1
|
Zhai J, Chen Z, Zhu Q, Guo Z, Sun X, Jiang L, Li J, Wang N, Yao X, Zhang C, Deng H, Wang S, Yang G. Curcumin inhibits PAT-induced renal ferroptosis via the p62/Keap1/Nrf2 signalling pathway. Toxicology 2024; 506:153863. [PMID: 38878878 DOI: 10.1016/j.tox.2024.153863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/24/2024]
Abstract
Patulin (PAT), the most common mycotoxin, is widespread in foods and beverages which poses a serious food safety issue to human health. Our previous research confirmed that exposure to PAT can lead to acute kidney injury (AKI). Curcumin is the most abundant active ingredient in turmeric rhizome with various biological activities. The aim of this study is to investigate whether curcumin can prevent the renal injury caused by PAT, and to explore potential mechanisms. In vivo, supplementation with curcumin attenuated PAT-induced ferroptosis. Mechanically, curcumin inhibited autophagy, led to the accumulation of p62 and its interaction with Keap1, promoted the nuclear translocation of nuclear factor E2 related factor 2 (Nrf2), and increased the expression of antioxidant stress factors in the process of ferroptosis. These results have also been confirmed in HKC cell experiments. Furthermore, knockdown of Nrf2 in HKC cells abrogated the protective effect of curcumin on ferroptosis. In conclusion, we confirmed that curcumin mitigated PAT-induced AKI by inhibiting ferroptosis via activation of the p62/Keap1/Nrf2 pathway. This study provides new potential targets and ideas for the prevention and treatment of PAT.
Collapse
Affiliation(s)
- Jianan Zhai
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Zhengguo Chen
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Qi Zhu
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Zhifang Guo
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Xiance Sun
- Department of Occupational & Environmental Health, Dalian Medical University, Dalian 116044, China
| | - Liping Jiang
- Department of Occupational & Environmental Health, Dalian Medical University, Dalian 116044, China
| | - Jing Li
- Department of Pathology, Dalian Medical University, Dalian 116044, China
| | - Ningning Wang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Xiaofeng Yao
- Department of Occupational & Environmental Health, Dalian Medical University, Dalian 116044, China
| | - Cong Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Haoyuan Deng
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Shaopeng Wang
- Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Guang Yang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China.
| |
Collapse
|
2
|
Preminger N, Schuldiner M. Beyond fission and fusion-Diving into the mysteries of mitochondrial shape. PLoS Biol 2024; 22:e3002671. [PMID: 38949997 PMCID: PMC11216622 DOI: 10.1371/journal.pbio.3002671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024] Open
Abstract
Mitochondrial shape and network formation have been primarily associated with the well-established processes of fission and fusion. However, recent research has unveiled an intricate and multifaceted landscape of mitochondrial morphology that extends far beyond the conventional fission-fusion paradigm. These less-explored dimensions harbor numerous unresolved mysteries. This review navigates through diverse processes influencing mitochondrial shape and network formation, highlighting the intriguing complexities and gaps in our understanding of mitochondrial architecture. The exploration encompasses various scales, from biophysical principles governing membrane dynamics to molecular machineries shaping mitochondria, presenting a roadmap for future research in this evolving field.
Collapse
Affiliation(s)
- Noga Preminger
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
3
|
Zhu Z, Li J, Shen S, Al-Furas H, Li S, Tong Y, Li Y, Zeng Y, Feng Q, Chen K, Ma N, Zhou F, Zhang Z, Li Z, Pang J, Ding K, Xu F. Targeting EGFR degradation by autophagosome degraders. Eur J Med Chem 2024; 270:116345. [PMID: 38564826 DOI: 10.1016/j.ejmech.2024.116345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/04/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
Several generations of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors have been developed for the treatment of non-small cell lung cancer (NSCLC) in clinic. However, emerging drug resistance mediated by new EGFR mutations or activations by pass, leads to malignant progression of NSCLC. Proteolysis targeting chimeras (PROTACs) have been utilized to overcome the drug resistance acquired by mutant EGFR, newly potent and selective degraders are still need to be developed for clinical applications. Herein, we developed autophagosome-tethering compounds (ATTECs) in which EGFR can be anchored to microtubule-associated protein-1 light chain-3B (LC3B) on the autophagosome with the assistance of the LC3 ligand GW5074. A series of EGFR-ATTECs have been designed and synthesized. Biological evaluations showed that these compounds could degrade EGFR and exhibited moderate inhibitory effects on certain NSCLC cell lines. The ATTEC 12c potently induced the degradation of EGFR with a DC50 value of 0.98 μM and a Dmax value of 81% in HCC827 cells. Mechanistic exploration revealed that the lysosomal pathway was mainly involved in this degradation. Compound 12c also exhibited promising inhibitory activity, as well as degradation efficiency in vivo. Our study highlights that EGFR-ATTECs could be developed as a new expandable EGFR degradation tool and also reveals a novel potential therapeutic strategy to prevent drug resistance acquired EGFR mutations.
Collapse
Affiliation(s)
- ZhongFeng Zhu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Jiaying Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Shujun Shen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Hawaa Al-Furas
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Shengrong Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Yichen Tong
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yi Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Yucheng Zeng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Qianyi Feng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Kaiyue Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Nan Ma
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Fengtao Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China.
| | - Zhang Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Zhengqiu Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Jiyan Pang
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Ke Ding
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China; State Key Laboratory of Chemical Biology, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, China.
| | - Fang Xu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of the People's Republic of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China.
| |
Collapse
|
4
|
Wang S, Zhang K, Song X, Huang Q, Lin S, Deng S, Qi M, Yang Y, Lu Q, Zhao D, Meng F, Li J, Lian Z, Luo C, Yao Y. TLR4 Overexpression Aggravates Bacterial Lipopolysaccharide-Induced Apoptosis via Excessive Autophagy and NF-κB/MAPK Signaling in Transgenic Mammal Models. Cells 2023; 12:1769. [PMID: 37443803 PMCID: PMC10340758 DOI: 10.3390/cells12131769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/25/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Gram-negative bacterial infections pose a significant threat to public health. Toll-like receptor 4 (TLR4) recognizes bacterial lipopolysaccharide (LPS) and induces innate immune responses, autophagy, and cell death, which have major impacts on the body's physiological homeostasis. However, the role of TLR4 in bacterial LPS-induced autophagy and apoptosis in large mammals, which are closer to humans than rodents in many physiological characteristics, remains unknown. So far, few reports focus on the relationship between TLR, autophagy, and apoptosis in large mammal levels, and we urgently need more tools to further explore their crosstalk. Here, we generated a TLR4-enriched mammal model (sheep) and found that a high-dose LPS treatment blocked autophagic degradation and caused strong innate immune responses and severe apoptosis in monocytes/macrophages of transgenic offspring. Excessive accumulation of autophagosomes/autolysosomes might contribute to LPS-induced apoptosis in monocytes/macrophages of transgenic animals. Further study demonstrated that inhibiting TLR4 downstream NF-κB or p38 MAPK signaling pathways reversed the LPS-induced autophagy activity and apoptosis. These results indicate that the elevated TLR4 aggravates LPS-induced monocytes/macrophages apoptosis by leading to lysosomal dysfunction and impaired autophagic flux, which is associated with TLR4 downstream NF-κB and MAPK signaling pathways. This study provides a novel TLR4-enriched mammal model to study its potential effects on autophagy activity, inflammation, oxidative stress, and cell death. These findings also enrich the biological functions of TLR4 and provide powerful evidence for bacterial infection.
Collapse
Affiliation(s)
- Sutian Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China (C.L.)
| | - Kunli Zhang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
| | - Xuting Song
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Qiuyan Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China (C.L.)
| | - Sen Lin
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Shoulong Deng
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Meiyu Qi
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150028, China
| | - Yecheng Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China (C.L.)
| | - Qi Lu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Duowei Zhao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Fanming Meng
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China (C.L.)
| | - Jianhao Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China (C.L.)
| | - Zhengxing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100083, China
| | - Chenglong Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China (C.L.)
| | - Yuchang Yao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
5
|
Kamihara T, Hirashiki A, Kokubo M, Shimizu A. Transcriptome Discovery of Genes in the Three Phases of Autophagy That Are Upregulated During Atrial Fibrillation. Circ Rep 2023; 5:114-122. [PMID: 37025933 PMCID: PMC10072901 DOI: 10.1253/circrep.cr-22-0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 04/08/2023] Open
Abstract
Background: Autophagy may contribute to the maintenance of atrial fibrillation (AF), but no previous study has concurrently surveyed all 3 phases of autophagy, namely autophagosome formation, lysosome formation, and autophagosome-lysosome fusion. Here we aimed to identify disorders involving various phases of autophagy during AF. Methods and Results: We used bioinformatic techniques to analyze publicly available DNA microarray datasets from the left atrium (LA) and right atrium (RA) of 7 patients with AF and 6 patients with normal sinus rhythm who underwent valvular surgeries. We compared gene expression levels in the LA (AF-LA) and RA of patients with AF with those in the LA and RA of patients with normal sinus rhythm. Several differentially expressed genes in the AF-LA sample were significantly associated with the Gene Ontogeny term 'Autophagy', indicating that the expression of autophagic genes was specifically altered in this dataset. In particular, the expression of genes known or suspected to be involved in autophagosome formation (autophagy related 5 [ATG5], autophagy related 10 [ATG10], autophagy related 12 [ATG12], and light chain 3B [LC3B]), lysosome formation (lysosomal associated membrane protein 1 [LAMP1] and lysosomal associated membrane protein 2 [LAMP2]), and autophagosome-lysosome fusion (synaptosome associated protein 29 [SNAP29], SNAP associated protein [SNAPIN], and syntaxin 17 [STX17]) was significantly upregulated in the LA-AF dataset. Conclusions: Autophagy is activated excessively in, and may perpetuate, AF.
Collapse
Affiliation(s)
- Takahiro Kamihara
- Department of Cardiology, National Center for Geriatrics and Gerontology Obu Japan
| | - Akihiro Hirashiki
- Department of Cardiology, National Center for Geriatrics and Gerontology Obu Japan
| | - Manabu Kokubo
- Department of Cardiology, National Center for Geriatrics and Gerontology Obu Japan
| | - Atsuya Shimizu
- Department of Cardiology, National Center for Geriatrics and Gerontology Obu Japan
| |
Collapse
|
6
|
D’Amico R, Impellizzeri D, Cordaro M, Siracusa R, Interdonato L, Marino Y, Crupi R, Gugliandolo E, Macrì F, Di Paola D, Peritore AF, Fusco R, Cuzzocrea S, Di Paola R. Complex Interplay between Autophagy and Oxidative Stress in the Development of Endometriosis. Antioxidants (Basel) 2022; 11:antiox11122484. [PMID: 36552692 PMCID: PMC9774576 DOI: 10.3390/antiox11122484] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Endometriosis (Endo) is a chronic gynecological disease. This paper aimed to evaluate the modulation of autophagy, oxidative stress and apoptosis with Açai Berries in a rat model of endometriosis. Endometriosis was induced with an intraperitoneal injection of minced uterus tissue from a donor rat into a recipient one. The abdominal high-frequency ultrasound (hfUS) analysis was performed at 7 and 14 days from the endometriosis induction to evaluate the growth of the lesion during the experiment. Seven days from the induction, once the lesions were implanted, an Açai Berry was administered daily by gavage for the next seven days. At the end of the experiment, the hfUS analysis showed a reduced lesion diameter in animals given the Açai Berry. A macroscopical and histological analysis confirmed this result. From the molecular point of view, Western blot analyses were conducted to evaluate the autophagy induction. Samples collected from the Endo group showed impaired autophagy, while the Açai Berry administration inhibited PI3K and AKT and ERK1/2 phosphorylation and promoted autophagy by inactivating mTOR. Additionally, Açai Berry administration dephosphorylated ATG1, promoting the activity of the ATG1/ULK1 complex that recruited Ambra1/Beclin1 and Atg9 to promote autophagosome nucleation and LC3II expression. Açai Berry administration also restored mitophagy, which increased Parkin cytosolic expression. The Açai Berry increased the expression of NRF2 in the nucleus and the expression of its downstream antioxidant proteins as NQO-1 and HO-1, thereby restoring the oxidative imbalance. It also restored the impaired apoptotic pathway by reducing BCL-2 and increasing BAX expression. This result was also confirmed by the TUNEL assay. Overall, our results displayed that Açai Berry administration was able to modulate autophagy, oxidative stress and apoptosis during endometriosis.
Collapse
Affiliation(s)
- Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Ylenia Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, Viale Annunzita, 98168 Messina, Italy
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, Viale Annunzita, 98168 Messina, Italy
| | - Francesco Macrì
- Department of Veterinary Sciences, University of Messina, Viale Annunzita, 98168 Messina, Italy
| | - Davide Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
- Correspondence:
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, n 31, 98166 Messina, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, Viale Annunzita, 98168 Messina, Italy
| |
Collapse
|
7
|
Zhang C, Ye L, Wang C, Xiong X, Li Y, Li P, Zhang X, Yu H. Toxic Effect of Combined Exposure of Microplastics and Copper on Goldfish (Carassius auratus): Insight from Oxidative Stress, Inflammation, Apoptosis and Autophagy in Hepatopancreas and Intestine. BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2022; 109:1029-1036. [PMID: 35908221 DOI: 10.1007/s00128-022-03585-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/07/2022] [Indexed: 06/15/2023]
Abstract
The enhancement of the toxic effect of microplastics (MPs) on heavy metals and its mechanism needs more in-depth and systematic research. In this study, the copper (Cu) accumulation, histological injury, and expression of genes involved in oxidative stress, inflammation, apoptosis, and autophagy of goldfish after single or combined exposure of MPs (1 mg/L) and Cu2+ (0.1 mg/L) for 7 days were determined. The results demonstrated that MPs enhanced the Cu accumulation in hepatopancreas and intestine of goldfish and induced more severe oxidative stress in the hepatopancreas and intestine of goldfish. Additionally, combined exposure of MPs and Cu induced inflammation, excessive apoptosis and insufficient autophagy in the hepatopancreas. Contrary, the inflammation and apoptosis were depressed in the intestine after combined exposure of MPs and Cu, which still requires further exploration. Hence, these findings provide further evidence for the threat of MPs and its adsorbed heavy metals.
Collapse
Affiliation(s)
- Cheng Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Limin Ye
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Chi Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaofan Xiong
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yanyao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Pengju Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaotian Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Haibo Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
8
|
Furuya F, Fujita Y, Matsuo N, Minamino H, Oguri Y, Isomura N, Ikeda K, Takesue K, Li Y, Kondo A, Mano F, Inagaki N. Liver autophagy-induced valine and leucine in plasma reflect the metabolic effect of sodium glucose co-transporter 2 inhibitor dapagliflozin. EBioMedicine 2022; 86:104342. [PMID: 36423374 PMCID: PMC9682354 DOI: 10.1016/j.ebiom.2022.104342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 09/14/2022] [Accepted: 10/19/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Sodium glucose co-transporter 2 (SGLT2) inhibitors are anti-diabetic drugs for type 2 diabetes that lower blood glucose levels and body weight. It is of special interest that SGLT2 inhibitors also improve liver metabolism and fatty liver. Liver is an important organ in regulation of energy metabolism, but the metabolic action of SGLT inhibitors in liver remains unclear. METHODS We investigated the factors associated with the beneficial effects of dapagliflozin, a SGLT2 inhibitor, in the liver after confirming its glucose-lowering and weight loss effects using an obesity and diabetes mouse model. We also performed clinical study of patients with type 2 diabetes to explore candidate biomarkers that reflect the beneficial action of dapagliflozin in the liver. FINDINGS In animal study, dapagliflozin induced autophagy in the liver (LC3-II to LC3-I expression ratio: P < 0·05 vs. control), and valine and leucine levels were increased in plasma (P < 0·01 vs. control) as well as in liver (P < 0·05 vs. control). Thus, increased plasma valine and leucine levels are potential biomarkers for improved liver metabolism. Clinical study found that valine and leucine levels were markedly higher in patients treated with dapagliflozin (valine: P < 0·05 vs. control, leucine: P < 0·01 vs. control) than those not treated after one week intervention. INTERPRETATION Dapagliflozin improves liver metabolism via hepatic autophagy, and plasma valine and leucine levels may reflect its metabolic effect. FUNDING AstraZeneca K.K., Ono Pharmaceutical Co., Ltd., Ministry of Education, Culture, Sports, Science and Technology (MEXT), Japan Society for the Promotion of Science (JSPS), Japan Agency for Medical Research and Development (AMED), Novo Nordisk Pharma Ltd., and Japan Foundation for Applied Enzymology, and MSD Life Science Foundation International.
Collapse
Affiliation(s)
| | - Yoshihito Fujita
- Corresponding author. Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | | | | | | | | | | | | | | | | | | | - Nobuya Inagaki
- Corresponding author. Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
9
|
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
|
10
|
Hua L, Zhang Q, Zhu X, Wang R, You Q, Wang L. Beyond Proteolysis-Targeting Chimeric Molecules: Designing Heterobifunctional Molecules Based on Functional Effectors. J Med Chem 2022; 65:8091-8112. [PMID: 35686733 DOI: 10.1021/acs.jmedchem.2c00316] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, with the successful development of proteolysis-targeting chimeric molecules (PROTACs), the potential of heterobifunctional molecules to contribute to reenvisioning drug design, especially small-molecule drugs, has been increasingly recognized. Inspired by PROTACs, diverse heterobifunctional molecules have been reported to simultaneously bind two or more molecules and bring them into proximity to interaction, such as ribonuclease-recruiting, autophagy-recruiting, lysosome-recruiting, kinase-recruiting, phosphatase-recruiting, glycosyltransferase-recruiting, and acetyltransferase-recruiting chimeras. On the basis of the heterobifunctional principle, more opportunities for advancing drug design by linking potential effectors to a protein of interest (POI) have emerged. Herein, we introduce heterobifunctional molecules other than PROTACs, summarize the limitations of existing molecules, list the main challenges, and propose perspectives for future research directions, providing insight into alternative design strategies based on substrate-proximity-based targeting.
Collapse
Affiliation(s)
- Liwen Hua
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, P. R. China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R.China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, P. R. China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R.China
| | - Xinyue Zhu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, P. R. China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R.China
| | - Ruoning Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, P. R. China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R.China
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, P. R. China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R.China
| |
Collapse
|
11
|
Qiu F, Yuan Y, Luo W, Gong YS, Zhang ZM, Liu ZM, Gao L. Asiatic acid alleviates ischemic myocardial injury in mice by modulating mitophagy- and glycophagy-based energy metabolism. Acta Pharmacol Sin 2022; 43:1395-1407. [PMID: 34522006 PMCID: PMC9160258 DOI: 10.1038/s41401-021-00763-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023]
Abstract
Myocardial infarction (MI) causes disturbances in myocardial energy metabolism, ultimately leading to a poor prognosis. Cytosolic glycogen autophagy (glycophagy) and mitochondrial autophagy (mitophagy) are upregulated in MI to optimize energy metabolism but to a limited extent. Asiatic acid (AA), a pentacyclic triterpene derived from the traditional Chinese herb Centella asiatica, displays anti-inflammatory, antioxidant, and antiapoptotic activities. AA has been found to alleviate focal cerebral and liver ischemic injury by reversing mitochondrial dysfunction. In this study, we investigated whether AA exerted cardioprotective effects against MI by activating glycophagy and mitophagy to improve the energy balance. In vitro cardioprotective effects were examined in neonatal mouse cardiomyocytes subjected to oxygen-glucose deprivation for 12 h. Treatment with AA (2-50 μM) significantly increased cell viability and improved the energy metabolism evidenced by increased ATP level and phosphocreatine/ATP ratio. In vivo cardioprotective effects were studied in a mouse model of MI. Administration of AA (5-125 mg·kg-1·d-1, ig) significantly reduced infarct size and ischemic myocardial injury, and improved cardiac function. AA treatment also promoted mitophagy and relieved mitochondrial edema evidenced by increased number of mitophagosomes in ischemic myocardium in vivo and increased mitochondria-light chain 3 (LC3)-II colocalization in ODG-treated cardiomyocytes in vitro. Mitophagy activation was accompanied by activation of the AMPK signaling pathway. Knockdown of AMPK abolished AA-activated mitophagy. Furthermore, we showed that glycophagy was upregulated in OGD cardiomyocytes evidenced by increased starch binding domain protein 1 (STBD1)-GABA type A receptor-associated protein-like 1(GABARAPL1) interaction and extracellular acidification rate, whereas AA treatment further promoted glycophagy accompanied by PI3K/Akt activation. PI3K inhibitor LY294002 or Akt inhibitor GSK690693 blocked the effects of AA on glycophagy and glycolysis. Finally, simultaneous inhibition of glycophagy and mitophagy abolished the cardioprotective effects and energy regulation of AA. These results demonstrate that AA protects ischemic cardiomyocytes by modulating glycophagy- and mitophagy-based energy metabolism through the PI3K/Akt and AMPK pathways.
Collapse
Affiliation(s)
- Fan Qiu
- grid.452753.20000 0004 1799 2798Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China ,grid.452753.20000 0004 1799 2798Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yi Yuan
- grid.452753.20000 0004 1799 2798Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Wei Luo
- grid.452753.20000 0004 1799 2798Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China ,grid.452753.20000 0004 1799 2798Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yan-shan Gong
- grid.452753.20000 0004 1799 2798Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China
| | - Zhong-ming Zhang
- grid.413389.40000 0004 1758 1622Department of Cardiovascular and Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China
| | - Zhong-min Liu
- grid.452753.20000 0004 1799 2798Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China ,grid.452753.20000 0004 1799 2798Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China ,grid.452753.20000 0004 1799 2798Shanghai Institute of Stem Cell Research and Clinical translation, Shanghai East Hospital, Tongji University, Shanghai 200120, China ,Shanghai Engineering Research Center for Stem Cell Clinical Treatment, Shanghai 200123, China
| | - Ling Gao
- grid.452753.20000 0004 1799 2798Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200123, China ,grid.452753.20000 0004 1799 2798Shanghai Institute of Stem Cell Research and Clinical translation, Shanghai East Hospital, Tongji University, Shanghai 200120, China ,Shanghai Engineering Research Center for Stem Cell Clinical Treatment, Shanghai 200123, China
| |
Collapse
|
12
|
Wang Y, Hu S, Shen L, Liu S, Wan L, Yang S, Hou M, Tian X, Zhang H, Xu KF. Dynamic Observation of Autophagy and Transcriptome Profiles in a Mouse Model of Bleomycin-Induced Pulmonary Fibrosis. Front Mol Biosci 2021; 8:664913. [PMID: 34395518 PMCID: PMC8358296 DOI: 10.3389/fmolb.2021.664913] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/30/2021] [Indexed: 11/13/2022] Open
Abstract
Pulmonary fibrosis is a group of progressive, fibrotic, and fatal lung diseases, and the role of autophagy in pulmonary fibrosis is controversial. In the current research, we dynamically observed a bleomycin-induced pulmonary fibrosis mouse model after 3, 7, 14, 21, and 28 days and investigated the expression of autophagy markers. We found that autophagy markers were not significantly changed on the indicated days in the mouse lung tissue. Then, RNA-Seq was used to analyze the gene expression and associated functions and pathways in fibrotic lung tissue on different days post-bleomycin. In addition, short time series expression miner (STEM) analysis was performed to explore the temporal post-bleomycin gene expression. Through STEM, continually up- or downregulated profiles did not demonstrate the critical role of autophagy in the development of fibrosis. Furthermore, gene ontology (GO) annotations showed that continually upregulated profiles were mainly related to fibrosis synthesis, extracellular space, and inflammation, while enriched pathways were mainly related to the PI3K-Akt signaling pathway, ECM-receptor interactions, and focal adhesion signaling pathway. For continually downregulated profiles, GO annotations mainly involved sarcomere organization, muscle contraction, and muscle fiber development. The enriched KEGG signaling pathways were the cAMP signaling pathway, cGMP-PKG signaling pathway, calcium signaling pathway, and cardiac muscle contraction. Moreover, we analyzed autophagy-related genes' expression in specific cells from a publicly available database of three human and one animal study of pulmonary fibrosis using single-cell sequencing technology. All results consistently demonstrated no critical role of autophagy in the pathogenesis of pulmonary fibrosis. In summary, autophagy may not critically and consistently change during the development of pulmonary fibrosis at different stages post-bleomycin in a mouse model. These continually up- or downregulated profiles, including gene profiles, and the corresponding functions and pathways may provide mechanistic insights into IPF therapy.
Collapse
Affiliation(s)
- Yani Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Siqi Hu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Lisha Shen
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Song Liu
- Medical Science Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Linyan Wan
- Department of Physiology, Institutes of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuhui Yang
- Department of Physiology, Institutes of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Mengjie Hou
- Department of Physiology, Institutes of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinlun Tian
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Hongbing Zhang
- Department of Physiology, Institutes of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Kai-Feng Xu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
13
|
Pei J, Wang G, Feng L, Zhang J, Jiang T, Sun Q, Ouyang L. Targeting Lysosomal Degradation Pathways: New Strategies and Techniques for Drug Discovery. J Med Chem 2021; 64:3493-3507. [PMID: 33764774 DOI: 10.1021/acs.jmedchem.0c01689] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A series of tools for targeted protein degradation are inspiring scientists to develop new drugs with advantages over traditional small-molecule drugs. Among these tools, proteolysis-targeting chimeras (PROTACs) are most representative of the technology based on proteasomes. However, the proteasome has little degradation effect on certain macromolecular proteins or aggregates, extracellular proteins, and organelles, which limits the application of PROTACs. Additionally, lysosomes play an important role in protein degradation. Therefore, lysosome-induced protein degradation drugs can directly regulate protein levels in vivo, achieve the goal of treating diseases, and provide new strategies for drug discovery. Lysosome-based degradation technology has the potential for clinical translation. In this review, strategies targeting lysosomal pathways and lysosome-based degradation techniques are summarized. In addition, lysosome-based degrading drugs are described, and the advantages and challenges are listed. Our efforts will certainly promote the design, discovery, and clinical application of drugs associated with this technology.
Collapse
Affiliation(s)
- Junping Pei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Lu Feng
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Tingting Jiang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
14
|
Jiao L, Shao Y, Yu Q, Li M, Wang Y, Gong M, Yang X, Liu T, Li Z, Liu H, Zhang Y, Tan Z, Sun L, Xuan L, Yin H, Zhang Y, Cai B, Zhang Y, Yang B. GDF11 replenishment protects against hypoxia-mediated apoptosis in cardiomyocytes by regulating autophagy. Eur J Pharmacol 2020; 885:173495. [DOI: 10.1016/j.ejphar.2020.173495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
|
15
|
Lechauve C, Keith J, Khandros E, Fowler S, Mayberry K, Freiwan A, Thom CS, Delbini P, Romero EB, Zhang J, Motta I, Tillman H, Cappellini MD, Kundu M, Weiss MJ. The autophagy-activating kinase ULK1 mediates clearance of free α-globin in β-thalassemia. Sci Transl Med 2020; 11:11/506/eaav4881. [PMID: 31434755 DOI: 10.1126/scitranslmed.aav4881] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 03/26/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022]
Abstract
In β-thalassemia, accumulated free α-globin forms intracellular precipitates that impair erythroid cell maturation and viability. Protein quality control systems mitigate β-thalassemia pathophysiology by degrading toxic free α-globin, although the associated mechanisms are poorly understood. We show that loss of the autophagy-activating Unc-51-like kinase 1 (Ulk1) gene in β-thalassemic mice reduces autophagic clearance of α-globin in red blood cell precursors and exacerbates disease phenotypes, whereas inactivation of the canonical autophagy-related 5 (Atg5) gene has relatively minor effects. Systemic treatment with the mTORC1 inhibitor rapamycin reduces α-globin precipitates and lessens pathologies in β-thalassemic mice via an ULK1-dependent pathway. Similarly, rapamycin reduces free α-globin accumulation in erythroblasts derived from CD34+ cells of β-thalassemic individuals. Our findings define a drug-regulatable pathway for ameliorating β-thalassemia.
Collapse
Affiliation(s)
- Christophe Lechauve
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Julia Keith
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Eugene Khandros
- Department of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stephanie Fowler
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kalin Mayberry
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abdullah Freiwan
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christopher S Thom
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paola Delbini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Emilio Boada Romero
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jingjing Zhang
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Irene Motta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Heather Tillman
- Departments of Pathology and Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - M Domenica Cappellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Mondira Kundu
- Departments of Pathology and Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
16
|
Toralova T, Kinterova V, Chmelikova E, Kanka J. The neglected part of early embryonic development: maternal protein degradation. Cell Mol Life Sci 2020; 77:3177-3194. [PMID: 32095869 PMCID: PMC11104927 DOI: 10.1007/s00018-020-03482-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 01/24/2020] [Accepted: 02/07/2020] [Indexed: 12/28/2022]
Abstract
The degradation of maternally provided molecules is a very important process during early embryogenesis. However, the vast majority of studies deals with mRNA degradation and protein degradation is only a very little explored process yet. The aim of this article was to summarize current knowledge about the protein degradation during embryogenesis of mammals. In addition to resuming of known data concerning mammalian embryogenesis, we tried to fill the gaps in knowledge by comparison with facts known about protein degradation in early embryos of non-mammalian species. Maternal protein degradation seems to be driven by very strict rules in terms of specificity and timing. The degradation of some maternal proteins is certainly necessary for the normal course of embryonic genome activation (EGA) and several concrete proteins that need to be degraded before major EGA have been already found. Nevertheless, the most important period seems to take place even before preimplantation development-during oocyte maturation. The defects arisen during this period seems to be later irreparable.
Collapse
Affiliation(s)
- Tereza Toralova
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - Veronika Kinterova
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic.
- Department of Veterinary Sciences, Czech University of Life Sciences in Prague, Prague, Czech Republic.
| | - Eva Chmelikova
- Department of Veterinary Sciences, Czech University of Life Sciences in Prague, Prague, Czech Republic
| | - Jiri Kanka
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| |
Collapse
|
17
|
Wang Z, Li C. Xenophagy in innate immunity: A battle between host and pathogen. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 109:103693. [PMID: 32243873 DOI: 10.1016/j.dci.2020.103693] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 06/11/2023]
Abstract
Autophagy is a fundamental bulk intracellular degradation and recycling process that directly eliminates intracellular microorganisms through "xenophagy" in various types of cells, especially in macrophages. Meanwhile, bacteria have evolved strategies and cellular self-defense mechanisms to prevent autophagosomal degradation and even attack the immune system of host. The lack of knowledge about the roles of autophagy in innate immunity severely limits our understanding of host defensive system and the development of farmed industry consisting of aquaculture. Increasing evidence in recent decades has shown the importance of autophagy. This review focuses on the triggering of xenophagy, targeting of invading pathogens to autophagosomes and elimination in the autophagolysosomes during pathogen infection. How the pathogen can escape from the xenophagy pathway was also discussed. Overall, we aim to reduce diseases and improve industrial production in aquaculture by providing theoretical and technical guidance on xenophagy.
Collapse
Affiliation(s)
- Zhenhui Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, PR China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
18
|
Leboutet R, Chen Y, Legouis R, Culetto E. Mitophagy during development and stress in C. elegans. Mech Ageing Dev 2020; 189:111266. [DOI: 10.1016/j.mad.2020.111266] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022]
|
19
|
Cho HM, Sun W. Molecular cross talk among the components of the regulatory machinery of mitochondrial structure and quality control. Exp Mol Med 2020; 52:730-737. [PMID: 32398745 PMCID: PMC7272630 DOI: 10.1038/s12276-020-0434-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial dysfunction critically impairs cellular health and often causes or affects the progression of several diseases, including neurodegenerative diseases and cancer. Thus, cells must have several ways to monitor the condition of mitochondrial quality and maintain mitochondrial health. Accumulating evidence suggests that the molecular machinery responding to spontaneous changes in mitochondrial morphology is associated with the routine mitochondrial quality control system. In this short review, we discuss recent progress made in linking mitochondrial structural dynamics and the quality control system. The health of mitochondria is important for cellular health, and is maintained by the same mechanisms that control their shape. Mitochondria continuously divide, fuse, elongate, and shrink, forming ever-changing networks inside cells. Damaged mitochondria produce toxic byproducts and have been implicated in neurodegenerative diseases and cancer. Although changes in mitochondrial structure are known to be related to cellular health, the detailed mechanisms are not well understood. In a review, Woong Sun and Hyo Min Cho at the Korea University College of Medicine, Seoul, detail how mitochondrial fusion, division, and recycling are controlled, what signals are used to dispose of damaged mitochondria, and how the shape-control mechanisms also regulate mitochondrial quality. This review will help us to more clearly understand the structure-function relationship of mitochondria.
Collapse
Affiliation(s)
- Hyo Min Cho
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea.
| |
Collapse
|
20
|
Ageta H, Tsuchida K. Post-translational modification and protein sorting to small extracellular vesicles including exosomes by ubiquitin and UBLs. Cell Mol Life Sci 2019; 76:4829-4848. [PMID: 31363817 PMCID: PMC11105257 DOI: 10.1007/s00018-019-03246-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/06/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023]
Abstract
Exosomes, a type of small extracellular vesicles (sEVs), are secreted membrane vesicles that are derived from various cell types, including cancer cells, mesenchymal stem cells, and immune cells via multivesicular bodies (MVBs). These sEVs contain RNAs (mRNA, miRNA, lncRNA, and rRNA), lipids, DNA, proteins, and metabolites, all of which mediate cell-to-cell communication. This communication is known to be implicated in a diverse set of diseases such as cancers and their metastases and degenerative diseases. The molecular mechanisms, by which proteins are modified and sorted to sEVs, are not fully understood. Various cellular processes, including degradation, transcription, DNA repair, cell cycle, signal transduction, and autophagy, are known to be associated with ubiquitin and ubiquitin-like proteins (UBLs). Recent studies have revealed that ubiquitin and UBLs also regulate MVBs and protein sorting to sEVs. Ubiquitin-like 3 (UBL3)/membrane-anchored Ub-fold protein (MUB) acts as a post-translational modification (PTM) factor to regulate efficient protein sorting to sEVs. In this review, we focus on the mechanism of PTM by ubiquitin and UBLs and the pathway of protein sorting into sEVs and discuss the potential biological significance of these processes.
Collapse
Affiliation(s)
- Hiroshi Ageta
- Division for Therapies Against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Kunihiro Tsuchida
- Division for Therapies Against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
21
|
AUTACs: Cargo-Specific Degraders Using Selective Autophagy. Mol Cell 2019; 76:797-810.e10. [DOI: 10.1016/j.molcel.2019.09.009] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 07/09/2019] [Accepted: 09/04/2019] [Indexed: 12/24/2022]
|
22
|
Cao Z, Wang Y, Long Z, He G. Interaction between autophagy and the NLRP3 inflammasome. Acta Biochim Biophys Sin (Shanghai) 2019; 51:1087-1095. [PMID: 31609412 DOI: 10.1093/abbs/gmz098] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Indexed: 12/15/2022] Open
Abstract
Autophagy, a metabolic pathway that plays an important role in maintaining the dynamic balance of cells, has two types, i.e. non-selective autophagy and selective autophagy. The role of non-selective autophagy is primarily to allow cells to circulate nutrients in an energy-limited environment, while selective autophagy primarily cleans up the organelles inside the cells to maintain the cell structure. The NLRP3 inflammasome is an innate immune response produced by the organism that can promote the secretion of interleukin-1β and interleukin-18 through caspase-1 activation and resist the damage of some pathogens. However, when the NLRP3 inflammasome is overactivated, it can cause various inflammatory diseases, such as inflammatory liver disease and inflammatory bowel disease. Many previous studies have shown that autophagy can inhibit the NLRP3 inflammasome, while in recent years, new studies have found that autophagy can also promote the NLRP3 inflammasome in some cases, and the NLRP3 inflammasome can, in turn, affect autophagy. In this review, the interaction between autophagy and the NLRP3 inflammasome is explored, and then the application of this interaction in disease treatment is discussed.
Collapse
Affiliation(s)
- Zhenrui Cao
- Chongqing Key Laboratory of Neurobiology, Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
| | - Yanhao Wang
- Chongqing Key Laboratory of Neurobiology, Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
| | - Zhimin Long
- Chongqing Key Laboratory of Neurobiology, Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
- Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| | - Guiqiong He
- Chongqing Key Laboratory of Neurobiology, Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
- Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
23
|
Owino CO, Chu JJH. Recent advances on the role of host factors during non-poliovirus enteroviral infections. J Biomed Sci 2019; 26:47. [PMID: 31215493 PMCID: PMC6582496 DOI: 10.1186/s12929-019-0540-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
Non-polio enteroviruses are emerging viruses known to cause outbreaks of polio-like infections in different parts of the world with several cases already reported in Asia Pacific, Europe and in United States of America. These outbreaks normally result in overstretching of health facilities as well as death in children under the age of five. Most of these infections are usually self-limiting except for the neurological complications associated with human enterovirus A 71 (EV-A71). The infection dynamics of these viruses have not been fully understood, with most inferences made from previous studies conducted with poliovirus.Non-poliovirus enteroviral infections are responsible for major outbreaks of hand, foot and mouth disease (HFMD) often associated with neurological complications and severe respiratory diseases. The myriad of disease presentations observed so far in children calls for an urgent need to fully elucidate the replication processes of these viruses. There are concerted efforts from different research groups to fully map out the role of human host factors in the replication cycle of these viral infections. Understanding the interaction between viral proteins and human host factors will unravel important insights on the lifecycle of this groups of viruses.This review provides the latest update on the interplay between human host factors/processes and non-polio enteroviruses (NPEV). We focus on the interactions involved in viral attachment, entry, internalization, uncoating, replication, virion assembly and eventual egress of the NPEV from the infected cells. We emphasize on the virus- human host interplay and highlight existing knowledge gaps that needs further studies. Understanding the NPEV-human host factors interactions will be key in the design and development of vaccines as well as antivirals against enteroviral infections. Dissecting the role of human host factors during NPEV infection cycle will provide a clear picture of how NPEVs usurp the human cellular processes to establish an efficient infection. This will be a boost to the drug and vaccine development against enteroviruses which will be key in control and eventual elimination of the viral infections.
Collapse
Affiliation(s)
- Collins Oduor Owino
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, National University of Singapore, Singapore, 117597, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
| |
Collapse
|
24
|
McCormick JJ, King KE, Dokladny K, Mermier CM. Effect of Acute Aerobic Exercise and Rapamycin Treatment on Autophagy in Peripheral Blood Mononuclear Cells of Adults With Prediabetes. Can J Diabetes 2019; 43:457-463. [PMID: 31213408 DOI: 10.1016/j.jcjd.2019.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/04/2019] [Accepted: 04/12/2019] [Indexed: 01/24/2023]
Abstract
OBJECTIVES Recently, a malfunction of the autophagic pathway has been implicated with impaired glucose metabolism and progression from prediabetes to type 2 diabetes. The aims of this study were to investigate the effect of exercise and rapamycin (RAPA) treatment on the autophagic process in peripheral blood mononuclear cells (PBMCs) from people with prediabetes compared with control subjects. METHODS Two groups matched for age and sex served as participants and included 6 participants with prediabetes (42.4±11.7 years) and 6 control subjects (44.4±11.9 years). Participants exercised at 50% of maximal oxygen consumption for 60 min with 5 min of rest interspersed every 20 min. PBMCs were isolated pre-exercise, immediately postexercise and 4 h after exercise recovery. Additional PBMCs were incubated for 24 h and either exposed to bafilomycin, rapamycin with bafilomycin (RAPA), or no treatment with vehicle (dimethyl sulfoxide). Proteins and mRNA were analyzed via western blot and quantitative real-time polymerase chain reaction, respectively. RESULTS Exercise increased autophagy immediately postexercise and recovered 4 h after exercise in control participants but not in participants with prediabetes. Autophagy increased in PBMCs from people with prediabetes and control participants after RAPA treatment; however, a significantly impaired autophagic response was observed in people with prediabetes when compared with control subjects. CONCLUSIONS Our results indicate an impairment in autophagic flux in PBMCs from people with prediabetes when compared with control subjects in response to both exercise and RAPA treatment. Future methods of autophagic upregulation should be investigated to spare malfunctions in autophagy in people with prediabetes.
Collapse
Affiliation(s)
- James J McCormick
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States.
| | - Kelli E King
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| | - Karol Dokladny
- Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, United States
| | - Christine M Mermier
- Department of Health, Exercise, and Sports Sciences, University of New Mexico, Albuquerque, New Mexico, United States
| |
Collapse
|
25
|
Abstract
Originally thought of as a stress response end point, the view of cellular senescence has since evolved into one encompassing a wide range of physiological and pathological functions, including both protumorignic and antitumorigenic features. It has also become evident that senescence is a highly dynamic and heterogenous process. Efforts to reconcile the beneficial and detrimental features of senescence suggest that physiological functions require the transient presence of senescent cells in the tissue microenvironment. Here, we propose the concept of a physiological "senescence life cycle," which has pathological consequences if not executed in its entirety.
Collapse
Affiliation(s)
- Adelyne Sue Li Chan
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| |
Collapse
|
26
|
Abstract
Enteroviruses (EVs) are the most common human pathogens worldwide. Recent international outbreaks in North America and South East Asia have emphasized the need for more effective anti-viral therapies. As obligate parasites, EVs rely on the host cellular machinery for effective viral propagation. Accumulating evidence has indicated that EVs subvert and disrupt the cellular autophagy pathway to facilitate productive infection, and consequently leading to host pathogenesis. Given that defective autophagy is a common factor in various human diseases, including neurodegeneration, cardiomyopathy, and metabolic disorders, a clear understanding of the relationship between EV infection and autophagy is warranted. In this review, we highlight recent advances in understanding the molecular mechanisms by which EVs exploit the autophagy pathway during different steps of viral life cycle, from entry, replication, and maturation to release. We also provide an overview of recent progress in EV subversion of the autophagy for immune evasion.
Collapse
Affiliation(s)
- Yasir Mohamud
- a Center for Heart Lung Innovation, St. Paul' s Hospital and Department of Pathology and Laboratory Medicine , University of British Columbia , Vancouver , BC , Canada
| | - Honglin Luo
- a Center for Heart Lung Innovation, St. Paul' s Hospital and Department of Pathology and Laboratory Medicine , University of British Columbia , Vancouver , BC , Canada
| |
Collapse
|
27
|
Mishra P, Ammanathan V, Manjithaya R. Chemical Biology Strategies to Study Autophagy. Front Cell Dev Biol 2018; 6:160. [PMID: 30538986 PMCID: PMC6277461 DOI: 10.3389/fcell.2018.00160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/06/2018] [Indexed: 11/13/2022] Open
Abstract
Growing amount of evidence in the last two decades highlight that macroautophagy (generally referred to as autophagy) is not only indispensable for survival in yeast but also equally important to maintain cellular quality control in higher eukaryotes as well. Importantly, dysfunctional autophagy has been explicitly shown to be involved in various physiological and pathological conditions such as cell death, cancer, neurodegenerative, and other diseases. Therefore, modulation and regulation of the autophagy pathway has emerged as an alternative strategy for the treatment of various disease conditions in the recent years. Several studies have shown genetic or pharmacological modulation of autophagy to be effective in treating cancer, clearing intracellular aggregates and pathogens. Understanding and controlling the autophagic flux, either through a genetic or pharmacological approach is therefore a highly promising approach and of great scientific interest as spatiotemporal and cell-tissue-organ level autophagy regulation is not clearly understood. Indeed, chemical biology approaches that identify small molecule effectors of autophagy have thus a dual benefit: the modulators act as tools to study and understand the process of autophagy, and may also have therapeutic potential. In this review, we discuss different strategies that have appeared to screen and identify potent small molecule modulators of autophagy.
Collapse
Affiliation(s)
- Piyush Mishra
- Autophagy Lab, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Veena Ammanathan
- Autophagy Lab, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Ravi Manjithaya
- Autophagy Lab, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| |
Collapse
|
28
|
Sánchez-Alegría K, Flores-León M, Avila-Muñoz E, Rodríguez-Corona N, Arias C. PI3K Signaling in Neurons: A Central Node for the Control of Multiple Functions. Int J Mol Sci 2018; 19:ijms19123725. [PMID: 30477115 PMCID: PMC6321294 DOI: 10.3390/ijms19123725] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/13/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022] Open
Abstract
Phosphoinositide 3-kinase (PI3K) signaling contributes to a variety of processes, mediating many aspects of cellular function, including nutrient uptake, anabolic reactions, cell growth, proliferation, and survival. Less is known regarding its critical role in neuronal physiology, neuronal metabolism, tissue homeostasis, and the control of gene expression in the central nervous system in healthy and diseased states. The aim of the present work is to review cumulative evidence regarding the participation of PI3K pathways in neuronal function, focusing on their role in neuronal metabolism and transcriptional regulation of genes involved in neuronal maintenance and plasticity or on the expression of pathological hallmarks associated with neurodegeneration.
Collapse
Affiliation(s)
- Karina Sánchez-Alegría
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510 México, DF, Mexico.
| | - Manuel Flores-León
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510 México, DF, Mexico.
| | - Evangelina Avila-Muñoz
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510 México, DF, Mexico.
| | - Nelly Rodríguez-Corona
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510 México, DF, Mexico.
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510 México, DF, Mexico.
| |
Collapse
|
29
|
Popat A, Patel AA, Warnes G. A Flow Cytometric Study of ER Stress and Autophagy. Cytometry A 2018; 95:672-682. [PMID: 30451364 DOI: 10.1002/cyto.a.23665] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/09/2018] [Accepted: 10/08/2018] [Indexed: 01/18/2023]
Abstract
The mechanistic link between ER stress, autophagy, and resultant cell death was investigated by the use of drugs Thapsigargin (Tg) and Chloroquine (CQ) with prior induction and or blockade of autophagy and apoptosis which modulated the ER stress response and resultant form of cell death. All these biological processes can be measured flow cytometrically allowing the determination of the type of cell death, G1 cell cycle arrest, cell cycle dependent measurement of ER stress transducer PERK, misfolded proteins, reticulophagy, and autophagy marker LC3B. Jurkat cells after Tg or CQ treatment became necrotic and apoptotic, showed G1 cell cycle arrest, autophagy, and ER stress. Prior induction of autophagy before ER stress increased levels of necrotic and apoptotic cell death. Autophagy was further up-regulated, while PERK was reduced or abrogated. CQ showed reduced levels of misfolded proteins and reticulophagy, while Tg showed no change in misfolded protein levels but increased reticulophagy and thus displayed more ER stress. Prior blockade of apoptosis before induction of ER stress resulted in cell survival except with high Tg levels which induced necrosis. Autophagy was up-regulated with modulation of PERK and reticulophagy levels with an abrogation of the misfolded protein response. Blockade of apoptosis with induction of autophagy before ER stress showed death by necrosis with high dose drugs and cell survival with low doses of drugs. CQ induced reduced levels G1 cell cycle arrest while it was maintained with Tg. Autophagy was also maintained with reduced levels of ER stress. These data demonstrates a profound link between the processes of ER stress, autophagy, and the resultant form of cell death all of which can be modulated depending upon the sequence and concentration of drugs employed. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- A Popat
- Flow Cytometry Core Facility, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary London University, London, England
| | - A A Patel
- Flow Cytometry Core Facility, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary London University, London, England
| | - G Warnes
- Flow Cytometry Core Facility, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary London University, London, England
| |
Collapse
|
30
|
Pham HQ, Yoshioka K, Mohri H, Nakata H, Aki S, Ishimaru K, Takuwa N, Takuwa Y. MTMR4, a phosphoinositide-specific 3'-phosphatase, regulates TFEB activity and the endocytic and autophagic pathways. Genes Cells 2018; 23:670-687. [PMID: 29962048 DOI: 10.1111/gtc.12609] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 02/05/2023]
Abstract
Phosphatidylinositol 3-phosphate (PI(3)P) is the predominant phosphoinositide species in early endosomes and autophagosomes, in which PI(3)P dictates traffic of these organelles. Phosphoinositide levels are tightly regulated by lipid-kinases and -phosphatases; however, a phosphatase that converts PI(3)P back to phosphatidylinositol in the endosomal and autophagosomal compartments is not fully understood. We investigated the subcellular distribution and functions of myotubularin-related protein-4 (MTMR4), which is distinct among other MTMRs in that it possesses a PI(3)P-binding FYVE domain, in lung alveolar epithelium-derived A549 cells. MTMR4 was localized mainly in late endosomes and autophagosomes. MTMR4 knockdown markedly suppressed the motility, fusion, and fission of PI(3)P-enriched structures, resulting in decreases in late endosomes, autophagosomes, and lysosomes, and enlargement of PI(3)P-enriched early and late endosomes. In amino acid- and serum-starved cells, MTMR4 knockdown decreased both autophagosomes and autolysosomes and markedly increased PI(3)P-containing autophagosomes and late endosomes, suggesting that the fusion with lysosomes of autophagosomes and late endosomes might be impaired. Notably, MTMR4 knockdown inhibited the nuclear translocation of starvation stress responsive transcription factor-EB (TFEB) with reduced expression of lysosome-related genes in starved cells. These findings indicate that MTMR4 is essential for the integrity of endocytic and autophagic pathways.
Collapse
Affiliation(s)
- Hoa Q Pham
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Kazuaki Yoshioka
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Hiromi Mohri
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Hiroki Nakata
- Department of Histology and Cell Biology, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Sho Aki
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Kazuhiro Ishimaru
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Japan
| | - Noriko Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Japan
- Department of Health and Medical Science, Ishikawa Prefectural Nursing University, Kahoku, Japan
| | - Yoh Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Japan
| |
Collapse
|
31
|
Lang A, Anand R, Altinoluk-Hambüchen S, Ezzahoini H, Stefanski A, Iram A, Bergmann L, Urbach J, Böhler P, Hänsel J, Franke M, Stühler K, Krutmann J, Scheller J, Stork B, Reichert AS, Piekorz RP. SIRT4 interacts with OPA1 and regulates mitochondrial quality control and mitophagy. Aging (Albany NY) 2018; 9:2163-2189. [PMID: 29081403 PMCID: PMC5680561 DOI: 10.18632/aging.101307] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 10/15/2017] [Indexed: 12/13/2022]
Abstract
The stress-responsive mitochondrial sirtuin SIRT4 controls cellular energy metabolism in a NAD+-dependent manner and is implicated in cellular senescence and aging. Here we reveal a novel function of SIRT4 in mitochondrial morphology/quality control and regulation of mitophagy. We report that moderate overexpression of SIRT4, but not its enzymatically inactive mutant H161Y, sensitized cells to mitochondrial stress. CCCP-triggered dissipation of the mitochondrial membrane potential resulted in increased mitochondrial ROS levels and autophagic flux, but surprisingly led to increased mitochondrial mass and decreased Parkin-regulated mitophagy. The anti-respiratory effect of elevated SIRT4 was accompanied by increased levels of the inner-membrane bound long form of the GTPase OPA1 (L-OPA1) that promotes mitochondrial fusion and thereby counteracts fission and mitophagy. Consistent with this, upregulation of endogenous SIRT4 expression in fibroblast models of senescence either by transfection with miR-15b inhibitors or by ionizing radiation increased L-OPA1 levels and mitochondrial fusion in a SIRT4-dependent manner. We further demonstrate that SIRT4 interacts physically with OPA1 in co-immunoprecipitation experiments. Overall, we propose that the SIRT4-OPA1 axis is causally linked to mitochondrial dysfunction and altered mitochondrial dynamics that translates into aging-associated decreased mitophagy based on an unbalanced mitochondrial fusion/fission cycle.
Collapse
Affiliation(s)
- Alexander Lang
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Ruchika Anand
- Institut für Biochemie und Molekularbiologie I, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Simone Altinoluk-Hambüchen
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Hakima Ezzahoini
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Anja Stefanski
- Molecular Proteomics Laboratory (BMFZ), Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Afshin Iram
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Laura Bergmann
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jennifer Urbach
- Institut für Biochemie und Molekularbiologie I, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Philip Böhler
- Institut für Molekulare Medizin I, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jan Hänsel
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Manuel Franke
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory (BMFZ), Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jean Krutmann
- IUF - Leibniz Institut für Umweltmedizinische Forschung, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jürgen Scheller
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Björn Stork
- Institut für Molekulare Medizin I, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Andreas S Reichert
- Institut für Biochemie und Molekularbiologie I, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Roland P Piekorz
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| |
Collapse
|
32
|
Ru YX, Dong SX, Li Y, Zhao SX, Liang HY, Zhu XF, Zheng YZ, Zhang FK. A novel anemia associated with membranous cytoplasm degeneration in 16 patients: an ultrastructural study. Ultrastruct Pathol 2018; 42:350-357. [PMID: 29913101 DOI: 10.1080/01913123.2018.1485807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Sixteen patients with mild anemia and hemolysis were difficult to be classified into any known category based on laboratory examinations and light microscopy. To make a definite diagnosis and investigate the pathomechanism, ultrastructural study was performed on erythroid cells from 16 patients. Transmission electron microscopy demonstrated a series of alterations of cytoplasm, including cytoplasm sequestration, membranous transformation, and degeneration in erythroblasts and reticulocytes at different stages. The affected erythroblasts were usually complicated with chromatin condensation, karyorrhexis, nuclear membrane lysis, and megaloblastic changes. The reticulocytes with the cytoplasm alterations had a huge size from 10 um to 15 um in diameter. The membranous cytoplasm degeneration revealed a unique pathomechanism of dyserythropoiesis and ineffective erythropoiesis in 16 patients with anemia, and suggested a novel anemia category though more details remained to be investigated.
Collapse
Affiliation(s)
- Yong-Xin Ru
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Shu-Xu Dong
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Yuan Li
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Shi-Xuan Zhao
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Hao-Yue Liang
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Xiao-Fan Zhu
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Yi-Zhou Zheng
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| | - Feng-Kui Zhang
- a State Key Laboratory of Experimental Hematology, Peking Union Medical College , Institute of Hematology & Blood Diseases Hospital , Beijing , China
| |
Collapse
|
33
|
Lee JN, Dutta RK, Maharjan Y, Liu ZQ, Lim JY, Kim SJ, Cho DH, So HS, Choe SK, Park R. Catalase inhibition induces pexophagy through ROS accumulation. Biochem Biophys Res Commun 2018; 501:696-702. [DOI: 10.1016/j.bbrc.2018.05.050] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/09/2018] [Indexed: 12/15/2022]
|
34
|
Liu Y, Liu HQ, Xiao JY, Ma KT, Wang XQY, Shen HJ, Luo JD. Autophagy is involved in the protective effect of endophilin A2 on H2O2-induced apoptosis in H9C2 cardiomyocytes. Biochem Biophys Res Commun 2018; 499:299-306. [DOI: 10.1016/j.bbrc.2018.03.151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 03/20/2018] [Indexed: 10/17/2022]
|
35
|
Kong EY, Cheng SH, Yu KN. Induction of autophagy and interleukin 6 secretion in bystander cells: metabolic cooperation for radiation-induced rescue effect? JOURNAL OF RADIATION RESEARCH 2018; 59:129-140. [PMID: 29385614 PMCID: PMC5951087 DOI: 10.1093/jrr/rrx101] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Indexed: 05/06/2023]
Abstract
We hypothesized that radiation-induced rescue effect (RIRE) shared similar mechanisms with 'metabolic cooperation', in which nutrient-deprived cancer cells prompted normal cells to provide nutrients. Our data demonstrated that X-ray irradiation induced autophagy in HeLa cells, which could last at least 18 h, and proved that the irradiated cells (IRCs) resorted to breaking down their own intracellular components to supply the molecules required for cell-repair enhancement (e.g. to activate the NF-κB pathway) in the absence of support from bystander unirradiated cells (UICs). Furthermore, autophagy accumulation in IRCs was significantly reduced when they were partnered with UICs, and more so with UICs with pre-induced autophagy before partnering (through starvation using Earle's Balanced Salt Solution), which showed that the autophagy induced in UICs supported the IRCs. Our results also showed that interleukin 6 (IL-6) was secreted by bystander UICs, particularly the UICs with pre-induced autophagy, when they were cultured in the medium having previously conditioned irradiated HeLa cells. It was established that autophagy could activate the signal transducer and activator of transcription 3 (STAT3) that was required for the IL-6 production in the autophagy process. Taken together, the metabolic cooperation of RIRE was likely initiated by the bystander factors released from IRCs, which induced autophagy and activated STAT3 to produce IL-6 in bystander UICs, and was finally manifested in the activation of the NF-κB pathway in IRCs by the IL-6 secreted by the UICs.
Collapse
Affiliation(s)
- Eva Yi Kong
- Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Shuk Han Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
- State Key Laboratory in Marine Pollution, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Kwan Ngok Yu
- Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
- State Key Laboratory in Marine Pollution, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong
- Corresponding author: Tel: +852-344-27812; Fax: +852-344-20538;
| |
Collapse
|
36
|
Antognelli C, Trapani E, Delle Monache S, Perrelli A, Daga M, Pizzimenti S, Barrera G, Cassoni P, Angelucci A, Trabalzini L, Talesa VN, Goitre L, Retta SF. KRIT1 loss-of-function induces a chronic Nrf2-mediated adaptive homeostasis that sensitizes cells to oxidative stress: Implication for Cerebral Cavernous Malformation disease. Free Radic Biol Med 2018; 115:202-218. [PMID: 29170092 PMCID: PMC5806631 DOI: 10.1016/j.freeradbiomed.2017.11.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 10/18/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
KRIT1 (CCM1) is a disease gene responsible for Cerebral Cavernous Malformations (CCM), a major cerebrovascular disease of proven genetic origin affecting 0.3-0.5% of the population. Previously, we demonstrated that KRIT1 loss-of-function is associated with altered redox homeostasis and abnormal activation of the redox-sensitive transcription factor c-Jun, which collectively result in pro-oxidative, pro-inflammatory and pro-angiogenic effects, suggesting a novel pathogenic mechanism for CCM disease and raising the possibility that KRIT1 loss-of-function exerts pleiotropic effects on multiple redox-sensitive mechanisms. To address this possibility, we investigated major redox-sensitive pathways and enzymatic systems that play critical roles in fundamental cytoprotective mechanisms of adaptive responses to oxidative stress, including the master Nrf2 antioxidant defense pathway and its downstream target Glyoxalase 1 (Glo1), a pivotal stress-responsive defense enzyme involved in cellular protection against glycative and oxidative stress through the metabolism of methylglyoxal (MG). This is a potent post-translational protein modifier that may either contribute to increased oxidative molecular damage and cellular susceptibility to apoptosis, or enhance the activity of major apoptosis-protective proteins, including heat shock proteins (Hsps), promoting cell survival. Experimental outcomes showed that KRIT1 loss-of-function induces a redox-sensitive sustained upregulation of Nrf2 and Glo1, and a drop in intracellular levels of MG-modified Hsp70 and Hsp27 proteins, leading to a chronic adaptive redox homeostasis that counteracts intrinsic oxidative stress but increases susceptibility to oxidative DNA damage and apoptosis, sensitizing cells to further oxidative challenges. While supporting and extending the pleiotropic functions of KRIT1, these findings shed new light on the mechanistic relationship between KRIT1 loss-of-function and enhanced cell predisposition to oxidative damage, thus providing valuable new insights into CCM pathogenesis and novel options for the development of preventive and therapeutic strategies.
Collapse
Affiliation(s)
| | - Eliana Trapani
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Science, University of L'Aquila, Italy
| | - Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Martina Daga
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Paola Cassoni
- Department of Medical Sciences, University of Torino, Italy
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Science, University of L'Aquila, Italy
| | - Lorenza Trabalzini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Italy
| | | | - Luca Goitre
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy.
| |
Collapse
|
37
|
Shimizu S. Biological Roles of Alternative Autophagy. Mol Cells 2018; 41:50-54. [PMID: 29370693 PMCID: PMC5792713 DOI: 10.14348/molcells.2018.2215] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 01/07/2023] Open
Abstract
Atg5 and Atg7 have long been considered as essential molecules for autophagy. However, we found that cells lacking these molecules still form autophagic vacuoles and perform autophagic protein degradation when subjected to certain stressors. During this unconventional autophagy pathway, autophagosomes appeared to be generated in a Rab9-dependent manner by the fusion of vesicles derived from the trans-Golgi and late endosomes. Therefore, mammalian autophagy can occur via at least two different pathways; the Atg5/Atg7-dependent conventional pathway and an Atg5/Atg7-independent alternative pathway.
Collapse
Affiliation(s)
- Shigeomi Shimizu
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510,
Japan
| |
Collapse
|
38
|
Ntsapi C, Lumkwana D, Swart C, du Toit A, Loos B. New Insights Into Autophagy Dysfunction Related to Amyloid Beta Toxicity and Neuropathology in Alzheimer's Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 336:321-361. [DOI: 10.1016/bs.ircmb.2017.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
39
|
From autophagy to mitophagy: the roles of P62 in neurodegenerative diseases. J Bioenerg Biomembr 2017; 49:413-422. [DOI: 10.1007/s10863-017-9727-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/20/2017] [Indexed: 12/31/2022]
|
40
|
Mishra P, Dauphinee AN, Ward C, Sarkar S, Gunawardena AHLAN, Manjithaya R. Discovery of pan autophagy inhibitors through a high-throughput screen highlights macroautophagy as an evolutionarily conserved process across 3 eukaryotic kingdoms. Autophagy 2017; 13:1556-1572. [PMID: 28792845 PMCID: PMC5612355 DOI: 10.1080/15548627.2017.1339002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Due to the involvement of macroautophagy/autophagy in different pathophysiological conditions such as infections, neurodegeneration and cancer, identification of novel small molecules that modulate the process is of current research and clinical interest. In this work, we developed a luciferase-based sensitive and robust kinetic high-throughput screen (HTS) of small molecules that modulate autophagic degradation of peroxisomes in the budding yeast Saccharomyces cerevisiae. Being a pathway-specific rather than a target-driven assay, we identified small molecule modulators that acted at key steps of autophagic flux. Two of the inhibitors, Bay11 and ZPCK, obtained from the screen were further characterized using secondary assays in yeast. Bay11 inhibited autophagy at a step before fusion with the vacuole whereas ZPCK inhibited the cargo degradation inside the vacuole. Furthermore, we demonstrated that these molecules altered the process of autophagy in mammalian cells as well. Strikingly, these molecules also modulated autophagic flux in a novel model plant, Aponogeton madagascariensis. Thus, using small molecule modulators identified by using a newly developed HTS autophagy assay, our results support that macroautophagy is a conserved process across fungal, animal and plant kingdoms.
Collapse
Affiliation(s)
- Piyush Mishra
- a Molecular Biology and Genetics Unit , Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bangalore , India
| | - Adrian N Dauphinee
- b Biology Department, Life Sciences Centre , Dalhousie University , Halifax , NS , Canada
| | - Carl Ward
- c Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences , University of Birmingham , Edgbaston, Birmingham , UK
| | - Sovan Sarkar
- c Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences , University of Birmingham , Edgbaston, Birmingham , UK
| | | | - Ravi Manjithaya
- a Molecular Biology and Genetics Unit , Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bangalore , India
| |
Collapse
|
41
|
Mishra P, Rai S, Manjithaya R. A novel dual luciferase based high throughput assay to monitor autophagy in real time in yeast S. cerevisiae. Biochem Biophys Rep 2017; 11:138-146. [PMID: 28955778 PMCID: PMC5614714 DOI: 10.1016/j.bbrep.2017.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 06/09/2017] [Accepted: 07/20/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Macroautophagy is a cellular response to starvation wherein superfluous and damaged cytoplasmic constituents are degraded to provide energy for survival and to maintain cellular homeostasis. Dysfunctional autophagy is attributed to disease progression in several pathological conditions and therefore, autophagy has appeared as a potential pharmacological target for such conditions. OBJECTIVE In search of potential drugs that modulate autophagy, identifying small molecule effectors of autophagy is the primary step. The conventional autophagy assays have a limitation that they cannot be scaled down to a high throughput format, therefore, novel sensitive assays are needed to discover new candidate molecules. Keeping this rationale in mind, a dual luciferase based assay was developed in the yeast S. cerevisiae that could measure both selective and general autophagy in real time. METHODS Firefly and Renilla luciferase reporter genes were cloned under POT-1 promoter. Using fatty acid medium the promoter was induced and the luciferase cargo was allowed to build up. The cells were then transferred to starvation conditions to stimulate autophagy and the degradation of luciferase markers was followed with time. RESULTS AND CONCLUSION The assay was more sensitive than conventional assays and could be scaled down to a 384 well format using an automated system. A good Z-factor score indicated that the assay is highly suitable for High Throughput Screening (HTS) of small molecule libraries. Screening of a small molecule library with our assay identified several known and novel modulators of autophagy.
Collapse
Affiliation(s)
- Piyush Mishra
- Autophagy Lab, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India
| | - Shashank Rai
- Autophagy Lab, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India
| | - Ravi Manjithaya
- Autophagy Lab, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India
| |
Collapse
|
42
|
Bajagic M, Archna A, Büsing P, Scrima A. Structure of the WD40-domain of human ATG16L1. Protein Sci 2017; 26:1828-1837. [PMID: 28685931 DOI: 10.1002/pro.3222] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/21/2017] [Accepted: 06/29/2017] [Indexed: 12/19/2022]
Abstract
Autophagy-related protein ATG16L1 is a component of the mammalian ATG12∼ATG5/ATG16L1 complex, which acts as E3-ligase to catalyze lipidation of LC3 during autophagosome biogenesis. The N-terminal part of ATG16L1 comprises the ATG5-binding site and coiled-coil dimerization domain, both also present in yeast ATG16 and essential for bulk and starvation induced autophagy. While absent in yeast ATG16, mammalian ATG16L1 further contains a predicted C-terminal WD40-domain, which has been shown to be involved in mediating interaction with diverse factors in the context of alternative functions of autophagy, such as inflammatory control and xenophagy. In this work, we provide detailed information on the domain boundaries of the WD40-domain of human ATG16L1 and present its crystal structure at a resolution of 1.55 Å.
Collapse
Affiliation(s)
- Milica Bajagic
- Structural Biology of Autophagy Group, Department of Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, 38124, Germany
| | - Archna Archna
- Structural Biology of Autophagy Group, Department of Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, 38124, Germany
| | - Petra Büsing
- Structural Biology of Autophagy Group, Department of Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, 38124, Germany
| | - Andrea Scrima
- Structural Biology of Autophagy Group, Department of Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, 38124, Germany
| |
Collapse
|
43
|
Abstract
Mitophagy is a mitochondrial quality control mechanism where damaged and surplus mitochondria are removed by autophagy. There are at least two different mammalian autophagy pathways: the Atg5-dependent conventional pathway and an Atg5-independent alternative pathway; the latter is involved in the erythrocyte mitophagy. In this chapter we describe the various experimental approaches to assess Atg5-indepedndent mitophagy in mammalian cells.
Collapse
|
44
|
Yang N, Dang S, Shi J, Wu F, Li M, Zhang X, Li Y, Jia X, Zhai S. Caffeic acid phenethyl ester attenuates liver fibrosis via inhibition of TGF-β1/Smad3 pathway and induction of autophagy pathway. Biochem Biophys Res Commun 2017; 486:22-28. [PMID: 28193525 DOI: 10.1016/j.bbrc.2017.02.057] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 02/09/2017] [Indexed: 02/06/2023]
Abstract
Caffeic acid phenethyl ester (CAPE) has been reported to possess the hepatoprotective effect. This study was to investigate the mechanism underlying CAPE against liver fibrosis in a liver fibrosis model induced by toxic carbon tetrachloride (CCl4) in male Sprague-Dawley rats and in vitro in CAPE (5 μM, 10 μM, 15 μM) treated hepatic stellate cells (HSC-T6). We found that CAPE treatment remarkably attenuated CCl4-induced liver fibrosis by blocking the activation of HSCs as determined by the expression alternation of transforming growth factor (TGF)-β1, phosphorylated Smad3 (p-Smad3), collage I, α-smooth muscle actin (α-SMA), matrix metalloproteinases (MMPs) 2, tissue inhibitor of matrix metalloproteinases (TIMPs) 1. The hepatoprotective effects of CAPE were also associated with upregulation of autophasomes in HSCs as determined by transmission electron microscopy (TEM) detection. The in vitro study further confrimed that CAPE attenuated liver fibrogenesis via inducing authophagic markers including LC3, ATG5, Beclin 1 expressions, while inhibiting AKT/mTOR signaling in HSC-T6 cells. Thus, the protective effects of CAPE against liver fibrosis might due to the inhibition of TGF-β1/Smad3 signaling and induction of authophagy in HSCs.
Collapse
Affiliation(s)
- Ning Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Shuangsuo Dang
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China.
| | - Juanjuan Shi
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Fengping Wu
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Mei Li
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Xin Zhang
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Yaping Li
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Xiaoli Jia
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Song Zhai
- Department of Infectious Diseases, The Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China.
| |
Collapse
|
45
|
Joffre C, Djavaheri-Mergny M, Pattingre S, Giuriato S. L’autophagie : le yin et le yang des cancers. Med Sci (Paris) 2017; 33:328-334. [DOI: 10.1051/medsci/20173303021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
46
|
Targeting autophagy as a strategy for drug discovery and therapeutic modulation. Future Med Chem 2017; 9:335-345. [DOI: 10.4155/fmc-2016-0210] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Autophagy is a self-protective mechanism of living cells or organisms under various stress conditions. Studies of human genetics and pathophysiology have implicated that alterations in autophagy affect the context of cellular homeostasis and disease-associated phenotypes. The molecular components of autophagy are currently being explored as new pharmacologic targets for drug development and therapeutic intervention of various diseases. Drugs that restore the normal autophagic pathways have the potential for effectively treating human disorders that depend on aberrations of autophagy. Here, we review the role of autophagy and its alterations in the pathogenesis of diverse diseases, and drug discovery strategies for modulating autophagy for therapeutic benefits as well as possible safety concerns and caveats associated with such approaches.
Collapse
|
47
|
Feng L, Zhang J, Zhu N, Ding Q, Zhang X, Yu J, Qiang W, Zhang Z, Ma Y, Huang D, Shen Y, Fang S, Yu Y, Wang H, Shen Y. Ubiquitin ligase SYVN1/HRD1 facilitates degradation of the SERPINA1 Z variant/α-1-antitrypsin Z variant via SQSTM1/p62-dependent selective autophagy. Autophagy 2017; 13:686-702. [PMID: 28121484 PMCID: PMC5388218 DOI: 10.1080/15548627.2017.1280207] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
SERPINA1/AAT/α-1-antitrypsin (serpin family A member 1) deficiency (SERPINA1/ AAT-D) is an autosomal recessive disorder characterized by the retention of misfolded SERPINA1/AAT in the endoplasmic reticulum (ER) of hepatocytes and a significant reduction of serum SERPINA1/AAT level. The Z variant of SERPINA1/AAT, containing a Glu342Lys (E342K) mutation (SERPINA1E342K/ATZ), the most common form of SERPINA1/AAT-D, is prone to misfolding and polymerization, which retains it in the ER of hepatocytes and leads to liver injury. Both proteasome and macroautophagy/autophagy pathways are responsible for disposal of SERPINA1E342K/ATZ after it accumulates in the ER. However, the mechanisms by which SERPINA1E342K/ATZ is selectively degraded by autophagy remain unknown. Here, we showed that ER membrane-spanning ubiquitin ligase (E3) SYVN1/HRD1 enhances the degradation of SERPINA1E342K/ATZ through the autophagy-lysosome pathway. We found that SYVN1 promoted SERPINA1E342K/ATZ, especially Triton X 100-insoluble SERPINA1E342K/ATZ clearance. However, the effect of SYVN1 in SERPINA1E342K/ATZ clearance was impaired after autophagy inhibition, as well as in autophagy-related 5 (atg5) knockout cells. On the contrary, autophagy induction enhanced SYVN1-mediated SERPINA1E342K/ATZ degradation. Further study showed that SYVN1 mediated SERPINA1E342K/ATZ ubiquitination, which is required for autophagic degradation of SERPINA1E342K/ATZ by promoting the interaction between SERPINA1E342K/ATZ and SQSTM1/p62 for formation of the autophagy complex. Interestingly, SYVN1-mediated lysine 48 (K48)-linked polyubiquitin chains that conjugated onto SERPINA1E342K/ATZ might predominantly bind to the ubiquitin-associated (UBA) domain of SQSTM1 and couple the ubiquitinated SERPINA1E342K/ATZ to the lysosome for degradation. In addition, autophagy inhibition attenuated the suppressive effect of SYVN1 on SERPINA1E342K/ATZ cytotoxicity, and the autophagy inducer rapamycin enhanced the suppressive effect of SYVN1 on SERPINA1E342K/ATZ-induced cell apoptosis. Therefore, this study proved that SYVN1 enhances SERPINA1E342K/ATZ degradation through SQSTM1-dependent autophagy and attenuates SERPINA1E342K/ATZ cytotoxicity.
Collapse
Affiliation(s)
- Lijie Feng
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Jin Zhang
- b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Na Zhu
- b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China.,c The 4th Affiliated Hospital, Anhui Medical University , Hefei, Anhui , China
| | - Qian Ding
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Xiaojie Zhang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Jishuang Yu
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Weimin Qiang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Zhetao Zhang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Yuyang Ma
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Dake Huang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China
| | - Yujun Shen
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Shengyun Fang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China.,d Center for Biomedical Engineering and Technology , University of Maryland , Baltimore , MD , USA
| | - Yifan Yu
- e Actuarial Science, School of Continuing Education , Columbia University , New York , NY , USA
| | - Haiping Wang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Yuxian Shen
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| |
Collapse
|
48
|
Kim MJ, Yoon JH, Ryu JH. Mitophagy: a balance regulator of NLRP3 inflammasome activation. BMB Rep 2017; 49:529-535. [PMID: 27439607 PMCID: PMC5227293 DOI: 10.5483/bmbrep.2016.49.10.115] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Indexed: 12/24/2022] Open
Abstract
The NLRP3 inflammasome is activated by a variety of external or host-derived stimuli and its activation initiates an inflammatory response through caspase-1 activation, resulting in inflammatory cytokine IL-1β maturation and secretion. The NLRP3 inflammasome activation is a kind of innate immune response, most likely mediated by myeloid cells acting as a host defense mechanism. However, if this activation is not properly regulated, excessive inflammation induced by overactivated NLRP3 inflammasome can be detrimental to the host, causing tissue damage and organ dysfunction, eventually causing several diseases. Previous studies have suggested that mitochondrial damage may be a cause of NLRP3 inflammasome activation and autophagy, which is a conserved self-degradation process that negatively regulates NLRP3 inflammasome activation. Recently, mitochondria-selective autophagy, termed mitophagy, has emerged as a central player for maintaining mitochondrial homeostasis through the elimination of damaged mitochondria, leading to the prevention of hyperinflammation triggered by NLRP3 inflammasome activation. In this review, we will first focus on the molecular mechanisms of NLRP3 inflammasome activation and NLRP3 inflammasome-related diseases. We will then discuss autophagy, especially mitophagy, as a negative regulator of NLPP3 inflammasome activation by examining recent advances in research. [BMB Reports 2016; 49(10): 529-535]
Collapse
Affiliation(s)
- Min-Ji Kim
- Research Center for Natural Human Defense System, Brain Korea 21 PLUS Project for Medical Science, and Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Joo-Heon Yoon
- Research Center for Natural Human Defense System, Brain Korea 21 PLUS Project for Medical Science, Department of Otorhinolaryngology, and The Airway Mucus Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ji-Hwan Ryu
- Brain Korea 21 PLUS Project for Medical Science and Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
49
|
Hayat M. Overview of Autophagy. AUTOPHAGY: CANCER, OTHER PATHOLOGIES, INFLAMMATION, IMMUNITY, INFECTION, AND AGING 2017:3-90. [DOI: 10.1016/b978-0-12-805420-8.00001-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
50
|
Hayat M. Overview of Autophagy. AUTOPHAGY: CANCER, OTHER PATHOLOGIES, INFLAMMATION, IMMUNITY, INFECTION, AND AGING 2017:1-122. [DOI: 10.1016/b978-0-12-812146-7.00001-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|