101
|
Pakpian N, Phopin K, Kitidee K, Govitrapong P, Wongchitrat P. Alterations in Mitochondrial Dynamic-related Genes in the Peripheral Blood of Alzheimer's Disease Patients. Curr Alzheimer Res 2020; 17:616-625. [PMID: 33023448 DOI: 10.2174/1567205017666201006162538] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 08/05/2020] [Accepted: 09/04/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Mitochondrial dysfunction is a pathological feature that manifests early in the brains of patients with Alzheimer's Disease (AD). The disruption of mitochondrial dynamics contributes to mitochondrial morphological and functional impairments. Our previous study demonstrated that the expression of genes involved in amyloid beta generation was altered in the peripheral blood of AD patients. OBJECTIVE The aim of this study was to further investigate the relative levels of mitochondrial genes involved in mitochondrial dynamics, including mitochondrial fission and fusion, and mitophagy in peripheral blood samples from patients with AD compared to healthy controls. METHODS The mRNA levels were analyzed by real-time polymerase chain reaction. Gene expression profiles were assessed in relation to cognitive performance. RESULTS Significant changes were observed in the mRNA expression levels of fission-related genes; Fission1 (FIS1) levels in AD subjects were significantly higher than those in healthy controls, whereas Dynamin- related protein 1 (DRP1) expression was significantly lower in AD subjects. The levels of the mitophagy-related genes, PTEN-induced kinase 1 (PINK1) and microtubule-associated protein 1 light chain 3 (LC3), were significantly increased in AD subjects and elderly controls compared to healthy young controls. The mRNA levels of Parkin (PARK2) were significantly decreased in AD. Correlations were found between the expression levels of FIS1, DRP1 and PARK2 and cognitive performance scores. CONCLUSION Alterations in mitochondrial dynamics in the blood may reflect impairments in mitochondrial functions in the central and peripheral tissues of AD patients. Mitochondrial fission, together with mitophagy gene profiles, might be potential considerations for the future development of blood-based biomarkers for AD.
Collapse
Affiliation(s)
- Nattaporn Pakpian
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Kamonrat Phopin
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Kuntida Kitidee
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | | | - Prapimpun Wongchitrat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom, Thailand
| |
Collapse
|
102
|
Abstract
Post-translational modifications of cellular substrates with ubiquitin and ubiquitin-like proteins (UBLs), including ubiquitin, SUMOs, and neural precursor cell-expressed developmentally downregulated protein 8, play a central role in regulating many aspects of cell biology. The UBL conjugation cascade is initiated by a family of ATP-dependent enzymes termed E1 activating enzymes and executed by the downstream E2-conjugating enzymes and E3 ligases. Despite their druggability and their key position at the apex of the cascade, pharmacologic modulation of E1s with potent and selective drugs has remained elusive until 2009. Among the eight E1 enzymes identified so far, those initiating ubiquitylation (UBA1), SUMOylation (SAE), and neddylation (NAE) are the most characterized and are implicated in various aspects of cancer biology. To date, over 40 inhibitors have been reported to target UBA1, SAE, and NAE, including the NAE inhibitor pevonedistat, evaluated in more than 30 clinical trials. In this Review, we discuss E1 enzymes, the rationale for their therapeutic targeting in cancer, and their different inhibitors, with emphasis on the pharmacologic properties of adenosine sulfamates and their unique mechanism of action, termed substrate-assisted inhibition. Moreover, we highlight other less-characterized E1s-UBA6, UBA7, UBA4, UBA5, and autophagy-related protein 7-and the opportunities for targeting these enzymes in cancer. SIGNIFICANCE STATEMENT: The clinical successes of proteasome inhibitors in cancer therapy and the emerging resistance to these agents have prompted the exploration of other signaling nodes in the ubiquitin-proteasome system including E1 enzymes. Therefore, it is crucial to understand the biology of different E1 enzymes, their roles in cancer, and how to translate this knowledge into novel therapeutic strategies with potential implications in cancer treatment.
Collapse
Affiliation(s)
- Samir H Barghout
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada (S.H.B., A.D.S.); Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada (S.H.B., A.D.S.); and Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt (S.H.B.)
| | - Aaron D Schimmer
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada (S.H.B., A.D.S.); Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada (S.H.B., A.D.S.); and Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt (S.H.B.)
| |
Collapse
|
103
|
Booth LA, Roberts JL, Dent P. The role of cell signaling in the crosstalk between autophagy and apoptosis in the regulation of tumor cell survival in response to sorafenib and neratinib. Semin Cancer Biol 2020; 66:129-139. [PMID: 31644944 PMCID: PMC7167338 DOI: 10.1016/j.semcancer.2019.10.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 09/23/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
The molecular mechanisms by which tumor cells survive or die following therapeutic interventions are complex. There are three broadly defined categories of cell death processes: apoptosis (Type I), autophagic cell death (Type II), and necrosis (Type III). In hematopoietic tumor cells, the majority of toxic stimuli cause these cells to undergo a death process called apoptosis; apoptosis specifically involves the cleavage of DNA into large defined pieces and their subsequent localization in vesicles. Thus, 'pure' apoptosis largely lacks inflammatory potential. In carcinomas, however, the mechanisms by which tumor cells ultimately die are considerably more complex. Although the machinery of apoptosis is engaged by toxic stimuli, other processes such as autophagy ("self-eating") and replicative cell death can lead to observations that do not simplistically correspond to any of the individual Type I-III formalized death categories. The 'hybrid' forms of cell death observed in carcinoma cells result in cellular materials being released into the extracellular space without packaging, which promotes inflammation, potentially leading to the accelerated re-growth of surviving tumor cells by macrophages. Drugs as single agents or in combinations can simultaneously initiate signaling via both apoptotic and autophagic pathways. Based on the tumor type and its oncogene drivers, as well as the drug(s) being used and the duration and intensity of the autophagosome signal, apoptosis and autophagy have the potential to act in concert to kill or alternatively that the actions of either pathway can act to suppress signaling by the other pathway. And, there also is evidence that autophagic flux, by causing lysosomal protease activation, with their subsequent release into the cytosol, can directly mediate killing. This review will discuss the interactive biology between apoptosis and autophagy in carcinoma cells. Finally, the molecular actions of the FDA-approved drugs neratinib and sorafenib, and how they enhance both apoptotic and toxic autophagic processes, alone or in combination with other agents, is discussed in a bench-to-bedside manner.
Collapse
Affiliation(s)
- Laurence A Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States
| | - Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States.
| |
Collapse
|
104
|
Golbabapour S, Bagheri-Lankarani K, Ghavami S, Geramizadeh B. Autoimmune Hepatitis and Stellate Cells: An Insight into the Role of Autophagy. Curr Med Chem 2020; 27:6073-6095. [PMID: 30947648 DOI: 10.2174/0929867326666190402120231] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 03/11/2019] [Accepted: 03/15/2019] [Indexed: 02/08/2023]
Abstract
Autoimmune hepatitis is a necroinflammatory process of liver, featuring interface hepatitis
by T cells, macrophages and plasma cells that invade to periportal parenchyma. In this process, a
variety of cytokines are secreted and liver tissues undergo fibrogenesis, resulting in the apoptosis of
hepatocytes. Autophagy is a complementary mechanism for restraining intracellular pathogens to
which the innate immune system does not provide efficient endocytosis. Hepatocytes with their
particular regenerative features are normally in a quiescent state, and, autophagy controls the accumulation
of excess products, therefore the liver serves as a basic model for the study of autophagy.
Impairment of autophagy in the liver causes the accumulation of damaged organelles, misfolded
proteins and exceeded lipids in hepatocytes as seen in metabolic diseases. In this review, we introduce
autoimmune hepatitis in association with autophagy signaling. We also discuss some genes and
proteins of autophagy, their regulatory roles in the activation of hepatic stellate cells and the importance
of lipophagy and tyrosine kinase in hepatic fibrogenesis. In order to provide a comprehensive
overview of the regulatory role of autophagy in autoimmune hepatitis, the pathway analysis of autophagy
in autoimmune hepatitis is also included in this article.
Collapse
Affiliation(s)
- Shahram Golbabapour
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Queen Elizabeth Hospital, Birmingham, B15 2WB, United Kingdom
| | - Kamran Bagheri-Lankarani
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Ghavami
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bita Geramizadeh
- Department of Pathology, Medical school of Shiraz University, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
105
|
Mei L, Qiu X, Jiang C, Yang A. Host Delipidation Mediated by Bacterial Effectors. Trends Microbiol 2020; 29:238-250. [PMID: 33092951 DOI: 10.1016/j.tim.2020.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022]
Abstract
Protein lipidation, the covalent attachment of a lipid moiety to a target protein, plays a critical role in many cellular processes in eukaryotic cells. Bacterial pathogens secrete various effectors to subvert the host signaling pathway as a mechanism of microbial pathogenesis. An increasing number of effectors from diverse bacterial pathogens function as cysteine proteases to cause irreversible delipidation of host lipidated proteins. This in turn results in disruption of crucial lipidation-mediated host signal transduction, thereby enabling pathogen survival and replication. In this review, we discuss the role of the bacterial effectors in interactions with the host and highlight our knowledge of irreversible host delipidation, with a focus on the common concerted biochemical mechanisms of the bacterial effectors.
Collapse
Affiliation(s)
- Ligang Mei
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaofeng Qiu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Chen Jiang
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing 401331, China; Ultrafast Transient Materials Science Center, Institute of Advanced Interdisciplinary Studies, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
106
|
Bindschedler A, Wacker R, Egli J, Eickel N, Schmuckli-Maurer J, Franke-Fayard BM, Janse CJ, Heussler VT. Plasmodium berghei sporozoites in nonreplicative vacuole are eliminated by a PI3P-mediated autophagy-independent pathway. Cell Microbiol 2020; 23:e13271. [PMID: 32979009 PMCID: PMC7757174 DOI: 10.1111/cmi.13271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/01/2022]
Abstract
The protozoan parasite Plasmodium, causative agent of malaria, invades hepatocytes by invaginating the host cell plasma membrane and forming a parasitophorous vacuole membrane (PVM). Surrounded by this PVM, the parasite undergoes extensive replication. Parasites inside a PVM provoke the Plasmodium‐associated autophagy‐related (PAAR) response. This is characterised by a long‐lasting association of the autophagy marker protein LC3 with the PVM, which is not preceded by phosphatidylinositol 3‐phosphate (PI3P)‐labelling. Prior to productive invasion, sporozoites transmigrate several cells and here we describe that a proportion of traversing sporozoites become trapped in a transient traversal vacuole, provoking a host cell response that clearly differs from the PAAR response. These trapped sporozoites provoke PI3P‐labelling of the surrounding vacuolar membrane immediately after cell entry, followed by transient LC3‐labelling and elimination of the parasite by lysosomal acidification. Our data suggest that this PI3P response is not only restricted to sporozoites trapped during transmigration but also affects invaded parasites residing in a compromised vacuole. Thus, host cells can employ a pathway distinct from the previously described PAAR response to efficiently recognise and eliminate Plasmodium parasites.
Collapse
Affiliation(s)
- Annina Bindschedler
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Rahel Wacker
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Jessica Egli
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Nina Eickel
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | - Blandine M Franke-Fayard
- Leiden malaria group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Chris J Janse
- Leiden malaria group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
107
|
Liu J, Guo ZN, Yan XL, Huang S, Ren JX, Luo Y, Yang Y. Crosstalk Between Autophagy and Ferroptosis and Its Putative Role in Ischemic Stroke. Front Cell Neurosci 2020; 14:577403. [PMID: 33132849 PMCID: PMC7566169 DOI: 10.3389/fncel.2020.577403] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022] Open
Abstract
Autophagy is a conserved process to maintains homeostasis via the degradation of toxic cell contents, which can either promote cell survival or accelerate cellular demise. Ferroptosis is a recently discovered iron-dependent cell death pathway associated with the accumulation of lethal reactive lipid species. In the past few years, an increasing number of studies have suggested the crosstalk between autophagy and ferroptosis. Ischemic stroke is a complex brain disease regulated by several cell death pathways, including autophagy and ferroptosis. However, the potential links between autophagy and ferroptosis in ischemic stroke have not yet been explored. In this review, we briefly overview the mechanisms of ferroptosis and autophagy, as well as their possible connections in ischemic stroke. The elucidation of crosstalk between different cell death pathways may provide insight into new future ischemic stroke therapies.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
| | - Xiu-Li Yan
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
| | - Shuo Huang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Jia-Xin Ren
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
| | - Yun Luo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| |
Collapse
|
108
|
Abdul Manap AS, Madhavan P, Vijayabalan S, Chia A, Fukui K. Explicating anti-amyloidogenic role of curcumin and piperine via amyloid beta (A β) explicit pathway: recovery and reversal paradigm effects. PeerJ 2020; 8:e10003. [PMID: 33062432 PMCID: PMC7532763 DOI: 10.7717/peerj.10003] [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: 03/10/2020] [Accepted: 08/30/2020] [Indexed: 12/28/2022] Open
Abstract
Previously, we reported the synergistic effects of curcumin and piperine in cell cultures as potential anti-cholinesterase and anti-amyloidogenic agents. Due to limited findings on the enrolment of these compounds on epigenetic events in AD, we aimed at elucidating the expression profiles of Aβ42-induced SH-SY5Y cells using microarray profiling. In this study, an optimized concentration of 35 µM of curcumin and piperine in combination was used to treat Aβ42 fibril and high-throughput microarray profiling was performed on the extracted RNA. This was then compared to curcumin and piperine used singularly at 49.11 µM and 25 µM, respectively. Our results demonstrated that in the curcumin treated group, from the top 10 upregulated and top 10 downregulated significantly differentially expressed genes (p < 0.05; fold change ≥ 2 or ≤ -2), there were five upregulated and three downregulated genes involved in the amyloidogenic pathway. While from top 10 upregulated and top 10 downregulated significantly differentially expressed genes (p < 0.05; fold change ≥ 2 or ≤ - 2) in the piperine treated group, there were four upregulated and three downregulated genes involved in the same pathway, whereas there were five upregulated and two downregulated genes involved (p < 0.05; fold change ≥ 2 or ≤ - 2) in the curcumin-piperine combined group. Four genes namely GABARAPL1, CTSB, RAB5 and AK5 were expressed significantly in all groups. Other genes such as ITPR1, GSK3B, PPP3CC, ERN1, APH1A, CYCS and CALM2 were novel putative genes that are involved in the pathogenesis of AD. We revealed that curcumin and piperine have displayed their actions against Aβ via the modulation of various mechanistic pathways. Alterations in expression profiles of genes in the neuronal cell model may explain Aβ pathology post-treatment and provide new insights for remedial approaches of a combined treatment using curcumin and piperine.
Collapse
Affiliation(s)
- Aimi Syamima Abdul Manap
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Shantini Vijayabalan
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Adeline Chia
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Koji Fukui
- Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Saitama, Japan
| |
Collapse
|
109
|
Gunata M, Parlakpinar H. A review of myocardial ischaemia/reperfusion injury: Pathophysiology, experimental models, biomarkers, genetics and pharmacological treatment. Cell Biochem Funct 2020; 39:190-217. [PMID: 32892450 DOI: 10.1002/cbf.3587] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/03/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases are known to be the most fatal diseases worldwide. Ischaemia/reperfusion (I/R) injury is at the centre of the pathology of the most common cardiovascular diseases. According to the World Health Organization estimates, ischaemic heart disease is the leading global cause of death, causing more than 9 million deaths in 2016. After cardiovascular events, thrombolysis, percutaneous transluminal coronary angioplasty or coronary bypass surgery are applied as treatment. However, after restoring coronary blood flow, myocardial I/R injury may occur. It is known that this damage occurs due to many pathophysiological mechanisms, especially increasing reactive oxygen types. Besides causing cardiomyocyte death through multiple mechanisms, it may be an important reason for affecting other cell types such as platelets, fibroblasts, endothelial and smooth muscle cells and immune cells. Also, polymorphonuclear leukocytes are associated with myocardial I/R damage during reperfusion. This damage may be insufficient in patients with co-morbidity, as it is demonstrated that it can be prevented by various endogenous antioxidant systems. In this context, the resulting data suggest that optimal cardioprotection may require a combination of additional or synergistic multi-target treatments. In this review, we discussed the pathophysiology, experimental models, biomarkers, treatment and its relationship with genetics in myocardial I/R injury. SIGNIFICANCE OF THE STUDY: This review summarized current information on myocardial ischaemia/reperfusion injury (pathophysiology, experimental models, biomarkers, genetics and pharmacological therapy) for researchers and reveals guiding data for researchers, especially in the field of cardiovascular system and pharmacology.
Collapse
Affiliation(s)
- Mehmet Gunata
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
110
|
Lyu N, Zhang J, Dai Y, Xiang J, Li Y, Xu J. Calcitriol inhibits apoptosis via activation of autophagy in hyperosmotic stress stimulated corneal epithelial cells in vivo and in vitro. Exp Eye Res 2020; 200:108210. [PMID: 32896533 DOI: 10.1016/j.exer.2020.108210] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/23/2020] [Accepted: 08/30/2020] [Indexed: 02/09/2023]
Abstract
BACKGROUND Previously, calcitriol has been demonstrated as a potential therapeutic agent for dry eye, whilst its role on corneal epithelium death remains unclear. This study aims to investigate the relationship between apoptosis and autophagy on dry eye related scenario, as well as the effect of calcitriol and its potential mechanism. METHODS In vitro, immortalized human corneal epithelial cells (iHCEC) were cultured in hyperosmotic medium with or without various concentrations of calcitriol and other reagents. In vivo, Wistar rats were applied with benzalkonium chloride to induce dry eye. Then rats were topically treated with calcitriol (10-6 M) for 14 days. Autophagy flux (LC3B-II and SQSTM1/P62) was examined by western blotting or immunostaining. To test cell apoptosis, western blotting for cleaved caspase-3, Annexin V/PI double staining and TUNEL assay were used. CCK-8 assay was performed to detect the cell viability. Small interfering RNA was used to knock down the expression of vitamin D receptor in iHCECs. RESULTS Autophagy activation could protect iHCECs against HS induced apoptosis in vitro, and calcitriol was able to augment autophagy flux via VDR signaling, shown as the remarkably elevated expression of LC3B-II, as well as the declined p62 expression. In vivo results further supported the protective role of calcitriol on corneal epithelium apoptosis through promoting autophagy in dry eye rats. CONCLUSION The current study indicated that autophagy was an adaptive change of corneal epithelial cells in response to hyperosmotic stress and calcitriol could prevent cells from apoptosis via further activation of autophagy through VDR pathway.
Collapse
Affiliation(s)
- Ning Lyu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Myopia, Fudan University, Shanghai, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jing Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Myopia, Fudan University, Shanghai, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yiqin Dai
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Myopia, Fudan University, Shanghai, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jun Xiang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Myopia, Fudan University, Shanghai, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yue Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Myopia, Fudan University, Shanghai, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jianjiang Xu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Myopia, Fudan University, Shanghai, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.
| |
Collapse
|
111
|
Bjedov I, Rallis C. The Target of Rapamycin Signalling Pathway in Ageing and Lifespan Regulation. Genes (Basel) 2020; 11:E1043. [PMID: 32899412 PMCID: PMC7565554 DOI: 10.3390/genes11091043] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
Ageing is a complex trait controlled by genes and the environment. The highly conserved mechanistic target of rapamycin signalling pathway (mTOR) is a major regulator of lifespan in all eukaryotes and is thought to be mediating some of the effects of dietary restriction. mTOR is a rheostat of energy sensing diverse inputs such as amino acids, oxygen, hormones, and stress and regulates lifespan by tuning cellular functions such as gene expression, ribosome biogenesis, proteostasis, and mitochondrial metabolism. Deregulation of the mTOR signalling pathway is implicated in multiple age-related diseases such as cancer, neurodegeneration, and auto-immunity. In this review, we briefly summarise some of the workings of mTOR in lifespan and ageing through the processes of transcription, translation, autophagy, and metabolism. A good understanding of the pathway's outputs and connectivity is paramount towards our ability for genetic and pharmacological interventions for healthy ageing and amelioration of age-related disease.
Collapse
Affiliation(s)
- Ivana Bjedov
- UCL Cancer Institute, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - Charalampos Rallis
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| |
Collapse
|
112
|
Hu SQ, Zhang QC, Meng QB, Hu AN, Zou JP, Li XL. Autophagy regulates exosome secretion in rat nucleus pulposus cells via the RhoC/ROCK2 pathway. Exp Cell Res 2020; 395:112239. [PMID: 32828826 DOI: 10.1016/j.yexcr.2020.112239] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
Our present study investigated whether exosome secretion of nucleus pulposus cells (NPCs) is regulated by autophagy. Different autophagic states of NPCs were induced by rapamycin (Rap), bafilomycin A1 (Baf) and other agents, and it was found that exosomes were secreted in an autophagy-dependent manner. Activation or inhibition of autophagy increased or decreased, respectively, the amount of exosomes that were released into the extracellular space. In addition, in order to confirm that Rap-promoted release of exosomes was mediated by autophagy rather than other pathways, we used autophagy associated gene 5 (ATG5) small-interfering RNA (siRNA) to silence the expression of ATG5 gene, which is indispensable for autophagy. The results showed that siRNA against ATG5 (siATG5) induced an accumulation of intraluminal vesicles (ILVs) in NPCs and a concomitant decrease in the amount of exosomes isolated from supernatant. Ras homolog gene (Rho) and Rho-associated coiled-coil forming protein kinase (ROCK) family molecules are capable of cytoskeletal remodeling and affecting vesicle transport. Therefore, we carried out targeted interventions and evaluated the effects of the RhoC/ROCK2 pathway on the secretion of exosomes within autophagic environment. Knockdown of RhoC and ROCK2 with corresponding siRNA significantly inhibited the secretion of exosomes originating from ILVs in NPCs, even when NPCs were subsequently treated with Rap. Taken together, our findings suggest that autophagy positively regulates expression levels of RhoC and ROCK2, and that the RhoC/ROCK2 pathway exerts a key function on NPCs-derived exosome secretion.
Collapse
Affiliation(s)
- Shun-Qi Hu
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qi-Chen Zhang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qing-Bing Meng
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - An-Nan Hu
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jia-Peng Zou
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xi-Lei Li
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
113
|
Tissue-Specific Impact of Autophagy Genes on the Ubiquitin-Proteasome System in C. elegans. Cells 2020; 9:cells9081858. [PMID: 32784405 PMCID: PMC7464313 DOI: 10.3390/cells9081858] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 12/30/2022] Open
Abstract
The ubiquitin–proteasome system (UPS) and the autophagy–lysosomal pathway (ALP) are the two main eukaryotic intracellular proteolytic systems involved in maintaining proteostasis. Several studies have reported on the interplay between the UPS and ALP, however it remains largely unknown how compromised autophagy affects UPS function in vivo. Here, we have studied the crosstalk between the UPS and ALP by investigating the tissue-specific effect of autophagy genes on the UPS at an organismal level. Using transgenic Caenorhabditis elegans expressing fluorescent UPS reporters, we show that the downregulation of the autophagy genes lgg-1 and lgg-2 (ATG8/LC3/GABARAP), bec-1 (BECLIN1), atg-7 (ATG7) and epg-5 (mEPG5) by RNAi decreases proteasomal degradation, concomitant with the accumulation of polyubiquitinated proteasomal substrates in a tissue-specific manner. For some of these genes, the changes in proteasomal degradation occur without a detectable alteration in proteasome tissue expression levels. In addition, the lgg-1 RNAi-induced reduction in proteasome activity in intestinal cells is not dependent on sqst-1/p62 accumulation. Our results illustrate that compromised autophagy can affect UPS in a tissue-specific manner, and demonstrate that UPS does not function as a direct compensatory mechanism in an animal. Further, a more profound understanding of the multilayered crosstalk between UPS and ALP can facilitate the development of therapeutic options for various disorders linked to dysfunction in proteostasis.
Collapse
|
114
|
Tan M, Zhang QB, Liu TH, Yang YY, Zheng JX, Zhou WJ, Xiong Q, Qing YF. Autophagy dysfunction may be involved in the pathogenesis of ankylosing spondylitis. Exp Ther Med 2020; 20:3578-3586. [PMID: 32855711 PMCID: PMC7444354 DOI: 10.3892/etm.2020.9116] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 07/14/2020] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to investigate the expression and significance of the mRNA of genes associated with autophagy and long non-coding RNA (lncRNA) GAS5 in peripheral blood mononuclear cells (PBMCs) of patients with ankylosing spondylitis (AS). The mRNA levels of microtubule-associated protein light chain 3 (LC3), Beclin1, autophagy-related gene (ATG)3, ATG5, ATG12, ATG 16 ligand 1 (ATG16L1) and lncRNA growth arrest-specific 5 (GAS5) in PBMCs from 60 patients with AS and 30 healthy controls (HC) were examined by reverse transcription-quantitative PCR. The correlations between the levels of LC3, Beclin1, ATG3, ATG5, ATG12 and ATG16L1 mRNA as well as lncRNA GAS5 levels with disease activity and laboratory parameters in patients with AS were determined by Spearman correlation analysis. In addition, the diagnostic value of lncRNA GAS5 for AS was explored through establishing a receiver operating characteristic (ROC) curve. The results indicated that, compared to the HCs, patients with AS had lower expression levels of LC3, ATG5, ATG12, ATG16L1 and lncRNA GAS5 in their PBMCs. Compared with those in patients with inactive AS, the levels of ATG5 and ATG12 were lower than those in patients with active AS. Of note, ATG5 and ATG12 mRNA levels were negatively correlated with disease activity indexes. lncRNA GAS5 was positively correlated with the expression of Beclin1, ATG3, ATG5, ATG12 and ATG16L1. The area under the ROC curve for the use of lncRNA GAS5 expression to diagnose AS was 0.808 with a 95% CI of 0.714-0.902. In conclusion, patients with AS had decreased expression of genes associated with autophagy and lncRNA GAS5. The extent of the reduction in ATG5 and ATG12 expression levels in patients with AS was correlated with the disease severity and activity. Furthermore, lncRNA GAS5 was a diagnostic indicator of AS.
Collapse
Affiliation(s)
- Min Tan
- Department of Geriatrics, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Quan-Bo Zhang
- Department of Geriatrics, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Tao-Hong Liu
- Department of Geriatrics, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yan-Yu Yang
- Department of Geriatrics, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Jian-Xiong Zheng
- Department of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Wen-Jun Zhou
- Department of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Qin Xiong
- Department of Geriatrics, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yu-Feng Qing
- Department of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
115
|
Huang Q, Liu Y, Zhang S, Yap YT, Li W, Zhang D, Gardner A, Zhang L, Song S, Hess RA, Zhang Z. Autophagy core protein ATG5 is required for elongating spermatid development, sperm individualization and normal fertility in male mice. Autophagy 2020; 17:1753-1767. [PMID: 32677505 DOI: 10.1080/15548627.2020.1783822] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Spermiogenesis is the longest phase of spermatogenesis, with dramatic morphological changes and a final step of spermiation, which involves protein degradation and the removal of excess cytoplasm; therefore, we hypothesized that macroautophagy/autophagy might be involved in the process. To test this hypothesis, we examined the function of ATG5, a core autophagy protein in male germ cell development. Floxed Atg5 and Stra8- iCre mice were crossed to conditionally inactivate Atg5 in male germ cells. In Atg5flox/flox; Stra8- iCre mutant mice, testicular expression of the autophagosome marker LC3A/B-II was significantly reduced, and expression of autophagy receptor SQSTM1/p62 was significantly increased, indicating a decrease in testicular autophagy activity. The fertility of mutant mice was dramatically reduced with about 70% being infertile. Sperm counts and motility were also significantly reduced compared to controls. Histological examination of the mutant testes revealed numerous, large residual bodies in the lumen of stages after their normal resorption within the seminiferous epithelium. The cauda epididymal lumen was filled with sloughed germ cells, large cytoplasmic bodies, and spermatozoa with disorganized heads and tails. Examination of cauda epididymal sperm by electron microscopy revealed misshapen sperm heads, a discontinuous accessory structure in the mid-piece and abnormal acrosome formation and loss of sperm individualization. Immunofluorescence staining of epididymal sperm showed abnormal mitochondria and acrosome distribution in the mutant mice. ATG5 was shown to induce autophagy by mediating multiple signals to maintain normal developmental processes. Our study demonstrated ATG5 is essential for male fertility and is involved in various aspects of spermiogenesis.Abbreviations: AKAP4: a-kinase anchoring protein 4; ATG5: autophagy-related 5; ATG7: autophagy-related 7; ATG10: autophagy-related 10; ATG12: autophagy-related 12; cKO: conditional knockout; DDX4: DEAD-box helicase 4; MAP1LC3/LC3/tg8: microtubule-associated protein 1 light chain 3; PBS: phosphate-buffered saline; PIWIL2/MILI: piwi like RNA-mediated gene silencing 2; RT-PCR: reverse transcription-polymerase chain reaction; SQSTM1/p62: sequestosome 1; TBC: tubulobulbar complexes; WT: wild type.
Collapse
Affiliation(s)
- Qian Huang
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, MI, USA
| | - Yunhao Liu
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Shiyang Zhang
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, MI, USA
| | - Yi Tian Yap
- Department of Physiology, Wayne State University, Detroit, MI, USA
| | - Wei Li
- Department of Physiology, Wayne State University, Detroit, MI, USA
| | - David Zhang
- School of Arts and Sciences, College of William and Mary, Williamsburg, VA, USA
| | - Ahmad Gardner
- Department of Physiology, Wayne State University, Detroit, MI, USA.,Detroit High School, Detroit, MI, USA
| | - Ling Zhang
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Shizheng Song
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Rex A Hess
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, IL, USA
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, MI, USA.,Department of Obstetrics/Gynecology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
116
|
Bonam SR, Bayry J, Tschan MP, Muller S. Progress and Challenges in The Use of MAP1LC3 as a Legitimate Marker for Measuring Dynamic Autophagy In Vivo. Cells 2020; 9:E1321. [PMID: 32466347 PMCID: PMC7291013 DOI: 10.3390/cells9051321] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 01/02/2023] Open
Abstract
Tremendous efforts have been made these last decades to increase our knowledge of intracellular degradative systems, especially in the field of autophagy. The role of autophagy in the maintenance of cell homeostasis is well documented and the existence of defects in the autophagic machinery has been largely described in diseases and aging. Determining the alterations occurring in the many forms of autophagy that coexist in cells and tissues remains complicated, as this cellular process is highly dynamic in nature and can vary from organ to organ in the same individual. Although autophagy is extensively studied, its functioning in different tissues and its links with other biological processes is still poorly understood. Several assays have been developed to monitor autophagy activity in vitro, ex vivo, and in vivo, based on different markers, the use of various inhibitors and activators, and distinct techniques. This review emphasizes the methods applied to measure (macro-)autophagy in tissue samples and in vivo via a protein, which centrally intervenes in the autophagy pathway, the microtubule-associated protein 1A/1B-light chain 3 (MAP1LC3), which is the most widely used marker and the first identified to associate with autophagosomal structures. These approaches are presented and discussed in terms of pros and cons. Some recommendations are provided to improve the reliability of the interpretation of results.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- CNRS, Biotechnology and Cell Signaling, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, 67412 Strasbourg University/Laboratory of Excellence Medalis, 67000 Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France;
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France;
| | - Mario P. Tschan
- Institute of Pathology, Division of Experimental Pathology, University of Bern, 3008 Bern, Switzerland;
| | - Sylviane Muller
- CNRS, Biotechnology and Cell Signaling, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, 67412 Strasbourg University/Laboratory of Excellence Medalis, 67000 Strasbourg, France
- Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University, 67000 Strasbourg, France
- University of Strasbourg Institute for Advanced Study, 67000 Strasbourg, France
| |
Collapse
|
117
|
Yun HR, Jo YH, Kim J, Shin Y, Kim SS, Choi TG. Roles of Autophagy in Oxidative Stress. Int J Mol Sci 2020; 21:ijms21093289. [PMID: 32384691 PMCID: PMC7246723 DOI: 10.3390/ijms21093289] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a catabolic process for unnecessary or dysfunctional cytoplasmic contents by lysosomal degradation pathways. Autophagy is implicated in various biological processes such as programmed cell death, stress responses, elimination of damaged organelles and development. The role of autophagy as a crucial mediator has been clarified and expanded in the pathological response to redox signalling. Autophagy is a major sensor of the redox signalling. Reactive oxygen species (ROS) are highly reactive molecules that are generated as by-products of cellular metabolism, principally by mitochondria. Mitochondrial ROS (mROS) are beneficial or detrimental to cells depending on their concentration and location. mROS function as redox messengers in intracellular signalling at physiologically low level, whereas excessive production of mROS causes oxidative damage to cellular constituents and thus incurs cell death. Hence, the balance of autophagy-related stress adaptation and cell death is important to comprehend redox signalling-related pathogenesis. In this review, we attempt to provide an overview the basic mechanism and function of autophagy in the context of response to oxidative stress and redox signalling in pathology.
Collapse
Affiliation(s)
- Hyeong Rok Yun
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
| | - Yong Hwa Jo
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Jieun Kim
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (H.R.Y.); (Y.S.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.S.K.); (T.G.C.); Tel.: +82-2-961-0524 (S.S.K.); +82-2-961-0287 (T.G.C.)
| | - Tae Gyu Choi
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.S.K.); (T.G.C.); Tel.: +82-2-961-0524 (S.S.K.); +82-2-961-0287 (T.G.C.)
| |
Collapse
|
118
|
Nakatogawa H. Mechanisms governing autophagosome biogenesis. Nat Rev Mol Cell Biol 2020; 21:439-458. [PMID: 32372019 DOI: 10.1038/s41580-020-0241-0] [Citation(s) in RCA: 457] [Impact Index Per Article: 114.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2020] [Indexed: 12/20/2022]
Abstract
Autophagosomes are double-membrane vesicles newly formed during autophagy to engulf a wide range of intracellular material and transport this autophagic cargo to lysosomes (or vacuoles in yeasts and plants) for subsequent degradation. Autophagosome biogenesis responds to a plethora of signals and involves unique and dynamic membrane processes. Autophagy is an important cellular mechanism allowing the cell to meet various demands, and its disruption compromises homeostasis and leads to various diseases, including metabolic disorders, neurodegeneration and cancer. Thus, not surprisingly, the elucidation of the molecular mechanisms governing autophagosome biogenesis has attracted considerable interest. Key molecules and organelles involved in autophagosome biogenesis, including autophagy-related (ATG) proteins and the endoplasmic reticulum, have been discovered, and their roles and relationships have been investigated intensely. However, several fundamental questions, such as what supplies membranes/lipids to build the autophagosome and how the membrane nucleates, expands, bends into a spherical shape and finally closes, have proven difficult to address. Nonetheless, owing to recent studies with new approaches and technologies, we have begun to unveil the mechanisms underlying these processes on a molecular level. We now know that autophagosome biogenesis is a highly complex process, in which multiple proteins and lipids from various membrane sources, supported by the formation of membrane contact sites, cooperate with biophysical phenomena, including membrane shaping and liquid-liquid phase separation, to ensure seamless segregation of the autophagic cargo. Together, these studies pave the way to obtaining a holistic view of autophagosome biogenesis.
Collapse
Affiliation(s)
- Hitoshi Nakatogawa
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.
| |
Collapse
|
119
|
Wang S, Li Y, Ma C. Atg3 promotes Atg8 lipidation via altering lipid diffusion and rearrangement. Protein Sci 2020; 29:1511-1523. [PMID: 32277540 DOI: 10.1002/pro.3866] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 12/20/2022]
Abstract
Atg3-catalyzed transferring of Atg8 to phosphatidylethanolamine (PE) in the phagophore membrane is essential for autophagy. Previous studies have demonstrated that this process requires Atg3 to interact with the phagophore membrane via its N-terminal amphipathic helix. In this study, by using combined biochemical and biophysical approaches, our data showed that in addition to binding to the membranes, Atg3 attenuates lipid diffusion and enriches lipid molecules with smaller headgroup. Our data suggest that Atg3 promotes Atg8 lipidation via altering lipid diffusion and rearrangement.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Li
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian, China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
120
|
Proteostasis Failure in Neurodegenerative Diseases: Focus on Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5497046. [PMID: 32308803 PMCID: PMC7140146 DOI: 10.1155/2020/5497046] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 03/03/2020] [Indexed: 12/11/2022]
Abstract
Protein homeostasis or proteostasis is an essential balance of cellular protein levels mediated through an extensive network of biochemical pathways that regulate different steps of the protein quality control, from the synthesis to the degradation. All proteins in a cell continuously turn over, contributing to development, differentiation, and aging. Due to the multiple interactions and connections of proteostasis pathways, exposure to stress conditions may cause various types of protein damage, altering cellular homeostasis and disrupting the entire network with additional cellular stress. Furthermore, protein misfolding and/or alterations during protein synthesis results in inactive or toxic proteins, which may overload the degradation mechanisms. The maintenance of a balanced proteome, preventing the formation of impaired proteins, is accomplished by two major catabolic routes: the ubiquitin proteasomal system (UPS) and the autophagy-lysosomal system. The proteostasis network is particularly important in nondividing, long-lived cells, such as neurons, as its failure is implicated with the development of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. These neurological disorders share common risk factors such as aging, oxidative stress, environmental stress, and protein dysfunction, all of which alter cellular proteostasis, suggesting that general mechanisms controlling proteostasis may underlay the etiology of these diseases. In this review, we describe the major pathways of cellular proteostasis and discuss how their disruption contributes to the onset and progression of neurodegenerative diseases, focusing on the role of oxidative stress.
Collapse
|
121
|
Echavarria-Consuegra L, Smit JM, Reggiori F. Role of autophagy during the replication and pathogenesis of common mosquito-borne flavi- and alphaviruses. Open Biol 2020; 9:190009. [PMID: 30862253 PMCID: PMC6451359 DOI: 10.1098/rsob.190009] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Arboviruses that are transmitted to humans by mosquitoes represent one of the most important causes of febrile illness worldwide. In recent decades, we have witnessed a dramatic re-emergence of several mosquito-borne arboviruses, including dengue virus (DENV), West Nile virus (WNV), chikungunya virus (CHIKV) and Zika virus (ZIKV). DENV is currently the most common mosquito-borne arbovirus, with an estimated 390 million infections worldwide annually. Despite a global effort, no specific therapeutic strategies are available to combat the diseases caused by these viruses. Multiple cellular pathways modulate the outcome of infection by either promoting or hampering viral replication and/or pathogenesis, and autophagy appears to be one of them. Autophagy is a degradative pathway generally induced to counteract viral infection. Viruses, however, have evolved strategies to subvert this pathway and to hijack autophagy components for their own benefit. In this review, we will focus on the role of autophagy in mosquito-borne arboviruses with emphasis on DENV, CHIKV, WNV and ZIKV, due to their epidemiological importance and high disease burden.
Collapse
Affiliation(s)
- Liliana Echavarria-Consuegra
- 1 Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen , Groningen , The Netherlands
| | - Jolanda M Smit
- 1 Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen , Groningen , The Netherlands
| | - Fulvio Reggiori
- 2 Department of Cell Biology, University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| |
Collapse
|
122
|
Allen EA, Baehrecke EH. Autophagy in animal development. Cell Death Differ 2020; 27:903-918. [PMID: 31988494 PMCID: PMC7206001 DOI: 10.1038/s41418-020-0497-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/19/2019] [Accepted: 01/07/2020] [Indexed: 01/13/2023] Open
Abstract
Macroautophagy (autophagy) delivers intracellular constituents to the lysosome to promote catabolism. During development in multiple organisms, autophagy mediates various cellular processes, including survival during starvation, programmed cell death, phagocytosis, organelle elimination, and miRNA regulation. Our current understanding of autophagy has been enhanced by developmental biology research during the last quarter of a century. Through experiments that focus on animal development, fundamental mechanisms that control autophagy and that contribute to disease were elucidated. Studies in embryos revealed specific autophagy molecules that mediate the removal of paternally derived mitochondria, and identified autophagy components that clear protein aggregates during development. Importantly, defects in mtDNA inheritance, or removal of paternal mtDNA via mitochondrial autophagy, can contribute to mitochondrial-associated disease. In addition, impairment of the clearance of protein aggregates by autophagy underlies neurodegenerative diseases. Experiments in multiple organisms also reveal conserved mechanisms of tissue remodeling that rely on the cooperation between autophagy and apoptosis to clear cell corpses, and defects in autophagy and apoptotic cell clearance can contribute to inflammation and autoimmunity. Here we provide an overview of key developmental processes that are mediated by autophagy in multiple animals.
Collapse
Affiliation(s)
- Elizabeth A Allen
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 423 Lazare Research Building, 364 Plantation St., Worcester, MA, 01655, USA
| | - Eric H Baehrecke
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 423 Lazare Research Building, 364 Plantation St., Worcester, MA, 01655, USA.
| |
Collapse
|
123
|
Suresh SN, Chakravorty A, Giridharan M, Garimella L, Manjithaya R. Pharmacological Tools to Modulate Autophagy in Neurodegenerative Diseases. J Mol Biol 2020; 432:2822-2842. [PMID: 32105729 DOI: 10.1016/j.jmb.2020.02.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/03/2020] [Accepted: 02/12/2020] [Indexed: 12/11/2022]
Abstract
Considerable evidences suggest a link between autophagy dysfunction, protein aggregation, and neurodegenerative diseases. Given that autophagy is a conserved intracellular housekeeping process, modulation of autophagy flux in various model organisms have highlighted its importance for maintaining proteostasis. In postmitotic cells such as neurons, compromised autophagy is sufficient to cause accumulation of ubiquitinated aggregates, neuronal dysfunction, degeneration, and loss of motor coordination-all hallmarks of neurodegenerative diseases. Reciprocally, enhanced autophagy flux augments cellular and organismal health, in addition to extending life span. These genetic studies not-withstanding a plethora of small molecule modulators of autophagy flux have been reported that alleviate disease symptoms in models of neurodegenerative diseases. This review summarizes the potential of such molecules to be, perhaps, one of the first autophagy drugs for treating these currently incurable diseases.
Collapse
Affiliation(s)
- S N Suresh
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Anushka Chakravorty
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India
| | - Mridhula Giridharan
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India
| | - Lakshmi Garimella
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India; Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India.
| |
Collapse
|
124
|
Kinzler MN, Zielke S, Kardo S, Meyer N, Kögel D, van Wijk SJL, Fulda S. STF-62247 and pimozide induce autophagy and autophagic cell death in mouse embryonic fibroblasts. Sci Rep 2020; 10:687. [PMID: 31959760 PMCID: PMC6971264 DOI: 10.1038/s41598-019-56990-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 12/20/2019] [Indexed: 01/14/2023] Open
Abstract
Induction of autophagy can have beneficial effects in several human diseases, e.g. cancer and neurodegenerative diseases (ND). Here, we therefore evaluated the potential of two novel autophagy-inducing compounds, i.e. STF-62247 and pimozide, to stimulate autophagy as well as autophagic cell death (ACD) using mouse embryonic fibroblasts (MEFs) as a cellular model. Importantly, both STF-62247 and pimozide triggered several hallmarks of autophagy in MEFs, i.e. enhanced levels of LC3B-II protein, its accumulation at distinct cytosolic sites and increase of the autophagic flux. Intriguingly, autophagy induction by STF-62247 and pimozide resulted in cell death that was significantly reduced in ATG5- or ATG7-deficient MEFs. Consistent with ACD induction, pharmacological inhibitors of apoptosis, necroptosis or ferroptosis failed to protect MEFs from STF-62247- or pimozide-triggered cell death. Interestingly, at subtoxic concentrations, pimozide stimulated fragmentation of the mitochondrial network, degradation of mitochondrial proteins (i.e. mitofusin-2 and cytochrome c oxidase IV (COXIV)) as well as a decrease of the mitochondrial mass, indicative of autophagic degradation of mitochondria by pimozide. In conclusion, this study provides novel insights into the induction of selective autophagy as well as ACD by STF-62247 and pimozide in MEFs.
Collapse
Affiliation(s)
- Maximilian N Kinzler
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstr. 3a, 60528, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| | - Svenja Zielke
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstr. 3a, 60528, Frankfurt, Germany
| | - Simon Kardo
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstr. 3a, 60528, Frankfurt, Germany
| | - Nina Meyer
- Experimental Neurosurgery, Goethe-University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Donat Kögel
- Experimental Neurosurgery, Goethe-University Hospital, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Sjoerd J L van Wijk
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstr. 3a, 60528, Frankfurt, Germany
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Komturstr. 3a, 60528, Frankfurt, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany.
- German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| |
Collapse
|
125
|
Herhaus L, Bhaskara RM, Lystad AH, Gestal‐Mato U, Covarrubias‐Pinto A, Bonn F, Simonsen A, Hummer G, Dikic I. TBK1-mediated phosphorylation of LC3C and GABARAP-L2 controls autophagosome shedding by ATG4 protease. EMBO Rep 2020; 21:e48317. [PMID: 31709703 PMCID: PMC6945063 DOI: 10.15252/embr.201948317] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 10/08/2019] [Accepted: 10/15/2019] [Indexed: 12/22/2022] Open
Abstract
Autophagy is a highly conserved catabolic process through which defective or otherwise harmful cellular components are targeted for degradation via the lysosomal route. Regulatory pathways, involving post-translational modifications such as phosphorylation, play a critical role in controlling this tightly orchestrated process. Here, we demonstrate that TBK1 regulates autophagy by phosphorylating autophagy modifiers LC3C and GABARAP-L2 on surface-exposed serine residues (LC3C S93 and S96; GABARAP-L2 S87 and S88). This phosphorylation event impedes their binding to the processing enzyme ATG4 by destabilizing the complex. Phosphorylated LC3C/GABARAP-L2 cannot be removed from liposomes by ATG4 and are thus protected from ATG4-mediated premature removal from nascent autophagosomes. This ensures a steady coat of lipidated LC3C/GABARAP-L2 throughout the early steps in autophagosome formation and aids in maintaining a unidirectional flow of the autophagosome to the lysosome. Taken together, we present a new regulatory mechanism of autophagy, which influences the conjugation and de-conjugation of LC3C and GABARAP-L2 to autophagosomes by TBK1-mediated phosphorylation.
Collapse
Affiliation(s)
- Lina Herhaus
- Institute of Biochemistry IISchool of MedicineGoethe UniversityFrankfurt am MainGermany
| | - Ramachandra M Bhaskara
- Department of Theoretical BiophysicsMax Planck Institute of BiophysicsFrankfurt am MainGermany
| | - Alf Håkon Lystad
- Department of Molecular MedicineFaculty of MedicineInstitute of Basic Medical Sciences and Centre for Cancer Cell ReprogrammingInstitute of Clinical MedicineUniversity of OsloOsloNorway
| | - Uxía Gestal‐Mato
- Institute of Biochemistry IISchool of MedicineGoethe UniversityFrankfurt am MainGermany
| | | | - Florian Bonn
- Institute of Biochemistry IISchool of MedicineGoethe UniversityFrankfurt am MainGermany
- Present address:
Immundiagnostik AGBensheimGermany
| | - Anne Simonsen
- Department of Molecular MedicineFaculty of MedicineInstitute of Basic Medical Sciences and Centre for Cancer Cell ReprogrammingInstitute of Clinical MedicineUniversity of OsloOsloNorway
| | - Gerhard Hummer
- Department of Theoretical BiophysicsMax Planck Institute of BiophysicsFrankfurt am MainGermany
- Institute for BiophysicsGoethe UniversityFrankfurt am MainGermany
| | - Ivan Dikic
- Institute of Biochemistry IISchool of MedicineGoethe UniversityFrankfurt am MainGermany
- Buchmann Institute for Molecular Life SciencesRiedberg Campus, Goethe University FrankfurtFrankfurt am MainGermany
| |
Collapse
|
126
|
Novel compounds for the modulation of mTOR and autophagy to treat neurodegenerative diseases. Cell Signal 2020; 65:109442. [DOI: 10.1016/j.cellsig.2019.109442] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022]
|
127
|
Tang BL. Syntaxin 16's Newly Deciphered Roles in Autophagy. Cells 2019; 8:cells8121655. [PMID: 31861136 PMCID: PMC6953085 DOI: 10.3390/cells8121655] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 12/16/2022] Open
Abstract
Syntaxin 16, a Qa-SNARE (soluble N-ethylmaleimide-sensitive factor activating protein receptor), is involved in a number of membrane-trafficking activities, particularly transport processes at the trans-Golgi network (TGN). Recent works have now implicated syntaxin 16 in the autophagy process. In fact, syntaxin 16 appears to have dual roles, firstly in facilitating the transport of ATG9a-containing vesicles to growing autophagosomes, and secondly in autolysosome formation. The former involves a putative SNARE complex between syntaxin 16, VAMP7 and SNAP-47. The latter occurs via syntaxin 16’s recruitment by Atg8/LC3/GABARAP family proteins to autophagosomes and endo-lysosomes, where syntaxin 16 may act in a manner that bears functional redundancy with the canonical autophagosome Qa-SNARE syntaxin 17. Here, I discuss these recent findings and speculate on the mechanistic aspects of syntaxin 16’s newly found role in autophagy.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore; ; Tel.: +65-6516-1040
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 119077, Singapore
| |
Collapse
|
128
|
Stead ER, Castillo-Quan JI, Miguel VEM, Lujan C, Ketteler R, Kinghorn KJ, Bjedov I. Agephagy - Adapting Autophagy for Health During Aging. Front Cell Dev Biol 2019; 7:308. [PMID: 31850344 PMCID: PMC6892982 DOI: 10.3389/fcell.2019.00308] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a major cellular recycling process that delivers cellular material and entire organelles to lysosomes for degradation, in a selective or non-selective manner. This process is essential for the maintenance of cellular energy levels, components, and metabolites, as well as the elimination of cellular molecular damage, thereby playing an important role in numerous cellular activities. An important function of autophagy is to enable survival under starvation conditions and other stresses. The majority of factors implicated in aging are modifiable through the process of autophagy, including the accumulation of oxidative damage and loss of proteostasis, genomic instability and epigenetic alteration. These primary causes of damage could lead to mitochondrial dysfunction, deregulation of nutrient sensing pathways and cellular senescence, finally causing a variety of aging phenotypes. Remarkably, advances in the biology of aging have revealed that aging is a malleable process: a mild decrease in signaling through nutrient-sensing pathways can improve health and extend lifespan in all model organisms tested. Consequently, autophagy is implicated in both aging and age-related disease. Enhancement of the autophagy process is a common characteristic of all principal, evolutionary conserved anti-aging interventions, including dietary restriction, as well as inhibition of target of rapamycin (TOR) and insulin/IGF-1 signaling (IIS). As an emerging and critical process in aging, this review will highlight how autophagy can be modulated for health improvement.
Collapse
Affiliation(s)
- Eleanor R Stead
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Jorge I Castillo-Quan
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, United States.,Department of Genetics, Harvard Medical School, Boston, MA, United States
| | | | - Celia Lujan
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Kerri J Kinghorn
- Institute of Healthy Ageing, University College London, London, United Kingdom.,Department of Genetics, Evolution and Environment, University College London, London, United Kingdom.,Institute of Neurology, University College London, London, United Kingdom
| | - Ivana Bjedov
- UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
129
|
Phadwal K, Feng D, Zhu D, MacRae VE. Autophagy as a novel therapeutic target in vascular calcification. Pharmacol Ther 2019; 206:107430. [PMID: 31647975 DOI: 10.1016/j.pharmthera.2019.107430] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2019] [Indexed: 02/07/2023]
Abstract
The autophagy pathway is a key regulator of cellular metabolism and homeostasis, and plays a critical role in maintaining normal vascular cell function. It is well recognised that autophagy can regulate endothelial cell homeostasis, vascular smooth muscle cell (VSMC) phenotype transition, and calcium (Ca2+) homeostasis in VSMCs. Emerging evidence has demonstrated that autophagy directly protects against vascular calcification (VC). Crosstalk between endosomes, dysfunctional mitochondria, autophagic vesicles and Ca2+ and phosphate (Pi) enriched matrix vesicles (MVs) may underpin the pathogenesis of VC. In this review, we summarize the current experimental evidence in understanding how autophagy maintains normal vascular cell function and its protective role against vascular calcification. We also discuss the underlying molecular and cellular mechanisms through which autophagy inhibits vascular calcification. Pharmacological modulation of autophagy may offer an exciting new strategy for the treatment of vascular calcification.
Collapse
Affiliation(s)
- Kanchan Phadwal
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Du Feng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511436, China.
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Diseases, The Second Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Vicky E MacRae
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| |
Collapse
|
130
|
Chen RH, Chen YH, Huang TY. Ubiquitin-mediated regulation of autophagy. J Biomed Sci 2019; 26:80. [PMID: 31630678 PMCID: PMC6802350 DOI: 10.1186/s12929-019-0569-y] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/01/2019] [Indexed: 02/01/2023] Open
Abstract
Autophagy is a major degradation pathway that utilizes lysosome hydrolases to degrade cellular constituents and is often induced under cellular stress conditions to restore cell homeostasis. Another prime degradation pathway in the cells is ubiquitin-proteasome system (UPS), in which proteins tagged by certain types of polyubiquitin chains are selectively recognized and removed by proteasome. Although the two degradation pathways are operated independently with different sets of players, recent studies have revealed reciprocal cross talks between UPS and autophagy at multiple layers. In this review, we summarize the roles of protein ubiquitination and deubiquitination in controlling the initiation, execution, and termination of bulk autophagy as well as the role of ubiquitination in signaling certain types of selective autophagy. We also highlight how dysregulation of ubiquitin-mediated autophagy pathways is associated with a number of human diseases and the potential of targeting these pathways for disease intervention.
Collapse
Affiliation(s)
- Ruey-Hwa Chen
- Academia Sinica, Institute of Biological Chemistry, Taipei, 115, Taiwan. .,Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, 100, Taiwan.
| | - Yu-Hsuan Chen
- Academia Sinica, Institute of Biological Chemistry, Taipei, 115, Taiwan.,Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, 100, Taiwan
| | - Tzu-Yu Huang
- Academia Sinica, Institute of Biological Chemistry, Taipei, 115, Taiwan.,Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, 100, Taiwan
| |
Collapse
|
131
|
Corona AK, Saulsbery HM, Corona Velazquez AF, Jackson WT. Enteroviruses Remodel Autophagic Trafficking through Regulation of Host SNARE Proteins to Promote Virus Replication and Cell Exit. Cell Rep 2019; 22:3304-3314. [PMID: 29562185 DOI: 10.1016/j.celrep.2018.03.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/15/2017] [Accepted: 02/28/2018] [Indexed: 11/30/2022] Open
Abstract
Enterovirus D68 (EV-D68) is a medically important respiratory plus-strand RNA virus of children that has been linked to acute flaccid myelitis. We have determined that EV-D68 induces autophagic signaling and membrane formation. Autophagy, a homeostatic degradative process that breaks down protein aggregates and damaged organelles, promotes replication of multiple plus-strand viruses. Induction of autophagic signals promotes EV-D68 replication, but the virus inhibits the downstream degradative steps of autophagy in multiple ways. EV-D68 proteases cleave a major autophagic cargo adaptor and the autophagic SNARE SNAP29, which reportedly regulates fusion between autophagosome to amphisome/autolysosome. Although the virus inhibits autophagic degradation, SNAP29 promotes virus replication early in infection. An orphan SNARE, SNAP47, is shown to have a previously unknown role in autophagy, and SNAP47 promotes the replication of EV-D68. Our study illuminates a mechanism for subversion of autophagic flux and redirection of the autophagic membranes to benefit EV-D68 replication.
Collapse
Affiliation(s)
- Abigail K Corona
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Avenue, Baltimore, MD 21201, USA
| | - Holly M Saulsbery
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Avenue, Baltimore, MD 21201, USA
| | - Angel F Corona Velazquez
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Avenue, Baltimore, MD 21201, USA
| | - William T Jackson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Avenue, Baltimore, MD 21201, USA.
| |
Collapse
|
132
|
Nitrogen deprivation elicits dimorphism, capsule biosynthesis and autophagy in Papiliotrema laurentii strain RY1. Micron 2019; 124:102708. [DOI: 10.1016/j.micron.2019.102708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 12/29/2022]
|
133
|
Xiao B, Hong L, Cai X, Mei S, Zhang P, Shao L. The true colors of autophagy in doxorubicin-induced cardiotoxicity. Oncol Lett 2019; 18:2165-2172. [PMID: 31452719 PMCID: PMC6676529 DOI: 10.3892/ol.2019.10576] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 06/13/2019] [Indexed: 12/22/2022] Open
Abstract
Patients with cancer receiving doxorubicin-based chemotherapy often have to stop taking the drug due to its cardiotoxicity and therefore lose out on the beneficial effects of its potent antitumor activity. Doxorubicin has been demonstrated to damage cardiomyocytes via various mechanisms, including accumulation of reactive oxygen species (ROS), DNA damage and autophagy dysfunction. The present review focuses on autophagy, describing the general process of autophagy and the controversy surrounding its role in doxorubicin-induced cardiotoxicity. In addition, the associations between autophagy and apoptosis, ROS, DNA damage and inflammatory processes are discussed. In the future, it will be useful to further elucidate the process of autophagy and reveal its association with various pathological processes to develop effective strategies of preventing doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Bin Xiao
- Department of Cardiology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China.,Medical Graduate School of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lang Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China.,Jiang Xi Provincial Institute of Cardiovascular Diseases, Nanchang, Jiangxi 330006, P.R. China
| | - Xinyong Cai
- Department of Cardiology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China.,Jiang Xi Provincial Institute of Cardiovascular Diseases, Nanchang, Jiangxi 330006, P.R. China
| | - Songbo Mei
- Department of Cardiology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China.,Medical Graduate School of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ping Zhang
- Department of Neurology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Liang Shao
- Department of Cardiology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China.,Jiang Xi Provincial Institute of Cardiovascular Diseases, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
134
|
Peng W, Minakaki G, Nguyen M, Krainc D. Preserving Lysosomal Function in the Aging Brain: Insights from Neurodegeneration. Neurotherapeutics 2019; 16:611-634. [PMID: 31183763 PMCID: PMC6694346 DOI: 10.1007/s13311-019-00742-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lysosomes are acidic, membrane-bound organelles that serve as the primary catabolic compartment of the cell. They are crucial to a variety of cellular processes from nutrient storage to autophagy. Given the diversity of lysosomal functions, it is unsurprising that lysosomes are also emerging as important players in aging. Lysosomal dysfunction is implicated in several aging-related neurodegenerative diseases including Alzheimer's, Parkinson's, amyotrophic lateral sclerosis/frontotemporal dementia, and Huntington's. Although the precise role of lysosomes in the aging brain is not well-elucidated, some insight into their function has been gained from our understanding of the pathophysiology of age-dependent neurodegenerative diseases. Therapeutic strategies targeting lysosomes and autophagic machinery have already been tested in several of these diseases with promising results, suggesting that improving lysosomal function could be similarly beneficial in preserving function in the aging brain.
Collapse
Affiliation(s)
- Wesley Peng
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA
| | - Georgia Minakaki
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA
| | - Maria Nguyen
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA
| | - Dimitri Krainc
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA.
| |
Collapse
|
135
|
Taguchi K, Elias BC, Qian S, Brooks CR. Quantifying autophagic flux in kidney tissue using structured illumination microscopy. Methods Cell Biol 2019; 153:231-253. [PMID: 31395381 PMCID: PMC10625164 DOI: 10.1016/bs.mcb.2019.05.004] [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] [Indexed: 11/06/2023]
Abstract
Kidney disease is estimated to affect 15% of the world's population. Autophagy is a key homeostatic pathway in eukaryotic cells, which has been linked to numerous pathological states. In the kidney, autophagy has been shown to modulate both acute and chronic injuries. Despite the importance of autophagy in kidney disease, few techniques to precisely monitor autophagic flux in kidney tissue are available. Here we describe an improved technique to quantify autophagic flux using an RFP-GFP-LC3 reporter mouse and super-resolution microscopy. Using structured illumination microscopy, we can resolve individual autophagosomes within kidney tubular cells. We describe the preparation of slides, staining, imaging and data processing. 3D surface rendering is utilized to categorize and quantify autophagosomes by number, size, fluorescence and autophagic flux in response to ischemia.
Collapse
Affiliation(s)
- Kensei Taguchi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Bertha C Elias
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Subo Qian
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Craig R Brooks
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States.
| |
Collapse
|
136
|
Perez Vidakovics MLA, Ure AE, Arrías PN, Romanowski V, Gómez RM. Junín virus induces autophagy in human A549 cells. PLoS One 2019; 14:e0218730. [PMID: 31216340 PMCID: PMC6583977 DOI: 10.1371/journal.pone.0218730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/07/2019] [Indexed: 12/21/2022] Open
Abstract
Autophagy, a highly regulated degradative process that promotes cellular homeostasis, is increasingly recognised as a fundamental component of the cellular response against viral infection. In this study, we investigated the role of autophagy during Junín virus (JUNV) multiplication using human A549 cells. We found that JUNV infection induces an increment of the LC3-II/LC3-I ratio, an accumulation of punctate pattern in RFP-LC3-transfected cells and the colocalisation of viral nucleoprotein and LC3 protein, suggesting autophagosome formation. JUNV infection also induced the degradation of the autophagy receptor p62, suggesting that complete autophagic flux was triggered. In addition, we showed that inhibition of autophagy with bafilomycin A1 or 3-methyladenine significantly reduces viral multiplication. Moreover, viral yield was increased when autophagy was induced using rapamycin. Furthermore, JUNV infection induced the colocalisation of p62, ATG16, RAB5, RAB7A and LAMP1 with the autophagosomal LC3 protein. That suggests that phagosomes undergo the maturation process during viral infection. Finally, we demonstrated that siRNA experiments targeting essential autophagy genes (ATG5, ATG7 and Beclin 1) reduce viral protein synthesis and viral yield. Overall, our results indicate that JUNV activates host autophagy machinery enhancing its multiplication.
Collapse
Affiliation(s)
| | - Agustín E. Ure
- Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Paula N. Arrías
- Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Víctor Romanowski
- Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Ricardo M. Gómez
- Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
137
|
Tan WD, Shen HM, Wong WF. Dysregulated autophagy in COPD: A pathogenic process to be deciphered. Pharmacol Res 2019; 144:1-7. [DOI: 10.1016/j.phrs.2019.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 10/27/2022]
|
138
|
Chen C, He J, Gao W, Wei Y, Liu G. Identification and Characterization of an Autophagy-Related Gene Acatg12 in Acremonium chrysogenum. Curr Microbiol 2019; 76:545-551. [PMID: 30899986 DOI: 10.1007/s00284-019-01650-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/08/2019] [Indexed: 01/28/2023]
Abstract
Autophagy is a highly conserved mechanism to overcome various stresses and recycle cytoplasmic components and organelles. Ubiquitin-like (UBL) protein Atg12 is a key protein involved in autophagosome formation through stimulation of Atg8 conjugation to phosphatidylethanolamine. Here, we describe the identification of the autophagy-related gene Acatg12, encoding an Atg12 homologous protein in the cephalosporin C producing fungus Acremonium chrysogenum. Disruption of Acatg12 impaired the delivery and degradation of eGFP-Atg8, indicating that the autophagic process was blocked. Meanwhile, conidiation was dramatically reduced in the Acatg12 disruption mutant (∆Acatg12). In contrast, cephalosporin C production was increased twofold in ∆Acatg12, but fungal growth was reduced after 6 days fermentation. Consistent with these results, the transcriptional level of the cephalosporin biosynthetic genes was increased in ∆Acatg12. The results extend our understanding of autophagy in filamentous fungi.
Collapse
Affiliation(s)
- Chang Chen
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100,101, China.,The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, 100864, China
| | - Jia He
- Department of Plant Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Wenyan Gao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100,101, China
| | - Yanmin Wei
- Department of Plant Science and Technology, Beijing University of Agriculture, Beijing, 102206, China.
| | - Gang Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100,101, China. .,The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, 100864, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
139
|
Abstract
Autophagy is an evolutionarily conserved self-digestion process which is essential to keep basal homeostasis in a cell. During this process, degradation and recycling of many cytoplasmic components including the long-lived, unnecessary or aggregated proteins and damaged organelles is achieved through lysosomal machinery. Autophagy has a critical role for lower eukaryotic organisms such as yeast to survive and adapt to nutrient starvation conditions. In addition to this primary function, autophagy appears as a crucial mechanism for cell differentiation and development enabling the cells to modify their content and morphology in response to environmental and hormonal cues. A recent study by Shang et al.1 shed more light on the molecular mechanisms of how autophagy regulates spermiogenesis.
Collapse
Affiliation(s)
- Nihan Ozturk
- Clinic of Urology, Pediatric Urology and Andrology, Section Molecular Andrology, Justus-Liebig-University Giessen, Schubertstrasse 81, 35392 Giessen, Germany
| | - Klaus Steger
- Clinic of Urology, Pediatric Urology and Andrology, Section Molecular Andrology, Justus-Liebig-University Giessen, Schubertstrasse 81, 35392 Giessen, Germany
| | - Undraga Schagdarsurengin
- Clinic of Urology, Pediatric Urology and Andrology, Section Molecular Andrology, Justus-Liebig-University Giessen, Schubertstrasse 81, 35392 Giessen, Germany
| |
Collapse
|
140
|
Harada K, Kotani T, Kirisako H, Sakoh-Nakatogawa M, Oikawa Y, Kimura Y, Hirano H, Yamamoto H, Ohsumi Y, Nakatogawa H. Two distinct mechanisms target the autophagy-related E3 complex to the pre-autophagosomal structure. eLife 2019; 8:43088. [PMID: 30810528 PMCID: PMC6405243 DOI: 10.7554/elife.43088] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/26/2019] [Indexed: 11/13/2022] Open
Abstract
In autophagy, Atg proteins organize the pre-autophagosomal structure (PAS) to initiate autophagosome formation. Previous studies in yeast revealed that the autophagy-related E3 complex Atg12-Atg5-Atg16 is recruited to the PAS via Atg16 interaction with Atg21, which binds phosphatidylinositol 3-phosphate (PI3P) produced at the PAS, to stimulate conjugation of the ubiquitin-like protein Atg8 to phosphatidylethanolamine. Here, we discover a novel mechanism for the PAS targeting of Atg12-Atg5-Atg16, which is mediated by the interaction of Atg12 with the Atg1 kinase complex that serves as a scaffold for PAS organization. While autophagy is partially defective without one of these mechanisms, cells lacking both completely lose the PAS localization of Atg12-Atg5-Atg16 and show no autophagic activity. As with the PI3P-dependent mechanism, Atg12-Atg5-Atg16 recruited via the Atg12-dependent mechanism stimulates Atg8 lipidation, but also has the specific function of facilitating PAS scaffold assembly. Thus, this study significantly advances our understanding of the nucleation step in autophagosome formation.
Collapse
Affiliation(s)
- Kumi Harada
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Tetsuya Kotani
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Hiromi Kirisako
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | | | - Yu Oikawa
- Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | | | | | - Hitoshi Nakatogawa
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
141
|
Aghaei M, Motallebnezhad M, Ghorghanlu S, Jabbari A, Enayati A, Rajaei M, Pourabouk M, Moradi A, Alizadeh AM, Khori V. Targeting autophagy in cardiac ischemia/reperfusion injury: A novel therapeutic strategy. J Cell Physiol 2019; 234:16768-16778. [PMID: 30807647 DOI: 10.1002/jcp.28345] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/17/2019] [Accepted: 01/22/2019] [Indexed: 12/31/2022]
Abstract
Acute myocardial infarction (AMI) is one of the leading causes of morbidity worldwide. Myocardial reperfusion is known as an effective therapeutic choice against AMI. However, reperfusion of blood flow induces ischemia/reperfusion (I/R) injury through different complex processes including ion accumulation, disruption of mitochondrial membrane potential, the formation of reactive oxygen species, and so forth. One of the processes that gets activated in response to I/R injury is autophagy. Indeed, autophagy acts as a "double-edged sword" in the pathology of myocardial I/R injury and there is a controversy about autophagy being beneficial or detrimental. On the basis of the autophagy effect and regulation on myocardial I/R injury, many studies targeted it as a therapeutic strategy. In this review, we discuss the role of autophagy in I/R injury and its targeting as a therapeutic strategy.
Collapse
Affiliation(s)
- Mehrdad Aghaei
- Rheumatology Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Morteza Motallebnezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Sajjad Ghorghanlu
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Jabbari
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ayesheh Enayati
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Pharmacognosy, Faculty of Pharmacy and Medicinal Plants Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Rajaei
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mona Pourabouk
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Alireza Moradi
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
142
|
Abstract
Classically, canonical autophagy has been considered a survival mechanism initiated in response to nutrient insufficiency. We now understand that autophagy functions in multiple scenarios where it is necessary to maintain homeostasis. Recent evidence has established that a variety of non-canonical functions for autophagy proteins are mechanistically and functionally distinct from autophagy. LC3-associated phagocytosis (LAP) is one such novel function for autophagy proteins and is a contributor to immune regulation and inflammatory responses across various cell and tissue types. Characterized by the conjugation of LC3 family proteins to phagosome membranes, LAP uses a portion of the canonical autophagy machinery, following ligation of surface receptors that recognize a variety of cargos including pathogens, dying cells, soluble ligands and protein aggregates. However, instead of affecting canonical autophagy, manipulation of the LAP pathway in vivo alters immune activation and inflammatory responses. In this Cell Science at a Glance article and the accompanying poster, we detail the divergence of this distinctive mechanism from that of canonical autophagy by comparing and contrasting shared and unique components of each pathway.
Collapse
Affiliation(s)
- Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Cancer Biology Program, St. Jude Pediatric Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Cancer Biology Program, St. Jude Pediatric Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
143
|
Liu R, Cheng Q, Song X, Wang H, Wang X, Wang L, Zhu B, Song L. A vital ubiquitin-conjugating enzyme CgUbe2g1 participated in regulation of immune response of Pacific oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 91:132-142. [PMID: 30389518 DOI: 10.1016/j.dci.2018.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 06/08/2023]
Abstract
As an important post-translational protein modification, ubiquitination has been demonstrated to play a vital role in immune response of vertebrates. Ubiquitin (Ub)-conjugating enzyme E2 is the "heart" of ubiquitination, which is responsible for Ub cellular signaling and substrate modification. In the present study, an Ub-conjugating enzyme E2 (designed as CgUbe2g1) was identified from oyster Crassostrea gigas, and its regulation in the immune response against lipopolysaccharide (LPS) stimulation was investigated. CgUbe2g1 encoded a polypeptide of 168 amino acids with the predicted molecular mass of 19.20 kDa and contained conserved catalytic 'Ubc' domains. It shared a higher similarity with the known UBC2G1 type E2s and was closely clustered with the type E2s identified from invertebrates in the phylogenetic assay. The mRNA transcripts of CgUbe2g1 were mainly distributed in hemocyte, mantle, hepatopancreas and male gonad of C. gigas. CgUbe2g1 protein was found to be colocalized with Ub around the nucleus of oyster hemocyte. The recombinant CgUbe2g1 protein (rCgUbe2g1) could activate the ubiquitination in vitro by binding both activated and un-activated Ub. The expressions of inflammation-related factors TNF-α and NF-κB in CgUbe2g1 transfected cells were both significantly up-regulated after LPS stimulation, which were 12.9-fold at 3 h (p < 0.01) and 2.3-fold at 6 h (p < 0.01) of that in negative control group, respectively. The phagocytic rate of hemocyte and the ROS level in hemocyte were both significantly decreased (p < 0.01), while the apoptosis rate was significantly increased (p < 0.01) after CgUbe2g1 mRNA was interfered. These results demonstrated that Ub-conjugating enzyme CgUbe2g1 was involved in the innate immune response of oyster against invading pathogen, which might play important roles in the activation of inflammatory response and regulation of cellular immune response.
Collapse
Affiliation(s)
- Rui Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Qi Cheng
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, China
| | - Xiaorui Song
- Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Hao Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Xiudan Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Beiwei Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, China
| | - Linsheng Song
- Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
144
|
Tong X, Gu J, Song R, Wang D, Sun Z, Sui C, Zhang C, Liu X, Bian J, Liu Z. Osteoprotegerin inhibit osteoclast differentiation and bone resorption by enhancing autophagy via AMPK/mTOR/p70S6K signaling pathway in vitro. J Cell Biochem 2019; 120:1630-1642. [PMID: 30256440 DOI: 10.1002/jcb.27468] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/18/2018] [Indexed: 01/24/2023]
Abstract
Osteoclasts are highly differentiated terminal cells formed by fusion of hematopoietic stem cells. Previously, osteoprotegerin (OPG) inhibit osteoclast differentiation and bone resorption by blocking receptor activator of nuclear factor-κB ligand (RANKL) binding to RANK indirect mechanism. Furthermore, autophagy plays an important role during osteoclast differentiation and function. However, whether autophagy is involved in OPG-inhibited osteoclast formation and bone resorption is not known. To elucidate the role of autophagy in OPG-inhibited osteoclast differentiation and bone resorption, we used primary osteoclast derived from mice bone marrow monocytes/macrophages (BMM) by induced M-CSF and RANKL. The results showed that autophagy-related proteins expression were upregulated; tartrate-resistant acid phosphatase-positive osteoclast number and bone resorption activity were decreased; LC3 puncta and autophagosomes number were increased and activated AMPK/mTOR/p70S6K signaling pathway. In addition, chloroquine (as the autophagy/lysosome inhibitor, CQ) or rapamycin (as the autophagy/lysosome inhibitor, Rap) attenuated osteoclast differentiation and bone resorption activity by OPG treatment via AMPK/mTOR/p70S6K signaling pathway. Our data demonstrated that autophagy plays a critical role in OPG inhibiting osteoclast differentiation and bone resorption via AMPK/mTOR/p70S6K signaling pathway in vitro.
Collapse
Affiliation(s)
- Xishuai Tong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Ruilong Song
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Dong Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Ziqiang Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Chen Sui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Chuang Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Xuezhong Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
145
|
Krieg S, Lüscher B, Vervoorts J, Dohmen M. Studying the Role of AMPK in Autophagy. Methods Mol Biol 2019; 1732:373-391. [PMID: 29480488 DOI: 10.1007/978-1-4939-7598-3_24] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
AMPK is an energy-sensing kinase and is required for the induction and progression of the autophagy process. In this chapter, we describe experimental approaches to study the steady state and flux of autophagy in response to AMPK activation. For this purpose, we provide detailed protocols for the measurement of general as well as AMPK-specific autophagy markers by immunoblot and immunofluorescence analysis.
Collapse
Affiliation(s)
- Sarah Krieg
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Jörg Vervoorts
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Marc Dohmen
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
146
|
Li W, Zhang L. Regulation of ATG and Autophagy Initiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1206:41-65. [PMID: 31776979 DOI: 10.1007/978-981-15-0602-4_2] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
ATG is involved in multiple processes of autophagosome formation, including the initial phase of autophagy. The mammalian autophagy complex-ULK1 complex is composed of ULK1, FIP200, ATG13 and ATG101, and the yeast autophagy initiation complex-ATG1 complex is composed of ATG1, ATG13, ATG17, ATG29 and ATG31. After this complex is activated, it binds and phosphorylates ATG9 on the vesicles. Then PI3KC3-C1 (yeast: ATG34: ATG15: ATG6: ATG14 or mammal animal: ATG34: ATG15: BECN1: ATG14L) is recruited to the PAS. Further, ATG12-ATG5-ATG16 complex is localized on PAS (Yeast) or localized on the outer surface of the membrane (mammal) and makes binding of ATG8 (LC3) with PE to form ATG8-PE complex, promoting autophagic membrane elongation, closure and formation autophagosome and autophagosome lysosome.
Collapse
Affiliation(s)
- Wen Li
- Department of Immunology, School of Basic Medical Science, Shandong University, 44# Wenhua Xi Road, Jinan, 250012, China
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Science, Shandong University, 44# Wenhua Xi Road, Jinan, 250012, China.
| |
Collapse
|
147
|
Autophagy: An Essential Degradation Program for Cellular Homeostasis and Life. Cells 2018; 7:cells7120278. [PMID: 30572663 PMCID: PMC6315530 DOI: 10.3390/cells7120278] [Citation(s) in RCA: 228] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 12/18/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a lysosome-dependent cellular degradation program that responds to a variety of environmental and cellular stresses. It is an evolutionarily well-conserved and essential pathway to maintain cellular homeostasis, therefore, dysfunction of autophagy is closely associated with a wide spectrum of human pathophysiological conditions including cancers and neurodegenerative diseases. The discovery and characterization of the kingdom of autophagy proteins have uncovered the molecular basis of the autophagy process. In addition, recent advances on the various post-translational modifications of autophagy proteins have shed light on the multiple layers of autophagy regulatory mechanisms, and provide novel therapeutic targets for the treatment of the diseases.
Collapse
|
148
|
Cao Y, Luo Y, Zou J, Ouyang J, Cai Z, Zeng X, Ling H, Zeng T. Autophagy and its role in gastric cancer. Clin Chim Acta 2018; 489:10-20. [PMID: 30472237 DOI: 10.1016/j.cca.2018.11.028] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/17/2018] [Accepted: 11/20/2018] [Indexed: 02/08/2023]
Abstract
Autophagy, which is tightly regulated by a series of autophagy-related genes (ATGs), is a vital intracellular homeostatic process through which defective proteins and organelles are degraded and recycled under starvation, hypoxia or other specific cellular stress conditions. For both normal cells and tumour cells, autophagy not only sustains cell survival but can also promote cell death. Autophagy-related signalling pathways include mTOR-dependent pathways, such as the AMPK/mTOR and PI3K/Akt/mTOR pathways, and non-mTOR dependent pathways, such as the P53 pathway. Additionally, autophagy plays a dual role in gastric carcinoma (GC), including a tumour-suppressor role and a tumour-promoter role. Long-term Helicobacter pylori infection can impair autophagy, which may eventually promote tumourigenesis of the gastric mucosa. Moreover, Beclin1, LC3 and P62/SQSTM1 are regarded as autophagy-related markers with GC prognostic value. Autophagy inhibitors and autophagy inducers show promise for GC treatment. This review describes research progress regarding autophagy and its significant role in gastric cancer.
Collapse
Affiliation(s)
- Yijing Cao
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study [Hunan Provincial Education Department document (Approval number: 2014-405)], Hengyang, Hunan 421001, PR China
| | - Yichen Luo
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study [Hunan Provincial Education Department document (Approval number: 2014-405)], Hengyang, Hunan 421001, PR China
| | - Juan Zou
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study [Hunan Provincial Education Department document (Approval number: 2014-405)], Hengyang, Hunan 421001, PR China
| | - Jun Ouyang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China
| | - Zhihong Cai
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study [Hunan Provincial Education Department document (Approval number: 2014-405)], Hengyang, Hunan 421001, PR China
| | - Xi Zeng
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study [Hunan Provincial Education Department document (Approval number: 2014-405)], Hengyang, Hunan 421001, PR China
| | - Hui Ling
- Key Laboratory of Tumor Cellular & Molecular Pathology (University of South China), College of Hunan Province, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, PR China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study [Hunan Provincial Education Department document (Approval number: 2014-405)], Hengyang, Hunan 421001, PR China.
| | - Tiebing Zeng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study [Hunan Provincial Education Department document (Approval number: 2014-405)], Hengyang, Hunan 421001, PR China; Institute of Pathogenic Biology, Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
149
|
Abstract
Mucociliary clearance is critically important in protecting the airways from infection and from the harmful effects of smoke and various inspired substances known to induce oxidative stress and persistent inflammation. An essential feature of the clearance mechanism involves regulation of the periciliary liquid layer on the surface of the airway epithelium, which is necessary for normal ciliary beating and maintenance of mucus hydration. The underlying ion transport processes associated with airway surface hydration include epithelial Na+ channel-dependent Na+ absorption occurring in parallel with CFTR and Ca2+-activated Cl- channel-dependent anion secretion, which are coordinately regulated to control the depth of the periciliary liquid layer. Oxidative stress is known to cause both acute and chronic effects on airway ion transport function, and an increasing number of studies in the past few years have identified an important role for autophagy as part of the physiological response to the damaging effects of oxidation. In this review, recent studies addressing the influence of oxidative stress and autophagy on airway ion transport pathways, along with results showing the potential of autophagy modulators in restoring the function of ion channels involved in transepithelial electrolyte transport necessary for effective mucociliary clearance, are presented.
Collapse
Affiliation(s)
- Scott M O'Grady
- Departments of Animal Science, Integrative Biology and Physiology, University of Minnesota , St. Paul, Minnesota
| |
Collapse
|
150
|
Mehrtash AB, Hochstrasser M. Ubiquitin-dependent protein degradation at the endoplasmic reticulum and nuclear envelope. Semin Cell Dev Biol 2018; 93:111-124. [PMID: 30278225 DOI: 10.1016/j.semcdb.2018.09.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 01/01/2023]
Abstract
Numerous nascent proteins undergo folding and maturation within the luminal and membrane compartments of the endoplasmic reticulum (ER). Despite the presence of various factors in the ER that promote protein folding, many proteins fail to properly fold and assemble and are subsequently degraded. Regulatory proteins in the ER also undergo degradation in a way that is responsive to stimuli or the changing needs of the cell. As in most cellular compartments, the ubiquitin-proteasome system (UPS) is responsible for the majority of the degradation at the ER-in a process termed ER-associated degradation (ERAD). Autophagic processes utilizing ubiquitin-like protein-conjugating systems also play roles in protein degradation at the ER. The ER is continuous with the nuclear envelope (NE), which consists of the outer nuclear membrane (ONM) and inner nuclear membrane (INM). While ERAD is known also to occur at the NE, only some of the ERAD ubiquitin-ligation pathways function at the INM. Protein degradation machineries in the ER/NE target a wide variety of substrates in multiple cellular compartments, including the cytoplasm, nucleoplasm, ER lumen, ER membrane, and the NE. Here, we review the protein degradation machineries of the ER and NE and the underlying mechanisms dictating recognition and processing of substrates by these machineries.
Collapse
Affiliation(s)
- Adrian B Mehrtash
- Department of Molecular, Cellular, & Developmental Biology, Yale University, New Haven, 06520, CT, USA.
| | - Mark Hochstrasser
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT, 06520, USA; Department of Molecular, Cellular, & Developmental Biology, Yale University, New Haven, 06520, CT, USA.
| |
Collapse
|