1
|
Zhang H, Meléndez A. Conserved components of the macroautophagy machinery in Caenorhabditis elegans. Genetics 2025; 229:iyaf007. [PMID: 40180610 PMCID: PMC12005284 DOI: 10.1093/genetics/iyaf007] [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: 03/29/2024] [Accepted: 12/13/2024] [Indexed: 04/05/2025] Open
Abstract
Macroautophagy involves the sequestration of cytoplasmic contents in a double-membrane autophagosome and its subsequent delivery to lysosomes for degradation and recycling. In Caenorhabditis elegans, autophagy participates in diverse processes such as stress resistance, cell fate specification, tissue remodeling, aging, and adaptive immunity. Genetic screens in C. elegans have identified a set of metazoan-specific autophagy genes that form the basis for our molecular understanding of steps unique to the autophagy pathway in multicellular organisms. Suppressor screens have uncovered multiple mechanisms that modulate autophagy activity under physiological conditions. C. elegans also provides a model to investigate how autophagy activity is coordinately controlled at an organismal level. In this chapter, we will discuss the molecular machinery, regulation, and physiological functions of autophagy, and also methods utilized for monitoring autophagy during C. elegans development.
Collapse
Affiliation(s)
- Hong Zhang
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Alicia Meléndez
- Department of Biology, Queens College, City University of New York, Flushing, NY 11367, USA
- Molecular, Cellular and Developmental Biology and Biochemistry Ph.D. Programs, The Graduate Center of the City University of New York, New York, NY 10016, USA
| |
Collapse
|
2
|
Wang Z, Zhang H. Phase-separated Condensates in Autophagosome Formation and Autophagy Regulation. J Mol Biol 2025:168964. [PMID: 39880155 DOI: 10.1016/j.jmb.2025.168964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
Biomacromolecules partition into numerous types of biological condensates or membrane-less organelles via liquid-liquid phase separation (LLPS). Newly formed liquid-like condensates may further undergo phase transition to convert into other material states, such as gel or solid states. Different biological condensates possess distinct material properties to fulfil their physiological functions in diverse cellular pathways and processes. Phase separation and condensates are extensively involved in the autophagy pathway. The autophagosome formation sites in both yeast and multicellular organisms are assembled as phase-separated condensates. TORC1, one of the core regulators of the autophagy-lysosome pathway, is subject to nonconventional regulation by multiple biological condensates. TFEB, the master transcription factor of the autophagy-lysosome pathway, phase separates to assemble liquid-like condensates involved in transcription of autophagic and lysosomal genes. The behaviors and transcriptional activity of TFEB condensates are governed by their material properties, thus suggesting novel autophagy intervention strategies. The phase separation process and the resulting condensates are emerging therapeutic targets for autophagy-related diseases.
Collapse
Affiliation(s)
- Zheng Wang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 PR China; School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006 PR China; Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang 330031 PR China.
| | - Hong Zhang
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101 PR China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049 PR China.
| |
Collapse
|
3
|
Huang F, Wang Y, Liu J, Cheng Y, Zhang X, Jiang H. Asperuloside alleviates osteoporosis by promoting autophagy and regulating Nrf2 activation. J Orthop Surg Res 2024; 19:855. [PMID: 39702357 DOI: 10.1186/s13018-024-05320-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Osteoporosis is a metabolic bone disease that has a common occurrence in postmenopausal women. Asperuloside (ASP) has been reported to exert anti-inflammatory and anti-oxidative effects in numerous diseases, such as rheumatoid arthritis and acute lung injury. However, whether ASP plays a role in osteoporosis has not been addressed. METHODS In vivo, ovariectomy (OVX) was used to induce mouse osteoporosis. Then, the mice were treated with 20 and 40 mg/kg ASP. In vitro, MC3T3-E1 cells were treated with 0, 1, 10, 20, 40 and 80 µM ASP. We chose 20 and 40 µM for further experiments due to no significant effects on cell viability. RESULTS The data indicated that ASP reduced osteoporosis in OVX mice and promoted osteogenic differentiation and mineralization in MC3T3-E1 cells. In addition, we explored that ASP protected against osteoporosis via inducing autophagy and activating Nrf2. CONCLUSION ASP alleviates OVX-induced osteoporosis by promoting autophagy and regulating Nrf2 activation.
Collapse
Affiliation(s)
- Fenglan Huang
- Department of Outpatient, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Yiteng Wang
- Department of Sports Medicine, Central Hospital of Dalian University of Technology, Dalian, 116021, China
| | - Jinzhu Liu
- Department of Orthopedics, Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - Ye Cheng
- Department of Outpatient, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Xiaonan Zhang
- Department of Orthopedics, Shenzhen Third People's Hospital, Shenzhen, 518112, China.
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China.
| | - Haoli Jiang
- Department of Orthopedics, Shenzhen Third People's Hospital, Shenzhen, 518112, China.
| |
Collapse
|
4
|
Anusha S, Negi PS. Tenebrio molitor (Mealworm) protein as a sustainable dietary strategy to improve health span in D-galactose-induced aged mice. Int J Biol Macromol 2024; 281:136610. [PMID: 39419135 DOI: 10.1016/j.ijbiomac.2024.136610] [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: 08/05/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024]
Abstract
Aging is an irreversible and continuous biological process involving intricate and interconnected mechanisms. The present work is focused on unravelling the anti-aging mechanisms of mealworm protein and protein-enriched fruit bar and vegetable soup in D-galactose-induced aged mice. Mealworm protein and enriched products significantly enhanced body weight, organ indices, and gut health. Behavioral assessments reflected enhanced neuroprotective effects. Mealworm protein and its enriched products demonstrated protective effects through anti-inflammatory activity with the highest reduction of TNFα (17.1 %), IL-6 (55.5 %), and IL-1β (75.1 %) levels and upregulated the anti-inflammatory marker (IL-4). Gene expression studies confirmed the induction of anti-aging effects by promoting metabolism, reducing cellular senescence, and enhancing anti-oxidant enzyme activity. The treatments extended telomere lengths by 3-4 times, further affirming the potential anti-aging efficacy of mealworm protein and its enriched products. Mealworm protein demonstrated positive effects on weight gain, anti-inflammatory responses, and telomere length; while fruit and vegetable products enhanced antioxidant activity, and positively influenced gut health. Further, a synergistic effect was observed by combining them, which resulted in improved overall anti-aging effect. The present work provides valuable insights into the multifaceted anti-aging mechanisms associated with mealworm protein and enriched products, highlighting their potential as functional foods with significant health-promoting effects.
Collapse
Affiliation(s)
- Siddaraju Anusha
- Department of Fruit and Vegetable Technology, CSIR-Central Food Technological Research Institute, Mysuru 570 020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Pradeep Singh Negi
- Department of Fruit and Vegetable Technology, CSIR-Central Food Technological Research Institute, Mysuru 570 020, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India.
| |
Collapse
|
5
|
Chen Y, Li X, Yang M, Liu SB. Research progress on morphology and mechanism of programmed cell death. Cell Death Dis 2024; 15:327. [PMID: 38729953 PMCID: PMC11087523 DOI: 10.1038/s41419-024-06712-8] [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: 12/18/2023] [Revised: 04/17/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024]
Abstract
Programmed cell death (PCD) is a basic process of life that is closely related to the growth, development, aging and disease of organisms and is one of the hotspots of life science research today. PCD is a kind of genetic control, autonomous and orderly important cell death that involves the activation, expression, and regulation of a series of genes. In recent years, with the deepening of research in this field, new mechanisms of multiple PCD pathways have been revealed. This article reviews and summarizes the multiple PCD pathways that have been discovered, analyses and compares the morphological characteristics and biomarkers of different types of PCD, and briefly discusses the role of various types of PCD in the diagnosis and treatment of different diseases, especially malignant tumors.
Collapse
Grants
- Jiangsu higher education institution innovative research team for science and technology (2021), Program of Jiangsu vocational college engineering technology research center (2023), Key technology progrom of Suzhou people’s livelihood technology projects (Grant No. SKY2021029), the Open Project of Jiangsu Biobank of Clinical Resources (TC2021B009), the Project of State Key Laboratory of Radiation Medicine and Protection, Soochow University, (No. GZK12023013), Programs of the Suzhou Vocational Health College (SZWZYTD202201), Qing‐Lan Project of Jiangsu Province in China (2021).
- Programs of the Suzhou Vocational Health College (szwzy 202210), Qing‐Lan Project of Jiangsu Province in China (2022).
- the Project of State Key Laboratory of Radiation Medicine and Protection, Soochow University, (No. GZK12023013)
Collapse
Affiliation(s)
- Yao Chen
- Suzhou Key Laboratory of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, China
| | - Xiaohua Li
- Department of Thyroid and Breast Surgery, Wuzhong People's Hospital of Suzhou City, Suzhou, China
| | - Minfeng Yang
- School of Public Health, Nantong University, Nantong, 226019, China.
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China.
| | - Song-Bai Liu
- Suzhou Key Laboratory of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, China.
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
6
|
Stringer JM, Alesi LR, Winship AL, Hutt KJ. Beyond apoptosis: evidence of other regulated cell death pathways in the ovary throughout development and life. Hum Reprod Update 2023; 29:434-456. [PMID: 36857094 PMCID: PMC10320496 DOI: 10.1093/humupd/dmad005] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/06/2022] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Regulated cell death is a fundamental component of numerous physiological processes; spanning from organogenesis in utero, to normal cell turnover during adulthood, as well as the elimination of infected or damaged cells throughout life. Quality control through regulation of cell death pathways is particularly important in the germline, which is responsible for the generation of offspring. Women are born with their entire supply of germ cells, housed in functional units known as follicles. Follicles contain an oocyte, as well as specialized somatic granulosa cells essential for oocyte survival. Follicle loss-via regulated cell death-occurs throughout follicle development and life, and can be accelerated following exposure to various environmental and lifestyle factors. It is thought that the elimination of damaged follicles is necessary to ensure that only the best quality oocytes are available for reproduction. OBJECTIVE AND RATIONALE Understanding the precise factors involved in triggering and executing follicle death is crucial to uncovering how follicle endowment is initially determined, as well as how follicle number is maintained throughout puberty, reproductive life, and ovarian ageing in women. Apoptosis is established as essential for ovarian homeostasis at all stages of development and life. However, involvement of other cell death pathways in the ovary is less established. This review aims to summarize the most recent literature on cell death regulators in the ovary, with a particular focus on non-apoptotic pathways and their functions throughout the discrete stages of ovarian development and reproductive life. SEARCH METHODS Comprehensive literature searches were carried out using PubMed and Google Scholar for human, animal, and cellular studies published until August 2022 using the following search terms: oogenesis, follicle formation, follicle atresia, oocyte loss, oocyte apoptosis, regulated cell death in the ovary, non-apoptotic cell death in the ovary, premature ovarian insufficiency, primordial follicles, oocyte quality control, granulosa cell death, autophagy in the ovary, autophagy in oocytes, necroptosis in the ovary, necroptosis in oocytes, pyroptosis in the ovary, pyroptosis in oocytes, parthanatos in the ovary, and parthanatos in oocytes. OUTCOMES Numerous regulated cell death pathways operate in mammalian cells, including apoptosis, autophagic cell death, necroptosis, and pyroptosis. However, our understanding of the distinct cell death mediators in each ovarian cell type and follicle class across the different stages of life remains the source of ongoing investigation. Here, we highlight recent evidence for the contribution of non-apoptotic pathways to ovarian development and function. In particular, we discuss the involvement of autophagy during follicle formation and the role of autophagic cell death, necroptosis, pyroptosis, and parthanatos during follicle atresia, particularly in response to physiological stressors (e.g. oxidative stress). WIDER IMPLICATIONS Improved knowledge of the roles of each regulated cell death pathway in the ovary is vital for understanding ovarian development, as well as maintenance of ovarian function throughout the lifespan. This information is pertinent not only to our understanding of endocrine health, reproductive health, and fertility in women but also to enable identification of novel fertility preservation targets.
Collapse
Affiliation(s)
- Jessica M Stringer
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lauren R Alesi
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Amy L Winship
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Karla J Hutt
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
7
|
Yuan M, Wang Y, Huang Z, Jing F, Qiao P, Zou Q, Li J, Cai Z. Impaired autophagy in amyloid-beta pathology: A traditional review of recent Alzheimer's research. J Biomed Res 2023; 37:30-46. [PMID: 36642915 PMCID: PMC9898044 DOI: 10.7555/jbr.36.20220145] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder. The major pathological changes in AD progression are the generation and accumulation of amyloid-beta (Aβ) peptides as well as the presence of abnormally hyperphosphorylated tau proteins in the brain. Autophagy is a conserved degradation pathway that eliminates abnormal protein aggregates and damaged organelles. Previous studies have suggested that autophagy plays a key role in the production and clearance of Aβ peptides to maintain a steady-state of Aβ peptides levels. However, a growing body of evidence suggests that autophagy is significantly impaired in the pathogenesis of AD, especially in Aβ metabolism. Therefore, this article reviews the latest studies concerning the mechanisms of autophagy, the metabolism of Aβ peptides, and the defective autophagy in the production and clearance of Aβ peptides. Here, we also summarize the established and new strategies for targeting autophagy in vivo and through clinical AD trials to identify gaps in our knowledge and to generate further questions.
Collapse
Affiliation(s)
- Minghao Yuan
- Chongqing Medical University, Chongqing 400042, China,Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China,Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Yangyang Wang
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China
| | - Zhenting Huang
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China
| | - Feng Jing
- Chongqing Medical University, Chongqing 400042, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China,Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Peifeng Qiao
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China
| | - Qian Zou
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China
| | - Jing Li
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China
| | - Zhiyou Cai
- Chongqing Medical University, Chongqing 400042, China,Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China,Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China,Zhiyou Cai, Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing 401147, China. Tel/Fax: +86-23-63515796/+86-23-63515796, E-mail:
| |
Collapse
|
8
|
Chen Y, Pan X, Zhao J, Li C, Lin Y, Wang Y, Liu X, Tian M. Icariin alleviates osteoarthritis through PI3K/Akt/mTOR/ULK1 signaling pathway. Eur J Med Res 2022; 27:204. [PMID: 36253872 PMCID: PMC9575285 DOI: 10.1186/s40001-022-00820-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVES This study aims to investigate the effects of Icariin (ICA) on interleukin-1β (IL-1β)-induced osteoarthritis (OA) and its potential mechanism of action. METHODS SW1353 chondrocytes were pretreated with ICA for 2 h, followed by stimulation with IL-1β to mimic OA. Expression levels of matrix metalloproteinases (MMP-3) and collagen II were determined using real-time PCR and Western blot assays. Autophagy activation (by ICA) or inhibition (by shRNA) was determined based on the expression levels of ULK1, Beclin-1, LC3-II/I, and p62, using Western blot analysis. The phosphorylation levels of PI3K, Akt, mTOR, and ULK1 were also detected using Western blot analysis. RESULTS IL-1β increased MMP-3 overproduction, induced collagen II degradation, and reduced the level of autophagy-associated proteins, including ULK1, Beclin-1, and LC3-II/I. In contrast, ICA pretreatment attenuated IL-1β-induced MMP-3 overproduction, increased collagen II expression, and induced expression of autophagy-related proteins. ICA also decreased PI3K, Akt, and mTOR phosphorylation, increased the production of ULK1, and induced autophagy. Short hairpin RNA-mediated knockdown of ULK1 led to activation of the PI3K/Akt/mTOR pathway, which reversed the protective effects of ICA. CONCLUSIONS Our findings indicate that ICA can induce autophagy by regulating the PI3K/AKT/mTOR/ULK1 signaling pathway. This study suggests that ICA may be effective for treating OA.
Collapse
Affiliation(s)
- Yan Chen
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi, 563006, China
| | - Xiaoli Pan
- Department of Rheumatology and Immunology Department, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China
| | - Jing Zhao
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi, 563006, China
| | - Chunyan Li
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi, 563006, China
| | - Yupei Lin
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi, 563006, China
| | - Yu Wang
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi, 563006, China
| | - Xu Liu
- Department of Rheumatology and Immunology Department, Peking University People's Hospital, Beijing, 100044, China
| | - Mei Tian
- Department of Rheumatology and Immunology Department, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China.
| |
Collapse
|
9
|
Zhou L, Xue X, Yang K, Feng Z, Liu M, Pastor-Pareja JC. Convergence of secretory, endosomal, and autophagic routes in trans-Golgi-associated lysosomes. J Cell Biol 2022; 222:213547. [PMID: 36239631 PMCID: PMC9577102 DOI: 10.1083/jcb.202203045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 08/17/2022] [Accepted: 09/23/2022] [Indexed: 12/15/2022] Open
Abstract
At the trans-Golgi, complex traffic connections exist to the endolysosomal system additional to the main Golgi-to-plasma membrane secretory route. Here, we investigated three hits in a Drosophila screen displaying secretory cargo accumulation in autophagic vesicles: ESCRT-III component Vps20, SNARE-binding Rop, and lysosomal pump subunit VhaPPA1-1. We found that Vps20, Rop, and lysosomal markers localize near the trans-Golgi. Furthermore, we document that the vicinity of the trans-Golgi is the main cellular location for lysosomes and that early, late, and recycling endosomes associate as well with a trans-Golgi-associated degradative compartment where basal microautophagy of secretory cargo and other materials occurs. Disruption of this compartment causes cargo accumulation in our hits, including Munc18 homolog Rop, required with Syx1 and Syx4 for Rab11-mediated endosomal recycling. Finally, besides basal microautophagy, we show that the trans-Golgi-associated degradative compartment contributes to the growth of autophagic vesicles in developmental and starvation-induced macroautophagy. Our results argue that the fly trans-Golgi is the gravitational center of the whole endomembrane system.
Collapse
Affiliation(s)
- Lingjian Zhou
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Xutong Xue
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Ke Yang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhi Feng
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Min Liu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - José C. Pastor-Pareja
- School of Life Sciences, Tsinghua University, Beijing, China,Tsinghua-Peking Center for Life Sciences, Beijing, China,Institute of Neurosciences, Consejo Superior de Investigaciones Científicas–Universidad Miguel Hernández, San Juan de Alicante, Spain
| |
Collapse
|
10
|
A Review of Signaling Transduction Mechanisms in Osteoclastogenesis Regulation by Autophagy, Inflammation, and Immunity. Int J Mol Sci 2022; 23:ijms23179846. [PMID: 36077242 PMCID: PMC9456406 DOI: 10.3390/ijms23179846] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoclastogenesis is an ongoing rigorous course that includes osteoclast precursors fusion and bone resorption executed by degradative enzymes. Osteoclastogenesis is controlled by endogenous signaling and/or regulators or affected by exogenous conditions and can also be controlled both internally and externally. More evidence indicates that autophagy, inflammation, and immunity are closely related to osteoclastogenesis and involve multiple intracellular organelles (e.g., lysosomes and autophagosomes) and certain inflammatory or immunological factors. Based on the literature on osteoclastogenesis induced by different regulatory aspects, emerging basic cross-studies have reported the emerging disquisitive orientation for osteoclast differentiation and function. In this review, we summarize the partial potential therapeutic targets for osteoclast differentiation and function, including the signaling pathways and various cellular processes.
Collapse
|
11
|
Lin H, Gao Y, Zhang C, Ma B, Wu M, Cui X, Wang H. Autophagy Regulation Influences β-Amyloid Toxicity in Transgenic Caenorhabditis elegans. Front Aging Neurosci 2022; 14:885145. [PMID: 35645788 PMCID: PMC9133694 DOI: 10.3389/fnagi.2022.885145] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/13/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive, neurodegenerative disease characterized by the accumulation of amyloid-beta (Aβ) proteins in the form of plaques that cause a proteostasis imbalance in the brain. Several studies have identified autophagy deficits in both AD patients and AD animal models. Here, we used transgenic Caenorhabditis elegans to study the relationship between autophagy flux and Aβ. We labeled autophagosomes with an advanced fluorescence reporter system, and used this to observe that human Aβ expression caused autophagosome accumulation in C. elegans muscle. The autophagy-related drugs chloroquine and 3-MA were employed to investigate the relationship between changes in autophagic flux and the toxicity of Aβ expression. We found that reducing autophagosome accumulation delayed Aβ-induced paralysis in the CL4176 strain of C. elegans, and alleviated Aβ-induced toxicity, thus having a neuroprotective effect. Finally, we used RNA-sequencing and proteomics to identify genes whose expression was affected by Aβ aggregation in C. elegans. We identified a series of enriched autophagy-related signal pathways, suggesting that autophagosome accumulation impairs Aβ protein homeostasis in nematodes. Thus, maintaining normal autophagy levels appears to be important in repairing the protein homeostasis imbalance caused by Aβ expression.
Collapse
|
12
|
Peña-Ramos O, Chiao L, Liu X, Yu X, Yao T, He H, Zhou Z. Autophagosomes fuse to phagosomes and facilitate the degradation of apoptotic cells in Caenorhabditis elegans. eLife 2022; 11:72466. [PMID: 34982028 PMCID: PMC8769646 DOI: 10.7554/elife.72466] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 01/03/2022] [Indexed: 12/17/2022] Open
Abstract
Autophagosomes are double-membrane intracellular vesicles that degrade protein aggregates, intracellular organelles, and other cellular components. During the development of the nematode Caenorhabditis elegans, many somatic and germ cells undergo apoptosis. These cells are engulfed and degraded by their neighboring cells. We discovered a novel role of autophagosomes in facilitating the degradation of apoptotic cells using a real-time imaging technique. Specifically, the double-membrane autophagosomes in engulfing cells are recruited to the surfaces of phagosomes containing apoptotic cells and subsequently fuse to phagosomes, allowing the inner vesicle to enter the phagosomal lumen. Mutants defective in the production of autophagosomes display significant defects in the degradation of apoptotic cells, demonstrating the importance of autophagosomes to this process. The signaling pathway led by the phagocytic receptor CED-1, the adaptor protein CED-6, and the large GTPase dynamin (DYN-1) promotes the recruitment of autophagosomes to phagosomes. Moreover, the subsequent fusion of autophagosomes with phagosomes requires the functions of the small GTPase RAB-7 and the HOPS complex. Further observations suggest that autophagosomes provide apoptotic cell-degradation activities in addition to and in parallel of lysosomes. Our findings reveal that, unlike the single-membrane, LC3-associated phagocytosis (LAP) vesicles reported to facilitate phagocytosis in mammals, it is the canonical double-membrane autophagosomes that facilitate the clearance of C. elegans apoptotic cells. These findings add autophagosomes to the collection of intracellular organelles that contribute to phagosome maturation, identify novel crosstalk between the autophagy and phagosome maturation pathways, and discover the upstream signaling molecules that initiate this crosstalk.
Collapse
Affiliation(s)
- Omar Peña-Ramos
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States
| | - Lucia Chiao
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States
| | - Xianghua Liu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States
| | - Xiaomeng Yu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States
| | - Tianyou Yao
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States
| | - Henry He
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States
| | - Zheng Zhou
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States
| |
Collapse
|
13
|
Zhao J, Li Z, Li J. The crystal structure of the FAM134B-GABARAP complex provides mechanistic insights into the selective binding of FAM134 to the GABARAP subfamily. FEBS Open Bio 2021; 12:320-331. [PMID: 34854256 PMCID: PMC8727931 DOI: 10.1002/2211-5463.13340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/07/2021] [Accepted: 11/30/2021] [Indexed: 01/18/2023] Open
Abstract
The mammalian Atg8 family (Atg8s proteins) consists of two subfamilies: GABARAP and LC3. All members can bind to the LC3‐interacting region (LIR) or Atg8‐interacting motif and participate in multiple steps of autophagy. The endoplasmic reticulum (ER) autophagy receptor FAM134B contains an LIR motif that can bind to Atg8s, but whether it can differentially bind to the two subfamilies and, if so, the structural basis for this preference remains unknown. Here, we found that FAM134B bound to the GABARAP subfamily more strongly than to the LC3 subfamily. We then solved the crystal structure of the FAM134B–GABARAP complex and demonstrated that FAM134B used both its LIR core and the C‐terminal helix to bind to GABARAP. We further showed that these properties might be conserved in FAM134A or FAM134C. The structure also allowed us to identify the structural determinants for the binding selectivity. Our work may be valuable for studying the differential functions of GABARAP and LC3 subfamilies in ER phagy in future.
Collapse
Affiliation(s)
- Junfeng Zhao
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhiwei Li
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianchao Li
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, China.,Department of Otorhinolaryngology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
14
|
Macroautophagy and Mitophagy in Neurodegenerative Disorders: Focus on Therapeutic Interventions. Biomedicines 2021; 9:biomedicines9111625. [PMID: 34829854 PMCID: PMC8615936 DOI: 10.3390/biomedicines9111625] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
Macroautophagy, a quality control mechanism, is an evolutionarily conserved pathway of lysosomal degradation of protein aggregates, pathogens, and damaged organelles. As part of its vital homeostatic role, macroautophagy deregulation is associated with various human disorders, including neurodegenerative diseases. There are several lines of evidence that associate protein misfolding and mitochondrial dysfunction in the etiology of Alzheimer’s, Parkinson’s, and Huntington’s diseases. Macroautophagy has been implicated in the degradation of different protein aggregates such as Aβ, tau, alpha-synuclein (α-syn), and mutant huntingtin (mHtt) and in the clearance of dysfunctional mitochondria. Taking these into consideration, targeting autophagy might represent an effective therapeutic strategy to eliminate protein aggregates and to improve mitochondrial function in these disorders. The present review describes our current understanding on the role of macroautophagy in neurodegenerative disorders and focuses on possible strategies for its therapeutic modulation.
Collapse
|
15
|
Huang L, Fu Q, Dai JM, Yan BC, Wang D, Puno PT, Yue J. High-content screening of diterpenoids from Isodon species as autophagy modulators and the functional study of their antiviral activities. Cell Biol Toxicol 2021; 37:695-713. [PMID: 33486680 DOI: 10.1007/s10565-021-09580-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/01/2021] [Indexed: 12/16/2022]
Abstract
Autophagy is a conserved lysosomal degradation process, and abnormal autophagy has been associated with various pathological processes, e.g., neurodegeneration, cancer, and pathogen infection. Small chemical modulators of autophagy show the potential to treat autophagy-associated diseases. Diterpenoids, nature products found in various plants, exhibit a wide range of bioactivity, and we have recently isolated and characterized over 150 diterpenoids from Isodon species distributed in China. Here, we applied a high-content fluorescence imaging-based assay to assess these diterpenoids' ability to affect autophagic flux in HeLa cells. We found that enanderinanin J, an ent-kauranoid dimer, is an autophagy inhibitor, manifested by its ability to increase lysosomal pH and inhibit the fusion between autophagosomes and lysosomes. Autophagy has been shown to be either positively or negatively involved in the life cycle of Zika virus (ZIKV), Japanese encephalitis virus (JEV), Dengue virus (DENV), and enterovirus-A71 (EV-A71). We found that enanderinanin J significantly inhibited the infection of ZIKV, DENV, JEV, or EV-A71. Interestingly, although ATG5 knockdown inhibited ZIKV or JEV infection, enanderinanin J further inhibited the infection of ZIKV or JEV in ATG5-knockdown cells. Taken together, our data indicate that enanderinanin J inhibits autophagosome-lysosome fusion and is a potential antiviral agent.
Collapse
Affiliation(s)
- Lihong Huang
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Qiang Fu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Jia-Meng Dai
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, China
| | - Bing-Chao Yan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, China
| | - Dawei Wang
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Pema-Tenzin Puno
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, China.
| | - Jianbo Yue
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
- City University of Hong Kong Chengdu Research Institute, Chengdu, China.
| |
Collapse
|
16
|
Agas D, Sabbieti MG. Autophagic Mediators in Bone Marrow Niche Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1376:61-75. [PMID: 34480334 DOI: 10.1007/5584_2021_666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The bone marrow serves as a reservoir for a multifunctional assortment of stem, progenitor, and mature cells, located in functional anatomical micro-areas termed niches. Within the niche, hematopoietic and mesenchymal progenies establish a symbiotic relationship characterized by interdependency and interconnectedness. The fine-tuned physical and molecular interactions that occur in the niches guarantee physiological bone turnover, blood cell maturation and egression, and moderation of inflammatory and oxidative intramural stressful conditions. The disruption of bone marrow niche integrity causes severe local and systemic pathological settings, and thus bone marrow inhabitants have been the object of extensive study. In this context, research has revealed the importance of the autophagic apparatus for niche homeostatic maintenance. Archetypal autophagic players such as the p62 and the Atg family proteins have been found to exert a variety of actions, some autophagy-related and others not; they moderate the essential features of mesenchymal and hematopoietic stem cells and switch their operational schedules. This chapter focuses on our current understanding of bone marrow functionality and the role of the executive autophagic apparatus in the niche framework. Autophagic mediators such as p62 and Atg7 are currently considered the most important orchestrators of stem and mature cell dynamics in the bone marrow.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| | | |
Collapse
|
17
|
Zhao X, Di Q, Yu J, Quan J, Xiao Y, Zhu H, Li H, Ling J, Chen W. USP19 (ubiquitin specific peptidase 19) promotes TBK1 (TANK-binding kinase 1) degradation via chaperone-mediated autophagy. Autophagy 2021; 18:891-908. [PMID: 34436957 DOI: 10.1080/15548627.2021.1963155] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
TBK1 (TANK-binding kinase 1) is an essential receptor protein required for the innate immune response, but the mechanisms underlying TBK1 stability, especially those regulated via autophagy, remain poorly understood. Here, we demonstrate that USP19 (ubiquitin specific peptidase 19) interacts with and promotes TBK1 lysosomal degradation via chaperone-mediated autophagy (CMA). We observed that TBK1 had a canonical CMA motif, knocking down key proteins involved in CMA (HSPA8/HSC70 or LAMP2A) or inhibiting CMA-prevented USP19-mediated TBK1 degradation. Furthermore, USP19 deficiency in macrophages caused an elevation of TBK1 and the activation of the type-I interferon signaling pathway after vesicular stomatitis virus (VSV) infection. Consistently, macrophage-specific usp19 knockout in mice resulted in attenuated VSV replication and resistance to VSV infection in vivo. Altogether, our results suggest that USP19 is a key regulator of TBK1 and uncovers a previously uncharacterized role for USP19 in CMA-mediated TBK1 degradation and infectious diseases.
Collapse
Affiliation(s)
- Xibao Zhao
- Guangdong Provincial Key Laboratory Of Regional Immunity And Diseases, Department Of Immunology, Shenzhen University School Of Medicine, Shenzhen, China
| | - Qianqian Di
- Guangdong Provincial Key Laboratory Of Regional Immunity And Diseases, Department Of Immunology, Shenzhen University School Of Medicine, Shenzhen, China
| | - Juan Yu
- Institute Of Immunology, Zhejiang University School Of Medicine, Hangzhou, China
| | - Jiazheng Quan
- Guangdong Provincial Key Laboratory Of Regional Immunity And Diseases, Department Of Immunology, Shenzhen University School Of Medicine, Shenzhen, China
| | - Yue Xiao
- Guangdong Provincial Key Laboratory Of Regional Immunity And Diseases, Department Of Immunology, Shenzhen University School Of Medicine, Shenzhen, China
| | - Huihui Zhu
- Institute Of Immunology, Zhejiang University School Of Medicine, Hangzhou, China
| | - Hongrui Li
- Institute Of Immunology, Zhejiang University School Of Medicine, Hangzhou, China
| | - Jing Ling
- Institute Of Immunology, Zhejiang University School Of Medicine, Hangzhou, China
| | - Weilin Chen
- Guangdong Provincial Key Laboratory Of Regional Immunity And Diseases, Department Of Immunology, Shenzhen University School Of Medicine, Shenzhen, China
| |
Collapse
|
18
|
Mesquita A, Glenn J, Jenny A. Differential activation of eMI by distinct forms of cellular stress. Autophagy 2021; 17:1828-1840. [PMID: 32559125 PMCID: PMC8386722 DOI: 10.1080/15548627.2020.1783833] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022] Open
Abstract
As one of the major, highly conserved catabolic pathways, autophagy delivers cytosolic components to lysosomes for degradation. It is essential for development, cellular homeostasis, and coping with stress. Reduced autophagy increases susceptibility to protein aggregation diseases and leads to phenotypes associated with aging. Of the three major forms of autophagy, macroautophagy (MA) can degrade organelles or aggregated proteins, and chaperone-mediated autophagy is specific for soluble proteins containing KFERQ-related targeting motifs. During endosomal microautophagy (eMI), cytoplasmic proteins are engulfed into late endosomes in an ESCRT machinery-dependent manner. eMI can be KFERQ-specific or occur in bulk and be induced by prolonged starvation. Its physiological regulation and function, however, are not understood. Here, we show that eMI in the Drosophila fat body, akin to the mammalian liver, is induced upon oxidative or genotoxic stress in an ESCRT and partially Hsc70-4-dependent manner. Interestingly, eMI activation is selective, as ER stress fails to elicit a response. Intriguingly, we find that reducing MA leads to a compensatory enhancement of eMI, suggesting a tight interplay between these degradative processes. Furthermore, we show that mutations in DNA damage response genes are sufficient to trigger eMI and that the response to oxidative stress is under the control of MAPK/JNK signaling. Our data suggest that, controlled by various signaling pathways, eMI allows an organ to react and adapt to specific types of stress and is thus likely critical to prevent disease.Abbreviations:Atg: autophagy-related; CMA: chaperone-mediated autophagy; DDR: DNA damage repair; Df: deficiency (deletion); (E)GFP: (enhanced) green fluorescent protein; eMI: endosomal microautophagy; ER: endoplasmatic reticulum; ESCRT: endosomal sorting complexes required for transport; Eto: etoposide; FLP: flipase; Hsc: heat shock cognate protein; LAMP2A: lysosomal-associated membrane protein 2A; LE: late endosome; MA: macroautophagy; MI: microautophagy; MVB: multivesicular body; PA: photoactivatable; Para: paraquat; ROS: reactive oxygen species; SEM: standard error of means; Tor: target of rapamycin [serine/threonine kinase]; UPR: unfolded protein response; Vps: vacuolar protein sorting.
Collapse
Affiliation(s)
- Ana Mesquita
- Department of Developmental and Molecular Biology, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, New York, NY, US
| | - James Glenn
- Department of Developmental and Molecular Biology, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, New York, NY, US
| | - Andreas Jenny
- Department of Developmental and Molecular Biology, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, New York, NY, US
- Department of Genetics, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, New York, NY, US
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine, New York, NY, US
| |
Collapse
|
19
|
Allen EA, Amato C, Fortier TM, Velentzas P, Wood W, Baehrecke EH. A conserved myotubularin-related phosphatase regulates autophagy by maintaining autophagic flux. J Cell Biol 2021; 219:152081. [PMID: 32915229 PMCID: PMC7594499 DOI: 10.1083/jcb.201909073] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 06/23/2020] [Accepted: 08/03/2020] [Indexed: 12/27/2022] Open
Abstract
Macroautophagy (autophagy) targets cytoplasmic cargoes to the lysosome for degradation. Like all vesicle trafficking, autophagy relies on phosphoinositide identity, concentration, and localization to execute multiple steps in this catabolic process. Here, we screen for phosphoinositide phosphatases that influence autophagy in Drosophila and identify CG3530. CG3530 is homologous to the human MTMR6 subfamily of myotubularin-related 3-phosphatases, and therefore, we named it dMtmr6. dMtmr6, which is required for development and viability in Drosophila, functions as a regulator of autophagic flux in multiple Drosophila cell types. The MTMR6 family member MTMR8 has a similar function in autophagy of higher animal cells. Decreased dMtmr6 and MTMR8 function results in autophagic vesicle accumulation and influences endolysosomal homeostasis.
Collapse
Affiliation(s)
- Elizabeth A Allen
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Clelia Amato
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Tina M Fortier
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Panagiotis Velentzas
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Will Wood
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Eric H Baehrecke
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
20
|
Noda NN, Wang Z, Zhang H. Liquid-liquid phase separation in autophagy. J Cell Biol 2021; 219:151909. [PMID: 32603410 PMCID: PMC7401820 DOI: 10.1083/jcb.202004062] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 01/05/2023] Open
Abstract
Liquid–liquid phase separation (LLPS) compartmentalizes and concentrates biomacromolecules into distinct condensates. Liquid-like condensates can transition into gel and solid states, which are essential for fulfilling their different functions. LLPS plays important roles in multiple steps of autophagy, mediating the assembly of autophagosome formation sites, acting as an unconventional modulator of TORC1-mediated autophagy regulation, and triaging protein cargos for degradation. Gel-like, but not solid, protein condensates can trigger formation of surrounding autophagosomal membranes. Stress and pathological conditions cause aberrant phase separation and transition of condensates, which can evade surveillance by the autophagy machinery. Understanding the mechanisms underlying phase separation and transition will provide potential therapeutic targets for protein aggregation diseases.
Collapse
Affiliation(s)
- Nobuo N Noda
- Institute of Microbial Chemistry (BIKAKEN), Tokyo, Japan
| | - Zheng Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
21
|
Furuta Y, Pena-Ramos O, Li Z, Chiao L, Zhou Z. Calcium ions trigger the exposure of phosphatidylserine on the surface of necrotic cells. PLoS Genet 2021; 17:e1009066. [PMID: 33571185 PMCID: PMC7904182 DOI: 10.1371/journal.pgen.1009066] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 02/24/2021] [Accepted: 01/18/2021] [Indexed: 11/18/2022] Open
Abstract
Intracellular Ca2+ level is under strict regulation through calcium channels and storage pools including the endoplasmic reticulum (ER). Mutations in certain ion channel subunits, which cause mis-regulated Ca2+ influx, induce the excitotoxic necrosis of neurons. In the nematode Caenorhabditis elegans, dominant mutations in the DEG/ENaC sodium channel subunit MEC-4 induce six mechanosensory (touch) neurons to undergo excitotoxic necrosis. These necrotic neurons are subsequently engulfed and digested by neighboring hypodermal cells. We previously reported that necrotic touch neurons actively expose phosphatidylserine (PS), an “eat-me” signal, to attract engulfing cells. However, the upstream signal that triggers PS externalization remained elusive. Here we report that a robust and transient increase of cytoplasmic Ca2+ level occurs prior to the exposure of PS on necrotic touch neurons. Inhibiting the release of Ca2+ from the ER, either pharmacologically or genetically, specifically impairs PS exposure on necrotic but not apoptotic cells. On the contrary, inhibiting the reuptake of cytoplasmic Ca2+ into the ER induces ectopic necrosis and PS exposure. Remarkably, PS exposure occurs independently of other necrosis events. Furthermore, unlike in mutants of DEG/ENaC channels, in dominant mutants of deg-3 and trp-4, which encode Ca2+ channels, PS exposure on necrotic neurons does not rely on the ER Ca2+ pool. Our findings indicate that high levels of cytoplasmic Ca2+ are necessary and sufficient for PS exposure. They further reveal two Ca2+-dependent, necrosis-specific pathways that promote PS exposure, a “two-step” pathway initiated by a modest influx of Ca2+ and further boosted by the release of Ca2+ from the ER, and another, ER-independent, pathway. Moreover, we found that ANOH-1, the worm homolog of mammalian phospholipid scramblase TMEM16F, is necessary for efficient PS exposure in thapsgargin-treated worms and trp-4 mutants, like in mec-4 mutants. We propose that both the ER-mediated and ER-independent Ca2+ pathways promote PS externalization through activating ANOH-1. Necrosis is a type of cell death that exhibits distinct morphological features such as cell and organelle swelling. Necrotic cells expose phosphatidylserine (PS)–a type of phospholipid—on their outer surfaces. Receptor molecules on phagocytes detect PS on necrotic cells and subsequently initiate the engulfment process. As necrosis is associated with stroke, cancer, neurodegenerative diseases, and heart diseases, studying necrotic cell clearance has important medical relevance. In the model organism the nematode C. elegans, we previously identified membrane proteins that promote the exposure of PS on necrotic cell surfaces by studying neurons that are induced to undergo necrosis by dominant mutations in ion channels. Here, in C. elegans, we have discovered that the necrotic insults trigger an increase of the cytoplasmic calcium ion (Ca2+), which in turn promotes PS externalization on necrotic cell surfaces. Furthermore, we have identified two different mechanisms that increase cytoplasmic Ca2+ levels, one dependent on the Ca2+ contribution from the endoplasmic reticulum (ER), the other independent of the ER. The Ca2+ signal targets ANOH-1, a worm homolog of mammalian proteins capable of externalizing PS, for promoting PS exposure on necrotic cells. Our findings reveal novel upstream regulatory mechanisms that promote necrotic cell clearance in animals.
Collapse
Affiliation(s)
- Yoshitaka Furuta
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- School of Pharmacy, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, Japan
| | - Omar Pena-Ramos
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Zao Li
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lucia Chiao
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Zheng Zhou
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
22
|
Rodrigues AFG, Ibelli AMG, Peixoto JDO, Cantão ME, de Oliveira HC, Savoldi IR, Souza MR, Mores MAZ, Carreño LOD, Ledur MC. Genes and SNPs Involved with Scrotal and Umbilical Hernia in Pigs. Genes (Basel) 2021; 12:genes12020166. [PMID: 33513662 PMCID: PMC7912685 DOI: 10.3390/genes12020166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/27/2022] Open
Abstract
Hernia is one of the most common defects in pigs. The most prevalent are the scrotal (SH), inguinal (IH) and umbilical (UH) hernias. We compared the inguinal ring transcriptome of normal and SH-affected pigs with the umbilical ring transcriptome of normal and UH-affected pigs to discover genes and pathways involved with the development of both types of hernia. A total of 13,307 transcripts was expressed in the inguinal and 13,302 in the umbilical ring tissues with 94.91% of them present in both tissues. From those, 35 genes were differentially expressed in both groups, participating in 108 biological processes. A total of 67 polymorphisms was identified in the inguinal ring and 76 in the umbilical ring tissue, of which 11 and 14 were novel, respectively. A single nucleotide polymorphism (SNP) with deleterious function was identified in the integrin α M (ITGAM) gene. The microtubule associated protein 1 light chain 3 γ (MAP1LC3C), vitrin (VIT), aggrecan (ACAN), alkaline ceramidase 2 (ACER2), potassium calcium-activated channel subfamily M α 1 (KCNMA1) and synaptopodin 2 (SYNPO2) genes are highlighted as candidates to trigger both types of hernia. We generated the first comparative study of the pig umbilical and inguinal ring transcriptomes, contributing to the understanding of the genetic mechanism involved with these two types of hernia in pigs and probably in other mammals.
Collapse
Affiliation(s)
- Ariene Fernanda Grando Rodrigues
- Programa de Pós-Graduação em Zootecnia, Departamento de Zootecnia, Centro de Educação Superior do Oeste (CEO), Universidade do Estado de Santa Catarina, UDESC, 89815-630 Chapecó, Brazil; (A.F.G.R.); (I.R.S.); (M.R.S.)
| | - Adriana Mércia Guaratini Ibelli
- Embrapa Suínos e Aves, Distrito de Tamanduá, 89715-899 Concórdia, Brazil; (A.M.G.I.); (J.d.O.P.); (M.E.C.); (M.A.Z.M.)
- Programa de Pós-Graduação em Ciências Veterinárias, Departamento de Ciências Veterinárias, Universidade Estadual do Centro-Oeste, 85015-430 Guarapuava, Brazil
| | - Jane de Oliveira Peixoto
- Embrapa Suínos e Aves, Distrito de Tamanduá, 89715-899 Concórdia, Brazil; (A.M.G.I.); (J.d.O.P.); (M.E.C.); (M.A.Z.M.)
- Programa de Pós-Graduação em Ciências Veterinárias, Departamento de Ciências Veterinárias, Universidade Estadual do Centro-Oeste, 85015-430 Guarapuava, Brazil
| | - Maurício Egídio Cantão
- Embrapa Suínos e Aves, Distrito de Tamanduá, 89715-899 Concórdia, Brazil; (A.M.G.I.); (J.d.O.P.); (M.E.C.); (M.A.Z.M.)
| | | | - Igor Ricardo Savoldi
- Programa de Pós-Graduação em Zootecnia, Departamento de Zootecnia, Centro de Educação Superior do Oeste (CEO), Universidade do Estado de Santa Catarina, UDESC, 89815-630 Chapecó, Brazil; (A.F.G.R.); (I.R.S.); (M.R.S.)
| | - Mayla Regina Souza
- Programa de Pós-Graduação em Zootecnia, Departamento de Zootecnia, Centro de Educação Superior do Oeste (CEO), Universidade do Estado de Santa Catarina, UDESC, 89815-630 Chapecó, Brazil; (A.F.G.R.); (I.R.S.); (M.R.S.)
- Programa de Pós-Graduação em Zootecnia, Departamento de Zootecnia, Universidade Federal do Rio Grande do Sul, UFRGS, 91540-000 Porto Alegre, Brazil
| | - Marcos Antônio Zanella Mores
- Embrapa Suínos e Aves, Distrito de Tamanduá, 89715-899 Concórdia, Brazil; (A.M.G.I.); (J.d.O.P.); (M.E.C.); (M.A.Z.M.)
| | | | - Mônica Corrêa Ledur
- Programa de Pós-Graduação em Zootecnia, Departamento de Zootecnia, Centro de Educação Superior do Oeste (CEO), Universidade do Estado de Santa Catarina, UDESC, 89815-630 Chapecó, Brazil; (A.F.G.R.); (I.R.S.); (M.R.S.)
- Embrapa Suínos e Aves, Distrito de Tamanduá, 89715-899 Concórdia, Brazil; (A.M.G.I.); (J.d.O.P.); (M.E.C.); (M.A.Z.M.)
- Correspondence: or ; Tel.: +55-49-3441-0411
| |
Collapse
|
23
|
Agas D, Sabbieti MG. Archetypal autophagic players through new lenses for bone marrow stem/mature cells regulation. J Cell Physiol 2021; 236:6101-6114. [PMID: 33492700 DOI: 10.1002/jcp.30296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022]
Abstract
The bone marrow landscape consists of specialized and stem/progenitor cells, which coordinate important tissue-related and systemic physiological features. Within the marrow cavity, stem/progenitor and differentiated hematopoietic and skeletal cells congregate into dynamic functional assemblies throughout specific anatomical regions, termed niches. There is a need for better understanding of the bone marrow microareas, through exploration of the intramural physical and molecular interactions of the distinctive cell populations. The elective liaisons established among the mesenchymal/stromal stem cell and hematopoietic stem cell lineage trees play a key role in orchestrating the stem/mature cell behavior and customized hierarchies within bone marrow cell populations. Recently, the autophagic apparatus has been discovered to be an important feature of bone marrow homeostasis. Autophagy-related factors involved in the labyrinthic and highly dynamic bone marrow workshop redesign the niche framework by coordinating the operational schedule of pluripotent stem and mature cells. The following report summarizes the most recent breakthroughs in our understanding of the intramural relationships between bone marrow cells and key autophagic mediators. Doubtless, the consideration of the autophagy-related and unrelated functions of main players, such as p62, Atg7, Atg5, and Beclin-1 remains a compelling task to thoroughly understand the complex relations between the heterogenic cell types that populate bone marrow.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Macerata, Italy
| | - Maria Giovanna Sabbieti
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Macerata, Italy
| |
Collapse
|
24
|
Functions of nuclear receptors SUMOylation. Clin Chim Acta 2021; 516:27-33. [PMID: 33476589 DOI: 10.1016/j.cca.2021.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/18/2022]
Abstract
The nuclear receptor superfamily is a family of ligand-activated transcription factors that play a key role in cell metabolism and human diseases. They can be modified after translation, such as acetylation, ubiquitination, phosphorylation and SUMOylation. Crosstalk between SUMO and ubiquitin, phosphorylation and acetylation regulates a variety of metabolic and physiological activities. Nuclear receptors play an important role in lipid metabolism, inflammation, bile acid homeostasis and autophagy. SUMOylation nuclear receptors can regulate their function and affect cell metabolism. It also provides a potential therapeutic target for atherosclerosis, tumor and other metabolic and inflammation-related diseases. This review focuses on the function of SUMOylation nuclear receptors.
Collapse
|
25
|
Duan X, Tong C. Autophagy in Drosophila and Zebrafish. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1208:333-356. [PMID: 34260032 DOI: 10.1007/978-981-16-2830-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Autophagy is a highly conserved cellular process that delivers cellular contents to the lysosome for degradation. It not only serves as a bulk degradation system for various cytoplasmic components but also functions selectively to clear damaged organelles, aggregated proteins, and invading pathogens (Feng et al., Cell Res 24:24-41, 2014; Galluzzi et al., EMBO J 36:1811-36, 2017; Klionsky et al., Autophagy 12:1-222, 2016). The malfunction of autophagy leads to multiple developmental defects and diseases (Mizushima et al., Nature 451:1069-75, 2008). Drosophila and zebrafish are higher metazoan model systems with sophisticated genetic tools readily available, which make it possible to dissect the autophagic processes and to understand the physiological functions of autophagy (Lorincz et al., Cells 6:22, 2017a; Mathai et al., Cells 6:21, 2017; Zhang and Baehrecke, Trends Cell Biol 25:376-87, 2015). In this chapter, we will discuss recent progress that has been made in the autophagic field by using these animal models. We will focus on the protein machineries required for autophagosome formation and maturation as well as the physiological roles of autophagy in both Drosophila and zebrafish.
Collapse
Affiliation(s)
- Xiuying Duan
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chao Tong
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China. .,The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
26
|
Chávez E, Velasco-Loyden G, Lozano-Rosas MG, Aguilar-Maldonado B, Muciño-Hernández G, Castro-Obregón S, Chagoya de Sánchez V. Role of autophagy in the chemopreventive effect of the IFC-305 compound in the sequential model of cirrhosis-hepatocellular carcinoma in the rat and in vitro. Am J Cancer Res 2020; 10:1844-1856. [PMID: 32642295 PMCID: PMC7339275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 05/30/2020] [Indexed: 06/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) can be originated from various etiologies and is preceded mostly by cirrhosis. Unfortunately, there is no effective treatment due to its late prognosis. Alterations in autophagy have been reported during the development and progression of HCC. Autophagy allows for the maintenance of a positive energy balance and the proper functioning of organelles through the selective degradation of cellular components. It has been demonstrated that autophagy suppresses spontaneous tumorigenesis in the liver. Therefore, autophagy has become a therapeutic target for effective HCC therapies. We have previously demonstrated that the adenosine-derived compound, IFC-305, has a chemopreventive effect on HCC, in addition to maintaining mitochondrial function in a sequential model of cirrhosis-HCC. Thus, the aim of this work was to determine if IFC-305 has an effect on autophagy in the sequential model of cirrhosis-HCC induced by diethylnitrosamine or in vitro in the HCC cell line HepG2 and mouse embryonic fibroblasts. The results of this work showed that IFC-305 modifies the levels of the BECN1, p62/SQSTM1 and LC3-II proteins that play an important role in the autophagic process. In vivo, IFC-305 regulates the levels of the PINK1 and PARKIN proteins that specifically mark mitochondria for repair or degradation. In the HepG2 cell line, its effect was accompanied by a decrease in cell viability. Interestingly, in nontumoral cells the time to autophagy induction was different compared to the HepG2 cells. This study suggests that autophagy induction may be part of the mechanism by which IFC-305 maintains mitochondrial function, thereby facilitating the prevention and reversal of HCC.
Collapse
Affiliation(s)
- Enrique Chávez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de MéxicoCiudad de México, México
| | - Gabriela Velasco-Loyden
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de MéxicoCiudad de México, México
| | - María Guadalupe Lozano-Rosas
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de MéxicoCiudad de México, México
| | - Beatriz Aguilar-Maldonado
- Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de MéxicoCiudad de México, México
| | - Gabriel Muciño-Hernández
- Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de MéxicoCiudad de México, México
| | - Susana Castro-Obregón
- Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de MéxicoCiudad de México, México
| | - Victoria Chagoya de Sánchez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de MéxicoCiudad de México, México
| |
Collapse
|
27
|
Zárraga-Granados G, Muciño-Hernández G, Sánchez-Carbente MR, Villamizar-Gálvez W, Peñas-Rincón A, Arredondo C, Andrés ME, Wood C, Covarrubias L, Castro-Obregón S. The nuclear receptor NR4A1 is regulated by SUMO modification to induce autophagic cell death. PLoS One 2020; 15:e0222072. [PMID: 32210435 PMCID: PMC7094859 DOI: 10.1371/journal.pone.0222072] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/08/2020] [Indexed: 12/31/2022] Open
Abstract
NR4A is a nuclear receptor protein family whose members act as sensors of cellular environment and regulate multiple processes such as metabolism, proliferation, migration, apoptosis, and autophagy. Since the ligand binding domains of these receptors have no cavity for ligand interaction, their function is most likely regulated by protein abundance and post-translational modifications. In particular, NR4A1 is regulated by protein abundance, phosphorylation, and subcellular distribution (nuclear-cytoplasmic translocation), and acts both as a transcription factor and as a regulator of other interacting proteins. SUMOylation is a post-translational modification that can affect protein stability, transcriptional activity, alter protein-protein interactions and modify intracellular localization of target proteins. In the present study we evaluated the role of SUMOylation as a posttranslational modification that can regulate the activity of NR4A1 to induce autophagy-dependent cell death. We focused on a model potentially relevant for neuronal cell death and demonstrated that NR4A1 needs to be SUMOylated to induce autophagic cell death. We observed that a triple mutant in SUMOylation sites has reduced SUMOylation, increased transcriptional activity, altered intracellular distribution, and more importantly, its ability to induce autophagic cell death is impaired.
Collapse
Affiliation(s)
- Gabriela Zárraga-Granados
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Gabriel Muciño-Hernández
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - María R. Sánchez-Carbente
- Biotechnology Research Center, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | - Wendy Villamizar-Gálvez
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Ana Peñas-Rincón
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Cristian Arredondo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María E. Andrés
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christopher Wood
- Laboratorio Nacional de Microscopía Avanzada, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico
| | - Luis Covarrubias
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico
| | - Susana Castro-Obregón
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
- * E-mail:
| |
Collapse
|
28
|
Miller DR, Cramer SD, Thorburn A. The interplay of autophagy and non-apoptotic cell death pathways. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 352:159-187. [PMID: 32334815 DOI: 10.1016/bs.ircmb.2019.12.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autophagy, the process of macromolecular degradation through the lysosome, has been extensively studied for the past decade or two. Autophagy can regulate cell death, especially apoptosis, through selective degradation of both positive and negative apoptosis regulators. However, multiple other programmed cell death pathways exist. As knowledge of these other types of cell death expand, it has been suggested that they also interact with autophagy. In this review, we discuss the molecular mechanisms that comprise three non-apoptotic forms of cell death (necroptosis, pyroptosis and ferroptosis) focusing on how the autophagy machinery regulates these different cell death mechanisms through (i) its degradative functions, i.e., true autophagy, and (ii) other non-degradative functions of the autophagy machinery such as serving as a signaling scaffold or by participating in other autophagy-independent cellular processes.
Collapse
Affiliation(s)
- Dannah R Miller
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Scott D Cramer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
29
|
Zhao YG, Zhang H. Core autophagy genes and human diseases. Curr Opin Cell Biol 2019; 61:117-125. [DOI: 10.1016/j.ceb.2019.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/11/2019] [Accepted: 08/04/2019] [Indexed: 02/06/2023]
|
30
|
Autophagy in bone homeostasis and the onset of osteoporosis. Bone Res 2019; 7:28. [PMID: 31666998 PMCID: PMC6804951 DOI: 10.1038/s41413-019-0058-7] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/28/2019] [Accepted: 06/02/2019] [Indexed: 02/06/2023] Open
Abstract
Autophagy is an evolutionarily conserved intracellular process, in which domestic cellular components are selectively digested for the recycling of nutrients and energy. This process is indispensable for cell homeostasis maintenance and stress responses. Both genetic and functional studies have demonstrated that multiple proteins involved in autophagic activities are critical to the survival, differentiation, and functioning of bone cells, including osteoblasts, osteocytes, and osteoclasts. Dysregulation at the level of autophagic activity consequently disturbs the balance between bone formation and bone resorption and mediates the onset and progression of multiple bone diseases, including osteoporosis. This review aims to introduce the topic of autophagy, summarize the understanding of its relevance in bone physiology, and discuss its role in the onset of osteoporosis and therapeutic potential.
Collapse
|
31
|
Lystad AH, Simonsen A. Mechanisms and Pathophysiological Roles of the ATG8 Conjugation Machinery. Cells 2019; 8:E973. [PMID: 31450711 PMCID: PMC6769624 DOI: 10.3390/cells8090973] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 12/18/2022] Open
Abstract
Since their initial discovery around two decades ago, the yeast autophagy-related (Atg)8 protein and its mammalian homologues of the light chain 3 (LC3) and γ-aminobutyric acid receptor associated proteins (GABARAP) families have been key for the tremendous expansion of our knowledge about autophagy, a process in which cytoplasmic material become targeted for lysosomal degradation. These proteins are ubiquitin-like proteins that become directly conjugated to a lipid in the autophagy membrane upon induction of autophagy, thus providing a marker of the pathway, allowing studies of autophagosome biogenesis and maturation. Moreover, the ATG8 proteins function to recruit components of the core autophagy machinery as well as cargo for selective degradation. Importantly, comprehensive structural and biochemical in vitro studies of the machinery required for ATG8 protein lipidation, as well as their genetic manipulation in various model organisms, have provided novel insight into the molecular mechanisms and pathophysiological roles of the mATG8 proteins. Recently, it has become evident that the ATG8 proteins and their conjugation machinery are also involved in intracellular pathways and processes not related to autophagy. This review focuses on the molecular functions of ATG8 proteins and their conjugation machinery in autophagy and other pathways, as well as their links to disease.
Collapse
Affiliation(s)
- Alf Håkon Lystad
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317 Oslo, Norway.
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317 Oslo, Norway.
| |
Collapse
|
32
|
Aas SN, Hamarsland H, Cumming KT, Rognlien SH, Aase OJ, Nordseth M, Karsrud S, Godager S, Tømmerbakke D, Handegard V, Raastad T. The impact of age and frailty on skeletal muscle autophagy markers and specific strength: A cross-sectional comparison. Exp Gerontol 2019; 125:110687. [PMID: 31404624 DOI: 10.1016/j.exger.2019.110687] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/18/2019] [Accepted: 08/08/2019] [Indexed: 01/06/2023]
Abstract
Aging is associated with reduced specific strength, defined as strength normalized to the cross-sectional area of a given muscle or muscle group. Dysregulated autophagy, impairing removal of dysfunctional proteins and organelles, is suggested as one of the underlying mechanisms. The aim of this study was to investigate levels of autophagic markers in skeletal muscle in groups known to differ in specific strength. Sixty-two volunteers were assigned to the following study groups: young, old non-frail, old pre-frail, and old frail individuals. Leg lean mass was assessed with dual-energy X-ray absorptiometry and quadriceps femoris muscle strength by isometric maximal voluntary contraction. The abundance of autophagic proteins within skeletal muscle cytosolic and membrane sub-fractions were determined by western blotting. In addition, the level of heat shock proteins and proteins involved in the regulation of protein synthesis were measured. The abundance of LC3-I was higher in old frail compared to young individuals. If the three elderly groups were pooled, the level of LC3-II was higher in old compared to young subjects. Pre-frail and frail elderly also displayed higher levels of certain heat shock proteins. No between-group differences were observed for p62, LC3-II/LC3-I ratio, or any of the anabolic signaling molecules. A negative correlation was observed between cytosolic LC3-I and specific strength. Higher levels of LC3-I in the frail elderly might represent attenuated autophagosome formation. However, higher LC3-II levels indicate an increased abundance of autophagosomes. These findings may therefore imply that both the process of autophagosome formation and autophagosome-lysosome fusion are affected in frail elderly. Higher levels of heat shock proteins might represent an auto-protective mechanism against increased levels of misfolded proteins, possibly due to inefficient degradation. In conclusion, the reduction in specific strength with aging and frailty may partly be caused by alterations in muscle protein quality control.
Collapse
Affiliation(s)
- Sigve Nyvik Aas
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway.
| | - Håvard Hamarsland
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | | | - Simen Helset Rognlien
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Ole Jølle Aase
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Martin Nordseth
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Stian Karsrud
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Sindre Godager
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Daniel Tømmerbakke
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Vilde Handegard
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Truls Raastad
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| |
Collapse
|
33
|
Stavoe AKH, Gopal PP, Gubas A, Tooze SA, Holzbaur ELF. Expression of WIPI2B counteracts age-related decline in autophagosome biogenesis in neurons. eLife 2019; 8:e44219. [PMID: 31309927 PMCID: PMC6634969 DOI: 10.7554/elife.44219] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/08/2019] [Indexed: 12/15/2022] Open
Abstract
Autophagy defects are implicated in multiple late-onset neurodegenerative diseases including Amyotrophic Lateral Sclerosis (ALS) and Alzheimer's, Huntington's, and Parkinson's diseases. Since aging is the most common shared risk factor in neurodegeneration, we assessed rates of autophagy in mammalian neurons during aging. We identified a significant decrease in the rate of constitutive autophagosome biogenesis during aging and observed pronounced morphological defects in autophagosomes in neurons from aged mice. While early stages of autophagosome formation were unaffected, we detected the frequent production of stalled LC3B-negative isolation membranes in neurons from aged mice. These stalled structures recruited the majority of the autophagy machinery, but failed to develop into LC3B-positive autophagosomes. Importantly, ectopically expressing WIPI2B effectively restored autophagosome biogenesis in aged neurons. This rescue is dependent on the phosphorylation state of WIPI2B at the isolation membrane, suggesting a novel therapeutic target in age-associated neurodegeneration.
Collapse
Affiliation(s)
- Andrea KH Stavoe
- Department of PhysiologyPerelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Pallavi P Gopal
- Department of PhysiologyPerelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Andrea Gubas
- Molecular Cell Biology of Autophagy LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| | - Sharon A Tooze
- Molecular Cell Biology of Autophagy LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| | - Erika LF Holzbaur
- Department of PhysiologyPerelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
34
|
Zheng H, Yuan C, Zhang H, Chen Y, Zhang H. The tissue- and developmental stage-specific involvement of autophagy genes in aggrephagy. Autophagy 2019; 16:589-599. [PMID: 31204564 DOI: 10.1080/15548627.2019.1632121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Genetic screens have identified two sets of genes that act at distinct steps of basal autophagy in higher eukaryotes: the pan-eukaryotic ATG genes and the metazoan-specific EPG genes. Very little is known about whether these core macroautophagy/autophagy genes are differentially employed during multicellular organism development. Here we analyzed the function of core autophagy genes in autophagic removal of SQST-1/SQSTM1 during C. elegans development. We found that loss of function of genes acting at distinct steps in the autophagy pathway causes different patterns of SQST-1 accumulation in different tissues and developmental stages. We also identified that the calpain protease clp-2 acts in a cell context-specific manner in SQST-1 degradation. clp-2 is required for degradation of SQST-1 in the hypodermis and neurons, but is dispensable in the body wall muscle and intestine. Our results indicate that autophagy genes are differentially employed in a tissue- and stage-specific manner during the development of multicellular organisms.Abbreviations: ATG: autophagy related; CLP: calpain family; EPG: ectopic PGL granules; ER: endoplasmic reticulum; ESCRT: endosomal sorting complex required for transport; GFP: green fluorescent protein; LGG-1/LC3: LC3, GABARAP and GATE-16 family; MIT: microtubule interacting and transport; PGL: P granule abnormality protein; SQST-1: sequestosome-related; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Hui Zheng
- Department of Immunology, Peking University School of Basic Medical Science, Beijing, P.R. China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Chongzhen Yuan
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Hui Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Yingyu Chen
- Department of Immunology, Peking University School of Basic Medical Science, Beijing, P.R. China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P.R. China
| |
Collapse
|
35
|
Gu H, Shi X, Liu C, Wang C, Sui N, Zhao Y, Gong J, Wang F, Zhang H, Li W, Zhao T. USP8 maintains embryonic stem cell stemness via deubiquitination of EPG5. Nat Commun 2019; 10:1465. [PMID: 30931944 PMCID: PMC6443784 DOI: 10.1038/s41467-019-09430-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 03/05/2019] [Indexed: 02/07/2023] Open
Abstract
Embryonic stem cells (ESCs) can propagate in an undifferentiated state indefinitely in culture and retain the potential to differentiate into any somatic lineage as well as germ cells. The catabolic process autophagy has been reported to be involved in ESC identity regulation, but the underlying mechanism is still largely unknown. Here we show that EPG5, a eukaryotic-specific autophagy regulator which mediates autophagosome/lysosome fusion, is highly expressed in ESCs and contributes to ESC identity maintenance. We identify that the deubiquitinating enzyme USP8 binds to the Coiled-coil domain of EPG5. Mechanistically, USP8 directly removes non-classical K63-linked ubiquitin chains from EPG5 at Lysine 252, leading to enhanced interaction between EPG5 and LC3. We propose that deubiquitination of EPG5 by USP8 guards the autophagic flux in ESCs to maintain their stemness. This work uncovers a novel crosstalk pathway between ubiquitination and autophagy through USP8-EPG5 interaction to regulate the stemness of ESCs.
Collapse
Affiliation(s)
- Haifeng Gu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xingxing Shi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chaoqun Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Qufu Normal University, Qufu, 273165, China
| | - Ning Sui
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Qufu Normal University, Qufu, 273165, China
| | - Yu Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiaqi Gong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fuping Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Hebei University, Baoding, 071002, China
| | - Hong Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
36
|
Wang Z, Zhang H. Phase Separation, Transition, and Autophagic Degradation of Proteins in Development and Pathogenesis. Trends Cell Biol 2019; 29:417-427. [PMID: 30826216 DOI: 10.1016/j.tcb.2019.01.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 01/03/2023]
Abstract
Phase separation and transition control the assembly and material states (liquid, gel like, or solid) of protein condensates to ensure that distinct cellular functions occur in a spatiotemporally controlled manner. The assembly and biophysical properties of condensates are precisely regulated by chaperone proteins, post-translational modifications (PTMs), and numerous cellular factors. Phase separation also triages misfolded and unwanted proteins for autophagic degradation. The concerted actions of receptor proteins, scaffold proteins, and PTMs determine the size, assembly rate, and material properties of condensates for efficient removal. Altered phase separation and transition affect the degradation of protein condensates, resulting in their accumulation under certain developmental and pathological conditions. Elucidation of the role of phase separation and transition in the degradation of disease-related protein condensates will provide insights into the molecular mechanism underlying the pathogenesis of various diseases.
Collapse
Affiliation(s)
- Zheng Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China.
| |
Collapse
|
37
|
Wang XJ, Zhou RJ, Zhang N, Jing Z. 20(S)-ginsenoside Rg3 sensitizes human non-small cell lung cancer cells to icotinib through inhibition of autophagy. Eur J Pharmacol 2019; 850:141-149. [PMID: 30772396 DOI: 10.1016/j.ejphar.2019.02.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 02/04/2023]
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have become a standard therapy for non-small cell lung cancer (NSCLC) patients with sensitive mutations. However, acquired resistance inevitably emerges after a median of 6-12 months. It has been demonstrated that autophagy plays an important role in EGFR-TKI resistance. 20(S)-ginsenoside Rg3 (Rg3) is proposed to sensitize the cancer cells to chemotherapy by inhibiting autophagy. We examined the ability of Rg3 to inhibit autophagy and increase the sensitivity of NSCLC cells to icotinib. We show that the induction of autophagy in response to icotinib contributes to the development of icotinib resistance. Rg3 is capable of inhibiting autophagic flux and enhancing the sensitivity of NSCLC cells to icotinib. The resistance to icotinib could also be reversed through Rg3-induced autophagy inhibition. Autophagy inhibition by Rg3 increases the therapeutic response in both icotinib-sensitive and icotinib-resistant NSCLC cells with an EGFR-activating mutation and may be an effective new treatment strategy for this disease.
Collapse
Affiliation(s)
- Xiao-Ju Wang
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou 34 Yanguan Lane, Hangzhou 310002, Zhejiang, PR China
| | - Rong-Jin Zhou
- Department of Pathology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, PR China
| | - Ni Zhang
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou 34 Yanguan Lane, Hangzhou 310002, Zhejiang, PR China
| | - Zhao Jing
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou 34 Yanguan Lane, Hangzhou 310002, Zhejiang, PR China.
| |
Collapse
|
38
|
Denton D, Xu T, Dayan S, Nicolson S, Kumar S. Crosstalk between Dpp and Tor signaling coordinates autophagy-dependent midgut degradation. Cell Death Dis 2019; 10:111. [PMID: 30737370 PMCID: PMC6368623 DOI: 10.1038/s41419-019-1368-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/06/2019] [Accepted: 01/14/2019] [Indexed: 11/17/2022]
Abstract
The majority of developmentally programmed cell death (PCD) is mediated by caspase-dependent apoptosis; however, additional modalities, including autophagy-dependent cell death, have important spatiotemporally restricted functions. Autophagy involves the engulfment of cytoplasmic components in a double membrane vesicle for delivery to the lysosome. An established model for autophagy-dependent PCD is Drosophila larval midgut removal during metamorphosis. Our previous work demonstrated that growth arrest is required to initiate autophagy-dependent midgut degradation and Target of rapamycin (Tor) limits autophagy induction. In further studies, we uncovered a role for Decapentaplegic (Dpp) in coordinating midgut degradation. Here, we provide new data to show that Dpp interacts with Tor during midgut degradation. Inhibiting Tor rescued the block in midgut degradation due to Dpp signaling. We propose that Dpp is upstream of Tor and down-regulation promotes growth arrest and autophagy-dependent midgut degradation. These findings underscore a relationship between Dpp and Tor signaling in the regulation of cell growth and tissue removal.
Collapse
Affiliation(s)
- Donna Denton
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA, 5001, Australia.
| | - Tianqi Xu
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA, 5001, Australia
| | - Sonia Dayan
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA, 5001, Australia
| | - Shannon Nicolson
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA, 5001, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA, 5001, Australia.
| |
Collapse
|
39
|
Gao H, Khawar MB, Li W. Autophagy in Reproduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1206:453-468. [PMID: 31776998 DOI: 10.1007/978-981-15-0602-4_21] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autophagy, a major degradation/recycling pathway, plays an essential role in cellular homeostasis maintenance, cell fate decision, and reproductive development. During reproduction, sperms and eggs, the specialized haploid gametes produced by the meiotic process of the germ cells in male and female respectively, are fused to form a new zygote that develops into fetus through embryogenesis and maternal-fetal crosstalk. Researches carried out in the past few years have proved that autophagy plays a key role in the regulation of reproduction process, and blockage of autophagy process likely contributes to reproductive abnormalities and even infertility. Here we summerize the recent progress in exploring the functional roles of autophagy in reproductive processes, such as spermatogenesis, folliculogenesis, fertilization, embryogenesis, and maternal-fetal crosstalk, in both animals and plants.
Collapse
Affiliation(s)
- Hui Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Muhammad Babar Khawar
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
40
|
Zhou H, Ma J, Shan Y, Qi X, Wang H, Jia L. A combination of chicken embryo extract and a nutritional supplement protect a rat model of aging againstd-galactose-induced dysfunction of mitochondria and autophagy. Food Funct 2019; 10:2774-2784. [DOI: 10.1039/c8fo01734d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aging process is usually associated with increased oxidative stress and deficiency of tissues and organs, which causes a decline in the life quality of individuals.
Collapse
Affiliation(s)
- Huimin Zhou
- College of Laboratory Medicine
- Dalian Medical University
- Dalian 116044
- China
- Department of Microbiology
| | - Jia Ma
- College of Laboratory Medicine
- Dalian Medical University
- Dalian 116044
- China
| | - Yujia Shan
- College of Laboratory Medicine
- Dalian Medical University
- Dalian 116044
- China
| | - Xia Qi
- College of Laboratory Medicine
- Dalian Medical University
- Dalian 116044
- China
| | - Huaxin Wang
- Department of Pathology
- Dalian Medical University
- Dalian 116044
- China
| | - Li Jia
- College of Laboratory Medicine
- Dalian Medical University
- Dalian 116044
- China
| |
Collapse
|
41
|
Zhao YG, Zhang H. Autophagosome maturation: An epic journey from the ER to lysosomes. J Cell Biol 2018; 218:757-770. [PMID: 30578282 PMCID: PMC6400552 DOI: 10.1083/jcb.201810099] [Citation(s) in RCA: 255] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 12/23/2022] Open
Abstract
Macroautophagy involves the sequestration of cytoplasmic contents in a double-membrane autophagosome and their delivery to lysosomes for degradation. In multicellular organisms, nascent autophagosomes fuse with vesicles originating from endolysosomal compartments before forming degradative autolysosomes, a process known as autophagosome maturation. ATG8 family members, tethering factors, Rab GTPases, and SNARE proteins act coordinately to mediate fusion of autophagosomes with endolysosomal vesicles. The machinery mediating autophagosome maturation is under spatiotemporal control and provides regulatory nodes to integrate nutrient availability with autophagy activity. Dysfunction of autophagosome maturation is associated with various human diseases, including neurodegenerative diseases, Vici syndrome, cancer, and lysosomal storage disorders. Understanding the molecular mechanisms underlying autophagosome maturation will provide new insights into the pathogenesis and treatment of these diseases.
Collapse
Affiliation(s)
- Yan G Zhao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Hong Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China .,National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
42
|
Velentzas PD, Zhang L, Das G, Chang TK, Nelson C, Kobertz WR, Baehrecke EH. The Proton-Coupled Monocarboxylate Transporter Hermes Is Necessary for Autophagy during Cell Death. Dev Cell 2018; 47:281-293.e4. [PMID: 30318245 DOI: 10.1016/j.devcel.2018.09.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/25/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023]
Abstract
Nutrient availability influences the production and degradation of materials that are required for cell growth and survival. Autophagy is a nutrient-regulated process that is used to degrade cytoplasmic materials and has been associated with human diseases. Solute transporters influence nutrient availability and sensing, yet we know little about how transporters influence autophagy. Here, we screen for solute transporters that are required for autophagy-dependent cell death and identify CG11665/hermes. We show that hermes is required for both autophagy during steroid-triggered salivary gland cell death and TNF-induced non-apoptotic eye cell death. hermes encodes a proton-coupled monocarboxylate transporter that preferentially transports pyruvate over lactate. mTOR signaling is elevated in hermes mutant cells, and decreased mTOR function suppresses the hermes salivary gland cell death phenotype. Hermes is most similar to human SLC16A11, a protein that was recently implicated in type 2 diabetes, thus providing a link between pyruvate, mTOR, autophagy, and possibly metabolic disorders.
Collapse
Affiliation(s)
- Panagiotis D Velentzas
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lejie Zhang
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Gautam Das
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Tsun-Kai Chang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Cancer Immunology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Charles Nelson
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - William R Kobertz
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
43
|
Abstract
Autophagy influences cell survival through maintenance of cell bioenergetics and clearance of protein aggregates and damaged organelles. Several lines of evidence indicate that autophagy is a multifaceted regulator of cell death, but controversy exists over whether autophagy alone can drive cell death under physiologically relevant circumstances. Here, we review the role of autophagy in cell death and examine how autophagy interfaces with other forms of cell death including apoptosis and necrosis.
Collapse
|
44
|
Issa AR, Sun J, Petitgas C, Mesquita A, Dulac A, Robin M, Mollereau B, Jenny A, Chérif-Zahar B, Birman S. The lysosomal membrane protein LAMP2A promotes autophagic flux and prevents SNCA-induced Parkinson disease-like symptoms in the Drosophila brain. Autophagy 2018; 14:1898-1910. [PMID: 29989488 PMCID: PMC6152503 DOI: 10.1080/15548627.2018.1491489] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 06/05/2018] [Accepted: 06/13/2018] [Indexed: 02/03/2023] Open
Abstract
The autophagy-lysosome pathway plays a fundamental role in the clearance of aggregated proteins and protection against cellular stress and neurodegenerative conditions. Alterations in autophagy processes, including macroautophagy and chaperone-mediated autophagy (CMA), have been described in Parkinson disease (PD). CMA is a selective autophagic process that depends on LAMP2A (lysosomal-associated membrane protein 2A), a mammal and bird-specific membrane glycoprotein that translocates cytosolic proteins containing a KFERQ-like peptide motif across the lysosomal membrane. Drosophila reportedly lack CMA and use endosomal microautophagy (eMI) as an alternative selective autophagic process. Here we report that neuronal expression of human LAMP2A protected Drosophila against starvation and oxidative stress, and delayed locomotor decline in aging flies without extending their lifespan. LAMP2A also prevented the progressive locomotor and oxidative defects induced by neuronal expression of PD-associated human SNCA (synuclein alpha) with alanine-to-proline mutation at position 30 (SNCAA30P). Using KFERQ-tagged fluorescent biosensors, we observed that LAMP2A expression stimulated selective autophagy in the adult brain and not in the larval fat body, but did not increase this process under starvation conditions. Noteworthy, we found that neurally expressed LAMP2A markedly upregulated levels of Drosophila Atg5, a key macroautophagy initiation protein, and that it increased the density of Atg8a/LC3-positive puncta, which reflects the formation of autophagosomes. Furthermore, LAMP2A efficiently prevented accumulation of the autophagy defect marker Ref(2)P/p62 in the adult brain under acute oxidative stress. These results indicate that LAMP2A can potentiate autophagic flux in the Drosophila brain, leading to enhanced stress resistance and neuroprotection. ABBREVIATIONS Act5C: actin 5C; a.E.: after eclosion; Atg5: autophagy-related 5; Atg8a/LC3: autophagy-related 8a; CMA: chaperone-mediated autophagy; DHE: dihydroethidium; elav: embryonic lethal abnormal vision; eMI: endosomal microautophagy; ESCRT: endosomal sorting complexes required for transport; GABARAP: GABA typeA receptor-associated protein; Hsc70-4: heat shock protein cognate 4; HSPA8/Hsc70: heat shock protein family A (Hsp70) member 8; LAMP2: lysosomal associated membrane protein 2; MDA: malondialdehyde; PA-mCherry: photoactivable mCherry; PBS: phosphate-buffered saline; PCR: polymerase chain reaction; PD: Parkinson disease; Ref(2)P/p62: refractory to sigma P; ROS: reactive oxygen species; RpL32/rp49: ribosomal protein L32; RT-PCR: reverse transcription polymerase chain reaction; SING: startle-induced negative geotaxis; SNCA/α-synuclein: synuclein alpha; SQSTM1/p62: sequestosome 1; TBS: Tris-buffered saline; UAS: upstream activating sequence.
Collapse
Affiliation(s)
- Abdul-Raouf Issa
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Jun Sun
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Céline Petitgas
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Ana Mesquita
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY, USA
| | - Amina Dulac
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Marion Robin
- ENSL, UCBL, CNRS, LBMC, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Bertrand Mollereau
- ENSL, UCBL, CNRS, LBMC, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Andreas Jenny
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY, USA
| | - Baya Chérif-Zahar
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Serge Birman
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| |
Collapse
|
45
|
Formation and maturation of autophagosomes in higher eukaryotes: a social network. Curr Opin Cell Biol 2018; 53:29-36. [DOI: 10.1016/j.ceb.2018.04.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 04/03/2018] [Accepted: 04/09/2018] [Indexed: 12/17/2022]
|
46
|
Abstract
This question of whether fungi undergo apoptosis-like programmed cell death can be separated into two questions. One question is about applying the term "apoptosis" to fungi, and the other is a more challenging question of whether fungi have evolved mechanisms that inflict self-injury. The answers to both questions depend on the definitions applied to "apoptosis" and "programmed cell death." Considering how these and other cell death terms originated and are currently defined for animals, some confusion arises when the terms are applied to fungi. While it is difficult to defend the concept of fungal apoptosis, the more interesting issue is whether fungi will eventually be found to encode programmed or extemporaneous self-destructive processes, as suggested by intriguing new findings.
Collapse
Affiliation(s)
- J Marie Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
47
|
Liu W, Duan X, Fang X, Shang W, Tong C. Mitochondrial protein import regulates cytosolic protein homeostasis and neuronal integrity. Autophagy 2018; 14:1293-1309. [PMID: 29909722 DOI: 10.1080/15548627.2018.1474991] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Neurodegeneration is characterized by protein aggregate deposits and mitochondrial malfunction. Reduction in Tom40 (translocase of outer membrane 40) expression, a key subunit of the translocase of the outer mitochondrial membrane complex, led to accumulation of ubiquitin (Ub)-positive protein aggregates engulfed by Atg8a-positive membranes. Other macroautophagy markers were also abnormally accumulated. Autophagy was induced but the majority of autophagosomes failed to fuse with lysosomes when Tom40 was downregulated. In Tom40 RNAi tissues, autophagosome-like (AL) structures, often not sealed, were 10 times larger than starvation induced autophagosomes. Atg5 downregulation abolished Tom40 RNAi induced AL structure formation, but the Ub-positive aggregates remained, whereas knock down of Syx17, a gene required for autophagosome-lysosome fusion, led to the disappearance of giant AL structures and accumulation of small autophagosomes and phagophores near the Ub-positive aggregates. The protein aggregates contained many mitochondrial preproteins, cytosolic proteins, and proteasome subunits. Proteasome activity and ATP levels were reduced and the ROS levels was increased in Tom40 RNAi tissues. The simultaneous inhibition of proteasome activity, reduction in ATP production, and increase in ROS, but none of these conditions alone, can mimic the imbalanced proteostasis phenotypes observed in Tom40 RNAi cells. Knockdown of ref(2)P or ectopic expression of Pink1 and park greatly reduced aggregate formation in Tom40 RNAi tissues. In nerve tissues, reduction in Tom40 activity leads to aggregate formation and neurodegeneration. Rather than diminishing the neurodegenerative phenotypes, overexpression of Pink1 enhanced them. We proposed that defects in mitochondrial protein import may be the key to linking imbalanced proteostasis and mitochondrial defects. ABBREVIATIONS AL: autophagosome-like; Atg12: Autophagy-related 12; Atg14: Autophagy-related 14; Atg16: Autophagy-related 16; Atg5: Autophagy-related 5; Atg6: Autophagy-related 6; Atg8a: Autophagy-related 8a; Atg9: Autophagy-related 9; ATP: adenosine triphosphate; Cas9: CRISPR associated protein 9; cDNA: complementary DNA; COX4: Cytochrome c oxidase subunit 4; CRISPR: clustered regularly interspaced short palindromic repeats; Cyt-c1: Cytochrome c1; DAPI: 4,6-diamidino-2-phenylindole dihydrochloride; Dcr-2: Dicer-2; FLP: Flippase recombination enzyme; FRT: FLP recombination target; GFP: green fluorescent protein; GO: gene ontology; gRNA: guide RNA; Hsp60: Heat shock protein 60A; HDAC6: Histone deacetylase 6; htt: huntingtin; Idh: Isocitrate dehydrogenase; IFA: immunofluorescence assay; Irp-1A: Iron regulatory protein 1A; kdn: knockdown; Marf: Mitochondrial assembly regulatory factor; MitoGFP: Mitochondrial-GFP; MS: mass spectrometry; MTPAP: mitochondrial poly(A) polymerase; Nmnat: Nicotinamide mononucleotide adenylyltransferase; OE: overexpression; Pink1/PINK1: PTEN-induced putative kinase 1; polyQ: polyglutamine; PRKN: parkin RBR E3 ubiquitin protein ligase; Prosα4: proteasome α4 subunit; Prosβ1: proteasome β1 subunit; Prosβ5: proteasome β5 subunit; Prosβ7: proteasome β7 subunit; ref(2)P: refractory to sigma P; RFP: red fluorescent protein; RNAi: RNA interference; ROS: reactive oxygen species; Rpn11: Regulatory particle non-ATPase 11; Rpt2: Regulatory particle triple-A ATPase 2; scu: scully; sicily: severe impairment of CI with lengthened youth; sesB: stress-sensitive B; Syx17: Syntaxin17; TEM: transmission electron microscopy; ttm50: tiny tim 50; Tom: translocase of the outer membrane; Tom20: translocase of outer membrane 20; Tom40: translocase of outer membrane 40; Tom70: translocase of outer membrane 70; UAS: upstream active sequence; Ub: ubiquitin; VNC: ventral nerve cord; ZFYVE1: zinc finger FYVE-type containing 1.
Collapse
Affiliation(s)
- Wei Liu
- a Life Sciences Institute and Innovation Center for Cell Signaling Network , Zhejiang University , Hangzhou , China
| | - Xiuying Duan
- a Life Sciences Institute and Innovation Center for Cell Signaling Network , Zhejiang University , Hangzhou , China
| | - Xuefei Fang
- a Life Sciences Institute and Innovation Center for Cell Signaling Network , Zhejiang University , Hangzhou , China
| | - Weina Shang
- a Life Sciences Institute and Innovation Center for Cell Signaling Network , Zhejiang University , Hangzhou , China
| | - Chao Tong
- a Life Sciences Institute and Innovation Center for Cell Signaling Network , Zhejiang University , Hangzhou , China
| |
Collapse
|
48
|
Thongrod S, Wanichanon C, Kankuan W, Siangcham T, Phadngam S, Morani F, Isidoro C, Sobhon P. Autophagy-Associated Shrinkage of the Hepatopancreas in Fasting Male Macrobrachium rosenbergii Is Rescued by Neuropeptide F. Front Physiol 2018; 9:613. [PMID: 29910737 PMCID: PMC5992412 DOI: 10.3389/fphys.2018.00613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/07/2018] [Indexed: 12/20/2022] Open
Abstract
Invertebrate neuropeptide F-I (NPF-I), much alike its mammalian homolog neuropeptide Y, influences several physiological processes, including circadian rhythms, cortical excitability, stress response, and food intake behavior. Given the role of autophagy in the metabolic stress response, we investigated the effect of NPF-1 on autophagy during fasting and feeding conditions in the hepatopancreas and muscle tissues of the male giant freshwater prawn Macrobrachium rosenbergii. Starvation up-regulated the expression of the autophagy marker LC3 in both tissues. Yet, based on the relative levels of the autophagosome-associated LC3-II isoform and of its precursor LC3-I, the hepatopancreas was more responsive than the muscle to starvation-induced autophagy. Injection of NPF-I inhibited the autophagosome formation in the hepatopancreas of fasting prawns. Relative to the body weight, the muscle weight was not affected, while that of the hepatopancreas decreased upon starvation and NPF-1 treatment could largely prevent such weight loss. Thus, the hepatopancreas is the reserve organ for the nutrient homeostasis during starvation and NPF-I plays a crucial role in the balancing of energy expenditure and energy intake during starvation by modulating autophagy.
Collapse
Affiliation(s)
- Sirorat Thongrod
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand.,Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Chaitip Wanichanon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Wilairat Kankuan
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand.,Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Tanapan Siangcham
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Suratchanee Phadngam
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Federica Morani
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand.,Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| |
Collapse
|
49
|
O'Keefe L, Denton D. Using Drosophila Models of Amyloid Toxicity to Study Autophagy in the Pathogenesis of Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5195416. [PMID: 29888266 PMCID: PMC5985114 DOI: 10.1155/2018/5195416] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/20/2018] [Accepted: 04/10/2018] [Indexed: 12/24/2022]
Abstract
Autophagy is a conserved catabolic pathway that involves the engulfment of cytoplasmic components such as large protein aggregates and organelles that are delivered to the lysosome for degradation. This process is important in maintaining neuronal function and raises the possibility of a role for autophagy in neurodegenerative diseases. Alzheimer's disease (AD) is the most prevalent form of these diseases and is characterized by the accumulation of amyloid plaques in the brain which arise due to the misfolding and aggregation of toxic peptides, including amyloid beta (Aβ). There is substantial evidence from both AD patients and animal models that autophagy is dysregulated in this disease. However, it remains to be determined whether this is protective or pathogenic as there is evidence that autophagy can act to promote the degradation as well as function in the generation of toxic Aβ peptides. Understanding the molecular details of the extensive crosstalk that occurs between the autophagic and endolysosomal cellular pathways is essential for identifying the molecular details of amyloid toxicity. Drosophila models that express the toxic proteins that aggregate in AD have been generated and have been shown to recapitulate hallmarks of the disease. Here we focus on what is known about the role of autophagy in amyloid toxicity in AD from mammalian models and how Drosophila models can be used to further investigate AD pathogenesis.
Collapse
Affiliation(s)
- Louise O'Keefe
- Department of Genetics and Evolution, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute, P.O. Box 11060, Adelaide, SA 5001, Australia
| | - Donna Denton
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia
| |
Collapse
|
50
|
Castets P, Frank S, Sinnreich M, Rüegg MA. "Get the Balance Right": Pathological Significance of Autophagy Perturbation in Neuromuscular Disorders. J Neuromuscul Dis 2018; 3:127-155. [PMID: 27854220 PMCID: PMC5271579 DOI: 10.3233/jnd-160153] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent research has revealed that autophagy, a major catabolic process in cells, is dysregulated in several neuromuscular diseases and contributes to the muscle wasting caused by non-muscle disorders (e.g. cancer cachexia) or during aging (i.e. sarcopenia). From there, the idea arose to interfere with autophagy or manipulate its regulatory signalling to help restore muscle homeostasis and attenuate disease progression. The major difficulty for the development of therapeutic strategies is to restore a balanced autophagic flux, due to the dynamic nature of autophagy. Thus, it is essential to better understand the mechanisms and identify the signalling pathways at play in the control of autophagy in skeletal muscle. A comprehensive analysis of the autophagic flux and of the causes of its dysregulation is required to assess the pathogenic role of autophagy in diseased muscle. Furthermore, it is essential that experiments distinguish between primary dysregulation of autophagy (prior to disease onset) and impairments as a consequence of the pathology. Of note, in most muscle disorders, autophagy perturbation is not caused by genetic modification of an autophagy-related protein, but rather through indirect alteration of regulatory signalling or lysosomal function. In this review, we will present the mechanisms involved in autophagy, and those ensuring its tight regulation in skeletal muscle. We will then discuss as to how autophagy dysregulation contributes to the pathogenesis of neuromuscular disorders and possible ways to interfere with this process to limit disease progression.
Collapse
Affiliation(s)
| | - Stephan Frank
- Institute of Pathology, Division of Neuropathology Basel University Hospital, Basel, Switzerland
| | - Michael Sinnreich
- Neuromuscular Research Center, Departments of Neurology and Biomedicine, Pharmazentrum, Basel, Switzerland
| | | |
Collapse
|