1
|
Woo K, Kim DH, Park HS, Oh MH, Lee JC, Choi CH. Acinetobacter baumannii OmpA hinders host autophagy via the CaMKK2-reliant AMPK-pathway. mBio 2025; 16:e0336924. [PMID: 39998213 PMCID: PMC11980379 DOI: 10.1128/mbio.03369-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Outer membrane protein A (OmpA) plays a vital role in the interactions between Acinetobacter baumannii and host cells. Autophagy is a defense mechanism that hinders the intracellular replication of bacteria, thereby safeguarding cells against microbial infections. While it has been observed that A. baumannii triggers cellular autophagy, the precise role of its virulence protein OmpA in this process remains uncertain. In this study, we investigated the effects of A. baumannii OmpA (AbOmpA) on autophagy and explored the underlying molecular mechanisms. We found that AbOmpA exerted its autophagy-suppressive effect through inhibition of CaMKK2 phosphorylation. Compared to the wild-type strain, the ompA-deletion mutant strain displayed considerably enhanced autophagy induction, via the AMPK-ULK1 pathway. AbOmpA hindered starvation-induced autophagy, while A. baumannii-Omp33 (AbOmp33) and Escherichia coli-OmpA (EcOmpA) did not. Importantly, we confirmed that exogenous AbOmpA suppressed autophagy through the CaMKK2-AMPK-ULK1 pathway during A. baumannii infection. These findings reveal a novel mechanism for AbOmpA-mediated autophagy evasion, providing new insights into the pathogenesis of A. baumannii infection.IMPORTANCEAcinetobacter baumannii is a significant clinical pathogen notorious for causing infections in hospitals. Its outer membrane protein A acts as a virulence factor and helps the bacteria evade host defenses. Autophagy is a defense mechanism that hinders the intracellular replication of bacteria. While it has been observed that A. baumannii triggers cellular autophagy, the precise role of its AbOmpA in this process remains uncertain. Our studies demonstrate the AbOmpA of A. baumannii inhibits the cellular defense process, autophagy, through the CaMKK2-AMPK-ULK1 signaling cascade, thereby enhancing bacterial survival. This insight into how AbOmpA bypasses autophagy sheds light on A. baumannii infection's novel virulence strategy and suggests possible treatments.
Collapse
Affiliation(s)
- Kyungho Woo
- Department of Microbiology, School of Medicine, Chungnam National University, Daejeon, South Korea
- Translational Immunology Institute, School of Medicine, Chungnam National University, Daejeon, South Korea
| | - Dong Ho Kim
- Department of Microbiology, School of Medicine, Chungnam National University, Daejeon, South Korea
- Translational Immunology Institute, School of Medicine, Chungnam National University, Daejeon, South Korea
| | - Ho-Sung Park
- Department of Microbiology, School of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, South Korea
- System Network Inflammation Control Research Center, School of Medicine, Chungnam National University, Daejeon, South Korea
| | - Man Hwan Oh
- Department of Microbiology, College of Science and Technology, Dankook University, Cheonan, South Korea
| | - Je Chul Lee
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Chul Hee Choi
- Department of Microbiology, School of Medicine, Chungnam National University, Daejeon, South Korea
- Translational Immunology Institute, School of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, South Korea
- System Network Inflammation Control Research Center, School of Medicine, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
2
|
Fan Q, Xiao K, A R, Gao LP, Wu YZ, Chen DD, Hu C, Jia XX, Liu CM, Liu X, Chen C, Shi Q, Dong XP. Accumulation of Prion Triggers the Enhanced Glycolysis via Activation of AMKP Pathway in Prion-Infected Rodent and Cell Models. Mol Neurobiol 2024; 61:9810-9834. [PMID: 37726499 DOI: 10.1007/s12035-023-03621-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023]
Abstract
Mitochondrial dysfunction is one of the hallmarks in the pathophysiology of prion disease and other neurodegenerative diseases. Various metabolic dysfunctions are identified and considered to contribute to the progression of some types of neurodegenerative diseases. In this study, we evaluated the status of glycolysis pathway in prion-infected rodent and cell models. The levels of the key enzymes, hexokinase (HK), phosphofructokinase (PFK), and pyruvate kinase (PK) were significantly increased, accompanying with markedly downregulated mitochondrial complexes. Double-stained IFAs revealed that the increased HK2 and PFK distributed widely in GFAP-, Iba1-, and NeuN-positive cells. We also identified increased levels of AMP-activated protein kinase (AMPK) and the downstream signaling. Changes of AMPK activity in prion-infected cells by the AMPK-specific inhibitor or activator induced the corresponding alterations not only in the downstream signaling, but also the expressions of three key kinases in glycolysis pathway and the mitochondrial complexes. Transient removal or complete clearance of prion propagation in the prion-infected cells partially but significantly reversed the increases of the key enzymes in glycolysis, the upregulation of AMPK signaling pathway, and the decreases of the mitochondrial complexes. Measurements of the cellular oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) showed lower OCR and higher ECAR in prion-infected cell line, which were sufficiently reversed by clearance of prion propagation. Those data indicate a metabolic reprogramming from oxidative phosphorylation to glycolysis in the brains during the progression of prion disease. Accumulation of PrPSc is critical for the switch to glycolysis, largely via activating AMPK pathway.
Collapse
Affiliation(s)
- Qin Fan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kang Xiao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruhan A
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li-Ping Gao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yue-Zhang Wu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dong-Dong Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chao Hu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiao-Xi Jia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chu-Mou Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xin Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Cao Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Qi Shi
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xiao-Ping Dong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.
- China Academy of Chinese Medical Sciences, Beijing, China.
- Shanghai Institute of Infectious Disease and Biosafety, Shanghai, China.
| |
Collapse
|
3
|
Althobaiti NA, Al-Abbas NS, Alsharif I, Albalawi AE, Almars AI, Basabrain AA, Jafer A, Ellatif SA, Bauthman NM, Almohaimeed HM, Soliman MH. Gadd45A-mediated autophagy regulation and its impact on Alzheimer's disease pathogenesis: Deciphering the molecular Nexus. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167353. [PMID: 39004381 DOI: 10.1016/j.bbadis.2024.167353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND The growth arrest and DNA damage-inducible 45 (Gadd45) gene has been implicated in various central nervous system (CNS) functions, both normal and pathological, including aging, memory, and neurodegenerative diseases. In this study, we examined whether Gadd45A deletion triggers pathways associated with neurodegenerative diseases including Alzheimer's disease (AD). METHODS Utilizing transcriptome data from AD-associated hippocampus samples, we identified Gadd45A as a pivotal regulator of autophagy. Comprehensive analyses, including Gene Ontology enrichment and protein-protein interaction network assessments, highlighted Cdkn1A as a significant downstream target of Gadd45A. Experimental validation confirmed Gadd45A's role in modulating Cdkn1A expression and autophagy levels in hippocampal cells. We also examined the effects of autophagy on hippocampal functions and proinflammatory cytokine secretion. Additionally, a murine model was employed to validate the importance of Gadd45A in neuroinflammation and AD pathology. RESULTS Our study identified 20 autophagy regulatory factors associated with AD, with Gadd45A emerging as a critical regulator. Experimental findings demonstrated that Gadd45A influences hippocampal cell fate by reducing Cdkn1A expression and suppressing autophagic activity. Comparisons between wild-type (WT) and Gadd45A knockout (Gadd45A-/-) mice revealed that Gadd45A-/- mice exhibited significant cognitive impairments, including deficits in working and spatial memory, increased Tau hyperphosphorylation, and elevated levels of kinases involved in Tau phosphorylation in the hippocampus. Additionally, Gadd45A-/- mice showed significant increases in pro-inflammatory cytokines and decreases autophagy markers in the brain. Neurotrophin levels and dendritic spine length were also reduced in Gadd45A-/- mice, likely contributing to the observed cognitive deficits. CONCLUSIONS These findings support the direct involvement of the Gadd45A gene in AD pathogenesis, and enhancing the expression of Gadd45A may represent a promising therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Norah A Althobaiti
- Biology Department, College of Science and Humanities, Shaqra University, Saudi Arabia
| | - Nouf S Al-Abbas
- Department of Biology, Jamoum University College, Umm Al-Qura University, 21955 Makkah, Saudi Arabia
| | - Ifat Alsharif
- Department of Biology, Jamoum University College, Umm Al-Qura University, 21955 Makkah, Saudi Arabia
| | - Aishah E Albalawi
- Faculty of Science, Department of Biology, University of Tabuk, Tabuk 47913, Saudi Arabia
| | - Amany I Almars
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ammar A Basabrain
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ayman Jafer
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sawsan Abd Ellatif
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), Alexandria 21934, Egypt
| | - Nuha M Bauthman
- Department of Obstetric & Gynecology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hailah M Almohaimeed
- Department of Basic Science, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mona H Soliman
- Botany and Microbiology Department, Faculty of Science, Cairo University, Giza 12613, Egypt; Biology Department, Faculty of Science, Taibah University, Al-Sharm, Yanbu El-Bahr, Yanbu 46429, Saudi Arabia.
| |
Collapse
|
4
|
Li Y, Lou N, Liu X, Zhuang X, Chen S. Exploring new mechanisms of Imeglimin in diabetes treatment: Amelioration of mitochondrial dysfunction. Biomed Pharmacother 2024; 175:116755. [PMID: 38772155 DOI: 10.1016/j.biopha.2024.116755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/23/2024] Open
Abstract
With the increasing prevalence of type 2 diabetes mellitus (T2DM), it has become critical to identify effective treatment strategies. In recent years, the novel oral hypoglycaemic drug Imeglimin has attracted much attention in the field of diabetes treatment. The mechanisms of its therapeutic action are complex and are not yet fully understood by current research. Current evidence suggests that pancreatic β-cells, liver, and skeletal muscle are the main organs in which Imeglimin lowers blood glucose levels and that it acts mainly by targeting mitochondrial function, thereby inhibiting hepatic gluconeogenesis, enhancing insulin sensitivity, promoting pancreatic β-cell function, and regulating energy metabolism. There is growing evidence that the drug also has a potentially volatile role in the treatment of diabetic complications, including metabolic cardiomyopathy, diabetic vasculopathy, and diabetic neuroinflammation. According to available clinical studies, its efficacy and safety profile are more evident than other hypoglycaemic agents, and it has synergistic effects when combined with other antidiabetic drugs, and also has potential in the treatment of T2DM-related complications. This review aims to shed light on the latest research progress in the treatment of T2DM with Imeglimin, thereby providing clinicians and researchers with the latest insights into Imeglimin as a viable option for the treatment of T2DM.
Collapse
Affiliation(s)
- Yilin Li
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China
| | - Nenngjun Lou
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China
| | - Xiaojing Liu
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China
| | - Xianghua Zhuang
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China; Multidisciplinary Innovation Center for Nephrology of the Second Hospital of Shandong University, Jinan 250033, China.
| | - Shihong Chen
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China; Multidisciplinary Innovation Center for Nephrology of the Second Hospital of Shandong University, Jinan 250033, China.
| |
Collapse
|
5
|
Griñán-Ferré C, Jarne-Ferrer J, Bellver-Sanchis A, Ribalta-Vilella M, Barroso E, Salvador JM, Jurado-Aguilar J, Palomer X, Vázquez-Carrera M, Pallàs M. Deletion of Gadd45a Expression in Mice Leads to Cognitive and Synaptic Impairment Associated with Alzheimer's Disease Hallmarks. Int J Mol Sci 2024; 25:2595. [PMID: 38473843 DOI: 10.3390/ijms25052595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/10/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
Gadd45 genes have been implicated in survival mechanisms, including apoptosis, autophagy, cell cycle arrest, and DNA repair, which are processes related to aging and life span. Here, we analyzed if the deletion of Gadd45a activates pathways involved in neurodegenerative disorders such as Alzheimer's Disease (AD). This study used wild-type (WT) and Gadd45a knockout (Gadd45a-/-) mice to evaluate AD progression. Behavioral tests showed that Gadd45a-/- mice presented lower working and spatial memory, pointing out an apparent cognitive impairment compared with WT animals, accompanied by an increase in Tau hyperphosphorylation and the levels of kinases involved in its phosphorylation in the hippocampus. Moreover, Gadd45a-/- animals significantly increased the brain's pro-inflammatory cytokines and modified autophagy markers. Notably, neurotrophins and the dendritic spine length of the neurons were reduced in Gadd45a-/- mice, which could contribute to the cognitive alterations observed in these animals. Overall, these findings demonstrate that the lack of the Gadd45a gene activates several pathways that exacerbate AD pathology, suggesting that promoting this protein's expression or function might be a promising therapeutic strategy to slow down AD progression.
Collapse
Affiliation(s)
- Christian Griñán-Ferré
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035 Barcelona, Spain
- Spanish Biomedical Research Center in Neurodegenerative Diseases (CIBERNED)-National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Júlia Jarne-Ferrer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035 Barcelona, Spain
| | - Aina Bellver-Sanchis
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035 Barcelona, Spain
| | - Marta Ribalta-Vilella
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035 Barcelona, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-National Institute of Health Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Jesús M Salvador
- Department of Immunology and Oncology, National Center for Biotechnology/CSIC, 28049 Madrid, Spain
| | - Javier Jurado-Aguilar
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-National Institute of Health Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-National Institute of Health Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-National Institute of Health Carlos III, 28029 Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Mercè Pallàs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, University of Barcelona, Avda. Joan XXIII 27, 08028 Barcelona, Spain
- Institute of Neurosciences of the University of Barcelona, University of Barcelona, 08035 Barcelona, Spain
- Spanish Biomedical Research Center in Neurodegenerative Diseases (CIBERNED)-National Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
6
|
Kato H, Iwashita K, Iwasa M, Kato S, Yamakage H, Suganami T, Tanaka M, Satoh-Asahara N. Imeglimin Exhibits Novel Anti-Inflammatory Effects on High-Glucose-Stimulated Mouse Microglia through ULK1-Mediated Suppression of the TXNIP-NLRP3 Axis. Cells 2024; 13:284. [PMID: 38334676 PMCID: PMC10854746 DOI: 10.3390/cells13030284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/10/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is an epidemiological risk factor for dementia and has been implicated in multifactorial pathologies, including neuroinflammation. In the present study, we aimed to elucidate the potential anti-inflammatory effects of imeglimin, a novel antidiabetic agent, on high-glucose (HG)-stimulated microglia. Mouse microglial BV2 cells were stimulated with HG in the presence or absence of imeglimin. We examined the effects of imeglimin on the levels of proinflammatory cytokines, intracellular reactive oxygen species (ROS), mitochondrial integrity, and components related to the inflammasome or autophagy pathways in these cells. Our results showed that imeglimin suppressed the HG-induced production of interleukin-1beta (IL-1β) by reducing the intracellular ROS levels, ameliorating mitochondrial dysfunction, and inhibiting the activation of the thioredoxin-interacting protein (TXNIP)-NOD-like receptor family pyrin domain containing 3 (NLRP3) axis. Moreover, the inhibitory effects of imeglimin on the TXNIP-NLRP3 axis depended on the imeglimin-induced activation of ULK1, which also exhibited novel anti-inflammatory effects without autophagy induction. These findings suggest that imeglimin exerted novel suppressive effects on HG-stimulated microglia through the ULK1-TXNIP-NLRP3 axis, and may, thereby, contribute to the development of innovative strategies to prevent T2DM-associated cognitive impairment.
Collapse
Affiliation(s)
- Hisashi Kato
- Department of Endocrinology, Metabolism and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, Kyoto 612-8555, Japan; (H.K.)
| | - Kaori Iwashita
- Department of Endocrinology, Metabolism and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, Kyoto 612-8555, Japan; (H.K.)
| | - Masayo Iwasa
- Department of Endocrinology, Metabolism and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, Kyoto 612-8555, Japan; (H.K.)
| | - Sayaka Kato
- Department of Endocrinology, Metabolism and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, Kyoto 612-8555, Japan; (H.K.)
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Hajime Yamakage
- Department of Endocrinology, Metabolism and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, Kyoto 612-8555, Japan; (H.K.)
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
- Department of Immunometabolism, Nagoya University Graduate School of Medicine, Nagoya 464-8601, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya 464-8601, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Nagoya University, Nagoya 464-8601, Japan
| | - Masashi Tanaka
- Department of Endocrinology, Metabolism and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, Kyoto 612-8555, Japan; (H.K.)
- Department of Rehabilitation, Health Science University, Minamitsuru-gun 401-0380, Japan
| | - Noriko Satoh-Asahara
- Department of Endocrinology, Metabolism and Hypertension Research, Clinical Research Institute, NHO Kyoto Medical Center, Kyoto 612-8555, Japan; (H.K.)
- Department of Metabolic Syndrome and Nutritional Science, Research Institute of Environmental Medicine, Nagoya University, Nagoya 466-8550, Japan
| |
Collapse
|
7
|
Zhen JB, Wang RB, Zhang YH, Sun F, Lin LH, Li ZX, Han Y, Lu YX. Effects of Trichinella spiralis and its serine protease inhibitors on autophagy of host small intestinal cells. Infect Immun 2023; 91:e0010323. [PMID: 37874164 PMCID: PMC10652968 DOI: 10.1128/iai.00103-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 10/25/2023] Open
Abstract
In eukaryotes, autophagy is induced as an innate defense mechanism against pathogenic microorganisms by self-degradation. Although trichinellosis is a foodborne zoonotic disease, there are few reports on the interplay between Trichinella spiralissurvival strategies and autophagy-mediated host defense. Therefore, this study focused on the association between T. spiralis and autophagy of host small intestinal cells. In this study, the autophagy-related indexes of host small intestinal cells after T. spiralis infection were detected using transmission electron microscopy, hematoxylin and eosin staining, immunohistochemistry, quantitative real-time polymerase chain reaction, and Western blotting. The results showed that autophagosomes and autolysosomes were formed in small intestinal cells, intestinal villi appeared edema, epithelial compactness was decreased, microtubule-associated protein 1A/1B-light chain 3B (LC3B) was expressed in lamina propria stromal cells of small intestine, and the expression of autophagy-related genes and proteins was changed significantly, indicating that T. spiralis induced autophagy of host small intestinal cells. Then, the effect of T. spiralis on autophagy-related pathways was explored by Western blotting. The results showed that the expression of autophagy-related pathway proteins was changed, indicating that T. spiralis regulated autophagy by affecting autophagy-related pathways. Finally, the roles of T. spiralis serine protease inhibitors (TsSPIs), such as T. spiralis Kazal-type SPI (TsKaSPI) and T. spiralis Serpin-type SPI (TsAdSPI), were further discussed in vitro and in vivo experiments. The results revealed that TsSPIs induced autophagy by influencing autophagy-related pathways, and TsAdSPI has more advantages. Overall, our results indicated that T. spiralis induced autophagy of host small intestinal cells, and its TsSPIs play an important role in enhancing autophagy flux by affecting autophagy-related pathways. These findings lay a foundation for further exploring the pathogenesis of intestinal dysfunction of host after T. spiralis infection, and also provide some experimental and theoretical basis for the prevention and treatment of trichinellosis.
Collapse
Affiliation(s)
- Jing-Bo Zhen
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Rui-Biao Wang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yu-Heng Zhang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Feng Sun
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Li-Hao Lin
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Zhi-Xin Li
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yang Han
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yi-Xin Lu
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| |
Collapse
|
8
|
Halle JL, Counts BR, Paez HG, Baumfalk DR, Zhang Q, Mohamed JS, Glazer ES, Puppa MJ, Smuder AJ, Alway SE, Carson JA. Recovery from FOLFOX chemotherapy-induced systemic and skeletal muscle metabolic dysfunction in mice. Am J Physiol Endocrinol Metab 2023; 325:E132-E151. [PMID: 37378624 PMCID: PMC10393342 DOI: 10.1152/ajpendo.00096.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/22/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
FOLFOX (5-fluorouracil, leucovorin, oxaliplatin) chemotherapy is used to treat colorectal cancer and can acutely induce metabolic dysfunction. However, the lasting effects on systemic and skeletal muscle metabolism after treatment cessation are poorly understood. Therefore, we investigated the acute and lasting effects of FOLFOX chemotherapy on systemic and skeletal muscle metabolism in mice. Direct effects of FOLFOX in cultured myotubes were also investigated. Male C57BL/6J mice completed four cycles (acute) of FOLFOX or PBS. Subsets were allowed to recover for 4 wk or 10 wk. Comprehensive Laboratory Animal Monitoring System (CLAMS) metabolic measurements were performed for 5 days before study endpoint. C2C12 myotubes were treated with FOLFOX for 24 hr. Acute FOLFOX attenuated body mass and body fat accretion independent of food intake or cage activity. Acute FOLFOX decreased blood glucose, oxygen consumption (V̇o2), carbon dioxide production (V̇co2), energy expenditure, and carbohydrate (CHO) oxidation. Deficits in V̇o2 and energy expenditure remained at 10 wk. CHO oxidation remained disrupted at 4 wk but returned to control levels after 10 wk. Acute FOLFOX reduced muscle COXIV enzyme activity, AMPK(T172), ULK1(S555), and LC3BII protein expression. Muscle LC3BII/I ratio was associated with altered CHO oxidation (r = 0.75, P = 0.03). In vitro, FOLFOX suppressed myotube AMPK(T172), ULK1(S555), and autophagy flux. Recovery for 4 wk normalized skeletal muscle AMPK and ULK1 phosphorylation. Our results provide evidence that FOLFOX disrupts systemic metabolism, which is not readily recoverable after treatment cessation. FOLFOX effects on skeletal muscle metabolic signaling did recover. Further investigations are warranted to prevent and treat FOLFOX-induced metabolic toxicities that negatively impact survival and life quality of patients with cancer.NEW & NOTEWORTHY The present study demonstrates that FOLFOX chemotherapy induces long-lasting deficits in systemic metabolism. Interestingly, FOLFOX modestly suppressed skeletal muscle AMPK and autophagy signaling in vivo and in vitro. The FOLFOX-induced suppression of muscle metabolic signaling recovered after treatment cessation, independent of systemic metabolic dysfunction. Future research should investigate if activating AMPK during treatment can prevent long-term toxicities to improve health and quality of life of patients with cancer and survivors.
Collapse
Affiliation(s)
- Jessica L Halle
- Integrative Muscle Biology Laboratory, Division of Regenerative and Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Brittany R Counts
- Integrative Muscle Biology Laboratory, Division of Regenerative and Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Hector G Paez
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Dryden R Baumfalk
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Quan Zhang
- Integrative Muscle Biology Laboratory, Division of Regenerative and Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Junaith S Mohamed
- Laboratory of Muscle and Nerve, Department of Diagnostic and Health Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Evan S Glazer
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Melissa J Puppa
- College of Health Sciences, The University of Memphis, Memphis, Tennessee, United States
| | - Ashley J Smuder
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, Florida, United States
| | - Stephen E Alway
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - James A Carson
- Integrative Muscle Biology Laboratory, Division of Regenerative and Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| |
Collapse
|
9
|
Zhang Y, Liu L, Hou X, Zhang Z, Zhou X, Gao W. Role of Autophagy Mediated by AMPK/DDiT4/mTOR Axis in HT22 Cells Under Oxygen and Glucose Deprivation/Reoxygenation. ACS OMEGA 2023; 8:9221-9229. [PMID: 36936290 PMCID: PMC10018509 DOI: 10.1021/acsomega.2c07280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Background: cerebral ischemia/reperfusion (I/R) injury is an important complication of ischemic stroke, and autophagy is one of the mechanisms of it. In this study, we aimed to determine the role and mechanism of autophagy in cerebral I/R injury. Methods: the oxygen and glucose deprivation/reoxygenation (OGD/R) method was used to model cerebral I/R injury in HT22 cells. CCK-8 and LDH were conducted to detect viability and damage of the cells, respectively. Apoptosis was measured by flow cytometry and Tunel staining. Autophagic vesicles of HT22 cells were assessed by transmission electron microscopy. Western blotting analysis was used to examine the protein expression involving AMPK/DDiT4/mTOR axis and autophagy-related proteins. 3-Methyladenine and rapamycin were, respectively, used to inhibit and activate autophagy, compound C and AICAR acted as AMPK inhibitor and activator, respectively, and were used to control the starting link of AMPK/DDiT4/mTOR axis. Results: autophagy was activated in HT22 cells after OGD/R was characterized by an increased number of autophagic vesicles, the expression of Beclin1 and LC3II/LC3I, and a decrease in the expression of P62. Rapamycin could increase the viability, reduce LDH leakage rate, and alleviate cell apoptosis in OGD/R cells by activating autophagy. 3-Methyladenine played an opposite role to rapamycin in OGD/R cells. The expression of DDiT4 and the ratio of p-AMPK/AMPK were increased after OGD/R in HT22 cells. While the ratio of p-mTOR/mTOR was reduced by OGD/R, AICAR effectively increased the number of autophagic vesicles, improved viability, reduced LDH leakage rate, and alleviated apoptosis in HT22 cells which suffered OGD/R. However, the effects of compound C in OGD/R HT22 cells were opposite to that of AICAR. Conclusions: autophagy is activated after OGD/R; autophagy activator rapamycin significantly enhanced the protective effect of autophagy on cells of OGD/R. AMPK/DDiT4/mTOR axis is an important pathway to activate autophagy, and AMPK/DDiT4/mTOR-mediated autophagy significantly alleviates cell damage caused by OGD/R.
Collapse
Affiliation(s)
| | | | | | | | | | - Weijuan Gao
- . Phone: 86 311 89926007. Fax: (86) 311 89926000
| |
Collapse
|
10
|
Wu S, Zhao K, Wang J, Liu N, Nie K, Qi L, Xia L. Recent advances of tanshinone in regulating autophagy for medicinal research. Front Pharmacol 2023; 13:1059360. [PMID: 36712689 PMCID: PMC9877309 DOI: 10.3389/fphar.2022.1059360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/19/2022] [Indexed: 01/15/2023] Open
Abstract
Initially described as an ancient and highly conserved catabolic biofunction, autophagy plays a significant role in disease pathogenesis and progression. As the bioactive ingredient of Salvia miltiorrhiza, tanshinone has recently shown profound effects in alleviating and treating various diseases by regulating autophagy. However, compared to the remarkable achievements in the known pharmacological effects of this traditional Chinese medicine, there is a lack of a concise and comprehensive review deciphering the mechanism by which tanshinone regulates autophagy for medicinal research. In this context, we concisely review the advances of tanshinone in regulating autophagy for medicinal research, including human cancer, the nervous system, and cardiovascular diseases. The pharmacological effects of tanshinone targeting autophagy involve the regulation of autophagy-related proteins, such as Beclin-1, LC3-II, P62, ULK1, Bax, ATG3, ATG5, ATG7, ATG9, and ATG12; the regulation of the PI3K/Akt/mTOR, MEK/ERK/mTOR, Beclin-1-related, and AMPK-related signaling pathways; the accumulation of reactive oxygen species (ROS); and the activation of AMPK. Notably, we found that tanshinone played a dual role in human cancers in an autophagic manner, which may provide a new avenue for potential clinical application. In brief, these findings on autophagic tanshinone and its derivatives provide a new clue for expediting medicinal research related to tanshinone compounds and autophagy.
Collapse
Affiliation(s)
- Sha Wu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of Traditional Chinese Medicine Regimen and Health of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kui Zhao
- College of Materials Science and Engineering, Southwest Forestry University, Kunming, Yunnan, China
| | - Jie Wang
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of Traditional Chinese Medicine Regimen and Health of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nannan Liu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of Traditional Chinese Medicine Regimen and Health of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kaidi Nie
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of Traditional Chinese Medicine Regimen and Health of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Luming Qi
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of Traditional Chinese Medicine Regimen and Health of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lina Xia
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Administration of Traditional Chinese Medicine Key Laboratory of Traditional Chinese Medicine Regimen and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of Traditional Chinese Medicine Regimen and Health of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
11
|
Ikutama R, Peng G, Tsukamoto S, Umehara Y, Trujillo-Paez JV, Yue H, Nguyen HLT, Takahashi M, Kageyama S, Komatsu M, Okumura K, Ogawa H, Ikeda S, Niyonsaba F. Cathelicidin LL-37 Activates Human Keratinocyte Autophagy through the P2X₇, Mechanistic Target of Rapamycin, and MAPK Pathways. J Invest Dermatol 2022; 143:751-761.e7. [PMID: 36455652 DOI: 10.1016/j.jid.2022.10.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/30/2022] [Accepted: 10/18/2022] [Indexed: 11/29/2022]
Abstract
Human cathelicidin LL-37 is a multifunctional antimicrobial peptide that exhibits antimicrobial and immunomodulatory activities. LL-37 regulates skin barrier function and was recently reported to activate autophagy in macrophages. Because autophagy deficiency is associated with skin diseases characterized by a dysfunctional epidermal barrier, we hypothesized that LL-37 might regulate the skin barrier through autophagy modulation. We showed that LL-37 activated autophagy in human keratinocytes and three-dimensional skin equivalent models as indicated by increases in LC3 puncta formation, decreases in p62, and autophagosome and autolysosome formation. LL-37‒induced autophagy was suppressed by P2X7 receptor, adenosine monophosphate‒activated protein kinase, and unc-51-like kinase 1 inhibitors, suggesting that the P2X7, adenosine monophosphate‒activated protein kinase, and unc-51-like kinase 1 pathways are involved. Moreover, LL-37 enhanced the phosphorylation of adenosine monophosphate‒activated protein kinase and unc-51-like kinase 1. In addition, LL-37‒mediated autophagy involves the mechanistic target of rapamycin and MAPK pathways. Interestingly, the LL-37‒induced distribution of tight junction proteins and improvement in the tight junction barrier were inhibited in autophagy-deficient keratinocytes and keratinocytes and skin models treated with autophagy inhibitors, indicating that the LL-37‒mediated tight junction barrier is associated with autophagy activation. Collectively, these findings suggest that LL-37 is a potential therapeutic target for skin diseases characterized by dysfunctional autophagy and skin barriers.
Collapse
Affiliation(s)
- Risa Ikutama
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ge Peng
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Saya Tsukamoto
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshie Umehara
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | | | - Hainan Yue
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hai Le Thanh Nguyen
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Miho Takahashi
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shun Kageyama
- Department of Physiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigaku Ikeda
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - François Niyonsaba
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Faculty of International Liberal Arts, Juntendo University, Tokyo, Japan.
| |
Collapse
|
12
|
Jia L, Lin XR, Guo WY, Huang M, Zhao Y, Zhang YS, Li J. Salvia chinensia Benth induces autophagy in esophageal cancer cells via AMPK/ULK1 signaling pathway. Front Pharmacol 2022; 13:995344. [PMID: 36120378 PMCID: PMC9478658 DOI: 10.3389/fphar.2022.995344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/08/2022] [Indexed: 12/24/2022] Open
Abstract
Salvia chinensia Benth (Shijianchuan in Chinese, SJC) has been used as a traditional anti-cancer herb. SJC showed good anti-esophageal cancer efficacy based on our clinical application. However, the current research on SJC is minimal, and its anti-cancer effect lacks scientific certification. This study aims to clarify the inhibitory effect of SJC on esophageal cancer and explore its underlying mechanism. Q-Orbitrap high-resolution LC/MS was used to identify the primary chemical constituents in SJC. Cell proliferation and colony formation assays showed that SJC could effectively inhibit the growth of esophageal tumor cells in vitro. To clarify its mechanism of action, proteomic and bioinformatic analyses were carried out by combining tandem mass labeling and two-dimensional liquid chromatography-mass spectrometry (LC-MS). Data are available via ProteomeXchange with identifier PXD035823. The results indicated that SJC could activate AMPK signaling pathway and effectively promote autophagy in esophageal cancer cells. Therefore, we further used western blotting to confirm that SJC activated autophagy in esophageal cancer cells through the AMPK/ULK1 signaling pathway. The results showed that P-AMPK and P-ULK1 were significantly up-regulated after the treatment with SJC. The ratio of autophagosomes marker proteins LC3II/I was significantly increased. In addition, the expression of the autophagy substrate protein P62 decreased with the degradation of autophagosomes. Using lentiviral transfection of fluorescent label SensGFP-StubRFP-LC3 protein and revalidation of LC3 expression before and after administration by laser confocal microscopy. Compared with the control group, the fluorescence expression of the SJC group was significantly enhanced, indicating that it promoted autophagy in esophageal cancer cells. Cell morphology and the formation of autophagosomes were observed by transmission electron microscopy. Our study shows that the tumor suppressor effect of SJC is related to promoting autophagy in esophageal tumor cells via the AMPK/ULK1 signaling pathway.
Collapse
Affiliation(s)
- Lei Jia
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xin-Rong Lin
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wen-Yan Guo
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ming Huang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yang Zhao
- Department of Traditional Chinese Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yu-Shuang Zhang
- Department of Traditional Chinese Medicine, Tumor Hospital of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jing Li
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Traditional Chinese Medicine, Tumor Hospital of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- *Correspondence: Jing Li,
| |
Collapse
|
13
|
Chen C, Shi Q, Xiao K, Zhou W, Gao C, Gao L, Han J, Wang J, Dong X. Activation of Innate Immunity and Autophagy in Brain Tissues with Prion Disease and Degradation of Abnormal PrPs in Cells - China's Studies. China CDC Wkly 2022; 4:735-740. [PMID: 36285112 PMCID: PMC9547741 DOI: 10.46234/ccdcw2022.153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/08/2022] [Indexed: 11/14/2022] Open
Abstract
Unlike infectious diseases caused by conventional microbes, there are no detectable specific humoral or cellular immunoresponses to prion infection. However, extensive and active gliosis is observable in affected brain regions along with significant deposits of scrapie-like prion protein (PrPSc). Here, we summarize our studies of vibrant activation of host non-specific immune components and autophagy in the microenvironment of prion infected brains. Activation of the brain's innate immunity and autophagy upon prion infection reflect non-specific host defense systems attempt to dispose of accumulated prions. Vibrant elevation of neuroinflammation leads to neuron injury.
Collapse
Affiliation(s)
- Cao Chen
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan City, Hubei Province, China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,China Academy of Chinese Medical Sciences, Beijing, China
| | - Kang Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wei Zhou
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chen Gao
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liping Gao
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jun Han
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jichun Wang
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,Division of Science and Technology, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China,Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan City, Hubei Province, China,China Academy of Chinese Medical Sciences, Beijing, China,Shanghai Institute of Infectious Disease and Biosafety, Shanghai Municipality, China,Xiaoping Dong,
| |
Collapse
|
14
|
Function and regulation of ULK1: From physiology to pathology. Gene 2022; 840:146772. [PMID: 35905845 DOI: 10.1016/j.gene.2022.146772] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/03/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022]
Abstract
The expression of ULK1, a core protein of autophagy, is closely related to autophagic activity. Numerous studies have shown that pathological abnormal expression of ULK1 is associated with various human diseases such as neurological disorders, infections, cardiovascular diseases, liver diseases and cancers. In addition, new advances in the regulation of ULK1 have been identified. Furthermore, targeting ULK1 as a therapeutic strategy for diseases is gaining attention as new corresponding activators or inhibitors are being developed. In this review, we describe the structure and regulation of ULK1 as well as the current targeted activators and inhibitors. Moreover, we highlight the pathological disorders of ULK1 expression and its critical role in human diseases.
Collapse
|
15
|
Prolonged proteasome inhibition antagonizes TGFβ1-dependent signalling by promoting the lysosomal-targeting of TGFβ receptors. Cell Signal 2022; 98:110414. [PMID: 35901932 DOI: 10.1016/j.cellsig.2022.110414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/05/2022] [Accepted: 07/15/2022] [Indexed: 01/18/2023]
Abstract
Impairing autophagy disrupts transforming growth factor beta 1 (TGFβ1) signalling and epithelial-mesenchymal transition (EMT) in non-small cell lung cancer (NSCLC). Since autophagy and proteasome-mediated degradation are interdependent, we investigated how prolonged downregulation of proteasomal catalytic activity affected TGFβ1-dependent signalling and EMT. Proteasome-dependent degradation was inhibited in A549 and H1299 NSCLC cells using MG132 and lactacystin, which are reversible and irreversible proteasome inhibitors, respectively. We observed that inhibiting proteasomal activity for 24 h decreased TGFβ-dependent nuclear accumulation of Smad2/3. Time course studies were then carried out to characterize the time frame of this observation. Short-term (< 8 h) proteasome inhibition resulted in increased receptor regulated Smad (R-Smad) phosphorylation and steady-state TGFβ receptor type II (TGFβRII) levels. However, prolonged (8-24 h) proteasome inhibition decreased TGFβ1-dependent R-Smad phosphorylation and steady-state TGFβRI and TGFβRII levels. Furthermore, proteasome inhibition blunted TGFβ-dependent E- to N-Cadherin shift, stress fiber formation, and increased cellular apoptosis via the TAK-1-TRAF6-p38 MAPK pathway. Interestingly, proteasome inhibition also increased autophagic flux, steady-state microtubule-associated protein light chain 3B-II and active uncoordinated 51-like autophagy activating kinase 1 levels, and co-localization of lysosomes with autophagy cargo proteins and autophagy-related proteins. Finally, we observed that proteasome inhibition increased TGFβRII endocytosis and trafficking to lysosomes and we conclude that prolonged proteasome inhibition disrupts TGFβ signalling outcomes through altered TGFβ receptor trafficking.
Collapse
|
16
|
Katz A, Gonen M, Shahar Y, Roichman A, Lerrer B, Cohen HY. Hypothalamus-Muscle Parallel Induction of Metabolic Pathways Following Physical Exercise. Front Neurosci 2022; 16:897005. [PMID: 35928013 PMCID: PMC9344923 DOI: 10.3389/fnins.2022.897005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
The modern lifestyle requires less physical activity and skills during our daily routine, leading to multiple pathologies related to physical disabilities and energy accessibility. Thus, exploring the mechanisms underlying the metabolic regulation of exercise is crucial. Here, we characterized the effect of forced and voluntary endurance exercises on three key metabolic signaling pathways, sirtuins, AMPK, and mTOR, across several metabolic tissues in mice: brain, muscles, and liver. Both voluntary and forced exercises induced AMPK with higher intensity in the first. The comparison between those metabolic tissues revealed that the hypothalamus and the hippocampus, two brain parts, showed different metabolic signaling activities. Strikingly, despite the major differences in the physiology of muscles and hypothalamic tissues, the hypothalamus replicates the metabolic response of the muscle in response to physical exercise. Specifically, muscles and hypothalamic tissues showed an increase and a decrease in AMPK and mTOR signaling, respectively. Overall, this study reveals new insight into the relation between the hypothalamus and muscles, which enhances the coordination within the muscle-brain axis and potentially improves the systemic response to physical activity performance and delaying health inactivity disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Haim Yosef Cohen
- The Mina & Everard Goodman Faculty of Life Sciences, The Sagol Center for Healthy Human Longevity, Bar-Ilan University, Ramat-Gan, Israel
| |
Collapse
|
17
|
Kang S, Dai A, Wang H, Ding PH. Interaction Between Autophagy and Porphyromonas gingivalis-Induced Inflammation. Front Cell Infect Microbiol 2022; 12:892610. [PMID: 35846745 PMCID: PMC9283780 DOI: 10.3389/fcimb.2022.892610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Autophagy is an immune homeostasis process induced by multiple intracellular and extracellular signals. Inflammation is a protective response to harmful stimuli such as pathogen microbial infection and body tissue damage. Porphyromonas gingivalis infection elicits both autophagy and inflammation, and dysregulation of autophagy and inflammation promotes pathology. This review focuses on the interaction between autophagy and inflammation caused by Porphyromonas gingivalis infection, aiming to elaborate on the possible mechanism involved in the interaction.
Collapse
|
18
|
Wang G, Liu Y, Zhu X, Lin K, Li M, Wu Z, Zhang R, Zheng Q, Li D, An T. Knockdown of miRNA-134-5p rescues dendritic deficits by promoting AMPK-mediated mitophagy in a mouse model of depression. Neuropharmacology 2022; 214:109154. [PMID: 35659969 DOI: 10.1016/j.neuropharm.2022.109154] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/23/2022] [Accepted: 05/29/2022] [Indexed: 12/28/2022]
Abstract
Neuronal dendrites and dendritic spines are essential for normal synaptic transmission and may be critically involved in the pathophysiology of various neurological disorders, including depression. Emerging data supports the role of mitochondria in dendritic protrusions in modulating the development and morphological plasticity of spines. Mitophagy, a mitochondria-specific form of autophagy, is the fundamental process of clearing damaged mitochondria to maintain cellular homeostasis. As a brain-specific microRNA, miR-134 is localized to the synaptodendritic compartment of hippocampal neurons and negatively regulates the development of dendritic spines. However, the role of miR-134 in mitophagy related to dendritic deficits in the pathophysiology of depression remains unclear. In this study, we showed that miR-134-5p knockdown abrogated depressive-like behavioral symptoms and corrected aberrant spine morphology in hippocampal neurons of chronic unpredictable mild stress (CUMS) mice. Moreover, knockdown of miR-134-5p triggered autophagy in dendrites, improved mitochondrial impairment, and induced the generation of autophagosomes in the hippocampus of CUMS mice. We further found that AMP-activated protein kinase (AMPK), which mediates the impairment of defective mitochondria via mitophagy, can bind directly to miR-134-5p and is negatively regulated by this miRNA. This study demonstrates that miR-134-5p exerts an enormous effect on dendritic deficits by promoting AMPK-mediated mitophagy and provides a potential new target for antidepressant drug research and development.
Collapse
Affiliation(s)
- Guoli Wang
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China; Collaborative Innovation Platform for Modernization and Industrialization of Regional Characteristic Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China
| | - Ying Liu
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China
| | - Xuejie Zhu
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China
| | - Kehao Lin
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China
| | - Mingkai Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China
| | - Zhenke Wu
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China
| | - Ronghua Zhang
- School of Pharmacy, Jinan University, Guangzhou, Guangdong, PR China
| | - Qiusheng Zheng
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China; Collaborative Innovation Platform for Modernization and Industrialization of Regional Characteristic Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China.
| | - Defang Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China; Collaborative Innovation Platform for Modernization and Industrialization of Regional Characteristic Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China.
| | - Tianyue An
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China; Collaborative Innovation Platform for Modernization and Industrialization of Regional Characteristic Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, PR China.
| |
Collapse
|
19
|
Belkozhayev AM, Al-Yozbaki M, George A, Niyazova RY, Sharipov KO, Byrne LJ, Wilson CM. Extracellular Vesicles, Stem Cells and the Role of miRNAs in Neurodegeneration. Curr Neuropharmacol 2022; 20:1450-1478. [PMID: 34414870 PMCID: PMC9881087 DOI: 10.2174/1570159x19666210817150141] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/16/2021] [Accepted: 06/14/2021] [Indexed: 11/22/2022] Open
Abstract
There are different modalities of intercellular communication governed by cellular homeostasis. In this review, we will explore one of these forms of communication called extracellular vesicles (EVs). These vesicles are released by all cells in the body and are heterogeneous in nature. The primary function of EVs is to share information through their cargo consisting of proteins, lipids and nucleic acids (mRNA, miRNA, dsDNA etc.) with other cells, which have a direct consequence on their microenvironment. We will focus on the role of EVs of mesenchymal stem cells (MSCs) in the nervous system and how these participate in intercellular communication to maintain physiological function and provide neuroprotection. However, deregulation of this same communication system could play a role in several neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, multiple sclerosis, prion disease and Huntington's disease. The release of EVs from a cell provides crucial information to what is happening inside the cell and thus could be used in diagnostics and therapy. We will discuss and explore new avenues for the clinical applications of using engineered MSC-EVs and their potential therapeutic benefit in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Ayaz M. Belkozhayev
- Al-Farabi Kazakh National University, Faculty of Biology and Biotechnology, Almaty, Republic of Kazakhstan
- Structural and Functional Genomics Laboratory of M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Republic of Kazakhstan
| | - Minnatallah Al-Yozbaki
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| | - Alex George
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
- Jubilee Centre for Medical Research, Jubilee Mission Medical College & Research Institute, Thrissur, Kerala, India
| | - Raigul Ye Niyazova
- Al-Farabi Kazakh National University, Faculty of Biology and Biotechnology, Almaty, Republic of Kazakhstan
| | - Kamalidin O. Sharipov
- Structural and Functional Genomics Laboratory of M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Republic of Kazakhstan
| | - Lee J. Byrne
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| | - Cornelia M. Wilson
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| |
Collapse
|
20
|
Huang JS, Li HJ, Guo ZX, Zhang JD, Wang WZ, Wang ZL, Amenyogbe E, Chen G. Identification and expression analysis of cobia (Rachycentron canadum) liver-related miRNAs under hypoxia stress. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:1951-1967. [PMID: 34633578 DOI: 10.1007/s10695-021-01017-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 09/09/2021] [Indexed: 06/13/2023]
Abstract
At present, due to the influence of global warming, seasonal change, diurnal variation, and eutrophication of the water body, hypoxia has become one of the major factors limiting the stable development of cobia (Rachycentron canadum) culture. In this study, the miRNAs involved in hypoxia stress were screened, and the target genes of miRNAs were annotated and analyzed. The results showed that a total of 184 conservative microRNA (miRNA) and 121 newly predicted miRNA were obtained by sequencing the liver of control (C) and hypoxic (dissolved oxygen, DO (2.64 ± 0.25) mg/L; 3 h) (S) groups. The pathways involved in energy metabolism included starch and sucrose metabolism (ko00500), glycosaminoglycan degradation (ko00531), and galactose metabolism (ko00052). The results indicate that the body maintains physiological activities by regulating some important pathways at the transcriptional level under hypoxia stress, such as the conversion of aerobic metabolism and anaerobic metabolism, the reduction of energy consumption, and the promotion of red blood cell proliferation to maintain the homeostasis of the body.
Collapse
Affiliation(s)
- Jian-Sheng Huang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, 524025, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524088, China
| | - Hong-Juan Li
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Zhi-Xiong Guo
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Jian-Dong Zhang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, 524025, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524088, China
| | - Wei-Zheng Wang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Zhong-Liang Wang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Eric Amenyogbe
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China.
| | - Gang Chen
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China.
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, 524025, China.
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524088, China.
| |
Collapse
|
21
|
Molecular mechanisms of mammalian autophagy. Biochem J 2021; 478:3395-3421. [PMID: 34554214 DOI: 10.1042/bcj20210314] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023]
Abstract
The ubiquitin-proteasome pathway (UPP) and autophagy play integral roles in cellular homeostasis. As part of their normal life cycle, most proteins undergo ubiquitination for some form of redistribution, localization and/or functional modulation. However, ubiquitination is also important to the UPP and several autophagic processes. The UPP is initiated after specific lysine residues of short-lived, damaged or misfolded proteins are conjugated to ubiquitin, which targets these proteins to proteasomes. Autophagy is the endosomal/lysosomal-dependent degradation of organelles, invading microbes, zymogen granules and macromolecules such as protein, carbohydrates and lipids. Autophagy can be broadly separated into three distinct subtypes termed microautophagy, chaperone-mediated autophagy and macroautophagy. Although autophagy was once thought of as non-selective bulk degradation, advancements in the field have led to the discovery of several selective forms of autophagy. Here, we focus on the mechanisms of primary and selective mammalian autophagy pathways and highlight the current knowledge gaps in these molecular pathways.
Collapse
|
22
|
Akhshi T, Trimble WS. A non-canonical Hedgehog pathway initiates ciliogenesis and autophagy. J Cell Biol 2021; 220:211568. [PMID: 33258871 PMCID: PMC7714386 DOI: 10.1083/jcb.202004179] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/19/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Primary cilia function as critical signaling hubs whose absence leads to severe disorders collectively known as ciliopathies; our knowledge of ciliogenesis remains limited. We show that Smo induces ciliogenesis through two distinct yet essential noncanonical Hh pathways in several cell types, including neurons. Surprisingly, ligand activation of Smo induces autophagy via an LKB1-AMPK axis to remove the satellite pool of OFD1. This is required, but not sufficient, for ciliogenesis. Additionally, Smo activates the Gαi-LGN-NuMA-dynein axis, causing accumulation of a portion of OFD1 at centrioles in early ciliogenesis. Both pathways are critical for redistribution of BBS4 from satellites to centrioles, which is also mediated by OFD1 centriolar translocation. Notably, different Smo agonists, which activate Smo distinctly, activate one or the other of these pathways; only in combination they recapitulate the activity of Hh ligand. These studies provide new insight into physiological stimuli (Hh) that activate autophagy and promote ciliogenesis and introduce a novel role for the Gαi-LGN-NuMA-dynein complex in this process.
Collapse
Affiliation(s)
- Tara Akhshi
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada
| | - William S Trimble
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada
| |
Collapse
|
23
|
Kim SY, Hwangbo H, Kim MY, Ji SY, Lee H, Kim GY, Kwon CY, Leem SH, Hong SH, Cheong J, Choi YH. Coptisine induces autophagic cell death through down-regulation of PI3K/Akt/mTOR signaling pathway and up-regulation of ROS-mediated mitochondrial dysfunction in hepatocellular carcinoma Hep3B cells. Arch Biochem Biophys 2020; 697:108688. [PMID: 33227289 DOI: 10.1016/j.abb.2020.108688] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/06/2020] [Accepted: 11/15/2020] [Indexed: 02/07/2023]
Abstract
Coptisine is isoquinoline alkaloid derived from Coptidis Rhizoma and is known to have potential anti-cancer activity toward various carcinomas. Targeting autophagy is one of the main approaches for cancer therapy, but whether the anti-cancer efficacy of coptisine involves autophagy is still unclear. Therefore, this study investigated the effect of coptisine on autophagy in hepatocellular carcinoma (HCC) Hep3B cells, and identified the underlying mechanism. Our results showed that coptisine increased cytotoxicity and autophagic vacuoles in a concentration-dependent manner. Furthermore, the expressions of light chain 3 (LC3)-I/II, Beclin-1 and autophagy genes were markedly increased by coptisine, while the expression of p62 decreased. In addition, we found that pretreatment with bafilomycin A1, an inhibitor of autophagosome-lysosome fusion, markedly reduced coptisine-mediated autophagic cell death, but 3-methyladenine, an inhibitor for autophagosome formation did not. Moreover, our results showed that although coptisine up-regulated AMP-activated protein kinase (AMPK) that partially induced LC3-I/II, coptisine-mediated AMPK signaling did not directly regulate autophagic cell death. Additionally, we found that coptisine suppressed the phosphorylation of phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR), and this effect was notably enhanced by PI3K inhibitor LY294002. Meanwhile, coptisine significantly increased both the production of mitochondrial reactive oxygen species (ROS) and the recruitment of mitophagy-regulated proteins to mitochondria. Furthermore, N-acetylcysteine, a potential ROS scavenger, substantially suppressed the expression of mitophagy-regulated proteins and LC3 puncta by coptisine. Overall, our results demonstrate that coptisine-mediated autophagic cell death was regulated by PI3K/Akt/mTOR signaling and mitochondrial ROS production associated with mitochondrial dysfunction. Taken together, these findings suggest that coptisine exerts its anti-cancer effects through induction of autophagy in HCC Hep3B cells.
Collapse
Affiliation(s)
- So Young Kim
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea; Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, 46241, Republic of Korea
| | - Hyun Hwangbo
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea; Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, 46241, Republic of Korea
| | - Min Yeong Kim
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea
| | - Seon Yeong Ji
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea
| | - Hyesook Lee
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | - Chan-Young Kwon
- Department of Oriental Neuropsychiatry, Dong-eui University College of Korean Medicine, Busan, 47227, Republic of Korea
| | - Sun-Hee Leem
- Department of Biological Science, College of Natural Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Su Hyun Hong
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea
| | - JaeHun Cheong
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, 46241, Republic of Korea.
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea.
| |
Collapse
|
24
|
Kwon I, Jang Y, Lee Y. Endurance Exercise-Induced Autophagy/Mitophagy Coincides with a Reinforced Anabolic State and Increased Mitochondrial Turnover in the Cortex of Young Male Mouse Brain. J Mol Neurosci 2020; 71:42-54. [PMID: 32535714 DOI: 10.1007/s12031-020-01624-6] [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] [Received: 11/10/2019] [Accepted: 06/08/2020] [Indexed: 01/07/2023]
Abstract
Autophagy/mitophagy, a cellular catabolic process necessary for sustaining normal cellular function, has emerged as a potential therapeutic strategy against numerous obstinate diseases. In this regard, endurance exercise (EXE)-induced autophagy/mitophagy (EIAM) has been considered as a potential health-enriching factor in various tissues including the brain; however, underlying mechanisms of EIAM in the brain has not been fully defined yet. This study investigated the molecular signaling nexus of EIAM pathways in the cortex of the brain. C57BL/6 young male mice were randomly assigned to a control group (CON, n = 12) and an endurance exercise group (EXE, n = 12). Our data demonstrated that exercise-induced autophagy coincided with an enhanced anabolic state (p-AKT, p-mTOR, and p-p70S6K); furthermore, mitophagy concurred with enhanced mitochondrial turnover: increases in both fission (DRP1, BNIP3, and PINK1) and fusion (OPA1 and MFN2) proteins. In addition, neither oxidative stress nor sirtuins (SIRT) 1 and 3 were associated with EIAM; instead, the activation of AMPK as well as a JNK-BCL2 axis was linked to EIAM promotion. Collectively, our results demonstrated that EXE-induced anabolic enrichment did not hinder autophagy/mitophagy and that the concurrent augmentation of mitochondrial fusion and fusion process contributed to sustaining mitophagy in the cortex of the brain. Our findings suggest that the EXE-induced concomitant potentiation of the catabolic and anabolic state is a unique molecular mechanism that simultaneously contributes to recycling and rebuilding the cellular structure, leading to upholding healthy cellular environment. Thus, the current study provides a novel autophagy/mitophagy mechanism, from which groundbreaking pharmacological strategies of autophagy can be developed.
Collapse
Affiliation(s)
- Insu Kwon
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, 11000 University Pkwy, Bldg.72, Pensacola, FL, 32514, USA
| | - Yongchul Jang
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, 11000 University Pkwy, Bldg.72, Pensacola, FL, 32514, USA
| | - Youngil Lee
- Molecular and Cellular Exercise Physiology Laboratory, Department of Movement Sciences and Health, Usha Kundu, MD College of Health, University of West Florida, 11000 University Pkwy, Bldg.72, Pensacola, FL, 32514, USA.
| |
Collapse
|
25
|
Wilkaniec A, Cieślik M, Murawska E, Babiec L, Gąssowska-Dobrowolska M, Pałasz E, Jęśko H, Adamczyk A. P2X7 Receptor is Involved in Mitochondrial Dysfunction Induced by Extracellular Alpha Synuclein in Neuroblastoma SH-SY5Y Cells. Int J Mol Sci 2020; 21:ijms21113959. [PMID: 32486485 PMCID: PMC7312811 DOI: 10.3390/ijms21113959] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022] Open
Abstract
The purinergic P2X7 receptor (P2X7R) belongs to a family of trimeric ion channels that are gated by extracellular adenosine 5′-triphosphate (ATP). Several studies have pointed to a role of P2X7R-dependent signalling in Parkinson's disease (PD)-related neurodegeneration. The pathology of (PD) is characterized by the formation of insoluble alpha-synuclein (α-Syn) aggregates—Lewy bodies, but the mechanisms underlying α-Syn-induced dopaminergic cell death are still partially unclear. Our previous studies indicate that extracellular α-Syn directly interact with neuronal P2X7R and induces intracellular free calcium mobilization in neuronal cells. The main objective of this study was to examine the involvement of P2X7R receptor in α-Syn-induced mitochondrial dysfunction and cell death. We found that P2X7R stimulation is responsible for α-Syn-induced oxidative stress and activation of the molecular pathways of programmed cell death. Exogenous α-Syn treatment led to P2X7R-dependent decrease in mitochondrial membrane potential as well as elevation of mitochondrial ROS production resulting in breakdown of cellular energy production. Moreover, P2X7R-dependent deregulation of AMP-activated protein kinase as well as decrease in parkin protein level could be responsible for α-Syn-induced mitophagy impairment and accumulation of dysfunctional mitochondria. P2X7R might be putative pharmacological targets in molecular mechanism of extracellular α-Syn toxicity.
Collapse
Affiliation(s)
- Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences Pawińskiego 5, 02-106 Warsaw, Poland; (M.C.); (L.B.); (M.G.-D.); (E.P.); (H.J.); (A.A.)
- Correspondence: ; Tel.: +48-22-608-66-00; Fax: +48-22-608-64-13
| | - Magdalena Cieślik
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences Pawińskiego 5, 02-106 Warsaw, Poland; (M.C.); (L.B.); (M.G.-D.); (E.P.); (H.J.); (A.A.)
| | - Emilia Murawska
- Department of Applied Microbiology, Institute of Microbiology, Warsaw University, Miecznikowa 1 Street, 02-096 Warsaw, Poland;
| | - Lidia Babiec
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences Pawińskiego 5, 02-106 Warsaw, Poland; (M.C.); (L.B.); (M.G.-D.); (E.P.); (H.J.); (A.A.)
| | - Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences Pawińskiego 5, 02-106 Warsaw, Poland; (M.C.); (L.B.); (M.G.-D.); (E.P.); (H.J.); (A.A.)
| | - Ewelina Pałasz
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences Pawińskiego 5, 02-106 Warsaw, Poland; (M.C.); (L.B.); (M.G.-D.); (E.P.); (H.J.); (A.A.)
| | - Henryk Jęśko
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences Pawińskiego 5, 02-106 Warsaw, Poland; (M.C.); (L.B.); (M.G.-D.); (E.P.); (H.J.); (A.A.)
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences Pawińskiego 5, 02-106 Warsaw, Poland; (M.C.); (L.B.); (M.G.-D.); (E.P.); (H.J.); (A.A.)
| |
Collapse
|
26
|
Xiang H, Zhang J, Lin C, Zhang L, Liu B, Ouyang L. Targeting autophagy-related protein kinases for potential therapeutic purpose. Acta Pharm Sin B 2020; 10:569-581. [PMID: 32322463 PMCID: PMC7161711 DOI: 10.1016/j.apsb.2019.10.003] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/06/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023] Open
Abstract
Autophagy, defined as a scavenging process of protein aggregates and damaged organelles mediated by lysosomes, plays a significant role in the quality control of macromolecules and organelles. Since protein kinases are integral to the autophagy process, it is critically important to understand the role of kinases in autophagic regulation. At present, intervention of autophagic processes by small-molecule modulators targeting specific kinases has becoming a reasonable and prevalent strategy for treating several varieties of human disease, especially cancer. In this review, we describe the role of some autophagy-related kinase targets and kinase-mediated phosphorylation mechanisms in autophagy regulation. We also summarize the small-molecule kinase inhibitors/activators of these targets, highlighting the opportunities of these new therapeutic agents.
Collapse
Key Words
- 4E-BP1, eukaryotic translation initiation factor 4E-binding protein
- AKT1, AKT serine/threonine kinase 1
- AMBRA1, autophagy/beclin-1 regulator 1
- AMPK, AMP-activated protein kinase
- ARF, auxin response factor gene
- ATG, autophagy-related protein
- Autophagy
- Autophagy-related kinase
- CaMKK2, calcium/calmodulin-dependent protein kinase kinase 2
- DAPK, death associated protein kinase
- FIP200, FAK family kinase-interacting protein of 200 kDa
- GAP, GTPase-activating protein
- GO, gene ontology
- GSK3α, glycogen synthase kinase 3 alpha
- HMGB1, high mobility group protein B1
- Human disease therapy
- JNK1, C-Jun N-terminal kinase
- LC3, microtubule-associated protein 1 light chain 3
- LKB1, serine/threonine-protein kinase stk11
- LPS, lipopolysaccharide
- LRRK2, leucine rich repeat kinase 2
- PD, Parkinson's disease
- PI, phosphatidylinositol
- PI3 kinase, phosphoinositide 3-kinase
- PI3P, phosphatidylinositol triphosphate
- PIM2, proviral insertion in murine lymphomas 2
- PINK1, PTEN-induced putative kinase 1
- PIP2, phosphatidylinositol-4,5-bisphosphate
- PKACα, a protein kinase cAMP-activated catalytic subunit alpha
- PKCα, protein kinase C alpha type
- PKD1, polycystin-1
- PPIs, protein–protein interactions
- PROTAC, proteolysis targeting chimeras
- PTMs, post-translational modifications
- Phosphorylation
- Protein kinases
- Rheb, the RAS homolog enriched in brain
- Small-molecule kinase inhibitors/activators
- TAK1, transforming growth factor activated kinase-1
- TFEB, transcription factor EB
- TNBC, triple-negative breast cancer
- TSC1/2, tuberous sclerosis complex proteins 1/2
- ULK complex, ULK1–mATG13–FIP200–ATG101 complex
- ULK1, unc-51-like kinase 1
- UVRAG, ultraviolet resistance-associated gene
- mTOR, mammalian target of rapamycin
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Honggang Xiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lan Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
27
|
Gao LP, Xiao K, Wu YZ, Chen DD, Yang XH, Shi Q, Dong XP. Enhanced Mitophagy Activity in Prion-Infected Cultured Cells and Prion-Infected Experimental Mice via a Pink1/Parkin-Dependent Mitophagy Pathway. ACS Chem Neurosci 2020; 11:814-829. [PMID: 32049477 DOI: 10.1021/acschemneuro.0c00039] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mitophagy is an important process for removing damaged mitochondria in cells, the dysfunction of which has been directly linked to an increasing number of neurodegenerative disorders. However, the details of mitophagy in prion diseases still need to be deeply explored. In this study, we identified more autophagosomes and large swelling mitochondria structures in the prion-infected cultured cell line SMB-S15 by transmission electron microscopy, accompanying the molecular evidence of activated autophagic flux. Western blots illustrated that the levels of Pink1 and Parkin, particularly in the mitochondrial fraction, were increased in SMB-S15 cells, whereas the levels of mitochondrial membrane proteins TIMM44, TOMM20, and TIMM23 were decreased. The amount of whole polyubiquitinated proteins decreased, but that of phosphor-polyubiquitinated proteins increased in SMB-S15 cells. The level of MFN2 in SMB-S15 cells were down-regulated, but its polyubiquitinated form was up-regulated. Knockdown of the expressions of Pink1 and Parkin by the individual SiRNAs in SMB-S15 cells reduced autophagic activity but did not seem to influence the expressions of TOMM20 and TIMM23. Moreover, we also demonstrated that the brain levels of Pink1 and Parkin in the mice infected with scrapie strains 139A and ME7 were remarkably increased at the terminal stage of the disease by Western blot and immunohistochemical (IHC) assays. Immunofluorescent assays revealed that Pink1 signals widely colocalized with GAFP-, Iba1-, and NeuN-positive cells in the brains of scrapie-infected mice. IHC assays with serial sections of the brain tissues infected with agents 139A and ME7 showed more Pink1- and Parkin-positive cells located at the areas with more PrPSc deposit. These results suggest an activated mitophagy in prion-infected cells and prion-infected experimental mice, probably via an enhanced Pink-Parkin pathway.
Collapse
Affiliation(s)
- Li-Ping Gao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Kang Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Yue-Zhang Wu
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Dong-Dong Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Xue-Hua Yang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
- China Academy of Chinese Medical Sciences,
Dongzhimeinei, South Road 16, Beijing 100700, China
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
- Center for Global Public Health, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
- China Academy of Chinese Medical Sciences,
Dongzhimeinei, South Road 16, Beijing 100700, China
| |
Collapse
|
28
|
Abstract
Prion disease, also known as transmissible spongiform encephalopathy (TES), is a fatal neurodegenerative disease caused by prion protein. The most important pathogenesis is related to changes in the conformation of cellular prion proteins (PrPC). The histopathological features of prion disease are spongiform degeneration, neuronal deficiency, glial activation and the deposition of amyloid-like PrPSc. Cellular prion protein, ubiquitously expressed in the brain and other tissues, is transformed into the PrP (PrPSc) isoform in the prion disease. In this chapter, we summarize the research progresses of prion disease, the structural organization and normal function of PrPC in the central nervous system. Moreover, the formation and transmissibility of prion aggregations (PrPSc) were also included. But we mainly focused on the function of PrPSc in autophagy. Several autophagic-related markers, such as p62 and LC3, are significantly upregulated in models of prion disease. Recent advances in the autophagic invention in prion disease and several pharmaceutical targets of autophagy were reviewed in this chapter. It is necessary to understand how the prion protein spread, transport and recycle, and what is the relationship between the clearance and autophagy.
Collapse
|
29
|
Maimaitiming A, Xiao K, Hu C, Chen J, Yang XH, Zhou DH, Gao LP, Dong XP, Shi Q. Aberrant Decrease of the Endogenous SIRT3 and Increases of Acetylated Proteins in Scrapie-Infected Cell Line SMB-S15 and in the Brains of Experimental Mice. ACS Chem Neurosci 2019; 10:4293-4302. [PMID: 31545894 DOI: 10.1021/acschemneuro.9b00341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The linkage between mitochondrial dysfunction and neurodegenerative diseases including prion diseases has been frequently reported. As the major deacetylase in mitochondria, SIRT3 plays a crucial part in regulating the function of many mitochondrial proteins. Although SIRT3 was reported to be linked to several neurodegenerative diseases, it is still unknown if SIRT3 is involved in prion diseases. In this study, we have presented a substantially declined status of mitochondrial SIRT3 in both the levels of cultured cells and an experimental rodent model during scrapie prion replication and infection. Such decreased SIRT3 activity led to a decreased deacetylating activity, resulting in increases of the acetylated forms of some substrates of SIRT3 in cells, such as SOD2 and ATP5β. Declined SOD2 and ATP5β activities subsequently caused an increase of intracellular ROS and a reduction of ATP. Furthermore, we have also proposed evidence that the activity of cellular SIRT3 is partially recovered when abnormal prion propagation in the cultured cells is removed by resveratrol. Those data emphasize a close connection between the prion replication and mitochondrial deacetylation due to SIRT3, thereby partially explaining mitochondrial dysfunction in prion diseases.
Collapse
Affiliation(s)
- Adalaiti Maimaitiming
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Kang Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Chao Hu
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Jia Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Xue-Hua Yang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Dong-Hua Zhou
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Li-Ping Gao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
- Center for Global Public Health, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Road 155, Beijing 102206, China
| |
Collapse
|
30
|
Iron dysregulation in vascular dementia: Focused on the AMPK/autophagy pathway. Brain Res Bull 2019; 153:305-313. [PMID: 31542426 DOI: 10.1016/j.brainresbull.2019.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/28/2019] [Accepted: 09/17/2019] [Indexed: 01/22/2023]
Abstract
Recent researches suggested that iron dysregulation play an important role in the pathogenesis of vascular dementia (VD). Iron deposition had been found in hippocampus in vascular dementia model in recent research. Nevertheless, the underlying mechanisms of iron deposition and its neurotoxicity in vascular dementia was still unclear. Thus, our research was aimed at whether the neurotoxicity of iron was associated with autophagy regulation. We established a chronic cerebral hypoperfusion model in the rat brain in order to mimic the vascular dementia using permanent bilateral common carotid artery occlusion (2VO). The preparation of iron overloaded rats model by intraperitoneal injection of iron dextran. Following, we tested the learning and memory function of each group using Morris Water Maze. Consequently, we analyzed the iron content and iron transport related molecules (TFR1, DMT1) in hippocampus. Furthermore, we examined the effect of iron deposition on autophagy-related molecules including AMPK, Beclin1 and LC3 and the number of autophagosomes in hippocampus. Last, we tested the apoptosis of neurons in hippocampus. We found that iron deposition in hippocampus in model groups which accompanied the decline of learning and memory function. And the expression of TFR1 and DMT1 were up-regulated in model groups. Moreover, iron deposition up-regulated the expression of AMPK, Beclin1 and LC3 and increase the number of autophagosomes in hippocampus. And the expression of Bax was up-regulated and Bcl-2 was down-regulated in iron deposition groups. To sum up, our data suggested that iron deposition increased AMPK/autophagy pathway associated molecules in the hippocampus and promoted neuronal apoptosis, which might be a new pathogenesis in vascular dementia.
Collapse
|
31
|
Chaurasia M, Gupta S, Das A, Dwarakanath B, Simonsen A, Sharma K. Radiation induces EIF2AK3/PERK and ERN1/IRE1 mediated pro-survival autophagy. Autophagy 2019; 15:1391-1406. [PMID: 30773986 PMCID: PMC6613886 DOI: 10.1080/15548627.2019.1582973] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 01/22/2019] [Accepted: 01/29/2019] [Indexed: 12/19/2022] Open
Abstract
Cellular effects of ionizing radiation include oxidative damage to macromolecules, unfolded protein response (UPR) and metabolic imbalances. Oxidative stress and UPR have been shown to induce macroautophagy/autophagy in a context-dependent manner and are crucial factors in determining the fate of irradiated cells. However, an in-depth analysis of the relationship between radiation-induced damage and autophagy has not been explored. In the present study, we investigated the relationship between radiation-induced oxidative stress, UPR and autophagy in murine macrophage cells. A close association was observed between radiation-induced oxidative burst, UPR and induction of autophagy, with the possible involvement of EIF2AK3/PERK (eukaryotic translation initiation factor 2 alpha kinase 3) and ERN1/IRE1 (endoplasmic reticulum [ER] to nucleus signaling 1). Inhibitors of either UPR or autophagy reduced the cell survival indicating the importance of these processes after radiation exposure. Moreover, modulation of autophagy affected lethality in the whole body irradiated C57BL/6 mouse. These findings indicate that radiation-induced autophagy is a pro-survival response initiated by oxidative stress and mediated by EIF2AK3 and ERN1. Abbreviations: ACTB: actin, beta; ATF6: activating transcription factor 6; ATG: autophagy-related; BafA1: bafilomycin A1; CQ: chloroquine; DBSA: 3,5-dibromosalicylaldehyde; EIF2AK3: eukaryotic translation initiation factor 2 alpha kinase 3; ERN1: endoplasmic reticulum (ER) to nucleus signaling 1; IR: ionizing radiation; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; 3-MA: 3-methyladenine; MTOR: mechanistic target of rapamycin kinase; NAC: N-acetyl-L-cysteine; PARP1: poly (ADP-ribose) polymerase family, member 1; 4-PBA: 4-phenylbutyrate; Rap: rapamycin; ROS: reactive oxygen species; UPR: unfolded protein response; XBP1: x-box binding protein 1.
Collapse
Affiliation(s)
- Madhuri Chaurasia
- Division of Metabolic Cell Signaling Research, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
- Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Swapnil Gupta
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Delhi, India
| | | | - Anne Simonsen
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kulbhushan Sharma
- Division of Metabolic Cell Signaling Research, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
32
|
Crocin induces anti-ischemia in middle cerebral artery occlusion rats and inhibits autophagy by regulating the mammalian target of rapamycin. Eur J Pharmacol 2019; 857:172424. [PMID: 31150648 DOI: 10.1016/j.ejphar.2019.172424] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 01/06/2023]
Abstract
Crocin, an active compound found in Gardenia jasminoides Ellis, has been shown to possess neuron-protective properties, but its potential mechanisms of action still remain poorly understood. In this study, the anti-ischemic effect and underlying mechanism of action of crocin were investigated in male rats with right middle cerebral artery occlusion/reperfusion. Computed tomography and magnetic resonance imaging were used to evaluate the area of infarction 24 h after reperfusion. Neurological scores were employed to evaluate nerve injury. Direct 2,3,5-triphenyltetrazolium chloride staining was used to calculate the infarct ratio 120 h after reperfusion. Finally, HT22 cells and Western blot were used to study the underlying mechanisms. Crocin showed a decreased infarct volume and neurological score in vivo, while the expression of LC3-II/I and AMP-activated protein kinase was remarkably down-regulated with increased levels of p62 and mammalian target of rapamycin (mTOR) expression. However, rapamycin significantly inhibited mTOR, which can impact the anti-ischemic effect of crocin in vitro. These results suggest that crocin may elicit an anti-ischemic effect probably through the mTOR pathway.
Collapse
|
33
|
López-Pérez Ó, Otero A, Filali H, Sanz-Rubio D, Toivonen JM, Zaragoza P, Badiola JJ, Bolea R, Martín-Burriel I. Dysregulation of autophagy in the central nervous system of sheep naturally infected with classical scrapie. Sci Rep 2019; 9:1911. [PMID: 30760781 PMCID: PMC6374525 DOI: 10.1038/s41598-019-38500-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/04/2018] [Indexed: 11/10/2022] Open
Abstract
Autophagy is a dynamic cellular mechanism involved in protein and organelle turnover through lysosomal degradation. Autophagy regulation modulates the pathologies associated with many neurodegenerative diseases. Using sheep naturally infected with scrapie as a natural animal model of prion diseases, we investigated the regulation of autophagy in the central nervous system (CNS) during the clinical phase of the disease. We present a gene expression and protein distribution analysis of different autophagy-related markers and investigate their relationship with prion-associated lesions in several areas of the CNS. Gene expression of autophagy markers ATG5 and ATG9 was downregulated in some areas of scrapie brains. In contrast, ATG5 protein accumulates in medulla oblongata and positively correlates with prion deposition and scrapie-related lesions. The accumulation of this protein and p62, a marker of autophagy impairment, suggests that autophagy is decreased in the late phases of the disease. However, the increment of LC3 proteins and the mild expression of p62 in basal ganglia and cerebellum, primarily in Purkinje cells, suggests that autophagy machinery is still intact in less affected areas. We hypothesize that specific cell populations of the CNS may display neuroprotective mechanisms against prion-induced toxicity through the induction of PrPSc clearance by autophagy.
Collapse
Affiliation(s)
- Óscar López-Pérez
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain.,Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain
| | - Alicia Otero
- Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain
| | - Hicham Filali
- Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain
| | - David Sanz-Rubio
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain
| | - Janne M Toivonen
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain
| | - Pilar Zaragoza
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain
| | - Juan J Badiola
- Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain
| | - Rosa Bolea
- Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain
| | - Inmaculada Martín-Burriel
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain. .,Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, Zaragoza, 50013, Spain.
| |
Collapse
|
34
|
Zhang Y, Liu S, Feng Q, Huang X, Wang X, Peng Y, Zhao Z, Liu Z. Perilaldehyde activates AMP-activated protein kinase to suppress the growth of gastric cancer via induction of autophagy. J Cell Biochem 2019; 120:1716-1725. [PMID: 30378150 DOI: 10.1002/jcb.27491] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/18/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND AND AIM Perillaldehyde (PAH), one of the major oil components in Perilla frutescens, is very critical to health maintenance, for a wide range of human chronic diseases, including cancers. AMP-activated protein kinase (AMPK) has been implicated in the activation of autophagy in distinct tissues. This study was designed to explore whether PAH prevents gastric cancer growth and to investigate the molecular mechanism. METHODS AND RESULTS In cultured mouse gastric cancer cell line MFCs and human gastric cancer cell lines GC9811-P, PAH activated AMPK by increasing the Thr172 phosphorylation and activity in a time-/concentration-dependent manner. Furthermore, incubation of MFCs with PAH also increased autophagy as determined by monodansylcadaverine (MDC) staining, which was reversed by AMPK inhibitor compound C. PAH further decreased MFCs cell survival, which was abolished by compound C or autophagy inhibitor 3-Methyladenine (3-MA). In vivo studies indicated that 4-week administration of PAH (100 mg/kg/day) suppressed the growth of gastric cancer and increased the levels of autophagy-related proteins, including beclin-1, LC3-II, cathepsin, caspase-3, p53, and cathepsin in tumors isolated from the xenograft model of gastric cancer in mice. Moreover, these anticancer effects produced by PAH were abolished by coadministration of compound C or 3-MA in vivo. CONCLUSIONS PAH increases AMPK phosphorylation and activity to induce gastric cancer cell autophagy to inhibit the growth of gastric cancer. In perspective, therapy of PAH should be applied to treat patients with gastric cancer.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Suosi Liu
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Qin Feng
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Xiuyun Huang
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Xiangyang Wang
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Ya Peng
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Zhihong Zhao
- Department of Neurology, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| | - Zhan Liu
- Department of Gastroenterology and Clinical Nutrition, The First Affiliated Hospital (People's Hospital of Hunan Province), Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
35
|
Hagio-Izaki K, Yasunaga M, Yamaguchi M, Kajiya H, Morita H, Yoneda M, Hirofuji T, Ohno J. Lipopolysaccharide induces bacterial autophagy in epithelial keratinocytes of the gingival sulcus. BMC Cell Biol 2018; 19:18. [PMID: 30165815 PMCID: PMC6117973 DOI: 10.1186/s12860-018-0168-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 08/13/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Interactions of resident bacteria and/or their producing lipopolysaccharide (LPS) with sulcular epithelial keratinocytes may be regulated by autophagy in the gingival sulcus. In this study, we investigated an induction of bacterial autophagy in exfoliative sulcular keratinocytes of the gingival sulcus and cultured keratinocytes treated with Porphyromonas gingivalis-originated LPS (PgLPS). RESULTS Exfoliative sulcular keratinocytes showed an induction of autophagy, in addition to increased expression of LPS-mediated factors including lipopolysaccharide-binding protein and toll-like receptors (TLRs), leading to co-localization of bacteria with autophagosomes. In contrast, exfoliative keratinocytes from the free gingiva did not show similar autophagy. Autophagy activity in human cultured keratinocyte cells (HaCaT) was induced by PgLPS, which was dependent partially on the AMP-activated protein kinase (AMPK) pathway via increased intracellular reactive oxygen species (ROS) and was in association with an activation of TLR4 signaling. After incubation of cultured keratinocytes with E.coli BioParticles following PgLPS stimulation, co-localization of bioparticles with autophagosomes was enhanced. Conversely, blockage of autophagy with 3-methyladenin and LPS-binding with polymyxin B led to significant reduction of co-localization of particles with autophagosomes. CONCLUSION These findings indicate that PgLPS-induced autophagy is at least partially responsible for interaction between bacteria and sulcular keratinocytes in the gingival sulcus.
Collapse
Affiliation(s)
- Kanako Hagio-Izaki
- Section of General Dentistry, Department of General Dentistry, Fukuoka Dental College, Fukuoka, Japan.,Research Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Madoka Yasunaga
- Research Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan.,Section of Orthodontics, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan
| | - Masahiro Yamaguchi
- Research Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan.,Section of Geriatric Dentistry, Department of General Dentistry, Fukuoka Dental College, Fukuoka, Japan
| | - Hiroshi Kajiya
- Research Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan.,Section of Cellular Physiology, Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Hiromitsu Morita
- Section of General Dentistry, Department of General Dentistry, Fukuoka Dental College, Fukuoka, Japan
| | - Masahiro Yoneda
- Section of General Dentistry, Department of General Dentistry, Fukuoka Dental College, Fukuoka, Japan
| | - Takao Hirofuji
- Section of General Dentistry, Department of General Dentistry, Fukuoka Dental College, Fukuoka, Japan
| | - Jun Ohno
- Research Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan.
| |
Collapse
|
36
|
Gao XL, Li JQ, Dong YT, Cheng EJ, Gong JN, Qin YL, Huang YQ, Yang JJ, Wang SJ, An DD. Upregulation of microRNA-335-5p reduces inflammatory responses by inhibiting FASN through the activation of AMPK/ULK1 signaling pathway in a septic mouse model. Cytokine 2018; 110:466-478. [PMID: 29866515 DOI: 10.1016/j.cyto.2018.05.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 04/28/2018] [Accepted: 05/17/2018] [Indexed: 12/22/2022]
Abstract
Sepsis, as a systemic inflammatory response syndrome (SIRS) subtype, is generally characterized by infection. Emerging evidence has highlighted dysregulated microRNAs (miRNAs) are involved in the progression of sepsis. The aim of the study was to investigate the effects of miR-335-5p on inflammatory responses in a septic mouse model. The hypothesis was subsequently asserted that the FASN gene and AMPK/ULK1 signaling pathway may participate in the regulation of miR-335-5p. A septic mouse model was established in order to validate the effect of miR-335-5p on the inflammatory response by means of suppressing the endogenous expression of FASN by siRNA against FASN in endothelial cells. A target prediction program and luciferase activity was employed to ascertain as to whether miR--335-5p targets FASN. The levels of inflammatory factors including IL-6 and IL-1β were determined by means of ELISA assay. RT-qPCR and western blot analysis were used to determine the AMPK/ULK1 signaling pathway-, apoptosis- and autophagy-related genes. Flow cytometry was employed in order to evaluate sepsis-induced cell apoptosis in response to miR-335-5p and FASN alternations. FASN was identified as a target gene of miR--335-5p. Gain- and loss-of-function studies revealed that miR-335-5p acted to enhance autophagy, reduce cell apoptosis, promote cell cycle entry in endothelial cells, and reduce inflammatory response through the modulation of pro- and anti-apoptotic factors in endothelial cells. The effect of miR-335-5p on endothelial cells was increased when FASN was suppressed by siRNA as well as when the AMPK/ULK1 signaling pathway was activated, suggesting that miR-335-5p influences sepsis by targeting and inhibiting FASN, and activating the AMPK/ULK1 signaling pathway. Our study provides evidence indicating that overexpressed miR-335-5p enhances autophagy by targeting FASN through activation of the AMPK/ULK1 signaling pathway working to alleviate the inflammatory response in septic mouse models, emphasizing the value of the functional upregulation of miR-335-5p as therapeutic strategy for sepsis.
Collapse
Affiliation(s)
- Xiao-Ling Gao
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Jian-Qiang Li
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Yan-Ting Dong
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Er-Jing Cheng
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Jian-Nan Gong
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Yi-Li Qin
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | | | | | | | - Dong-Dong An
- Shanxi Medical University, Taiyuan 030000, PR China
| |
Collapse
|
37
|
Shi Q, Li JL, Ma Y, Gao LP, Xiao K, Wang J, Zhou W, Chen C, Guo YJ, Dong XP. Decrease of RyR2 in the prion infected cell line and in the brains of the scrapie infected mice models and the patients of human prion diseases. Prion 2018; 12:175-184. [PMID: 29676187 DOI: 10.1080/19336896.2018.1465162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
The levels of ryanodine receptors (RyRs) are usually increased in the brains of human Alzheimer disease (AD) and AD animal models. To evaluate the underlying alteration of brain RyRs in prion disease, scrapie infected cell line SMB-S15 and its infected mice were tested. RyR2 specific Western blots revealed markedly decreased RyR2 levels both in the cells and in the brains of infected mice. Assays of the brain samples of other scrapie (agents 139A and ME7) infected mice collected at different time-points during incubation period showed time-dependent decreases of RyR2. Immunofluorescent assays (IFA) verified that the expression of RyR2 locates predominantly in cytoplasm of SMB cells and overlapped with the neurons in the brain slices of mice. Furthermore, significant down-regulation of RyR2 was also detected in the postmortem cortical brains of the patients of various types of human prion diseases, including sporadic Creutzfeldt-Jakob disease (sCJD), fatal familial insomnia (FFI) and G114V-genetic CJD. Our data here propose the evidences of remarkably decreased brain RyR2 at terminal stages of both human prion diseases and prion infected rodent models. It also highlights that the therapeutic strategy with antagonist of RyRs in AD may not be suitable for prion disease.
Collapse
Affiliation(s)
- Qi Shi
- a State Key Laboratory for Infectious Disease Prevention and Control , Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Jian-Le Li
- a State Key Laboratory for Infectious Disease Prevention and Control , Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China.,b Department of Neurology , Beijing Friendship Hospital, Capital Medical University , Xicheng District, Beijing , People's Republic of China
| | - Yue Ma
- a State Key Laboratory for Infectious Disease Prevention and Control , Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Li-Ping Gao
- a State Key Laboratory for Infectious Disease Prevention and Control , Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Kang Xiao
- a State Key Laboratory for Infectious Disease Prevention and Control , Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Jing Wang
- a State Key Laboratory for Infectious Disease Prevention and Control , Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Wei Zhou
- a State Key Laboratory for Infectious Disease Prevention and Control , Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Cao Chen
- a State Key Laboratory for Infectious Disease Prevention and Control , Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Yan-Jun Guo
- b Department of Neurology , Beijing Friendship Hospital, Capital Medical University , Xicheng District, Beijing , People's Republic of China
| | - Xiao-Ping Dong
- a State Key Laboratory for Infectious Disease Prevention and Control , Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| |
Collapse
|
38
|
Tian X, Yu C, Shi L, Li D, Chen X, Xia D, Zhou J, Xu W, Ma C, Gu L, An Y. MicroRNA-199a-5p aggravates primary hypertension by damaging vascular endothelial cells through inhibition of autophagy and promotion of apoptosis. Exp Ther Med 2018; 16:595-602. [PMID: 30116316 PMCID: PMC6090226 DOI: 10.3892/etm.2018.6252] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 04/13/2018] [Indexed: 12/19/2022] Open
Abstract
The present study investigated the expression of microRNA (miRNA or miR)-199a-5p in the peripheral blood of patients with primary hypertension, and examined its mechanism of action in vascular endothelial cell injury induced by hypertension. A total of 57 patients with primary hypertension, who were treated at the Affiliated Hospital of Qingdao University (Qingdao, China) between December 2014 and November 2015 were included in the present study. Peripheral blood was collected from all patients. The expression of miR-199a-5p was measured using reverse-transcription quantitative polymerase chain reaction analysis. Human umbilical vein endothelial cells (HUVECs) were divided into negative control, miR-199a-5p mimics and rescue (co-transfected with miR-199a-5p mimics and inhibitor) groups. After transfection, the proliferation and apoptosis of HUVECs were evaluated by a Cell Counting Kit-8 assay, a bromodeoxyuridine incorporation assay and flow cytometry. Western blot analysis was used to determine the expression of proteins involved in autophagy-associated and adenosine monophosphate kinase (AMPK)/unc-51 like autophagy activating kinase 1 (ULK1) signaling pathways. Laser scanning confocal microscopy and electron microscopy were used to observe the autophagy of HUVECs. The expression of miR-199a-5p was elevated in peripheral blood of patients with hypertension, and was correlated with the progression of hypertension. Overexpression of miR-199a-5p inhibited the proliferation and promoted the apoptosis of HUVECs. Upon expression of miR-199a-5p, the transition between microtubule-associated proteins 1A/1B light chain 3B (LC3B)I and LC3BII proteins was inhibited, the expression of p62 protein was upregulated. In addition, miR-199a-5p decreased the numbers of autophagosomes and autolysosomes in HUVECs. The present study demonstrated that expression of miR-199a-5p is positively correlated with the severity of hypertension. Expression of miR-199a-5p aggravated vascular endothelial injury by inhibiting autophagy and promoting the apoptosis of HUVECs via downregulation of the AMPK/ULK1 signaling pathway.
Collapse
Affiliation(s)
- Xintao Tian
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Chunpeng Yu
- Department of Intervention, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Lei Shi
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Dan Li
- Department of Cardiovascular Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Xiaoxue Chen
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Di Xia
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Jingwei Zhou
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Wanqun Xu
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Chengtai Ma
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Lihua Gu
- Department of Emergency Internal Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Yi An
- Department of Cardiovascular Medicine, Laoshan District of The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| |
Collapse
|
39
|
Taniguchi K, Matsumura K, Ii H, Kageyama S, Ashihara E, Chano T, Kawauchi A, Yoshiki T, Nakata S. Depletion of gamma-glutamylcyclotransferase in cancer cells induces autophagy followed by cellular senescence. Am J Cancer Res 2018; 8:650-661. [PMID: 29736310 PMCID: PMC5934555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/16/2018] [Indexed: 06/08/2023] Open
Abstract
Gamma-glutamylcyclotransferase (GGCT) was originally identified as a protein highly expressed in bladder cancer tissues by proteomic analysis, and its higher expression in a variety of cancers compared to normal tissues have been shown. Depletion of GGCT in various cancer cells results in antiproliferative effects both in vitro and in vivo; thus it is considered a promising therapeutic target. Although it has been shown that knockdown of GGCT induces cellular senescence and non-apoptotic cell death, associated with upregulation of cyclin-dependent kinase inhibitors (CDKIs) including p21WAF1/CIP1, the cellular events that follow GGCT depletion are not fully understood. Here, we show that GGCT depletion induced autophagy in MCF7 breast and PC3 prostate cancer cells. Conversely, overexpression of GGCT in NIH3T3 fibroblast under conditions of serum deprivation inhibited autophagy and increased proliferation. Simultaneous knockdown of autophagy related-protein 5, a critical effector of autophagy, along with GGCT in MCF7 and PC3 cells led to significant attenuation of the multiple cellular responses, including upregulation of CDKIs, increased numbers of senescence-associated β-galactosidase positive senescent cells, and growth inhibition. Furthermore, we show that autophagy-promoting signaling cascades including activation of the AMPK-ULK1 pathway and/or inactivation of the mTORC2-Akt pathway were triggered in GGCT-depleted cells. These results indicate that autophagy plays an important role in the growth inhibition of cancer cells caused by GGCT depletion.
Collapse
Affiliation(s)
- Keiko Taniguchi
- Department of Clinical Oncology, Kyoto Pharmaceutical UniversityMisasagi-Nakauchicho 5, Yamashina-ku, Kyoto 607-8414, Japan
| | - Kengo Matsumura
- Department of Clinical Pharmacology and Therapeutics, Kyoto University HospitalShogoin-kawaharacho 54, Sakyoku, Kyoto 606-8507, Japan
| | - Hiromi Ii
- Department of Clinical Oncology, Kyoto Pharmaceutical UniversityMisasagi-Nakauchicho 5, Yamashina-ku, Kyoto 607-8414, Japan
| | - Susumu Kageyama
- Department of Urology, Shiga University of Medical ScienceTsukinowa-cho, Seta, Otsu, Shiga 520-2192, Japan
| | - Eishi Ashihara
- Department of Clinical and Translational Physiology, Kyoto Pharmaceutical UniversityMisasagi-Nakauchicho 5, Yamashina-ku, Kyoto 607-8414, Japan
| | - Tokuhiro Chano
- Department of Clinical Laboratory Medicine, Shiga University of Medical ScienceTsukinowa-cho, Seta, Otsu, Shiga 520-2192, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical ScienceTsukinowa-cho, Seta, Otsu, Shiga 520-2192, Japan
| | - Tatsuhiro Yoshiki
- Department of Clinical Oncology, Kyoto Pharmaceutical UniversityMisasagi-Nakauchicho 5, Yamashina-ku, Kyoto 607-8414, Japan
- Department of Urology, Shiga University of Medical ScienceTsukinowa-cho, Seta, Otsu, Shiga 520-2192, Japan
| | - Susumu Nakata
- Department of Clinical Oncology, Kyoto Pharmaceutical UniversityMisasagi-Nakauchicho 5, Yamashina-ku, Kyoto 607-8414, Japan
| |
Collapse
|
40
|
Zhang L, Ouyang L, Guo Y, Zhang J, Liu B. UNC-51-like Kinase 1: From an Autophagic Initiator to Multifunctional Drug Target. J Med Chem 2018; 61:6491-6500. [PMID: 29509411 DOI: 10.1021/acs.jmedchem.7b01684] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
UNC-51-like kinase 1 (ULK1), known as an ortholog of the yeast Atg1, is the serine-threonine kinase and the autophagic initiator in mammals. Accumulating evidence has recently revealed the kinase domain structure of ULK1 and its post-translational modifications, as well as further elucidated its regulatory autophagic pathways and associations with diverse human diseases. Interestingly, a series of small molecules have been recently reported to target ULK1 or ULK1-modulating autophagy, which may provide a clue on exploiting them as novel candidate drugs. Taken together, this review discusses how ULK1 acts as an autophagic initiator for modulation of its intricate mechanisms, as well as how ULK1 becomes a multifunctional target for potential therapeutic applications.
Collapse
Affiliation(s)
- Lan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Yongzhi Guo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| |
Collapse
|
41
|
Huang Z, Huang X, Wang Q, Jiang R, Sun G, Xu Y, Wu Q. Extract of Euryale ferox Salisb
exerts antidepressant effects and regulates autophagy through the adenosine monophosphate-activated protein kinase-UNC-51-like kinase 1 pathway. IUBMB Life 2018; 70:300-309. [DOI: 10.1002/iub.1731] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/02/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Zhiheng Huang
- School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Nanjing China
| | - Xiaoyan Huang
- School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Nanjing China
| | - Qian Wang
- School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Nanjing China
| | - Ruizhi Jiang
- School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Nanjing China
| | - Guangda Sun
- School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Nanjing China
| | - Yiming Xu
- School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Nanjing China
| | - Qinan Wu
- School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Nanjing China
| |
Collapse
|
42
|
Zhang RQ, Chen C, Xiao LJ, Sun J, Ma Y, Yang XD, Xu XF, Xiao K, Shi Q, Chen ZB, Dong XP. Aberrant alterations of the expressions and S-nitrosylation of calmodulin and the downstream factors in the brains of the rodents during scrapie infection. Prion 2018; 11:352-367. [PMID: 28968141 DOI: 10.1080/19336896.2017.1367082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The aberrant alterations of calmodulin (CaM) and its downstream substrates have been reported in some neurodegenerative diseases, but rarely described in prion disease. In this study, the potential changes of Ca2+/CaM and its associated agents in the brains of scrapie agent 263K-infected hamsters and the prion infected cell line SMB-S15 were evaluated by various methodologies. We found that the level of CaM in the brains of 263K-infected hamsters started to increase at early stage and maintained at high level till terminal stage. The increased CaM mainly accumulated in the regions of cortex, thalamus and cerebellum of 263K-infected hamsters and well localization of CaM with NeuN positive cells. However, the related kinases such as total and phosphorylated forms of CaMKII and CaMKIV, as well as the downstream proteins such as CREB and BDNF in the brain of 263K-infected hamsters were decreased. Further analysis showed a remarkable increase of S-nitrosylated (SNO) form of CaM in the brains of 263K-infected hamsters. Dynamic analysis of S-nitrosylated CaM showed the SNO form of CaM abnormally increases in a time-dependent manner during prion infection. Compared with that of the normal partner cell line SMB-PS, the CaM level in SMB-S15 cells was increased, meanwhile, the downstream proteins, such as CaMKII, p-CaMKII, CREB, as well as BDNF, were also increased, especially in the nucleic fraction. No SNO-CaM was detected in the cell lines SMB-S15 and SMB-PS. Our data indicate an aberrant increase of CaM during prion infection in vivo and in vitro.
Collapse
Affiliation(s)
- Ren-Qing Zhang
- a College of Life Science and Technology , Heilongjiang Bayi Agricultural University , Daqing , People's Republic of China.,b State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Cao Chen
- b State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Li-Jie Xiao
- a College of Life Science and Technology , Heilongjiang Bayi Agricultural University , Daqing , People's Republic of China
| | - Jing Sun
- b State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Yue Ma
- b State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Xiao-Dong Yang
- b State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Xiao-Feng Xu
- b State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Kang Xiao
- b State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Qi Shi
- b State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| | - Zhi-Bao Chen
- a College of Life Science and Technology , Heilongjiang Bayi Agricultural University , Daqing , People's Republic of China
| | - Xiao-Ping Dong
- b State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention , Beijing , People's Republic of China
| |
Collapse
|
43
|
Ma Y, Shi Q, Wang J, Xiao K, Sun J, Lv Y, Guo M, Zhou W, Chen C, Gao C, Zhang BY, Dong XP. Reduction of NF-κB (p65) in Scrapie-Infected Cultured Cells and in the Brains of Scrapie-Infected Rodents. ACS Chem Neurosci 2017; 8:2535-2548. [PMID: 28783945 DOI: 10.1021/acschemneuro.7b00273] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Transcription factor NF-κB functions as a pleiotropic regulator of target genes controlling physiological function as well as pathological processes of many different diseases, including some neurodegenerative diseases. However, the role of NF-κB in the pathogenesis of prion disease remains ambiguous. In this study, the status of NF-κB (p65) in a prion-infected cell line SMB-S15 was first evaluated. Significantly lower levels of p65 and the phosphorylated form of p65 (p-p65) were detected in SMB-S15 cells, compared with its normal partner cell line SMB-PS. Markedly slower responses of the NF-κB system to the stimulation of TNF-α were observed in SMB-S15 cells. Removal of PrPSc replication in SMB-S15 cells rescued the expression and activity of NF-κB. However, overexpression of p65 in SMB-S15 cells did not influence the propagation of PrPSc. Moreover, significant decline of p65 level was also observed in the brain tissues of mice infected with the lysates of SMB-S15 cells and hamsters infected with scrapie agent 263K at terminal stage. Immunofluorescence assays (IFAs) on brain sections from either normal or scrapie-infected rodents revealed colocalization of p65 with neuronal nuclear (NeuN) protein positive cells but not with glial fibrillary acidic protein (GFAP) positive cells. Assays of the agents involving in the regulation of NF-κB showed down-regulated phosphoinositide 3-kinase (PI3K) and protein kinase B (PKB/Akt) both in SMB-S15 cells and in the brains of scrapie-infected rodents. Those data indicate a remarkable repression of the classical NF-κB pathway during prion infection both in vitro and in vivo. The alteration of NF-κB (p65) shows close association with the replication and accumulation of PrPSc in the cells.
Collapse
Affiliation(s)
- Yue Ma
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Jing Wang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Kang Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Jing Sun
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Yan Lv
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Man Guo
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Wei Zhou
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Cao Chen
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Chen Gao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Bao-Yun Zhang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
- Key Laboratory
of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People’s Republic of China
| |
Collapse
|
44
|
Sun S, Xuan F, Ge X, Zhu J, Zhang W. Dynamic mRNA and miRNA expression analysis in response to hypoxia and reoxygenation in the blunt snout bream (Megalobrama amblycephala). Sci Rep 2017; 7:12846. [PMID: 28993687 PMCID: PMC5634510 DOI: 10.1038/s41598-017-12537-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/12/2017] [Indexed: 12/24/2022] Open
Abstract
Adaptation to hypoxia is a complex process involving various pathways and regulation mechanisms. A better understanding of the genetic influence on these mechanisms could permit selection for hypoxia-sensitive fish. To aid this understanding, an integrated analysis of miRNA and mRNA expression was performed in Megalobrama amblycephala under four acute hypoxia and reoxygenation stages. A number of significantly differentially-expressed miRNAs and genes associated with oxidative stress were identified, and their functional characteristics were revealed by GO function and KEGG pathway analysis. They were found to be involved in HIF-1 pathways known to affect energy metabolism and apoptosis. MiRNA-mRNA interaction pairs were detected from comparison of expression between the four different stages. The function annotation results also showed that many miRNA-mRNA interaction pairs were likely to be involved in regulating hypoxia stress. As a unique resource for gene expression and regulation during hypoxia and reoxygenation, this study could provide a starting point for further studies to better understand the genetic background of hypoxia stress.
Collapse
Affiliation(s)
- Shengming Sun
- Key Laboratory of Genetic Breeding and Aquaculture Biology of Freshwater Fishes, Ministry of Agriculture, Freshwater Fisheries Research Centre, Chinese Academy of Fishery Sciences, Wuxi, 214081, P.R. China
| | - Fujun Xuan
- Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng City, Jiangsu Province, 224002, P.R. China
| | - Xianping Ge
- Key Laboratory of Genetic Breeding and Aquaculture Biology of Freshwater Fishes, Ministry of Agriculture, Freshwater Fisheries Research Centre, Chinese Academy of Fishery Sciences, Wuxi, 214081, P.R. China.
| | - Jian Zhu
- Key Laboratory of Genetic Breeding and Aquaculture Biology of Freshwater Fishes, Ministry of Agriculture, Freshwater Fisheries Research Centre, Chinese Academy of Fishery Sciences, Wuxi, 214081, P.R. China.
| | - Wuxiao Zhang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, P.R. China
| |
Collapse
|
45
|
Abstract
Cells constantly adapt their metabolism to meet their energy needs and respond to nutrient availability. Eukaryotes have evolved a very sophisticated system to sense low cellular ATP levels via the serine/threonine kinase AMP-activated protein kinase (AMPK) complex. Under conditions of low energy, AMPK phosphorylates specific enzymes and growth control nodes to increase ATP generation and decrease ATP consumption. In the past decade, the discovery of numerous new AMPK substrates has led to a more complete understanding of the minimal number of steps required to reprogramme cellular metabolism from anabolism to catabolism. This energy switch controls cell growth and several other cellular processes, including lipid and glucose metabolism and autophagy. Recent studies have revealed that one ancestral function of AMPK is to promote mitochondrial health, and multiple newly discovered targets of AMPK are involved in various aspects of mitochondrial homeostasis, including mitophagy. This Review discusses how AMPK functions as a central mediator of the cellular response to energetic stress and mitochondrial insults and coordinates multiple features of autophagy and mitochondrial biology.
Collapse
|
46
|
Majumder P, Chakrabarti O. Lysosomal Quality Control in Prion Diseases. Mol Neurobiol 2017; 55:2631-2644. [PMID: 28421536 DOI: 10.1007/s12035-017-0512-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/04/2017] [Indexed: 11/28/2022]
Abstract
Prion diseases are transmissible, familial or sporadic. The prion protein (PrP), a normal cell surface glycoprotein, is ubiquitously expressed throughout the body. While loss of function of PrP does not elicit apparent phenotypes, generation of misfolded forms of the protein or its aberrant metabolic isoforms has been implicated in a number of neurodegenerative disorders such as scrapie, kuru, Creutzfeldt-Jakob disease, fatal familial insomnia, Gerstmann-Sträussler-Scheinker and bovine spongiform encephalopathy. These diseases are all phenotypically characterised by spongiform vacuolation of the adult brain, hence collectively termed as late-onset spongiform neurodegeneration. Misfolded form of PrP (PrPSc) and one of its abnormal metabolic isoforms (the transmembrane CtmPrP) are known to be disease-causing agents that lead to progressive loss of structure or function of neurons culminating in neuronal death. The aberrant forms of PrP utilise and manipulate the various intracellular quality control mechanisms during pathogenesis of these diseases. Amongst these, the lysosomal quality control machinery emerges as one of the primary targets exploited by the disease-causing isoforms of PrP. The autophagosomal-lysosomal degradation pathway is adversely affected in multiple ways in prion diseases and may hence be regarded as an important modulator of neurodegeneration. Some of the ESCRT pathway proteins have also been shown to be involved in the manifestation of disease phenotype. This review discusses the significance of the lysosomal quality control pathway in affecting transmissible and familial types of prion diseases.
Collapse
Affiliation(s)
- Priyanka Majumder
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Sector-1, Block-AF, Bidhannagar, Kolkata, West Bengal, 700064, India
| | - Oishee Chakrabarti
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Sector-1, Block-AF, Bidhannagar, Kolkata, West Bengal, 700064, India.
| |
Collapse
|
47
|
Tao YF, Li ZH, Du WW, Xu LX, Ren JL, Li XL, Fang F, Xie Y, Li M, Qian GH, Li YH, Li YP, Li G, Wu Y, Feng X, Wang J, He WQ, Hu SY, Lu J, Pan J. Inhibiting PLK1 induces autophagy of acute myeloid leukemia cells via mammalian target of rapamycin pathway dephosphorylation. Oncol Rep 2017; 37:1419-1429. [PMID: 28184925 PMCID: PMC5364848 DOI: 10.3892/or.2017.5417] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/30/2017] [Indexed: 02/07/2023] Open
Abstract
Decreased autophagy is accompanied by the development of a myeloproliferative state or acute myeloid leukemia (AML). AML cells are often sensitive to autophagy‑inducing stimuli, prompting the idea that targeting autophagy can be useful in AML cytotoxic therapy. AML NB4 cells overexpressing microtubule-associated protein 1 light chain 3-green fluorescent protein were screened with 69 inhibitors to analyze autophagy activity. AML cells were treated with the polo-like kinase 1 (PLK1) inhibitors RO3280 and BI2536 before autophagy analysis. Cleaved LC3 (LC3-II) and the phosphorylation of mammalian target of rapamycin (mTOR), adenosine monophosphate-activated protein kinase, and Unc-51-like kinase 1 during autophagy was detected with western blotting. Autophagosomes were detected using transmission electron microscopy. Several inhibitors had promising autophagy inducer effects: BI2536, MLN0905, SK1-I, SBE13 HCL and RO3280. Moreover, these inhibitors all targeted PLK1. Autophagy activity was increased in the NB4 cells treated with RO3280 and BI2536. Inhibition of PLK1 expression in NB4, K562 and HL-60 leukemia cells with RNA interference increased LC3-II and autophagy activity. The phosphorylation of mTOR was reduced significantly in NB4 cells treated with RO3280 and BI2536, and was also reduced significantly when PLK1 expression was downregulated in the NB4, K562 and HL-60 cells. We demonstrate that PLK1 inhibition induces AML cell autophagy and that it results in mTOR dephosphorylation. These results may provide new insights into the molecular mechanism of PLK1 in regulating autophagy.
Collapse
MESH Headings
- Animals
- Autophagy
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cell Cycle Proteins/antagonists & inhibitors
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Proliferation
- Child
- Female
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice
- Neoplasm Staging
- Phosphorylation
- Prognosis
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Survival Rate
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
- Polo-Like Kinase 1
Collapse
Affiliation(s)
- Yan-Fang Tao
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Zhi-Heng Li
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Wei-Wei Du
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Li-Xiao Xu
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Jun-Li Ren
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Xiao-Lu Li
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Fang Fang
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Yi Xie
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Mei Li
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Guang-Hui Qian
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Yan-Hong Li
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Yi-Ping Li
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Gang Li
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Yi Wu
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Xing Feng
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Jian Wang
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Wei-Qi He
- CAM-SU Genomic Resource Center, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Shao-Yan Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Jun Lu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| | - Jian Pan
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, Jiangsu 215003, P.R. China
| |
Collapse
|
48
|
Cheng J, Zhang T, Ji H, Tao K, Guo J, Wei W. Functional characterization of AMP-activated protein kinase signaling in tumorigenesis. Biochim Biophys Acta Rev Cancer 2016; 1866:232-251. [PMID: 27681874 DOI: 10.1016/j.bbcan.2016.09.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
AMP-activated protein kinase (AMPK) is a ubiquitously expressed metabolic sensor among various species. Specifically, cellular AMPK is phosphorylated and activated under certain stressful conditions, such as energy deprivation, in turn to activate diversified downstream substrates to modulate the adaptive changes and maintain metabolic homeostasis. Recently, emerging evidences have implicated the potential roles of AMPK signaling in tumor initiation and progression. Nevertheless, a comprehensive description on such topic is still in scarcity, especially in combination of its biochemical features with mouse modeling results to elucidate the physiological role of AMPK signaling in tumorigenesis. Hence, we performed this thorough review by summarizing the tumorigenic role of each component along the AMPK signaling, comprising of both its upstream and downstream effectors. Moreover, their functional interplay with the AMPK heterotrimer and exclusive efficacies in carcinogenesis were chiefly explained among genetically altered mice models. Importantly, the pharmaceutical investigations of AMPK relevant medications have also been highlighted. In summary, in this review, we not only elucidate the potential functions of AMPK signaling pathway in governing tumorigenesis, but also potentiate the future targeted strategy aiming for better treatment of aberrant metabolism-associated diseases, including cancer.
Collapse
Affiliation(s)
- Ji Cheng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Tao Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hongbin Ji
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science, Shanghai 200031, People's Republic of China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China.
| | - Jianping Guo
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
49
|
Zhao L, Cui L, Jiang X, Zhang J, Zhu M, Jia J, Zhang Q, Zhang J, Zhang D, Huang Y. Extracellular pH regulates autophagy via the AMPK-ULK1 pathway in rat cardiomyocytes. FEBS Lett 2016; 590:3202-12. [PMID: 27531309 DOI: 10.1002/1873-3468.12359] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/02/2016] [Accepted: 08/10/2016] [Indexed: 12/20/2022]
Abstract
Various pathological conditions contribute to pH fluctuations and affect the functions of vital organs such as the heart. In this study, we show that in rat cardiomyocytes, acidic extracellular pH (pHe) inhibits autophagy, whereas alkaline pHe stimulates it. We also find that adenosine monophosphate-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR) and Unc-51-like kinase 1 (ULK1) are very sensitive to pHe changes. Furthermore, by interfering with AMPK, mTOR or ULK1 activity, we demonstrate that the AMPK-ULK1 pathway, but not the mTOR pathway, plays a crucial role on pHe-regulated autophagy and cardiomyocyte viability. These data provide a potential therapeutic strategy against cardiomyocyte injury triggered by pH fluctuations.
Collapse
Affiliation(s)
- Liping Zhao
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Lin Cui
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xupin Jiang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Junhui Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Minghua Zhu
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiezhi Jia
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qiong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiaping Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Dongxia Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
50
|
Wang H, Tian C, Sun J, Chen LN, Lv Y, Yang XD, Xiao K, Wang J, Chen C, Shi Q, Shao QX, Dong XP. Overexpression of PLK3 Mediates the Degradation of Abnormal Prion Proteins Dependent on Chaperone-Mediated Autophagy. Mol Neurobiol 2016; 54:4401-4413. [PMID: 27344333 DOI: 10.1007/s12035-016-9985-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 06/14/2016] [Indexed: 12/19/2022]
Abstract
Polo-like kinase 3 (PLK3) is the main cause of cell cycle reentry-related neuronal apoptosis which has been implicated in the pathogenesis of prion diseases. Previous work also showed the regulatory activity of exogenous PLK3 on the degradation of PrP (prion protein) mutants and pathogenic PrPSc; however, the precise mechanisms remain unknown. In this study, we identified that the overexpression of PLK3-mediated degradation of PrP mutant and PrPSc was repressed by lysosome rather than by proteasomal and macroautophagy inhibitors. Core components of chaperone-mediated autophagy (CMA) effectors, lysosome-associated membrane protein type 2A (LAMP2a), and heat shock cognate protein 70 (Hsc70) are markedly decreased in the HEK293T cells expressing PrP mutant and scrapie-infected cell line SMB-S15. Meanwhile, PrP mutant showed ability to interact with LAMP2a and Hsc70. Overexpression of PLK3 sufficiently increased the cellular levels of LAMP2a and Hsc70, accompanying with declining the accumulations of PrP mutant and PrPSc. The kinase domain (KD) of PLK3 was responsible for elevating LAMP2a and Hsc70. Knockdown of endogenous PLK3 enhanced the activity of macroautophagy in the cultured cells. Moreover, time-dependent reductions of LAMP2a and Hsc70 were also observed in the brain tissues of hamster-adapted scrapie agent 263K-infected hamsters, indicating an impairment of CMA during prion infection. Those data indicate that the overexpression of PLK3-mediated degradation of abnormal PrP is largely dependent on CMA pathway.
Collapse
Affiliation(s)
- Hui Wang
- Department of Immunology and Key Laboratory of Laboratory Medicine of Jiangsu Province, Medical School, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.,State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China
| | - Chan Tian
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China
| | - Jing Sun
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China
| | - Li-Na Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China
| | - Yan Lv
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China
| | - Xiao-Dong Yang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China
| | - Kang Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China
| | - Jing Wang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China
| | - Cao Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China
| | - Qi-Xiang Shao
- Department of Immunology and Key Laboratory of Laboratory Medicine of Jiangsu Province, Medical School, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing, 102206, China. .,Chinese Academy of Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|