1
|
Iibushi J, Nozawa T, Toh H, Nakagawa I. ATG9B regulates bacterial internalization via actin rearrangement. iScience 2024; 27:109623. [PMID: 38706859 PMCID: PMC11066431 DOI: 10.1016/j.isci.2024.109623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/16/2024] [Accepted: 03/26/2024] [Indexed: 05/07/2024] Open
Abstract
Invasive bacterial pathogens are internalized by host cells through endocytosis, which is regulated by a cascade of actin rearrangement signals triggered by host cell receptors or bacterial proteins delivered into host cells. However, the molecular mechanisms that mediate actin rearrangement to promote bacterial invasion are not fully understood. Here, we show that the autophagy-related (ATG) protein ATG9B regulates the internalization of various bacteria by controlling actin rearrangement. ATG knockout screening and knockdown experiments in HeLa cells identified ATG9B as a critical factor for bacterial internalization. In particular, cells with ATG9B knockdown exhibited an accumulation of actin filaments and phosphorylated LIM kinase and cofilin, suggesting that ATG9B is involved in actin depolymerization. Furthermore, the kinase activity of Unc-51-like autophagy-activating kinase 1 was found to regulate ATG9B localization and actin remodeling. These findings revealed a newly discovered function of ATG proteins in bacterial infection rather than autophagy-mediated immunity.
Collapse
Affiliation(s)
- Junpei Iibushi
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| | - Takashi Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| | - Hirotaka Toh
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| |
Collapse
|
2
|
Ke L, Lin X, Luo Y, Tao S, Yan C, He Y, Wu Y, Liu N, Qin Y. Autophagy core protein BECN1 is vital for spermatogenesis and male fertility in mice†. Biol Reprod 2024; 110:599-614. [PMID: 37975917 DOI: 10.1093/biolre/ioad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/17/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
Mammalian spermatogenesis is a highly complex multi-step biological process, and autophagy has been demonstrated to be involved in the process of spermatogenesis. Beclin-1/BECN1, a core autophagy factor, plays a critical role in many biological processes and diseases. However, its function in spermatogenesis remains largely unclear. In the present study, germ cell-specific Beclin 1 (Becn1) knockout mice were generated and were conducted to determine the role of Becn1 in spermatogenesis and fertility of mice. Results indicate that Becn1 deficiency leads to reduced sperm motility and quantity, partial failure of spermiation, actin network disruption, excessive residual cytoplasm, acrosome malformation, and aberrant mitochondrial accumulation of sperm, ultimately resulting in reduced fertility in male mice. Furthermore, inhibition of autophagy was observed in the testes of germ cell-specific Becn1 knockout mice, which may contribute to impaired spermiogenesis and reduced fertility. Collectively, our results reveal that Becn1 is essential for fertility and spermiogenesis in mice.
Collapse
Affiliation(s)
- Lu Ke
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xinyi Lin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yuchuan Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Siming Tao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chang Yan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yifeilong He
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yingjie Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ning Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Yinghe Qin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
3
|
Kundu S, Jaiswal M, Babu Mullapudi V, Guo J, Kamat M, Basso KB, Guo Z. Investigation of Glycosylphosphatidylinositol (GPI)-Plasma Membrane Interaction in Live Cells and the Influence of GPI Glycan Structure on the Interaction. Chemistry 2024; 30:e202303047. [PMID: 37966101 PMCID: PMC10922586 DOI: 10.1002/chem.202303047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/05/2023] [Accepted: 11/15/2023] [Indexed: 11/16/2023]
Abstract
Glycosylphosphatidylinositols (GPIs) need to interact with other components in the cell membrane to transduce transmembrane signals. A bifunctional GPI probe was employed for photoaffinity-based proximity labelling and identification of GPI-interacting proteins in the cell membrane. This probe contained the entire core structure of GPIs and was functionalized with photoreactive diazirine and clickable alkyne to facilitate its crosslinking with proteins and attachment of an affinity tag. It was disclosed that this probe was more selective than our previously reported probe containing only a part structure of the GPI core for cell membrane incorporation and an improved probe for studying GPI-cell membrane interaction. Eighty-eight unique membrane proteins, many of which are related to GPIs/GPI-anchored proteins, were identified utilizing this probe. The proteomics dataset is a valuable resource for further analyses and data mining to find new GPI-related proteins and signalling pathways. A comparison of these results with those of our previous probe provided direct evidence for the profound impact of GPI glycan structure on its interaction with the cell membrane.
Collapse
Affiliation(s)
- Sayan Kundu
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Mohit Jaiswal
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | | | - Jiatong Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Manasi Kamat
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Kari B Basso
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
- UF Health Cancer Centre, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
4
|
Ye J, Zhang J, Zhu Y, Wang L, Jiang X, Liu B, He G. Targeting autophagy and beyond: Deconvoluting the complexity of Beclin-1 from biological function to cancer therapy. Acta Pharm Sin B 2023; 13:4688-4714. [PMID: 38045051 PMCID: PMC10692397 DOI: 10.1016/j.apsb.2023.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/05/2023] [Accepted: 08/02/2023] [Indexed: 12/05/2023] Open
Abstract
Beclin-1 is the firstly-identified mammalian protein of the autophagy machinery, which functions as a molecular scaffold for the assembly of PI3KC3 (class III phosphatidylinositol 3 kinase) complex, thus controlling autophagy induction and other cellular trafficking events. Notably, there is mounting evidence establishing the implications of Beclin-1 in diverse tumorigenesis processes, including tumor suppression and progression as well as resistance to cancer therapeutics and CSC (cancer stem-like cell) maintenance. More importantly, Beclin-1 has been confirmed as a potential target for the treatment of multiple cancers. In this review, we provide a comprehensive survey of the structure, functions, and regulations of Beclin-1, and we discuss recent advances in understanding the controversial roles of Beclin-1 in oncology. Moreover, we focus on summarizing the targeted Beclin-1-regulating strategies in cancer therapy, providing novel insights into a promising strategy for regulating Beclin-1 to improve cancer therapeutics in the future.
Collapse
Affiliation(s)
- Jing Ye
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanghui Zhu
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lian Wang
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease Related Molecular Network, Chengdu 610041, China
| | - Xian Jiang
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Liu
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gu He
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease Related Molecular Network, Chengdu 610041, China
| |
Collapse
|
5
|
Derkaczew M, Martyniuk P, Hofman R, Rutkowski K, Osowski A, Wojtkiewicz J. The Genetic Background of Abnormalities in Metabolic Pathways of Phosphoinositides and Their Linkage with the Myotubular Myopathies, Neurodegenerative Disorders, and Carcinogenesis. Biomolecules 2023; 13:1550. [PMID: 37892232 PMCID: PMC10605126 DOI: 10.3390/biom13101550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/16/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Myo-inositol belongs to one of the sugar alcohol groups known as cyclitols. Phosphatidylinositols are one of the derivatives of Myo-inositol, and constitute important mediators in many intracellular processes such as cell growth, cell differentiation, receptor recycling, cytoskeletal organization, and membrane fusion. They also have even more functions that are essential for cell survival. Mutations in genes encoding phosphatidylinositols and their derivatives can lead to many disorders. This review aims to perform an in-depth analysis of these connections. Many authors emphasize the significant influence of phosphatidylinositols and phosphatidylinositols' phosphates in the pathogenesis of myotubular myopathies, neurodegenerative disorders, carcinogenesis, and other less frequently observed diseases. In our review, we have focused on three of the most often mentioned groups of disorders. Inositols are the topic of many studies, and yet, there are no clear results of successful clinical trials. Analysis of the available literature gives promising results and shows that further research is still needed.
Collapse
Affiliation(s)
- Maria Derkaczew
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
- Students’ Scientific Club of Pathophysiologists, Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Piotr Martyniuk
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
- Students’ Scientific Club of Pathophysiologists, Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Robert Hofman
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
- Students’ Scientific Club of Pathophysiologists, Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Krzysztof Rutkowski
- Students’ Scientific Club of Pathophysiologists, Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
- The Nicolaus Copernicus Municipal Polyclinical Hospital in Olsztyn, 10-045 Olsztyn, Poland
| | - Adam Osowski
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| |
Collapse
|
6
|
Nicco C, Thomas M, Guillermet J, Havard M, Laurent-Tchenio F, Doridot L, Dautry F, Batteux F, Tchenio T. Mechanistic target of rapamycin (mTOR) regulates self-sustained quiescence, tumor indolence, and late clinical metastasis in a Beclin-1-dependent manner. Cell Cycle 2023; 22:542-564. [PMID: 36123968 PMCID: PMC9928463 DOI: 10.1080/15384101.2022.2123187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Self-sustained quiescence (SSQ) has been characterized as a stable but reversible non-proliferative cellular state that limits the cloning of cultured cancer cells. By developing refined clonogenic assays, we showed here that cancer cells in SSQ can be selected with anticancer agents and that culture at low cell density induced SSQ in pancreas and prostate adenocarcinoma cells. Pre-culture of cells in 3D or their pretreatment with pharmacological inhibitors of mechanistic target of rapamycin (mTOR) synergize with low cell density for induction of SSQ in a Beclin-1-dependent manner. Dissociated pancreatic adenocarcinoma (PAAD) cells rendered defective for SSQ by down-regulating Beclin-1 expression exhibit higher tumor growth rate when injected subcutaneously into mice. Conversely, dissociated PAAD cells in SSQ promote the formation of small indolent tumors that eventually transitioned to a rapid growth phase. Ex vivo clonogenic assays showed that up to 40% of clonogenic cancer cells enzymatically dissociated from resected fast-growing tumors could enter SSQ, suggesting that SSQ could significantly impact the proliferation of cancer cells that are naturally dispersed from tumors. Remarkably, the kinetics of clinical metastatic recurrence in 124 patients with pancreatic adenocarcinoma included in the TGCA-PAAD project could be predicted from Beclin-1 and Cyclin-A2 mRNA levels in their primary tumor, Cyclin A2 mRNA being a marker of both cell proliferation and mTOR complex 1 activity. Overall, our data show that SSQ is likely to promote the late development of clinical metastases and suggest that identifying new agents targeting cancer cells in SSQ could help improve patient survival.
Collapse
Affiliation(s)
- Carole Nicco
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| | - Marine Thomas
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| | - Julie Guillermet
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm U1037, CNRS U5071, Université Toulouse III, Toulouse, France
| | - Maryline Havard
- Laboratory of Biology and Applied Pharmacology (LBPA), CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Fanny Laurent-Tchenio
- Laboratory of Biology and Applied Pharmacology (LBPA), CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Ludivine Doridot
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| | - François Dautry
- Laboratory of Biology and Applied Pharmacology (LBPA), CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Frédéric Batteux
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| | - Thierry Tchenio
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| |
Collapse
|
7
|
Zhao L, Wang H, Liu S, Xi T, Wang L, Li Y, Chen L, Jianping R, Liang KX. Inhibition of autophagy reduces the rate of fluoride-induced LS8 apoptosis via regulating ATG5 and ATG7. J Biochem Mol Toxicol 2023; 37:e23280. [PMID: 36536498 DOI: 10.1002/jbt.23280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 10/27/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Excessive fluoride affects ameloblast differentiation and tooth development. The fate of fluorinated ameloblasts is determined by multiple signaling pathways in response to a range of stimuli. Both autophagy and apoptosis are involved in the regulation of dental fluorosis as well as in protein synthesis and enamel mineralization. Emerging evidence suggests that autophagy and apoptosis are interconnected and that their interaction greatly influences cell death. However, the effect of autophagy on apoptosis in fluoride-treated ameloblasts is unclear. Here, we employed an in vitro cellular model of fluorosis in mouse ameloblast-like LS8 cells and induced autophagy using sodium fluoride (NaF). Our findings suggest that NaF treatment induces autophagy in LS8 cells, and ATG5 and ATG7 are important molecules involved in this process. We also showed that NaF treatment reduced cell viability in Atg5/7 siRNA and autophagy inhibitor-treated LS8 cells. More importantly, NaF-induced apoptosis can be reversed by inhibiting early stage of autophagy. In conclusion, our study shows that autophagy is closely related to dental fluorosis, and inhibition of autophagy, especially ATG5/7, reduces fluoride-induced cell death and apoptosis.
Collapse
Affiliation(s)
- Lin Zhao
- Ningxia Key Laboratory of Cranio-maxillofacial Deformities, College of Stomatology, Ningxia Medical University, Yinchuan, China
| | - Han Wang
- Ningxia Key Laboratory of Cranio-maxillofacial Deformities, College of Stomatology, Ningxia Medical University, Yinchuan, China
| | - Sijia Liu
- Ningxia Key Laboratory of Cranio-maxillofacial Deformities, College of Stomatology, Ningxia Medical University, Yinchuan, China
| | - Tao Xi
- Ningxia Key Laboratory of Cranio-maxillofacial Deformities, College of Stomatology, Ningxia Medical University, Yinchuan, China
| | - Liyuan Wang
- Ningxia Key Laboratory of Cranio-maxillofacial Deformities, College of Stomatology, Ningxia Medical University, Yinchuan, China
- Stomatological Hospital of the General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yang Li
- Ningxia Key Laboratory of Cranio-maxillofacial Deformities, College of Stomatology, Ningxia Medical University, Yinchuan, China
| | - Lu Chen
- Department of Preventive Dentistry, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Stomatology Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Ruan Jianping
- Department of Preventive Dentistry, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Stomatology Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Kristina Xiao Liang
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
8
|
Hasan KMM, Haque MA. Autophagy and Its Lineage-Specific Roles in the Hematopoietic System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:8257217. [PMID: 37180758 PMCID: PMC10171987 DOI: 10.1155/2023/8257217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 05/16/2023]
Abstract
Autophagy is a dynamic process that regulates the selective and nonselective degradation of cytoplasmic components, such as damaged organelles and protein aggregates inside lysosomes to maintain tissue homeostasis. Different types of autophagy including macroautophagy, microautophagy, and chaperon-mediated autophagy (CMA) have been implicated in a variety of pathological conditions, such as cancer, aging, neurodegeneration, and developmental disorders. Furthermore, the molecular mechanism and biological functions of autophagy have been extensively studied in vertebrate hematopoiesis and human blood malignancies. In recent years, the hematopoietic lineage-specific roles of different autophagy-related (ATG) genes have gained more attention. The evolution of gene-editing technology and the easy access nature of hematopoietic stem cells (HSCs), hematopoietic progenitors, and precursor cells have facilitated the autophagy research to better understand how ATG genes function in the hematopoietic system. Taking advantage of the gene-editing platform, this review has summarized the roles of different ATGs at the hematopoietic cell level, their dysregulation, and pathological consequences throughout hematopoiesis.
Collapse
Affiliation(s)
- Kazi Md Mahmudul Hasan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia 7003, Bangladesh
- Department of Neurology, David Geffen School of Medicine, The University of California, 710 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Md Anwarul Haque
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia 7003, Bangladesh
| |
Collapse
|
9
|
Kunduri G, Acharya U, Acharya JK. Lipid Polarization during Cytokinesis. Cells 2022; 11:3977. [PMID: 36552741 PMCID: PMC9776629 DOI: 10.3390/cells11243977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
The plasma membrane of eukaryotic cells is composed of a large number of lipid species that are laterally segregated into functional domains as well as asymmetrically distributed between the outer and inner leaflets. Additionally, the spatial distribution and organization of these lipids dramatically change in response to various cellular states, such as cell division, differentiation, and apoptosis. Division of one cell into two daughter cells is one of the most fundamental requirements for the sustenance of growth in all living organisms. The successful completion of cytokinesis, the final stage of cell division, is critically dependent on the spatial distribution and organization of specific lipids. In this review, we discuss the properties of various lipid species associated with cytokinesis and the mechanisms involved in their polarization, including forward trafficking, endocytic recycling, local synthesis, and cortical flow models. The differences in lipid species requirements and distribution in mitotic vs. male meiotic cells will be discussed. We will concentrate on sphingolipids and phosphatidylinositols because their transbilayer organization and movement may be linked via the cytoskeleton and thus critically regulate various steps of cytokinesis.
Collapse
Affiliation(s)
- Govind Kunduri
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | | | | |
Collapse
|
10
|
Liu Y, Bao Z, Lin Z, Xue Q. Transcriptomic identification of key genes in Pacific oysters Crassostrea gigas responding to major abiotic and biotic stressors. FISH & SHELLFISH IMMUNOLOGY 2022; 131:1027-1039. [PMID: 36372203 DOI: 10.1016/j.fsi.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/09/2022] [Accepted: 11/06/2022] [Indexed: 06/16/2023]
Abstract
Oysters are commercially important intertidal filter-feeding species. Mass mortality events of oysters often occur due to environmental stresses, such as exposure to fluctuating temperatures, salinity, and air, as well as to metal pollution and pathogen infection. Here, RNA-seq data were used to identify shared and specific responsive genes by differential gene expression analysis and weighted gene co-expression network analysis. A total of 18 up-regulated and 10 down-regulated shared responsive genes were identified corresponding to five different stressors. Total 27 stressor-specific genes for temperature, 11 for salinity, 80 for air exposure, 51 for metal pollution, and 636 for Vibrio mediterranei pathogen stress were identified in oysters. Elongin-β was identified as a crucial gene for thermal stress response. Some HSP70s were determined to be shared responsive genes while others were specific to thermal tolerance. The proteins encoded by these stress-related genes should be further investigated to characterize their physiological functions. In addition, the uncharacterized proteins and ncRNAs that were identified may be involved in species-specific stress-response and regulatory mechanisms. This study identified specific genes related to stressors relevant to oyster cultivation. These findings provide useful information for new selective breeding strategies using a data driven method.
Collapse
Affiliation(s)
- Youli Liu
- Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ningbo, 315604, China; Zhejiang Key Laboratory of Aquatic Germplasm Resource, Zhejiang Wanli University, Ningbo, 315100, China; College of Marine Life Sciences, Ocean University of China, Qingdao, 266100, China
| | - Zhenmin Bao
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266100, China
| | - Zhihua Lin
- Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ningbo, 315604, China; Zhejiang Key Laboratory of Aquatic Germplasm Resource, Zhejiang Wanli University, Ningbo, 315100, China.
| | - Qinggang Xue
- Ninghai Institute of Mariculture Breeding and Seed Industry, Zhejiang Wanli University, Ningbo, 315604, China; Zhejiang Key Laboratory of Aquatic Germplasm Resource, Zhejiang Wanli University, Ningbo, 315100, China.
| |
Collapse
|
11
|
Yin H, Shan Y, Xia T, Ji Y, Yuan L, You Y, You B. Emerging Roles of Lipophagy in Cancer Metastasis. Cancers (Basel) 2022; 14:cancers14184526. [PMID: 36139685 PMCID: PMC9496701 DOI: 10.3390/cancers14184526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Metastasis is the main cause of death in patients with malignant tumors worldwide. Mounting evidence suggests lipid droplet metabolism is involved in the process of metastasis. As a mechanism to selectively degrade lipid droplets, the current research on lipophagy and tumor metastasis is quite limited. This review summarizes the crosstalk among lipophagy, tumor lipid metabolism and cancer metastasis, which will provide a new reference for the development of effective targeted drugs. Abstract Obesity is a prominent risk factor for certain types of tumor progression. Adipocytes within tumor stroma contribute to reshaping tumor microenvironment (TME) and the metabolism and metastasis of tumors through the production of cytokines and adipokines. However, the crosstalk between adipocytes and tumor cells remains a major gap in this field. Known as a subtype of selective autophagy, lipophagy is thought to contribute to lipid metabolism by breaking down intracellular lipid droplets (LDs) and generating free fatty acids (FAs). The metastatic potential of cancer cells closely correlates with the lipid degradation mechanisms, which are required for energy generation, signal transduction, and biosynthesis of membranes. Here, we discuss the recent advance in the understanding of lipophagy with tumor lipid metabolism and review current studies on the roles of lipoghagy in the metastasis of certain human malignancies. Additionally, the novel candidate drugs targeting lipophagy are integrated for effective treatment strategies.
Collapse
Affiliation(s)
- Haimeng Yin
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
- Medical School, Nantong University, Qixiu Road 19, Nantong 226001, China
| | - Ying Shan
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
- Medical School, Nantong University, Qixiu Road 19, Nantong 226001, China
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
| | - Tian Xia
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
- Medical School, Nantong University, Qixiu Road 19, Nantong 226001, China
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
| | - Yan Ji
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
- Medical School, Nantong University, Qixiu Road 19, Nantong 226001, China
| | - Ling Yuan
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
- Medical School, Nantong University, Qixiu Road 19, Nantong 226001, China
| | - Yiwen You
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
- Medical School, Nantong University, Qixiu Road 19, Nantong 226001, China
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
- Correspondence: (Y.Y.); (B.Y.)
| | - Bo You
- Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
- Medical School, Nantong University, Qixiu Road 19, Nantong 226001, China
- Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Xisi Road 20, Nantong 226001, China
- Correspondence: (Y.Y.); (B.Y.)
| |
Collapse
|
12
|
Nrf2 Pathway and Autophagy Crosstalk: New Insights into Therapeutic Strategies for Ischemic Cerebral Vascular Diseases. Antioxidants (Basel) 2022; 11:antiox11091747. [PMID: 36139821 PMCID: PMC9495910 DOI: 10.3390/antiox11091747] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Cerebrovascular disease is highly prevalent and has a complex etiology and variable pathophysiological activities. It thus poses a serious threat to human life and health. Currently, pathophysiological research on cerebrovascular diseases is gradually improving, and oxidative stress and autophagy have been identified as important pathophysiological activities that are gradually attracting increasing attention. Many studies have found some effects of oxidative stress and autophagy on cerebrovascular diseases, and studies on the crosstalk between the two in cerebrovascular diseases have made modest progress. However, further, more detailed studies are needed to determine the specific mechanisms. This review discusses nuclear factor erythroid 2-related factor 2 (Nrf2) molecules, which are closely associated with oxidative stress and autophagy, and the crosstalk between them, with the aim of providing clues for studying the two important pathophysiological changes and their crosstalk in cerebrovascular diseases as well as exploring new target treatments.
Collapse
|
13
|
Zhu K, Wu Y, He P, Fan Y, Zhong X, Zheng H, Luo T. PI3K/AKT/mTOR-Targeted Therapy for Breast Cancer. Cells 2022; 11:2508. [PMID: 36010585 PMCID: PMC9406657 DOI: 10.3390/cells11162508] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 12/25/2022] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB/AKT) and mechanistic target of rapamycin (mTOR) (PAM) pathways play important roles in breast tumorigenesis and confer worse prognosis in breast cancer patients. The inhibitors targeting three key nodes of these pathways, PI3K, AKT and mTOR, are continuously developed. For breast cancer patients to truly benefit from PAM pathway inhibitors, it is necessary to clarify the frequency and mechanism of abnormal alterations in the PAM pathway in different breast cancer subtypes, and further explore reliable biomarkers to identify the appropriate population for precision therapy. Some PI3K and mTOR inhibitors have been approved by regulatory authorities for the treatment of specific breast cancer patient populations, and many new-generation PI3K/mTOR inhibitors and AKT isoform inhibitors have also been shown to have good prospects for cancer therapy. This review summarizes the changes in the PAM signaling pathway in different subtypes of breast cancer, and the latest research progress about the biomarkers and clinical application of PAM-targeted inhibitors.
Collapse
Affiliation(s)
- Kunrui Zhu
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yanqi Wu
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Ping He
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yu Fan
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Xiaorong Zhong
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Hong Zheng
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Ting Luo
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| |
Collapse
|
14
|
Abstract
Lysosomes exert pleiotropic functions to maintain cellular homeostasis and degrade autophagy cargo. Despite the great advances that have boosted our understanding of autophagy and lysosomes in both physiology and pathology, their function in mitosis is still controversial. During mitosis, most organelles are reshaped or repurposed to allow the correct distribution of chromosomes. Mitotic entry is accompanied by a reduction in sites of autophagy initiation, supporting the idea of an inhibition of autophagy to protect the genetic material against harmful degradation. However, there is accumulating evidence revealing the requirement of selective autophagy and functional lysosomes for a faithful chromosome segregation. Degradation is the most-studied lysosomal activity, but recently described alternative functions that operate in mitosis highlight the lysosomes as guardians of mitotic progression. Because the involvement of autophagy in mitosis remains controversial, it is important to consider the specific contribution of signalling cascades, the functions of autophagic proteins and the multiple roles of lysosomes, as three entangled, but independent, factors controlling genomic stability. In this Review, we discuss the latest advances in this area and highlight the therapeutic potential of targeting autophagy for drug development.
Collapse
Affiliation(s)
- Eugenia Almacellas
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Caroline Mauvezin
- Department of Biomedicine, Faculty of Medicine, University of Barcelona c/ Casanova, 143 08036 Barcelona, Spain.,August Pi i Sunyer Biomedical Research Institute (IDIBAPS), c/ Rosselló, 149-153 08036 Barcelona, Spain
| |
Collapse
|
15
|
Rakowski M, Porębski S, Grzelak A. Nutraceuticals as Modulators of Autophagy: Relevance in Parkinson’s Disease. Int J Mol Sci 2022; 23:ijms23073625. [PMID: 35408992 PMCID: PMC8998447 DOI: 10.3390/ijms23073625] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 12/29/2022] Open
Abstract
Dietary supplements and nutraceuticals have entered the mainstream. Especially in the media, they are strongly advertised as safe and even recommended for certain diseases. Although they may support conventional therapy, sometimes these substances can have unexpected side effects. This review is particularly focused on the modulation of autophagy by selected vitamins and nutraceuticals, and their relevance in the treatment of neurodegenerative diseases, especially Parkinson’s disease (PD). Autophagy is crucial in PD; thus, the induction of autophagy may alleviate the course of the disease by reducing the so-called Lewy bodies. Hence, we believe that those substances could be used in prevention and support of conventional therapy of neurodegenerative diseases. This review will shed some light on their ability to modulate the autophagy.
Collapse
Affiliation(s)
- Michał Rakowski
- The Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, 90-237 Lodz, Poland
- Cytometry Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (S.P.); (A.G.)
- Correspondence:
| | - Szymon Porębski
- Cytometry Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (S.P.); (A.G.)
| | - Agnieszka Grzelak
- Cytometry Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (S.P.); (A.G.)
| |
Collapse
|
16
|
de la Cruz-Ojeda P, Flores-Campos R, Navarro-Villarán E, Muntané J. The Role of Non-Coding RNAs in Autophagy During Carcinogenesis. Front Cell Dev Biol 2022; 10:799392. [PMID: 35309939 PMCID: PMC8926078 DOI: 10.3389/fcell.2022.799392] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Macroautophagy (autophagy herein) is a cellular stress response and a survival pathway involved in self-renewal and quality control processes to maintain cellular homeostasis. The alteration of autophagy has been implicated in numerous diseases such as cancer where it plays a dual role. Autophagy serves as a tumor suppressor in the early phases of cancer formation with the restoration of homeostasis and eliminating cellular altered constituents, yet in later phases, autophagy may support and/or facilitate tumor growth, metastasis and may contribute to treatment resistance. Key components of autophagy interact with either pro- and anti-apoptotic factors regulating the proximity of tumor cells to apoptotic cliff promoting cell survival. Autophagy is regulated by key cell signaling pathways such as Akt (protein kinase B, PKB), mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) involved in cell survival and metabolism. The expression of critical members of upstream cell signaling, as well as those directly involved in the autophagic and apoptotic machineries are regulated by microRNAs (miRNAs) and long non-coding RNAs (lncRNAs). Consequently, non-coding RNAs play a relevant role in carcinogenesis and treatment response in cancer. The review is an update of the current knowledge in the regulation by miRNA and lncRNA of the autophagic components and their functional impact to provide an integrated and comprehensive regulatory network of autophagy in cancer.
Collapse
Affiliation(s)
- Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain.,Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD o Ciberehd), Institute of Health Carlos III, Madrid, Spain
| | - Rocío Flores-Campos
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain.,Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD o Ciberehd), Institute of Health Carlos III, Madrid, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain.,Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.,Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD o Ciberehd), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
17
|
Targeting the endo-lysosomal autophagy pathway to treat inflammatory bowel diseases. J Autoimmun 2022; 128:102814. [PMID: 35298976 DOI: 10.1016/j.jaut.2022.102814] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 01/18/2023]
Abstract
Inflammatory bowel disease (IBD) is a serious public health problem in Western society with a continuing increase in incidence worldwide. Safe, targeted medicines for IBD are not yet available. Autophagy, a vital process implicated in normal cell homeostasis, provides a potential point of entry for the treatment of IBDs, as several autophagy-related genes are associated with IBD risk. We conducted a series of experiments in three distinct mouse models of colitis to test the effectiveness of therapeutic P140, a phosphopeptide that corrects autophagy dysfunctions in other autoimmune and inflammatory diseases. Colitis was experimentally induced in mice by administering dextran sodium sulfate and 2,4,6 trinitrobenzene sulfonic acid. Transgenic mice lacking both il-10 and iRhom2 - involved in tumor necrosis factor α secretion - were also used. In the three models investigated, P140 treatment attenuated the clinical and histological severity of colitis. Post-treatment, altered expression of several macroautophagy and chaperone-mediated autophagy markers, and of pro-inflammatory mediators was corrected. Our results demonstrate that therapeutic intervention with an autophagy modulator improves colitis in animal models. These findings highlight the potential of therapeutic peptide P140 for use in the treatment of IBD.
Collapse
|
18
|
Zhou H, Ling H, Li Y, Jiang X, Cheng S, Zubeir GM, Xia Y, Qin X, Zhang J, Zou Z, Chen C. Downregulation of beclin 1 restores arsenite-induced impaired autophagic flux by improving the lysosomal function in the brain. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 229:113066. [PMID: 34929507 DOI: 10.1016/j.ecoenv.2021.113066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 06/14/2023]
Abstract
Arsenite is a toxic metalloid that causes various adverse effects in the brain. However, the underlying mechanisms of arsenite-induced neurotoxicity remain poorly understood. In this study, both adult beclin 1+/+ and beclin 1+/- mice were employed to establish a model of chronic arsenite exposure by treating with arsenite via drinking water for 6 months. The results clearly demonstrated that exposure to arsenite profoundly caused damage to the cerebral cortex, induced autophagy and impaired autophagic flux in the cerebral cortex. Heterozygous disruption of beclin 1 in animals remarkably alleviated the neurotoxic effects of arsenite. To verify the results obtained in the animals, a permanent U251 cell line was used. After treating of cells with arsenite, similar phenomenon was also observed, showing the significant elevation in the expression levels of autophagy-related genes. Importantly, lysosomal dysfunction caused by arsenite was observed in vitro and in vivo. Either knockdown of beclin 1 in cells or heterozygous disruption of beclin 1 in animals remarkably alleviated the lysosomal dysfunction induced by arsenite. These findings indicate that downregulation of beclin 1 could restore arsenite-induced impaired autophagic flux possibly through improving lysosomal function, and correct that regulation of autophagy via beclin 1 would be an alternative approach for the treatment of arsenite neurotoxicity.
Collapse
Affiliation(s)
- Hongmei Zhou
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Hong Ling
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yunlong Li
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Shuqun Cheng
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | | | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory disease, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China; Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, People's Republic of China; Dongsheng Lung-Brain Disease Joint Lab, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
19
|
Class III PI3K Biology. Curr Top Microbiol Immunol 2022; 436:69-93. [DOI: 10.1007/978-3-031-06566-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Liu Y, Wei CH, Li C, Chen WZ, Zhu Y, Xu RS. Phosphoinositide-3-kinase regulatory subunit 4 participates in the occurrence and development of amyotrophic lateral sclerosis by regulating autophagy. Neural Regen Res 2021; 17:1609-1616. [PMID: 34916448 PMCID: PMC8771104 DOI: 10.4103/1673-5374.330621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The development of amyotrophic lateral sclerosis (ALS) may be related to the abnormal alterations of multiple proteins. Our previous study revealed that the expression of phosphoinositide-3-kinase regulatory subunit 4 (PIK3R4) was decreased in ALS. However, the role of PIK3R4 in ALS pathogenesis remains unknown. This study was the first to find that transfection of PC12 cells with small interfering RNA against the PIK3R4 gene significantly decreased the expression levels of PIK3R4 and the autophagy-related proteins p62 and LC3. Additionally, in vivo experiments revealed that the PIK3R4 protein was extensively expressed in the anterior horn, posterior horn, central canal, and areas surrounding the central canal in cervical, thoracic, and lumbar segments of the spinal cord in adult mice. PIK3R4 protein was mainly expressed in the neurons within the spinal lumbar segments. PIK3R4 and p62 expression levels were significantly decreased at both the pre-onset and onset stages of ALS disease in Tg(SOD1*G93A)1Gur mice compared with control mice, but these proteins were markedly increased at the progression stage. LC3 protein expression did not change during progression of ALS. These findings suggest that PIK3R4 likely participates in the prevention of ALS progression. This study was approved by the Ethics Committee for Animal Care and Use of Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University (approval No. 2020025) on March 26, 2020.
Collapse
Affiliation(s)
- Yue Liu
- Department of Neurology, First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Cai-Hui Wei
- Department of Neurology, First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Cheng Li
- Department of Neurology, First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Wen-Zhi Chen
- Department of Neurology, First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yu Zhu
- Department of Neurology, First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Ren-Shi Xu
- Department of Neurology, First Affiliated Hospital of Nanchang Medical College, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
21
|
Ren H, Yin X, Su C, Guo M, Wang XF, Na L, Lin Y, Wang X. Equine lentivirus counteracts SAMHD1 restriction by Rev-mediated degradation of SAMHD1 via the BECN1-dependent lysosomal pathway. Autophagy 2021; 17:2800-2817. [PMID: 33172327 PMCID: PMC8525956 DOI: 10.1080/15548627.2020.1846301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 10/16/2020] [Accepted: 10/30/2020] [Indexed: 02/09/2023] Open
Abstract
The innate immune restriction factor SAMHD1 can inhibit diverse viruses in myeloid cells. Mechanistically, SAMHD1 inhibits lentiviral replication including HIV-1 by depleting the nucleotide pool to interfere with their reverse transcription. Equine infectious anemia virus (EIAV) is an ancient lentivirus that preferentially attacks macrophages. However, the mechanism by which EIAV successfully establishes infection in macrophages with functional SAMHD1 remains unclear. Here, we demonstrate that while equine SAMDH1 can limit EIAV replication in equine macrophages at the reverse transcription stage, the antiviral effect is counteracted by the well-known transcriptional regulator Rev, which downregulates equine SAMHD1 through the lysosomal pathway. Remarkably, Rev hijacks BECN1 (beclin 1) and PIK3C3 to mediate SAMHD1 degradation in a canonical macroautophagy/autophagy-independent pathway. Our study illustrates that equine lentiviral Rev possesses important functions in evading cellular innate immunity in addition to its RNA regulatory function, and may provide new insights into the co-evolutionary arms race between SAMHD1 and lentiviruses.Abbreviations:3-MA: 3-methyladenine; AA: amino acid; ACTB: actin beta; AD: activation domain; ATG: autophagy related; Baf A1: bafilomycin A1; BD: binding domain; BECN1: beclin 1; BH3: BCL2-homology-3 domain; BiFC: bimolecular fluorescence complementation; CCD: coiled-coil domain; class III PtdIns3K: class III phosphatidylinositol 3-kinase; CQ: chloroquine; Co-IP: co-immunoprecipitation; dNTPase: dGTP-stimulated deoxynucleoside triphosphate triphosphohydrolase; ECD: evolutionarily conserved domain; EIAV: equine infectious anemia virus; eMDMs: equine monocyte-derived macrophages; GFP: green fluorescent protein; HD: histidine-aspartic; HIV-1: human immunodeficiency virus-1; hpi: hours post infection; hpt: hours post transfection; KO: knockout; LAMP2: lysosomal associated membrane protein 2; LMB: leptomycin B; PMA: phorbol 12-myristate 13-acetate; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; ND: unknown non-essential domain; NES: nuclear export signal; NLS: localization signal; NS: statistically non-significant; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; RBD: RNA binding domain; RT: reverse transcriptase; siRNAs: small interfering RNAs; SAMHD1: SAM and HD domain containing deoxynucleoside triphosphate triphosphohydrolase 1; SIV: simian immunodeficiency virus; VN: C-terminal residues of Venus 174 to 238; VC: N-terminal residues 2 to 173 of Venus.
Collapse
Affiliation(s)
- Huiling Ren
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xin Yin
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chao Su
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Miaomiao Guo
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xue-Feng Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Lei Na
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yuezhi Lin
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiaojun Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
22
|
Hu Z, Sankar DS, Vu B, Leytens A, Vionnet C, Wu W, Stumpe M, Martínez-Martínez E, Stork B, Dengjel J. ULK1 phosphorylation of striatin activates protein phosphatase 2A and autophagy. Cell Rep 2021; 36:109762. [PMID: 34592149 DOI: 10.1016/j.celrep.2021.109762] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/16/2021] [Accepted: 09/06/2021] [Indexed: 01/18/2023] Open
Abstract
The evolutionarily conserved ULK1 kinase complex acts as gatekeeper of canonical autophagy and regulates induction of autophagosome biogenesis. To better understand control of ULK1 and analyze whether ULK1 has broader functions that are also linked to the later steps of autophagy, we perform comprehensive phosphoproteomic analyses. Combining in vivo with in vitro data, we identify numerous direct ULK1 target sites within autophagy-relevant proteins that are critical for autophagosome maturation and turnover. In addition, we highlight an intimate crosstalk between ULK1 and several phosphatase complexes. ULK1 is not only a PP2A target but also directly phosphorylates the regulatory PP2A subunit striatin, activating PP2A and serving as positive feedback to promote autophagy-dependent protein turnover. Thus, ULK1 and phosphatase activities are tightly coordinated to robustly regulate protein degradation by autophagy.
Collapse
Affiliation(s)
- Zehan Hu
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | | | - Bich Vu
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Alexandre Leytens
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Christine Vionnet
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Wenxian Wu
- Institute of Molecular Medicine I, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Michael Stumpe
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | | | - Björn Stork
- Institute of Molecular Medicine I, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland.
| |
Collapse
|
23
|
Guo P, Ma Y, Deng G, Li L, Gong Y, Yang F, You Y. CYR61, regulated by miR-22-3p and MALAT1, promotes autophagy in HK-2 cell inflammatory model. Transl Androl Urol 2021; 10:3486-3500. [PMID: 34532273 PMCID: PMC8421830 DOI: 10.21037/tau-21-623] [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: 06/17/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Background Renal tubular epithelial cells play an important role in renal function and are a major site of injury from inflammation. Emerging evidence suggests that CYR61 is involved in the regulation of autophagy. However, there are few studies on CYR61 in nephropathy and associated inflammation. This study aimed to clarify how CYR61 regulates autophagy in human renal epithelial cells while in an inflammatory state and regulates the upstream pathway of CYR61 levels. Methods The human renal tubular epithelial cells (HK-2) cell line treated by lipopolysaccharide (LPS) was used as an inflammatory model of human epithelial cells. Short hairpin RNA (shRNA) was used to down-regulate CYR61, and the changes in the transcription and expression levels of related molecules, as well as the morphological changes of HK-2 cells, were detected by quantitative real time-PCR (qRT-PCR), western blot (WB), and transmission electron microscopy. Either CYR61 or MALAT1 were up-regulated by overexpression vectors, or MALAT1 was down-regulated by miR-22-3p mimics. Subsequently, the levels of CYR61, MALAT1, related inflammatory factors, and autophagy factors were measured by qPCR, WB, and enzyme-linked immunosorbent assay (ELISA). Cell apoptosis was detected by flow cytometry and acridine-orange assay. Results We observed that down-regulation of CYR61 could down-regulate 1B-light chain 3 (LC3) level and inhibit autophagy in the LPS-induced inflammation model of HK-2 cells. The expression levels of CYR61, Beclin1, Atg5, LC3, interleukin 6 (IL-6), and tumor necrosis factor-α (TNF-α) were significantly increased by upregulating CYR61 or MALAT1 by overexpression vector, while the expression level of p62 was significantly decreased, intracellular reactive oxygen species (ROS) content was increased, and the proportion of autophagy and apoptosis was increased. The use of miR-22-3p mimics significantly reversed the changes induced by up-regulation of CYR61 or MALAT1 at the molecular and cellular levels. Conclusions Our data indicated that CYR61 positively regulates autophagy of HK-2 cells under an inflammatory state, and was negatively regulated by miR-22-3p, while miR-22-3p and MALAT1 were negatively regulated by each other.
Collapse
Affiliation(s)
- Pengwei Guo
- Department of Nephrology, Jinan University, Guangzhou, China.,Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yanfei Ma
- Department of Gland Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Gao Deng
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Lingling Li
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yunxia Gong
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Fafen Yang
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yanwu You
- Department of Nephrology, Jinan University, Guangzhou, China.,Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
24
|
Din SRU, Nisar MA, Ramzan MN, Saleem MZ, Ghayas H, Ahmad B, Batool S, Kifayat K, Guo X, Huang M, Zhong M. Latcripin-7A from Lentinula edodes C 91-3 induces apoptosis, autophagy, and cell cycle arrest at G1 phase in human gastric cancer cells via inhibiting PI3K/Akt/mTOR signaling. Eur J Pharmacol 2021; 907:174305. [PMID: 34224698 DOI: 10.1016/j.ejphar.2021.174305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 11/28/2022]
Abstract
Gastric cancer (G.C) is one of the most lethal cancer types worldwide. Current treatment requires surgery along with chemotherapy, which causes obstacles for speedy recovery. The discovery of novel drugs is needed for better treatment of G.C with minimum side effects. Latcripin-7A (LP-7A) is a newly discovered peptide extracted from Lentinula edodes. It is recently studied for its anti-cancer activity. In this study, LP-7A was modeled using a phyre2 server. Anti-proliferation effects of LP-7A on G.C cells were examined via CCK-8, colony formation, and morphology assay. Apoptosis of LP-7A treated G.C cells was evaluated via Hoechst Stain, western blot and flow cytometry. Autophagy was assessed via acridine orange staining and western blot. The cell cycle was assessed via flow cytometry assay and western blot. Pathway was studied via western blot and STRING database. Anti-migratory effects of LP-7A treated G.C cells were analyzed via wound healing, western blot, and migration and invasion assay. LP-7A effectively inhibited the growth of G.C cells by inhibiting the PI3K/Akt/mTOR pathway. G.C cells treated with LP-7A arrested the cell cycle at the G1 phase, contributing to the inhibition of migration and invasion. Furthermore, LP-7A induced apoptosis and autophagy in gastric cancer cells. These results indicated that LP-7A is a promising anti-cancer agent. It affected the proliferation and growth of G.C cells (SGC-7901 and BGC-823) by inducing apoptosis, autophagy, and inhibiting cell cycle at the G1 phase in G.C cells.
Collapse
Affiliation(s)
- Syed Riaz Ud Din
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Muhammad Azhar Nisar
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Muhammad Noman Ramzan
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Muhammad Zubair Saleem
- Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China; School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122, PR China.
| | - Hassan Ghayas
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China.
| | - Bashir Ahmad
- Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China; Department of Biology, The University of Haripur, Pakistan.
| | - Samana Batool
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China; Department of Microbiology and Molecular Genetics, University of Okara, 56300, Pakistan.
| | - Kashif Kifayat
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Xiaorong Guo
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Min Huang
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Mintao Zhong
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| |
Collapse
|
25
|
Vieira PH, Benjamim CF, Atella G, Ramos I. VPS38/UVRAG and ATG14, the variant regulatory subunits of the ATG6/Beclin1-PI3K complexes, are crucial for the biogenesis of the yolk organelles and are transcriptionally regulated in the oocytes of the vector Rhodnius prolixus. PLoS Negl Trop Dis 2021; 15:e0009760. [PMID: 34492013 PMCID: PMC8448300 DOI: 10.1371/journal.pntd.0009760] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/17/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
In insects the reserve proteins are stored in the oocytes into endocytic-originated vesicles named yolk organelles. VPS38/UVRAG and ATG14 are the variant regulatory subunits of two class-III ATG6/Beclin1 PI3K complexes that regulate the recruitment of the endocytic (complex II) and autophagic (complex I) machineries. In a previous work from our group, we found that the silencing of ATG6/Beclin1 resulted in the formation of yolk-deficient oocytes due to defects in the endocytosis of the yolk proteins. Because ATG6/Beclin1 is present in the two above-described PI3K complexes, we could not identify the contributions of each complex to the yolk defective phenotypes. To address this, here we investigated the role of the variant subunits VPS38/UVRAG (complex II, endocytosis) and ATG14 (complex I, autophagy) in the biogenesis of the yolk organelles in the insect vector of Chagas Disease Rhodnius prolixus. Interestingly, the silencing of both genes phenocopied the silencing of ATG6/Beclin1, generating 1) accumulation of yolk proteins in the hemolymph; 2) white, smaller, and yolk-deficient oocytes; 3) abnormal yolk organelles in the oocyte cortex; and 4) unviable F1 embryos. However, we found that the similar phenotypes were the result of a specific cross-silencing effect among the PI3K subunits where the silencing of VPS38/UVRAG and ATG6/Beclin1 resulted in the specific silencing of each other, whereas the silencing of ATG14 triggered the silencing of all three PI3K components. Because the silencing of VPS38/UVRAG and ATG6/Beclin1 reproduced the yolk-deficiency phenotypes without the cross silencing of ATG14, we concluded that the VPS38/UVRAG PI3K complex II was the major contributor to the previously observed phenotypes in silenced insects. Altogether, we found that class-III ATG6/Beclin1 PI3K complex II (VPS38/UVRAG) is essential for the yolk endocytosis and that the subunits of both complexes are under an unknown transcriptional regulatory system.
Collapse
Affiliation(s)
- Priscila H Vieira
- Laboratório de Bioquímica de Insetos, Instituto de Bioquímica Médica Leopoldo de Meis. Universidade Federal do Rio de Janeiro, Brazil
| | - Claudia F Benjamim
- Laboratório de Imunologia Molecular e Celular, Instituto de Biofísica Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro, Brazil
| | - Georgia Atella
- Laboratório de de Bioquímica de Lipídeos e Lipoproteínas, Instituto de Bioquímica Médica Leopoldo de Meis. Universidade Federal do Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular-INCT-EM/CNPq. Rio de Janeiro, Brazil
| | - Isabela Ramos
- Laboratório de Bioquímica de Insetos, Instituto de Bioquímica Médica Leopoldo de Meis. Universidade Federal do Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular-INCT-EM/CNPq. Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Tran S, Fairlie WD, Lee EF. BECLIN1: Protein Structure, Function and Regulation. Cells 2021; 10:cells10061522. [PMID: 34204202 PMCID: PMC8235419 DOI: 10.3390/cells10061522] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
BECLIN1 is a well-established regulator of autophagy, a process essential for mammalian survival. It functions in conjunction with other proteins to form Class III Phosphoinositide 3-Kinase (PI3K) complexes to generate phosphorylated phosphatidylinositol (PtdIns), lipids essential for not only autophagy but other membrane trafficking processes. Over the years, studies have elucidated the structural, biophysical, and biochemical properties of BECLIN1, which have shed light on how this protein functions to allosterically regulate these critical processes of autophagy and membrane trafficking. Here, we review these findings and how BECLIN1’s diverse protein interactome regulates it, as well as its impact on organismal physiology.
Collapse
Affiliation(s)
- Sharon Tran
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - W. Douglas Fairlie
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
- Correspondence: (W.D.F.); (E.F.L.)
| | - Erinna F. Lee
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
- Correspondence: (W.D.F.); (E.F.L.)
| |
Collapse
|
27
|
Bischoff ME, Zang Y, Chu J, Price AD, Ehmer B, Talbot NJ, Newbold MJ, Paul A, Guan JL, Plas DR, Meller J, Czyzyk-Krzeska MF. Selective MAP1LC3C (LC3C) autophagy requires noncanonical regulators and the C-terminal peptide. J Cell Biol 2021; 220:212107. [PMID: 33988680 PMCID: PMC8129795 DOI: 10.1083/jcb.202004182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 03/02/2021] [Accepted: 04/19/2021] [Indexed: 12/01/2022] Open
Abstract
LC3s are canonical proteins necessary for the formation of autophagosomes. We have previously established that two paralogs, LC3B and LC3C, have opposite activities in renal cancer, with LC3B playing an oncogenic role and LC3C a tumor-suppressing role. LC3C is an evolutionary late gene present only in higher primates and humans. Its most distinct feature is a C-terminal 20-amino acid peptide cleaved in the process of glycine 126 lipidation. Here, we investigated mechanisms of LC3C-selective autophagy. LC3C autophagy requires noncanonical upstream regulatory complexes that include ULK3, UVRAG, RUBCN, PIK3C2A, and a member of ESCRT, TSG101. We established that postdivision midbody rings (PDMBs) implicated in cancer stem-cell regulation are direct targets of LC3C autophagy. LC3C C-terminal peptide is necessary and sufficient to mediate LC3C-dependent selective degradation of PDMBs. This work establishes a new noncanonical human-specific selective autophagic program relevant to cancer stem cells.
Collapse
Affiliation(s)
- Megan E Bischoff
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Yuanwei Zang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH.,Department of Urology, Qilu Hospital, Shandong University, Jinan, People's Republic of China
| | - Johnson Chu
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Adam D Price
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Birgit Ehmer
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Nicholas J Talbot
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Michael J Newbold
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Anurag Paul
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - David R Plas
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Jarek Meller
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH.,Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.,Department of Biomedical Informatics, University of Cincinnati College of Medicine, Cincinnati, OH.,Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Maria F Czyzyk-Krzeska
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH.,Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH.,Cincinnati Veterans Affairs Medical Center, Department of Veterans Affairs, Cincinnati, OH
| |
Collapse
|
28
|
Wang H, Ding Z, Gou M, Hu J, Wang Y, Wang L, Wang Y, Di T, Zhang X, Hao X, Wang X, Yang Y, Qian W. Genome-wide identification, characterization, and expression analysis of tea plant autophagy-related genes (CsARGs) demonstrates that they play diverse roles during development and under abiotic stress. BMC Genomics 2021; 22:121. [PMID: 33596831 PMCID: PMC7891152 DOI: 10.1186/s12864-021-07419-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/31/2021] [Indexed: 12/12/2022] Open
Abstract
Background Autophagy, meaning ‘self-eating’, is required for the degradation and recycling of cytoplasmic constituents under stressful and non-stressful conditions, which helps to maintain cellular homeostasis and delay aging and longevity in eukaryotes. To date, the functions of autophagy have been heavily studied in yeast, mammals and model plants, but few studies have focused on economically important crops, especially tea plants (Camellia sinensis). The roles played by autophagy in coping with various environmental stimuli have not been fully elucidated to date. Therefore, investigating the functions of autophagy-related genes in tea plants may help to elucidate the mechanism governing autophagy in response to stresses in woody plants. Results In this study, we identified 35 C. sinensis autophagy-related genes (CsARGs). Each CsARG is highly conserved with its homologues from other plant species, except for CsATG14. Tissue-specific expression analysis demonstrated that the abundances of CsARGs varied across different tissues, but CsATG8c/i showed a degree of tissue specificity. Under hormone and abiotic stress conditions, most CsARGs were upregulated at different time points during the treatment. In addition, the expression levels of 10 CsARGs were higher in the cold-resistant cultivar ‘Longjing43’ than in the cold-susceptible cultivar ‘Damianbai’ during the CA period; however, the expression of CsATG101 showed the opposite tendency. Conclusions We performed a comprehensive bioinformatic and physiological analysis of CsARGs in tea plants, and these results may help to establish a foundation for further research investigating the molecular mechanisms governing autophagy in tea plant growth, development and response to stress. Meanwhile, some CsARGs could serve as putative molecular markers for the breeding of cold-resistant tea plants in future research. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07419-2.
Collapse
Affiliation(s)
- Huan Wang
- College of Horticulture, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zhaotang Ding
- College of Horticulture, Qingdao Agricultural University, Qingdao, 266109, China
| | - Mengjie Gou
- College of Horticulture, Qingdao Agricultural University, Qingdao, 266109, China
| | - Jianhui Hu
- College of Horticulture, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yu Wang
- College of Horticulture, Qingdao Agricultural University, Qingdao, 266109, China
| | - Lu Wang
- National Center for Tea Plant Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, China.,Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, 310008, China
| | - Yuchun Wang
- College of Agriculture and Food Science, Zhejiang A&F University, Hangzhou, 311300, China
| | - Taimei Di
- National Center for Tea Plant Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, China.,Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, 310008, China
| | - Xinfu Zhang
- College of Horticulture, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xinyuan Hao
- National Center for Tea Plant Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, China.,Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, 310008, China
| | - Xinchao Wang
- National Center for Tea Plant Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, China.,Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, 310008, China
| | - Yajun Yang
- National Center for Tea Plant Improvement, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou, 310008, China.,Key Laboratory of Tea Biology and Resources Utilization, Ministry of Agriculture, Hangzhou, 310008, China
| | - Wenjun Qian
- College of Horticulture, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
29
|
Hu YJ, Zhong JT, Gong L, Zhang SC, Zhou SH. Autophagy-Related Beclin 1 and Head and Neck Cancers. Onco Targets Ther 2020; 13:6213-6227. [PMID: 32669852 PMCID: PMC7335767 DOI: 10.2147/ott.s256072] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Beclin 1, a positive regulator of autophagy, behaves as a double-edged sword in tumorigenesis. Beclin 1 contributes to tumor suppression by removing defective or damaged organelles and other cellular components; however, its activity can also stimulate cancer initiation and progression. In head and neck cancer, Beclin 1 overexpression promotes autophagy, which limits DNA damage and chromosomal instability and increases necrosis and inflammation by impacting apoptotic and autophagic pathways. This paper reviews the relationship between Beclin 1, carcinogenesis and head and neck cancer prognosis.
Collapse
Affiliation(s)
- Yang-Jie Hu
- Department of Otolaryngology, The Affiliated Cixi Hospital of Wenzhou Medical University, Cixi 315300, Zhejiang, People's Republic of China.,Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, People's Republic of China
| | - Jiang-Tao Zhong
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, People's Republic of China
| | - Liang Gong
- Department of Otolaryngology, The Affiliated Cixi Hospital of Wenzhou Medical University, Cixi 315300, Zhejiang, People's Republic of China
| | - Si-Cong Zhang
- Department of Otolaryngology, The Affiliated Cixi Hospital of Wenzhou Medical University, Cixi 315300, Zhejiang, People's Republic of China
| | - Shui-Hong Zhou
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, People's Republic of China
| |
Collapse
|
30
|
Birgisdottir ÅB, Johansen T. Autophagy and endocytosis – interconnections and interdependencies. J Cell Sci 2020; 133:133/10/jcs228114. [DOI: 10.1242/jcs.228114] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
ABSTRACT
Autophagy and endocytosis are membrane-vesicle-based cellular pathways for degradation and recycling of intracellular and extracellular components, respectively. These pathways have a common endpoint at the lysosome, where their cargo is degraded. In addition, the two pathways intersect at different stages during vesicle formation, fusion and trafficking, and share parts of the molecular machinery. Accumulating evidence shows that autophagy is dependent upon endocytosis and vice versa. The emerging joint network of autophagy and endocytosis is of vital importance for cellular metabolism and signaling, and thus also highly relevant in disease settings. In this Review, we will discuss examples of how the autophagy machinery impacts on endocytosis and cell signaling, and highlight how endocytosis regulates the different steps in autophagy in mammalian cells. Finally, we will focus on the interplay of these pathways in the quality control of their common endpoint, the lysosome.
Collapse
Affiliation(s)
- Åsa B. Birgisdottir
- The Heart and Lung Clinic, University Hospital of North Norway, 9037 Tromsø, Norway
- Clinical Cardiovascular Research Group, Department of Clinical Medicine, University of Tromsø –The Arctic University of Norway, 9037 Tromsø, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø–The Arctic University of Norway, 9037 Tromsø, Norway
| |
Collapse
|
31
|
Autophagy-Independent Functions of the Autophagy Machinery. Cell 2020; 177:1682-1699. [PMID: 31199916 PMCID: PMC7173070 DOI: 10.1016/j.cell.2019.05.026] [Citation(s) in RCA: 606] [Impact Index Per Article: 121.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/11/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023]
Abstract
Macroautophagy (herein referred to as autophagy) is an evolutionary ancient mechanism that culminates with the lysosomal degradation of superfluous or potentially dangerous cytosolic entities. Over the past 2 decades, the molecular mechanisms underlying several variants of autophagy have been characterized in detail. Accumulating evidence suggests that most, if not all, components of the molecular machinery for autophagy also mediate autophagy-independent functions. Here, we discuss emerging data on the non-autophagic functions of autophagy-relevant proteins.
Collapse
|
32
|
Rizzelli F, Malabarba MG, Sigismund S, Mapelli M. The crosstalk between microtubules, actin and membranes shapes cell division. Open Biol 2020; 10:190314. [PMID: 32183618 PMCID: PMC7125961 DOI: 10.1098/rsob.190314] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
Mitotic progression is orchestrated by morphological and mechanical changes promoted by the coordinated activities of the microtubule (MT) cytoskeleton, the actin cytoskeleton and the plasma membrane (PM). MTs assemble the mitotic spindle, which assists sister chromatid separation, and contact the rigid and tensile actomyosin cortex rounded-up underneath the PM. Here, we highlight the dynamic crosstalk between MTs, actin and cell membranes during mitosis, and discuss the molecular connections between them. We also summarize recent views on how MT traction forces, the actomyosin cortex and membrane trafficking contribute to spindle positioning in isolated cells in culture and in epithelial sheets. Finally, we describe the emerging role of membrane trafficking in synchronizing actomyosin tension and cell shape changes with cell-substrate adhesion, cell-cell contacts and extracellular signalling events regulating proliferation.
Collapse
Affiliation(s)
| | - Maria Grazia Malabarba
- IEO, Istituto Europeo di Oncologia IRCCS, Milan, Italy
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Milan, Italy
| | - Sara Sigismund
- IEO, Istituto Europeo di Oncologia IRCCS, Milan, Italy
- Dipartimento di Oncologia ed Emato-oncologia, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
33
|
Pandey M, Saleem S, Nautiyal H, Pottoo FH, Javed MN. PINK1/Parkin in Neurodegenerative Disorders. ADVANCES IN MEDICAL DIAGNOSIS, TREATMENT, AND CARE 2020:282-301. [DOI: 10.4018/978-1-7998-1317-0.ch011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
PTEN-induced kinase 1 (PINK1), a mitochondrial serine/threonine-protein kinase encoded by the PINK1 gene, is thought to protect cells from stress-induced mitochondrial dysfunction. The activity of PINK1 facilitates the binding of Parkin protein with depolarized mitochondria to induce autophagy. Mutations of PINK1causes a type of autosomal recessive early-onset Parkinson's disease. Cell depends on the surveillance systems or mechanisms like protein quality control to handle the alterations in the proteins that are induced because of these mutations. These mutant proteins are found to be pathogenic and are reported to be related to various neurodegenerative disorders. This chapter focuses on the role of PINK1/Parkin in mitochondria quality control and its subsequent effect in neurodegeneration.
Collapse
Affiliation(s)
- Mukesh Pandey
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, India
| | - Shakir Saleem
- Department of Public Health, College of Health Science, Saudi Electronic University, Riyadh, Saudi Arabia
| | - Himani Nautiyal
- Department of Pharmacology, Siddhartha Institute of Pharmacy, India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Saudi Arabia
| | | |
Collapse
|
34
|
He F, Nichols RM, Kailasam L, Wensel TG, Agosto MA. Critical Role for Phosphatidylinositol-3 Kinase Vps34/PIK3C3 in ON-Bipolar Cells. Invest Ophthalmol Vis Sci 2019; 60:2861-2874. [PMID: 31260037 PMCID: PMC6607926 DOI: 10.1167/iovs.19-26586] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Purpose Phosphatidylinositol-3-phosphate (PI(3)P), and Vps34, the type III phosphatidylinositol 3-kinase primarily responsible for its production, are important for function and survival of sensory neurons, where they have key roles in membrane processing events, such as autophagy, endosome processing, and fusion of membranes bearing ubiquitinated cargos with lysosomes. We examined their roles in the most abundant class of secondary neurons in the vertebrate retina, the ON-bipolar cells (ON-BCs). Methods A conditional Vps34 knockout mouse line was generated by crossing Vps34 floxed mice with transgenic mice expressing Cre recombinase in ON-BCs. Structural changes in the retina were determined by immunofluorescence and electron microscopy, and bipolar cell function was determined by electroretinography. Results Vps34 deletion led to selective death of ON-BCs, a thinning of the inner nuclear layer, and a progressive decline of electroretinogram b-wave amplitudes. There was no evidence for loss of other retinal neurons, or disruption of rod-horizontal cell contacts in the outer plexiform layer. Loss of Vps34 led to aberrant accumulation of membranes positive for autophagy markers LC3, p62, and ubiquitin, accumulation of endosomal membranes positive for Rab7, and accumulation of lysosomes. Similar effects were observed in Purkinje cells of the cerebellum, leading to severe and progressive ataxia. Conclusions These results support an essential role for PI(3)P in fusion of autophagosomes with lysosomes and in late endosome maturation. The cell death resulting from Vps34 knockout suggests that these processes are essential for the health of ON-BCs.
Collapse
Affiliation(s)
- Feng He
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States
| | - Ralph M Nichols
- Department of Ophthalmology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States
| | - Lavanya Kailasam
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States
| | - Theodore G Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States.,Department of Ophthalmology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States
| | - Melina A Agosto
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
35
|
Downregulation of class II phosphoinositide 3-kinase PI3K-C2β delays cell division and potentiates the effect of docetaxel on cancer cell growth. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:472. [PMID: 31752944 PMCID: PMC6873561 DOI: 10.1186/s13046-019-1472-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023]
Abstract
Background Alteration of signalling pathways regulating cell cycle progression is a common feature of cancer cells. Several drugs targeting distinct phases of the cell cycle have been developed but the inability of many of them to discriminate between normal and cancer cells has strongly limited their clinical potential because of their reduced efficacy at the concentrations used to limit adverse side effects. Mechanisms of resistance have also been described, further affecting their efficacy. Identification of novel targets that can potentiate the effect of these drugs or overcome drug resistance can provide a useful strategy to exploit the anti-cancer properties of these agents to their fullest. Methods The class II PI3K isoform PI3K-C2β was downregulated in prostate cancer PC3 cells and cervical cancer HeLa cells using selective siRNAs and the effect on cell growth was determined in the absence or presence of the microtubule-stabilizing agent/anti-cancer drug docetaxel. Mitosis progression was monitored by time-lapse microscopy. Clonogenic assays were performed to determine the ability of PC3 and HeLa cells to form colonies upon PI3K-C2β downregulation in the absence or presence of docetaxel. Cell multi-nucleation was assessed by immunofluorescence. Tumour growth in vivo was assessed using a xenograft model of PC3 cells upon PI3K-C2β downregulation and in combination with docetaxel. Results Downregulation of PI3K-C2β delays mitosis progression in PC3 and HeLa cells, resulting in reduced ability to form colonies in clonogenic assays in vitro. Compared to control cells, PC3 cells lacking PI3K-C2β form smaller and more compact colonies in vitro and they form tumours more slowly in vivo in the first weeks after cells implant. Stable and transient PI3K-C2β downregulation potentiates the effect of low concentrations of docetaxel on cancer cell growth. Combination of PI3K-C2β downregulation and docetaxel almost completely prevents colonies formation in clonogenic assays in vitro and strongly inhibits tumour growth in vivo. Conclusions These data reveal a novel role for the class II PI3K PI3K-C2β during mitosis progression. Furthermore, data indicate that blockade of PI3K-C2β might represent a novel strategy to potentiate the effect of docetaxel on cancer cell growth.
Collapse
|
36
|
Matthew-Onabanjo AN, Janusis J, Mercado-Matos J, Carlisle AE, Kim D, Levine F, Cruz-Gordillo P, Richards R, Lee MJ, Shaw LM. Beclin 1 Promotes Endosome Recruitment of Hepatocyte Growth Factor Tyrosine Kinase Substrate to Suppress Tumor Proliferation. Cancer Res 2019; 80:249-262. [PMID: 31744816 DOI: 10.1158/0008-5472.can-19-1555] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/01/2019] [Accepted: 11/13/2019] [Indexed: 12/24/2022]
Abstract
Beclin 1 has nonautophagic functions that include its ability to regulate endocytic receptor trafficking. However, the contribution of this function to tumor suppression is poorly understood. Here, we provide in vivo evidence that Beclin 1 suppresses tumor proliferation by regulating the endocytic trafficking and degradation of the EGFR and transferrin (TFR1) receptors. Beclin 1 promoted endosomal recruitment of hepatocyte growth factor tyrosine kinase substrate (HRS), which was necessary for sorting surface receptors to intraluminal vesicles for signal silencing and lysosomal degradation. In tumors with low Beclin 1 expression, endosomal HRS recruitment was diminished and receptor function was sustained. Collectively, our results demonstrate a novel role for Beclin 1 in impeding tumor growth by coordinating the regulation of key growth factor and nutrient receptors. These data provide an explanation for how low levels of Beclin 1 facilitate tumor proliferation and contribute to poor cancer outcomes. SIGNIFICANCE: Beclin 1 controls the trafficking fate of growth regulatory receptors to suppress tumor proliferation.
Collapse
Affiliation(s)
- Asia N Matthew-Onabanjo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts.,Medical Scientist Training Program, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jenny Janusis
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jose Mercado-Matos
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts.,Medical Scientist Training Program, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Anne E Carlisle
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Dohoon Kim
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Fayola Levine
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Peter Cruz-Gordillo
- Medical Scientist Training Program, University of Massachusetts Medical School, Worcester, Massachusetts.,Program in Systems Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Ryan Richards
- Program in Systems Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Michael J Lee
- Program in Systems Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Leslie M Shaw
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts.
| |
Collapse
|
37
|
Autophagy: A novel mechanism of chemoresistance in cancers. Biomed Pharmacother 2019; 119:109415. [DOI: 10.1016/j.biopha.2019.109415] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022] Open
|
38
|
Pu Q, Lin P, Wang Z, Gao P, Qin S, Cui L, Wu M. Interaction among inflammasome, autophagy and non-coding RNAs: new horizons for drug. PRECISION CLINICAL MEDICINE 2019; 2:166-182. [PMID: 31598387 PMCID: PMC6770284 DOI: 10.1093/pcmedi/pbz019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/22/2019] [Accepted: 08/25/2019] [Indexed: 02/07/2023] Open
Abstract
Autophagy and inflammasomes are shown to interact in various situations including
infectious disease, cancer, diabetes and neurodegeneration. Since multiple layers of
molecular regulators contribute to the interplay between autophagy and inflammasome
activation, the detail of such interplay remains largely unknown. Non-coding RNAs
(ncRNAs), which have been implicated in regulating an expanding list of cellular processes
including immune defense against pathogens and inflammatory response in cancer and
metabolic diseases, may join in the crosstalk between inflammasomes and autophagy in
physiological or disease conditions. In this review, we summarize the latest research on
the interlink among ncRNAs, inflammasomes and autophagy and discuss the emerging role of
these three in multiple signaling transduction pathways involved in clinical conditions.
By analyzing these intriguing interconnections, we hope to unveil the mechanism
inter-regulating these multiple processes and ultimately discover potential drug targets
for some refractory diseases.
Collapse
Affiliation(s)
- Qinqin Pu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Lin
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Zhihan Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Pan Gao
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shugang Qin
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Luqing Cui
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA
| |
Collapse
|
39
|
Autophagy differentially regulates macrophage lipid handling depending on the lipid substrate (oleic acid vs. acetylated-LDL) and inflammatory activation state. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:158527. [PMID: 31520777 DOI: 10.1016/j.bbalip.2019.158527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/01/2019] [Accepted: 09/08/2019] [Indexed: 12/17/2022]
Abstract
The regulation of lipid droplet (LD) dynamics by autophagy in naïve macrophages is complex: Inhibiting autophagosome initiation steps attenuates oleic acid (OA) induced LD (OA-LD) biogenesis, whereas interfering with later-autophagosome maturation/lysosomal steps accelerates OA-LD biogenesis rate, but not OA-LD degradation. Here we hypothesized that regulation of macrophage lipid handling by autophagy may be lipid-substrate and activation-state-specific. Using automated quantitative live-cell imaging, initial LD biogenesis rate was ~30% slower when the lipid source was acetylated low density lipoprotein (acLDL) compared to OA. Yet, both were similarly affected by triacsin-C, an inhibitor of acyl-CoA synthase, which inhibited, and etomoxir, an inhibitor of acylcarnitine palmitoyl transferase (fatty acid oxidation), which augmented, LD biogenesis rates. An autophagy inducing peptide, Tat-Beclin1, enhanced the degradation, and inhibited (by 37%) the biogenesis of acLDL induced LD (acLDL-LD). Yet, Tat-Beclin1 increased OA-LD biogenesis rate by 70%. When macrophages were pre-activated with LPS + INFG they exhibited increased autophagosome number and area, and reduced BECN1 and ATG14 protein levels, which associated with a markedly attenuated autophagic flux. Concomitantly, OA-LD and acLDL-LD biogenesis rates increased 3 and 7.4-fold, respectively, but could not be further modulated by Tat-Beclin1, as observed in non-activated/naïve macrophages. We propose that macrophage autophagy, and/or components of its machinery, differentially regulate LD/foam-cell biogenesis depending on the lipid-source, and that inflammatory activation uncouples autophagy from LD biogenesis.
Collapse
|
40
|
Wong M, Ganapathy AS, Suchanec E, Laidler L, Ma T, Nighot P. Intestinal epithelial tight junction barrier regulation by autophagy-related protein ATG6/beclin 1. Am J Physiol Cell Physiol 2019; 316:C753-C765. [PMID: 30892937 DOI: 10.1152/ajpcell.00246.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A defective tight junction (TJ) barrier is a key pathogenic factor for inflammatory bowel disease. Previously, we have shown that autophagy, a cell survival mechanism, enhances intestinal epithelial TJ barrier function. Autophagy-related protein-6 (ATG6/beclin 1), a key protein in the autophagy pathway, also plays a role in the endocytic pathway. The constitutive role of beclin 1 in the intestinal TJ barrier is not known. In Caco-2 cells, beclin 1 was found to be coimmunoprecipitated with the TJ protein occludin and colocalized with occludin on the membrane. Treatment of Caco-2 cells with beclin 1 peptide [transactivating regulatory protein (Tat)-beclin 1] reduced TJ barrier function. Activation of beclin 1 increased occludin endocytosis and reduced total occludin protein level. In contrast, beclin 1 siRNA transfection enhanced Caco-2 TJ barrier function. In pharmacologic and genetic autophagy inhibition studies, the constitutive function of beclin 1 in the TJ barrier was found to be autophagy independent. However, de novo induction of autophagy with starvation or rapamycin prevented Tat-beclin 1-induced increase in TJ permeability and reduction in occludin level. Induction of autophagy also resulted in reduced beclin 1-occludin association. In mouse colon, beclin 1 colocalized with occludin on the epithelial membrane. Perfusion of mouse colon with beclin 1 peptide caused an increase in colonic TJ permeability that was prevented by in vivo induction of autophagy. These findings show that beclin 1 plays a constitutive, autophagy-independent role in the regulation of intestinal TJ barrier function via endocytosis of occludin. Autophagy terminates constitutive beclin 1 function in the TJ barrier and enhances the TJ barrier.
Collapse
Affiliation(s)
- Morgan Wong
- Department of Internal Medicine, University of New Mexico School of Medicine , Albuquerque, New Mexico
| | | | - Eric Suchanec
- Department of Medicine, College of Medicine, Pennsylvania State University , Hershey, Pennsylvania
| | - Laura Laidler
- Department of Internal Medicine, University of New Mexico School of Medicine , Albuquerque, New Mexico
| | - Thomas Ma
- Department of Medicine, College of Medicine, Pennsylvania State University , Hershey, Pennsylvania
| | - Prashant Nighot
- Department of Medicine, College of Medicine, Pennsylvania State University , Hershey, Pennsylvania
| |
Collapse
|
41
|
Noguchi S, Honda S, Saitoh T, Matsumura H, Nishimura E, Akira S, Shimizu S. Beclin 1 regulates recycling endosome and is required for skin development in mice. Commun Biol 2019; 2:37. [PMID: 30701202 PMCID: PMC6347619 DOI: 10.1038/s42003-018-0279-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023] Open
Abstract
Beclin 1 is a key regulator of autophagy and endocytosis. However, its autophagy-independent functions remain poorly understood. Here, we report that Beclin 1 regulates recycling endosome and is required for skin development in vivo. We first established keratinocyte-specific Beclin 1-knockout mice and found that these mutant mice died owing to severe impairment of epidermal barrier. Beclin 1 plays a role in autophagy and the endocytic pathway in cooperation with Atg14 and UVRAG, respectively, and keratinocyte-specific Atg14-knockout mice do not show any abnormal phenotypes, suggesting that Beclin 1 has a role in skin development via the endocytic pathway. Furthermore, we found that Beclin 1 deficiency causes mislocalization of integrins via a defect of recycling endosome, abnormal cell detachment of basal cells and their immature differentiation, and abnormal skin development. These results provide the first genetic evidence showing the roles of Beclin 1 in recycling endosome and skin development.
Collapse
Affiliation(s)
- Saori Noguchi
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Shinya Honda
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Tatsuya Saitoh
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871 Japan
- Division of Inflammation Biology, Institute for Enzyme Research, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, 770-8503 Japan
| | - Hiroyuki Matsumura
- Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Emi Nishimura
- Stem Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Shizuo Akira
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka, 565-0871 Japan
| | - Shigeomi Shimizu
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| |
Collapse
|
42
|
Shi B, Ma M, Zheng Y, Pan Y, Lin X. mTOR and Beclin1: Two key autophagy-related molecules and their roles in myocardial ischemia/reperfusion injury. J Cell Physiol 2019; 234:12562-12568. [PMID: 30618070 DOI: 10.1002/jcp.28125] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022]
Abstract
Autophagy is the general term of lysosomal degradation of substances in cells, which is considered the key to maintaining the normal structure and function of the heart. It also has a correlation with several heart diseases, in particular, myocardial ischemia/reperfusion (I/R) injury. At the stage of myocardial ischemia, autophagy degrades nonfunctional cytoplasmic proteins providing the critical nutrients for the critical life activities, thereby suppressing cell apoptosis and necrosis. However, autophagy is likely to affect the heart negatively in the reperfusion stage. Mammalian target of rapamycin (mTOR) and Beclin1 are two vital autophagy-related molecules in myocardial I/R injury playing significant roles in different stages. In the ischemia stage, mTOR plays its roles through AMPK/mTOR and phosphoinositide 3-kinase/Akt/mTOR pathway, whereas Beclin1 plays its roles through its upregulation in the reperfusion stage. A possible interaction between mTOR and Beclin1 has been reported recently, and further studies need to be done to find the underlying interaction between the two molecules in myocardial I/R injury.
Collapse
Affiliation(s)
- Binhao Shi
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Clinical Medical, Anhui Medical University, Hefei, China
| | - Mengqing Ma
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yitian Zheng
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Clinical Medical, Anhui Medical University, Hefei, China
| | - Yanyan Pan
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Clinical Medical, Anhui Medical University, Hefei, China
| | - Xianhe Lin
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
43
|
Zheng K, He Z, Kitazato K, Wang Y. Selective Autophagy Regulates Cell Cycle in Cancer Therapy. Theranostics 2019; 9:104-125. [PMID: 30662557 PMCID: PMC6332805 DOI: 10.7150/thno.30308] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 10/30/2018] [Indexed: 12/21/2022] Open
Abstract
Aberrant function of cell cycle regulators results in uncontrolled cell proliferation, making them attractive therapeutic targets in cancer treatment. Indeed, survival of many cancers exclusively relies on these proteins, and several specific inhibitors are in clinical use. Although the ubiquitin-proteasome system is responsible for the periodic quality control of cell cycle proteins during cell cycle progression, increasing evidence clearly demonstrates the intimate interaction between cell cycle regulation and selective autophagy, important homeostasis maintenance machinery. However, these studies have often led to divergent rather than unifying explanations due to complexity of the autophagy signaling network, the inconsistent functions between general autophagy and selective autophagy, and the different characteristics of autophagic substrates. In this review, we highlight current data illustrating the contradictory and important role of cell cycle proteins in regulating autophagy. We also focus on how selective autophagy acts as a central mechanism to maintain orderly DNA repair and genome integrity by degrading specific cell cycle proteins, regulating cell division, and promoting DNA damage repair. We further discuss the ways in which selective autophagy may impact the cell cycle regulators, since failure to appropriately remove these can interfere with cell death-related processes, including senescence and autophagy-related cell death. Imbalanced cell proliferation is typically utilized by cancer cells to acquire resistance. Finally, we discuss the possibility of a potent anticancer therapeutic strategy that targets selective autophagy or autophagy and cell cycle together.
Collapse
|
44
|
Hu A, Li X, He J, Gong X, Wu Z, Ning P. Classical swine fever virus-Shimen infection upregulates SH3GLB1 expression in porcine alveolar macrophages. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2018.1552839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Aoxue Hu
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, PR China
| | - Xuepeng Li
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, PR China
| | - Jun He
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, PR China
| | - Xiaocheng Gong
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, PR China
| | - Zhongxing Wu
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, PR China
| | - Pengbo Ning
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, PR China
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi’an, Shaanxi, PR China
| |
Collapse
|
45
|
Yang C, Wu S, Wu X, Zhou X, Jin S, Jiang H. Silencing circular RNA UVRAG inhibits bladder cancer growth and metastasis by targeting the microRNA-223/fibroblast growth factor receptor 2 axis. Cancer Sci 2018; 110:99-106. [PMID: 30387298 PMCID: PMC6317955 DOI: 10.1111/cas.13857] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 12/19/2022] Open
Abstract
Circular RNA UVRAG (circUVRAG), a type of non-coding RNA, is derived and cyclized by part of the exon from the UVRAG gene. However, the role of circUVRAG in bladder cancer (BLCA) has not been reported. The purpose of the present study was therefore to characterize the role of circUVRAG in BLCA. Bioinformatics analysis showed interactive relationships among circUVRAG, microRNA-223 (miR-223), and fibroblast growth factor receptor 2 (FGFR2). Quantitative real-time PCR was used to detect the expression of circUVRAG in BLCA cell lines. UM-UC-3 cells were stably transfected with siRNA against circUVRAG, and cell proliferation and migration ability were tested using the CCK8 assay, clone formation, and Transwell assays in vitro. Tumor xenograft formation and metastasis were determined using nude mice. Fluorescence in situ hybridization was used to confirm the subcellular localization of circUVRAG, and the luciferase reporter assay was used to confirm the relationships among circUVRAG, miR-223, and FGFR2. Results showed that circUVRAG was upregulated in BLCA cell lines. Downregulation of circUVRAG expression suppressed proliferation and metastasis both in vitro and in vivo. Downregulation of circUVRAG suppressed FGFR2 expression by "sponging" miR-223, which was confirmed by rescue experiments and luciferase reporter assay. Overall, the results showed that downregulation of circUVRAG suppressed the aggressive biological phenotype of BLCA. Taken together, silencing circular RNA UVRAG inhibited bladder cancer growth and metastasis by targeting the miR-223/FGFR2 axis, which may provide a potential biomarker and therapeutic target for the management of BLCA.
Collapse
Affiliation(s)
- Chen Yang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Siqi Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaobo Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xuejian Zhou
- Department of Urology, The Fifth People's Hospital of Shanghai, Shanghai, China
| | - Shengming Jin
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
46
|
Grimaldi M, Bo VD, Ferrari B, Roda E, De Luca F, Veneroni P, Barni S, Verri M, De Pascali SA, Fanizzi FP, Bernocchi G, Bottone MG. Long-term effects after treatment with platinum compounds, cisplatin and [Pt(O,O'-acac)(γ-acac)(DMS)]: Autophagy activation in rat B50 neuroblastoma cells. Toxicol Appl Pharmacol 2018; 364:1-11. [PMID: 30529627 DOI: 10.1016/j.taap.2018.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/26/2018] [Accepted: 12/05/2018] [Indexed: 01/01/2023]
Abstract
Cisplatin (cisPt), among the best known components of multi-drug front-line therapies used for the treatments of solid tumors, such as the childhood neuroblastoma, acts through DNA linking. Nevertheless, the cisPt effectiveness is compromised by the onset of severe side effects, including neurotoxicity that results in neurodegeneration, cell death, and drug-resistance. In the field of experimental oncology, aimed at overcoming cytotoxicity and chemoresistance, great efforts are devoted to the synthesis of new platinum-based drugs, such as [Pt(O,O'-acac)(γ-acac)(DMS)] (PtAcacDMS), which shows a specific reactivity with sulfur residues of enzymes involved in apoptosis. Autophagy, an evolutionary conserved degradation pathway for recycling of cytoplasmic components, represents one of the mechanisms adopted by cancer cells which contribute to drug-resistance. In the present study, standard acute (48 h-exposure) and long-term effects (7 day-recovery after treatment or 7 day-recovery followed by reseeding and 96 h-growth), of cisPt and PtAcacDMS (40 and 10 μM, respectively) were investigated in vitro employing rat B50 neuroblastoma as a cancer model. Using fluorescence and electron microscopy, as well as biochemical techniques, our data highlight a key role of the autophagic process in B50 cells. Specifically, long-term effects caused by cisPt lead to inhibition of the apoptotic process and paralleled by the activation of autophagy, thus evidencing that autophagy has a protective role after cisPt exposure, allowing cells to survive. Whereas, long-term effects produced by PtAcacDMS lead toward both apoptosis and autophagy activation. In conclusion, autophagy may represents an alternative cell death pathway, circumventing drug-resistance strategies employed by cancer cells to survive chemoterapy.
Collapse
Affiliation(s)
- Maddalena Grimaldi
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Italy
| | - Veronica Dal Bo
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Italy
| | - Beatrice Ferrari
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Italy
| | - Elisa Roda
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Italy; Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, ICS Maugeri Spa, IRCCS Pavia, Via Maugeri 10, Pavia, Italy
| | - Fabrizio De Luca
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Italy
| | - Paola Veneroni
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Italy
| | - Sergio Barni
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Italy
| | - Manuela Verri
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Italy
| | - Sandra A De Pascali
- General and Inorganic Chemistry Laboratory, Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Lecce, Italy
| | - Francesco P Fanizzi
- General and Inorganic Chemistry Laboratory, Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Lecce, Italy
| | - Graziella Bernocchi
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Italy
| | - Maria G Bottone
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Italy.
| |
Collapse
|
47
|
Ohashi Y, Tremel S, Williams RL. VPS34 complexes from a structural perspective. J Lipid Res 2018; 60:229-241. [PMID: 30397185 PMCID: PMC6358306 DOI: 10.1194/jlr.r089490] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/24/2018] [Indexed: 02/07/2023] Open
Abstract
VPS34 phosphorylates phosphatidylinositol to produce PtdIns3P and is the progenitor of the phosphoinositide 3-kinase (PI3K) family. VPS34 has a simpler domain organization than class I PI3Ks, which belies the complexity of its quaternary organization, with the enzyme always functioning within larger assemblies. PtdIns3P recruits specific recognition modules that are common in protein-sorting pathways, such as autophagy and endocytic sorting. It is best characterized in two heterotetramers, complexes I and II. Complex I is composed of VPS34, VPS15, Beclin 1, and autophagy-related gene (ATG)14L, whereas complex II replaces ATG14L with UVRAG. Because VPS34 can form a component of several distinct complexes, it enables independent regulation of various pathways that are controlled by PtdIns3P. Complexes I and II are critical for early events in autophagy and endocytic sorting, respectively. Autophagy has a complex association with cancer. In early stages, it inhibits tumorigenesis, but in later stages, it acts as a survival factor for tumors. Recently, various disease-associated somatic mutations were found in genes encoding complex I and II subunits. Lipid kinase activities of the complexes are also influenced by posttranslational modifications (PTMs). Mapping PTMs and somatic mutations on three-dimensional models of the complexes suggests mechanisms for how these affect VPS34 activity.
Collapse
Affiliation(s)
- Yohei Ohashi
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Shirley Tremel
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Roger L Williams
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| |
Collapse
|
48
|
Menon MB, Dhamija S. Beclin 1 Phosphorylation - at the Center of Autophagy Regulation. Front Cell Dev Biol 2018; 6:137. [PMID: 30370269 PMCID: PMC6194997 DOI: 10.3389/fcell.2018.00137] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/26/2018] [Indexed: 01/07/2023] Open
Abstract
Autophagy is a tightly regulated catabolic process wherein cells under stress sequester cytosolic constituents like damaged proteins and organelles in double-membrane vesicles called autophagosomes. The autophagosomes degrade their cargo by lysosomal proteolysis generating raw materials for the biosynthesis of vital macromolecules. One of the initial steps in the assembly of autophagosomes from pre-autophagic structures is the recruitment and activation of the class III phosphatidylinositol 3-kinase complex consisting of Beclin 1 (BECN1), VPS34, VPS15, and ATG14 proteins. Several pieces of evidence indicate that the phosphorylation and ubiquitination of BECN1 at an array of residues fine-tune the responses to diverse autophagy modulating stimuli and helps in maintaining the balance between pro-survival autophagy and pro-apoptotic responses. In this mini-review, we will discuss the importance of distinct BECN1 phosphorylation events, the diverse signaling pathways and kinases involved and their role in the regulation of autophagy.
Collapse
Affiliation(s)
- Manoj B. Menon
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany,*Correspondence: Manoj B. Menon,
| | - Sonam Dhamija
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center – University of Freiburg, Freiburg, Germany,Division of RNA Biology and Cancer, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
49
|
Vantaggiato C, Panzeri E, Castelli M, Citterio A, Arnoldi A, Santorelli FM, Liguori R, Scarlato M, Musumeci O, Toscano A, Clementi E, Bassi MT. ZFYVE26/SPASTIZIN and SPG11/SPATACSIN mutations in hereditary spastic paraplegia types AR-SPG15 and AR-SPG11 have different effects on autophagy and endocytosis. Autophagy 2018; 15:34-57. [PMID: 30081747 DOI: 10.1080/15548627.2018.1507438] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
ZFYVE26/Spastizin and SPG11/Spatacsin encode 2 large proteins that are mutated in hereditary autosomal-recessive spastic paraplegia/paraparesis (HSP) type 15 (AR-SPG15) and type 11 (AR-SPG11), respectively. We previously have reported that AR-SPG15-related ZFYVE26 mutations lead to autophagy defects with accumulation of immature autophagosomes. ZFYVE26 and SPG11 were found to be part of a complex including the AP5 (adaptor related protein complex 5) and to have a critical role in autophagic lysosomal reformation with identification of autophagic and lysosomal defects in cells with both AR-SPG15- and AR-SPG11-related mutations. In spite of these similarities between the 2 proteins, here we report that ZFYVE26 and SPG11 are differently involved in autophagy and endocytosis. We found that both ZFYVE26 and SPG11 interact with RAB5A and RAB11, 2 proteins regulating endosome trafficking and maturation, but only ZFYVE26 mutations affected RAB protein interactions and activation. ZFYVE26 mutations lead to defects in the fusion between autophagosomes and endosomes, while SPG11 mutations do not affect this step and lead to a milder autophagy defect. We thus demonstrate that ZFYVE26 and SPG11 affect the same cellular physiological processes, albeit at different levels: both proteins have a role in autophagic lysosome reformation, but only ZFYVE26 acts at the intersection between endocytosis and autophagy, thus representing a key player in these 2 processes. Indeed expression of the constitutively active form of RAB5A in cells with AR-SPG15-related mutations partially rescues the autophagy defect. Finally the model we propose demonstrates that autophagy and the endolysosomal pathway are central processes in the pathogenesis of these complicated forms of hereditary spastic paraparesis. Abbreviations: ALR, autophagic lysosome reformation; AP5, adaptor related protein complex 5; AR, autosomal-recessive; HSP, hereditary spastic paraplegia/paraparesis; ATG14, autophagy related 14; BafA, bafilomycin A1; BECN1, beclin 1; EBSS, Earle balanced salt solution; EEA1, early endosome antigen 1; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; GDP, guanosine diphosphate; GFP, green fluorescent protein; GTP, guanosine triphosphate; HSP, hereditary spastic paraplegias; LBPA, lysobisphosphatidic acid; MAP1LC3B/LC3B, microtubule associated protein 1 light chain 3 beta; MVBs, multivesicular bodies; PIK3C3, phosphatidylinositol 3-kinase, catalytic subunit type 3; PIK3R4, phosphoinositide-3-kinase regulatory subunit 4; PtdIns3P, phosphatidylinositol-3-phosphate; RFP, red fluorescent protein; RUBCN, RUN and cysteine rich domain containing beclin 1 interacting protein; shRNA, short hairpin RNA; SQSTM1/p62, sequestosome 1; TCC: thin corpus callosum; TF, transferrin; UVRAG, UV radiation resistance associated.
Collapse
Affiliation(s)
- Chiara Vantaggiato
- a Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology , Bosisio Parini , Lecco , Italy
| | - Elena Panzeri
- a Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology , Bosisio Parini , Lecco , Italy
| | - Marianna Castelli
- a Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology , Bosisio Parini , Lecco , Italy
| | - Andrea Citterio
- a Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology , Bosisio Parini , Lecco , Italy
| | - Alessia Arnoldi
- a Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology , Bosisio Parini , Lecco , Italy
| | | | - Rocco Liguori
- c Department of Biomedical and Neuromotor Sciences , University of Bologna; IRCCS Institute of Neurological Sciences , Bologna , Italy
| | - Marina Scarlato
- d Dept. of Neurosciences and Institute of Experimental Neurology (INSpe) , San Raffaele Scientific Institute , Milan , Italy
| | - Olimpia Musumeci
- e Department of Clinical and Experimental Medicine , University of Messina , Messina , Italy
| | - Antonio Toscano
- e Department of Clinical and Experimental Medicine , University of Messina , Messina , Italy
| | - Emilio Clementi
- a Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology , Bosisio Parini , Lecco , Italy.,f Unit of Clinical Pharmacology, CNR Institute of Neuroscience, Department of Biomedical and Clinical Sciences , University Hospital "Luigi Sacco", Università di Milano , Milan , Italy
| | - Maria Teresa Bassi
- a Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology , Bosisio Parini , Lecco , Italy
| |
Collapse
|
50
|
Soukup SF, Vanhauwaert R, Verstreken P. Parkinson's disease: convergence on synaptic homeostasis. EMBO J 2018; 37:embj.201898960. [PMID: 30065071 DOI: 10.15252/embj.201898960] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/07/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease, the second most common neurodegenerative disorder, affects millions of people globally. There is no cure, and its prevalence will double by 2030. In recent years, numerous causative genes and risk factors for Parkinson's disease have been identified and more than half appear to function at the synapse. Subtle synaptic defects are thought to precede blunt neuronal death, but the mechanisms that are dysfunctional at synapses are only now being unraveled. Here, we review recent work and propose a model where different Parkinson proteins interact in a cell compartment-specific manner at the synapse where these proteins regulate endocytosis and autophagy. While this field is only recently emerging, the work suggests that the loss of synaptic homeostasis may contribute to neurodegeneration and is a key player in Parkinson's disease.
Collapse
Affiliation(s)
- Sandra-Fausia Soukup
- VIB-KU Leuven Center for Brain& Disease Research, Leuven, Belgium .,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Roeland Vanhauwaert
- VIB-KU Leuven Center for Brain& Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Patrik Verstreken
- VIB-KU Leuven Center for Brain& Disease Research, Leuven, Belgium .,Department of Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|