the pathogenesis of vitiligo.
Collapse
Affiliation(s)
- Yi-Lei Li
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Rui-Qun Qi
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yang Yang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - He-Xiao Wang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hang-Hang Jiang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zheng-Xiu Li
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Bi-Huan Xiao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Song Zheng
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yu-Xiao Hong
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jiu-Hong Li
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hong-Duo Chen
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xing-Hua Gao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
44
|
Li X, Zhou Y, Zhang X, Cao X, Wu C, Guo P. Cordycepin stimulates autophagy in macrophages and prevents atherosclerotic plaque formation in ApoE -/- mice. Oncotarget 2017; 8:94726-94737. [PMID: 29212261 PMCID: PMC5706907 DOI: 10.18632/oncotarget.21886] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/21/2017] [Indexed: 12/12/2022] Open
Abstract
Autophagy in macrophages plays a key role in the pathogenesis and progression of atherosclerosis and has become a potential therapeutic target. Here we show that cordycepin (Cpn), a natural derivative of adenosine, markedly reduced atherosclerotic plaque and ameliorated associated symptoms such as dyslipidemia, hyperglycemia and inflammation in ApoE-/- mice. Supplementation of Cpn dose-dependently inhibited oxLDL-elicited foam cell formation and modulated intracellular cholesterol homeostasis by inhibiting cholesterol uptake and promoting cholesterol efflux in RAW264.7 macrophages. Notably, Cpn exhibited significant stimulating effect on macrophage autophagy, as estimated by western blotting, immunofluorescent staining and autophagic vacuoles observation by transmission electron microscopy. The inhibitive effects of Cpn on foam cell formation were dramatically deteriorated in the presence of various autophagy inhibitors, suggesting that autophagy participate, at least in part, in the atheroprotective role of Cpn. Further investigations using different autophagy inhibitors and specific siRNAs for AMP-activated protein kinase (AMPK) gamma1 subunit indicated that Cpn may stimulate macrophage autophagy through AMPK-mTOR pathway. Together, our results demonstrated Cpn as a potential therapeutic agent for the prevention and treatment of atherosclerosis, and the autophagic activity presents a novel mechanism for Cpn-mediated atheroprotection.
Collapse
Affiliation(s)
- Xin Li
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, P.R. China
| | - Yue Zhou
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, P.R. China
| | - Xue Zhang
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, P.R. China
| | - Xiaoxue Cao
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, P.R. China
| | - Chongming Wu
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, P.R. China
| | - Peng Guo
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, P.R. China
| |
Collapse
|
45
|
Chen HJC, Ip SW, Lin FD. Simultaneous Mass Spectrometric Analysis of Methylated and Ethylated Peptides in Human Hemoglobin: Correlation with Cigarette Smoking. Chem Res Toxicol 2017; 30:2074-2083. [DOI: 10.1021/acs.chemrestox.7b00234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Hauh-Jyun Candy Chen
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Ming-Hsiung, Chia-Yi 62142, Taiwan
| | - Sun Wai Ip
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Ming-Hsiung, Chia-Yi 62142, Taiwan
| | - Fu-Di Lin
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Ming-Hsiung, Chia-Yi 62142, Taiwan
| |
Collapse
|
46
|
An Z, Qiao F, Lu Q, Ma Y, Liu Y, Lu F, Xu Z. Interleukin-6 downregulated vascular smooth muscle cell contractile proteins via ATG4B-mediated autophagy in thoracic aortic dissection. Heart Vessels 2017; 32:1523-1535. [PMID: 28965202 DOI: 10.1007/s00380-017-1054-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/22/2017] [Indexed: 01/03/2023]
Abstract
Interleukin-6 (IL-6) overexpression played an important role in the pathogenesis of thoracic aortic dissection (TAD). Our previous study found enhanced autophagy accompanying with contractile proteins α smooth muscle actin (α-SMA) and smooth muscle 22α (SM22α) degradation in TAD aortic vascular smooth muscle cells (VSMCs). Autophagy is an important way for intracellular proteins degradation, while IL-6 has been found as a contributing factor of autophagy in some cancers. These indicated IL-6 might contribute to the occurrence of TAD by promoting autophagy-induced contractile proteins degradation, which has not been investigated. The aim of the present study is to verify this hypothesis and investigate the mechanism of it. We collected 10 TAD and 10 control aortic specimens from patients underwent TAD surgical repair and coronary artery bypass grafting, respectively. Quantitative real-time polymerase chain reaction was used to detect mRNA expression. Protein expression level was assessed by enzyme-linked immunosorbent assay, western blot, and immunohistochemistry. Microtubule-associated protein 1 light chain 3 beta overexpression adenovirus with green and red fluorescent protein tags and transmission electron microscopy were used to detect autophagy level in VSMCs. 3-Methyladenine (3-MA) and chloroquine were used to block autophagy in human VSMCs. Experiment results showed that the expression of IL-6 was significantly increased accompanying with up-regulated autophagy in TAD aortic wall compared with controls. In vitro results showed that IL-6 stimulation decreased the expression of VSMCs contractile proteins α-SMA and SM22α accompanying with up-regulated autophagy. Blocking autophagy with 3-MA or chloroquine inhibited IL-6 induced α-SMA and SM22α degradation. Further investigation showed that autophagy-related 4B cysteine peptidase (ATG4B) was significantly overexpressed in TAD aortic wall and played important role in IL-6 induced autophagy up-regulation. ATG4B knockdown blocked IL-6-induced autophagy and α-SMA and SM22α degradation, while ATG4B overexpression partly replaced the function of IL-6 in human VSMCs. In conclusion, our study demonstrated that IL-6 downregulated expression of VSMCs contractile proteins α-SMA and SM22α via enhancing ATG4B-mediated autophagy in TAD.
Collapse
Affiliation(s)
- Zhao An
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Fan Qiao
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Qijue Lu
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ye Ma
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Yang Liu
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China
| | - Fanglin Lu
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China.
| | - Zhiyun Xu
- Department of Cardiovascular Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Rd, Shanghai, 200433, China.
| |
Collapse
|