1
|
Niu X, You Q, Hou K, Tian Y, Wei P, Zhu Y, Gao B, Ashrafizadeh M, Aref AR, Kalbasi A, Cañadas I, Sethi G, Tergaonkar V, Wang L, Lin Y, Kang D, Klionsky DJ. Autophagy in cancer development, immune evasion, and drug resistance. Drug Resist Updat 2025; 78:101170. [PMID: 39603146 DOI: 10.1016/j.drup.2024.101170] [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: 07/16/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Macroautophagy/autophagy is a highly conserved evolutionary mechanism involving lysosomes for the degradation of cytoplasmic components including organelles. The constitutive, basal level of autophagy is fundamental for preserving cellular homeostasis; however, alterations in autophagy can cause disease pathogenesis, including cancer. The role of autophagy in cancer is particularly complicated, since this process acts both as a tumor suppressor in precancerous stages but facilitates tumor progression during carcinogenesis and later stages of cancer progression. This shift between anti-tumor and pro-tumor roles may be influenced by genetic and environmental factors modulating key pathways such as those involving autophagy-related proteins, the PI3K-AKT-MTOR axis, and AMPK, which often show dysregulation in tumors. Autophagy regulates various cellular functions, including metabolism of glucose, glutamine, and lipids, cell proliferation, metastasis, and several types of cell death (apoptosis, ferroptosis, necroptosis and immunogenic cell death). These multifaceted roles demonstrate the potential of autophagy to affect DNA damage repair, cell death pathways, proliferation and survival, which are critical in determining cancer cells' response to chemotherapy. Therefore, targeting autophagy pathways presents a promising strategy to combat chemoresistance, as one of the major reasons for the failure in cancer patient treatment. Furthermore, autophagy modulates immune evasion and the function of immune cells such as T cells and dendritic cells, influencing the tumor microenvironment and cancer's biological behavior. However, the therapeutic targeting of autophagy is complex due to its dual role in promoting survival and inducing cell death in cancer cells, highlighting the need for strategies that consider both the beneficial and detrimental effects of autophagy modulation in cancer therapy. Hence, both inducers and inhibitors of autophagy have been introduced for the treatment of cancer. This review emphasizes the intricate interplay between autophagy, tumor biology, and immune responses, offering insights into potential therapeutic approaches that deploy autophagy in the cancer suppression.
Collapse
Affiliation(s)
- Xuegang Niu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Qi You
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Kaijian Hou
- School of Public Health(Long Hu people hospital), Shantou University, Shantou, 515000, Guangdong, China
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, IL 60532, USA
| | - Penghui Wei
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Bin Gao
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Amir Reza Aref
- VitroVision Department, DeepkinetiX, Inc, Boston, MA, USA
| | - Alireza Kalbasi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Israel Cañadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A⁎STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Lingzhi Wang
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Yuanxiang Lin
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Dezhi Kang
- Department of Neurosurgery, Neurosurgery Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Pan C, Zhao H, Cai X, Wu M, Qin B, Li J. The connection between autophagy and ferroptosis in AKI: recent advances regarding selective autophagy. Ren Fail 2024; 46:2379601. [PMID: 39099238 DOI: 10.1080/0886022x.2024.2379601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
Acute kidney injury (AKI) is a significant issue in public health, displaying a high occurrence rate and mortality rate. Ferroptosis, a form of programmed cell death (PCD), is characterized by iron accumulation and intensified lipid peroxidation. Recent studies have demonstrated the pivotal significance of ferroptosis in AKI caused by diverse stimuli, including ischemia-reperfusion injury (IRI), sepsis and toxins. Autophagy, a multistep process that targets damaged organelles and macromolecules for degradation and recycling, also plays an essential role in AKI. Previous research has demonstrated that autophagy deletion in proximal tubules could aggravate tubular injury and renal function loss, indicating the protective function of autophagy in AKI. Consequently, finding ways to stimulate autophagy has become a crucial therapeutic strategy. The recent discovery of the role of selective autophagy in influencing ferroptosis has identified new therapeutic targets for AKI and has highlighted the importance of understanding the cross-talk between autophagy and ferroptosis. This study aims to provide an overview of the signaling pathways involved in ferroptosis and autophagy, focusing on the mechanisms and functions of selective autophagy and autophagy-dependent ferroptosis. We hope to establish a foundation for future investigations into the interaction between autophagy and ferroptosis in AKI as well as other diseases.
Collapse
Affiliation(s)
- Chunyu Pan
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hairui Zhao
- Department of Nephrology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaojing Cai
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manyi Wu
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bowen Qin
- Department of Nephrology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Junhua Li
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Nephrology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Yazaki E, Uehara T, Sakamoto H, Inagaki Y. Dinotoms possess two evolutionary distinct autophagy-related ubiquitin-like conjugation systems. Protist 2024; 175:126067. [PMID: 39341116 DOI: 10.1016/j.protis.2024.126067] [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: 03/11/2024] [Revised: 08/29/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024]
Abstract
Autophagy is an intracellular degradation mechanism by which cytoplasmic materials are delivered to and degraded in the lysosome-fused autophagosome (autolysosome) and proposed to have been established at an early stage of eukaryotic evolution. Dinoflagellates harboring endosymbiotic diatoms (so-called "dinotoms"), which retain their own nuclei and mitochondria in addition to plastids, have been investigated as an intermediate toward the full integration of a eukaryotic phototroph into the host-controlled organelle (i.e., plastid) through endosymbiosis. Pioneering studies systematically evaluated the degree of host governance on several metabolic pathways in the endosymbiotic diatoms (ESDs). However, little attention has been paid to the impact of the endosymbiotic lifestyle on the autophagy operated in the ESDs. In this study, we searched for ATG3, ATG4, ATG5, ATG7, ATG8, ATG10, and ATG12, which are required for autophagosome formation, in the RNA-seq data from dinotoms Durinskia baltica and Kryptoperidinium foliaceum. We detected two evolutionally distinct sets of the ATG proteins in the dinotom species, one affiliated with the dinoflagellate homologs and the other with the diatom homologs in phylogenetic analyses. The results suggest that the ATG proteins descended from the diatom taken up by the dinoflagellate host persist for autophagosome formation and, most likely, autophagy.
Collapse
Affiliation(s)
- Euki Yazaki
- Research Center for Advanced Analysis, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan; RIKEN iTHEMS, Wako, Saitama, Japan.
| | - Tadaaki Uehara
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hirokazu Sakamoto
- Department of Infection and Host Defense, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Yuji Inagaki
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan; Center for Computational Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
4
|
Ke PY, Yeh CT. Functional Role of Hepatitis C Virus NS5A in the Regulation of Autophagy. Pathogens 2024; 13:980. [PMID: 39599533 PMCID: PMC11597459 DOI: 10.3390/pathogens13110980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Many types of RNA viruses, including the hepatitis C virus (HCV), activate autophagy in infected cells to promote viral growth and counteract the host defense response. Autophagy acts as a catabolic pathway in which unnecessary materials are removed via the lysosome, thus maintaining cellular homeostasis. The HCV non-structural 5A (NS5A) protein is a phosphoprotein required for viral RNA replication, virion assembly, and the determination of interferon (IFN) sensitivity. Recently, increasing evidence has shown that HCV NS5A can induce autophagy to promote mitochondrial turnover and the degradation of hepatocyte nuclear factor 1 alpha (HNF-1α) and diacylglycerol acyltransferase 1 (DGAT1). In this review, we summarize recent progress in understanding the detailed mechanism by which HCV NS5A triggers autophagy, and outline the physiological significance of the balance between host-virus interactions.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry and Molecular Biology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| |
Collapse
|
5
|
Warwick AM, Bomze HM, Wang L, Hao Y, Stinnett SS, Gospe SM. Hypoxia-mediated rescue of retinal ganglion cells deficient in mitochondrial complex I is independent of the hypoxia-inducible factor pathway. Sci Rep 2024; 14:24114. [PMID: 39406814 PMCID: PMC11480089 DOI: 10.1038/s41598-024-75916-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Continuous exposure to environmental hypoxia (11% O2) has been shown to markedly slow the progressive degeneration of retinal ganglion cells (RGCs) in a mouse model of mitochondrial optic neuropathy with RGC-specific deletion of the key mitochondrial complex I accessory subunit ndufs4. As a first step toward identifying the therapeutic mechanism of hypoxia in this model, we conducted a series of experiments to investigate the role of the hypoxia-inducible factor (HIF) regulatory pathway in RGC neuroprotection. Vglut2-Cre; ndufs4loxP/loxP mice were crossed with strains bearing floxed alleles of the negative HIF regulatory vhl or of the two major HIF α-subunit isoforms, Hif1α and Hif2α. Deletion of vhl within ndufs4-deficient RGCs failed to prevent RGC degeneration under normoxia, indicating that HIF activation is not sufficient to achieve RGC rescue. Furthermore, the rescue of ndufs4-deficient RGCs by hypoxia remained robust despite genetic inactivation of Hif1α and Hif2α. Our findings demonstrate that the HIF pathway is entirely dispensable to the rescue of RGCs by hypoxia. Future efforts to uncover key HIF-independent molecular pathways induced by hypoxia in this mouse model may be of therapeutic relevance to mitochondrial optic neuropathies such as Leber hereditary optic neuropathy.
Collapse
Affiliation(s)
- Alexander M Warwick
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Howard M Bomze
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Luyu Wang
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Ying Hao
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sandra S Stinnett
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sidney M Gospe
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Ophthalmology, Box 3712 Med Center, Duke University, 2351 Erwin Road, Durham, NC, 27710, USA.
| |
Collapse
|
6
|
Wei XH, Liao LY, Yin YX, Xu Q, Xie SS, Liu M, Gao LB, Chen HQ, Zhou R. Overexpression of long noncoding RNA DUXAP8 inhibits ER-phagy through activating AKT/mTOR signaling and contributes to preeclampsia. Cell Mol Life Sci 2024; 81:336. [PMID: 39120751 PMCID: PMC11335266 DOI: 10.1007/s00018-024-05385-y] [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: 04/05/2024] [Revised: 07/09/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Preeclampsia (PE) is a life-threatening pregnancy-specific complication with controversial mechanisms and no effective treatment except delivery is available. Currently, increasing researchers suggested that PE shares pathophysiologic features with protein misfolding/aggregation disorders, such as Alzheimer disease (AD). Evidences have proposed defective autophagy as a potential source of protein aggregation in PE. Endoplasmic reticulum-selective autophagy (ER-phagy) plays a critical role in clearing misfolded proteins and maintaining ER homeostasis. However, its roles in the molecular pathology of PE remain unclear. We found that lncRNA DUXAP8 was upregulated in preeclamptic placentae and significantly correlated with clinical indicators. DUXAP8 specifically binds to PCBP2 and inhibits its ubiquitination-mediated degradation, and decreased levels of PCBP2 reversed the activation effect of DUXAP8 overexpression on AKT/mTOR signaling pathway. Function experiments showed that DUXAP8 overexpression inhibited trophoblastic proliferation, migration, and invasion of HTR-8/SVneo and JAR cells. Moreover, pathological accumulation of swollen and lytic ER (endoplasmic reticulum) was observed in DUXAP8-overexpressed HTR8/SVneo cells and PE placental villus trophoblast cells, which suggesting that ER clearance ability is impaired. Further studies found that DUXAP8 overexpression impaired ER-phagy and caused protein aggregation medicated by reduced FAM134B and LC3II expression (key proteins involved in ER-phagy) via activating AKT/mTOR signaling pathway. The increased level of FAM134B significantly reversed the inhibitory effect of DUXAP8 overexpression on the proliferation, migration, and invasion of trophoblasts. In vivo, DUXAP8 overexpression through tail vein injection of adenovirus induced PE-like phenotypes in pregnant rats accompanied with activated AKT/mTOR signaling, decreased expression of FAM134B and LC3-II proteins and increased protein aggregation in placental tissues. Our study reveals the important role of lncRNA DUXAP8 in regulating trophoblast biological behaviors through FAM134B-mediated ER-phagy, providing a new theoretical basis for understanding the pathogenesis of PE.
Collapse
Affiliation(s)
- Xiao-Hong Wei
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Ling-Yun Liao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Yang-Xue Yin
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Qin Xu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Shuang-Shuang Xie
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Min Liu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Lin-Bo Gao
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Hong-Qin Chen
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) , Ministry of Education, West China Second University Hospital, Sichuan University, NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, P.R. China.
| |
Collapse
|
7
|
Yamada K, St Croix C, Stolz DB, Tyurina YY, Tyurin VA, Bradley LR, Kapralov AA, Deng Y, Zhou X, Wei Q, Liao B, Fukuda N, Sullivan M, Trudeau J, Ray A, Kagan VE, Zhao J, Wenzel SE. Compartmentalized mitochondrial ferroptosis converges with optineurin-mediated mitophagy to impact airway epithelial cell phenotypes and asthma outcomes. Nat Commun 2024; 15:5818. [PMID: 38987265 PMCID: PMC11237105 DOI: 10.1038/s41467-024-50222-2] [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: 07/11/2023] [Accepted: 07/03/2024] [Indexed: 07/12/2024] Open
Abstract
A stable mitochondrial pool is crucial for healthy cell function and survival. Altered redox biology can adversely affect mitochondria through induction of a variety of cell death and survival pathways, yet the understanding of mitochondria and their dysfunction in primary human cells and in specific disease states, including asthma, is modest. Ferroptosis is traditionally considered an iron dependent, hydroperoxy-phospholipid executed process, which induces cytosolic and mitochondrial damage to drive programmed cell death. However, in this report we identify a lipoxygenase orchestrated, compartmentally-targeted ferroptosis-associated peroxidation process which occurs in a subpopulation of dysfunctional mitochondria, without promoting cell death. Rather, this mitochondrial peroxidation process tightly couples with PTEN-induced kinase (PINK)-1(PINK1)-Parkin-Optineurin mediated mitophagy in an effort to preserve the pool of functional mitochondria and prevent cell death. These combined peroxidation processes lead to altered epithelial cell phenotypes and loss of ciliated cells which associate with worsened asthma severity. Ferroptosis-targeted interventions of this process could preserve healthy mitochondria, reverse cell phenotypic changes and improve disease outcomes.
Collapse
Affiliation(s)
- Kazuhiro Yamada
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka, 545-8585, Japan
| | - Claudette St Croix
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Donna B Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Yulia Y Tyurina
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Vladimir A Tyurin
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Laura R Bradley
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Alexander A Kapralov
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Yanhan Deng
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiuxia Zhou
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Qi Wei
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Bo Liao
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Otolaryngology-Head & Neck Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Nobuhiko Fukuda
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Mara Sullivan
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - John Trudeau
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Anuradha Ray
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Jinming Zhao
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| | - Sally E Wenzel
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
8
|
Liu J, Li H, Dong Q, Liang Z. Multi omics analysis of mitophagy subtypes and integration of machine learning for predicting immunotherapy responses in head and neck squamous cell carcinoma. Aging (Albany NY) 2024; 16:10579-10614. [PMID: 38913914 PMCID: PMC11236326 DOI: 10.18632/aging.205964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/29/2024] [Indexed: 06/26/2024]
Abstract
Mitophagy serves as a critical mechanism for tumor cell death, significantly impacting the progression of tumors and their treatment approaches. There are significant challenges in treating patients with head and neck squamous cell carcinoma, underscoring the importance of identifying new targets for therapy. The function of mitophagy in head and neck squamous carcinoma remains uncertain, thus investigating its impact on patient outcomes and immunotherapeutic responses is especially crucial. We initially analyzed the differential expression, prognostic value, intergene correlations, copy number variations, and mutation frequencies of mitophagy-related genes at the pan-cancer level. Through unsupervised clustering, we divided head and neck squamous carcinoma into three subtypes with distinct prognoses, identified the signaling pathway features of each subtype using ssGSEA, and characterized subtype B as having features of an immune desert using various immune infiltration calculation methods. Using multi-omics data, we identified the genomic variation characteristics, mutated gene pathway features, and drug sensitivity features of the mitophagy subtypes. Utilizing a combination of 10 machine learning algorithms, we have developed a prognostic scoring model called Mitophagy Subgroup Risk Score (MSRS), which is used to predict patient survival and the response to immune checkpoint blockade therapy. Simultaneously, we applied MSRS to single-cell analysis to explore intercellular communication. Through laboratory experiments, we validated the biological function of SLC26A9, one of the genes in the risk model. In summary, we have explored the significant role of mitophagy in head and neck tumors through multi-omics data, providing new directions for clinical treatment.
Collapse
Affiliation(s)
- Junzhi Liu
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Huimin Li
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qiuping Dong
- Laboratory of Cancer Cell Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zheng Liang
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
9
|
Lacy SM, Taubitz RJ, Urban ND, Turowski SN, Smith ED, Helms AS, Michele DE, Truttmann MC. FICD deficiency protects mice from hypertrophy-induced heart failure via BiP-mediated activation of the UPR ER and ER-phagy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596287. [PMID: 38853840 PMCID: PMC11160590 DOI: 10.1101/2024.05.28.596287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Cardiomyocytes require the HSP70 chaperone BiP to maintain proteostasis in the endoplasmic reticulum (ER) following cardiac stress. The adenylyl transferase (AMPylase) FICD is increasingly recognized to regulate BiP activity through the post-translational addition of an adenosine monophosphate moiety to BiP surface residues. However, the physiological impact of FICD-mediated BiP regulation in the context of cardiovascular health is unknown. Here, we find that FICD deficiency prevents pressure overload-associated heart failure, hypertrophy, and fibrosis, and that FICD knockout mice maintain normal cardiac function after cardiac pressure overload. At a cellular level, we observe that FICD-mediated BiP AMPylation blunts the induction of the unfolded protein response (UPR ER ) and impairs BiP interaction with FAM134B, an ER-phagy receptor, thus limiting ER-phagy induction under stress. In contrast, FICD loss significantly increases BiP-dependent UPR ER induction and ER-phagy in stressed cardiomyocytes. We also uncover cell type-specific consequences of FICD activity in response to ER stress, positioning FICD as a critical proteostasis regulator in cardiac tissue. Our results highlight a novel regulatory paradigm controlling stress resilience in cardiomyocytes and offer a rationale to consider FICD as a therapeutic target to treat cardiac hypertrophy.
Collapse
|
10
|
Zhu Y, Hui Q, Zhang Z, Fu H, Qin Y, Zhao Q, Li Q, Zhang J, Guo L, He W, Han C. Advancements in the study of synaptic plasticity and mitochondrial autophagy relationship. J Neurosci Res 2024; 102:e25309. [PMID: 38400573 DOI: 10.1002/jnr.25309] [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: 10/09/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
Synapses serve as the points of communication between neurons, consisting primarily of three components: the presynaptic membrane, synaptic cleft, and postsynaptic membrane. They transmit signals through the release and reception of neurotransmitters. Synaptic plasticity, the ability of synapses to undergo structural and functional changes, is influenced by proteins such as growth-associated proteins, synaptic vesicle proteins, postsynaptic density proteins, and neurotrophic growth factors. Furthermore, maintaining synaptic plasticity consumes more than half of the brain's energy, with a significant portion of this energy originating from ATP generated through mitochondrial energy metabolism. Consequently, the quantity, distribution, transport, and function of mitochondria impact the stability of brain energy metabolism, thereby participating in the regulation of fundamental processes in synaptic plasticity, including neuronal differentiation, neurite outgrowth, synapse formation, and neurotransmitter release. This article provides a comprehensive overview of the proteins associated with presynaptic plasticity, postsynaptic plasticity, and common factors between the two, as well as the relationship between mitochondrial energy metabolism and synaptic plasticity.
Collapse
Affiliation(s)
- Yousong Zhu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinlong Hui
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Zheng Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Hao Fu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Yali Qin
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qiong Zhao
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinqing Li
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Junlong Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Lei Guo
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Wenbin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Cheng Han
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
11
|
Liang X, Wang Y, Li S, Fan J, Zhou F, Li X, Li S, Li Y. Enhancing Rab7 Activity by Inhibiting TBC1D5 Expression Improves Mitophagy in Alzheimer's Disease Models. J Alzheimers Dis 2024; 100:279-296. [PMID: 38848175 DOI: 10.3233/jad-231300] [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: 06/09/2024]
Abstract
Background Mitochondrial dysfunction exists in Alzheimer's disease (AD) brain, and damaged mitochondria need to be removed by mitophagy. Small GTPase Rab7 regulates the fusion of mitochondria and lysosome, while TBC1D5 inhibits Rab7 activation. However, it is not clear whether the regulation of Rab7 activity by TBC1D5 can improve mitophagy and inhibit AD progression. Objective To investigate the role of TBC1D5 in mitophagy and its regulatory mechanism for Rab7, and whether activation of mitophagy can inhibit the progression of AD. Methods Mitophagy was determined by western blot and immunofluorescence. The morphology and quantity of mitochondria were tracked by TEM. pCMV-Mito-AT1.03 was employed to detect the cellular ATP. Amyloid-β secreted by AD cells was detected by ELISA. Co-immunoprecipitation was used to investigate the binding partner of the target protein. Golgi-cox staining was applied to observe neuronal morphology of mice. The Morris water maze test and Y-maze were performed to assess spatial learning and memory, and the open field test was measured to evaluate motor function and anxiety-like phenotype of experimental animals. Results Mitochondrial morphology was impaired in AD models, and TBC1D5 was highly expressed. Knocking down TBC1D5 increased the expression of active Rab7, promoted the fusion of lysosome and autophagosome, thus improving mitophagy, and improved the morphology of hippocampal neurons and the impaired behavior in AD mice. Conclusions Knocking down TBC1D5 increased Rab7 activity and promoted the fusion of autophagosome and lysosome. Our study provided insights into the mechanisms that bring new possibilities for AD therapy targeting mitophagy.
Collapse
Affiliation(s)
- Xiao Liang
- School of Medicine, Chongqing University, Chongqing, P.R. China
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Yangyang Wang
- School of Medicine, Chongqing University, Chongqing, P.R. China
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Siyu Li
- School of Medicine, Chongqing University, Chongqing, P.R. China
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Jianing Fan
- School of Medicine, Chongqing University, Chongqing, P.R. China
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Fanlin Zhou
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, P.R. China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer - iCQBC, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Xiaoju Li
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, P.R. China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer - iCQBC, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Shijie Li
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, P.R. China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer - iCQBC, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Yu Li
- School of Medicine, Chongqing University, Chongqing, P.R. China
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, P.R. China
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer - iCQBC, Chongqing University Cancer Hospital, Chongqing, P.R. China
| |
Collapse
|
12
|
Udagawa O. Oocyte Health and Quality: Implication of Mitochondria-related Organelle Interactions. Results Probl Cell Differ 2024; 73:25-42. [PMID: 39242373 DOI: 10.1007/978-3-031-62036-2_2] [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: 09/09/2024]
Abstract
Among factors like hormonal imbalance and uterine condition, oocyte quality is regarded as one of the key factors involved in age-related decline in the reproductive capacity. Here, are discussions about the functions played by organelles within the oocyte in forming the next generation that is more suitable for survival. Many insights on the adaptation to aging and maintenance of quality can be obtained from: interactions between mitochondria and other organelles that enable the long life of primordial oocytes; characteristics of organelle interactions after breaking dormancy from primary oocytes to mature oocytes; and characteristics of interactions between mitochondria and other organelles of aged oocytes collected during the ovulatory cycle from elderly individuals and animals. This information would potentially be beneficial to the development of future therapeutic methods or agents.
Collapse
Affiliation(s)
- Osamu Udagawa
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
13
|
Lin L, Zhao Y, Zheng Q, Zhang J, Li H, Wu W. Epigenetic targeting of autophagy for cancer: DNA and RNA methylation. Front Oncol 2023; 13:1290330. [PMID: 38148841 PMCID: PMC10749975 DOI: 10.3389/fonc.2023.1290330] [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: 09/07/2023] [Accepted: 11/28/2023] [Indexed: 12/28/2023] Open
Abstract
Autophagy, a crucial cellular mechanism responsible for degradation and recycling of intracellular components, is modulated by an intricate network of molecular signals. Its paradoxical involvement in oncogenesis, acting as both a tumor suppressor and promoter, has been underscored in recent studies. Central to this regulatory network are the epigenetic modifications of DNA and RNA methylation, notably the presence of N6-methyldeoxyadenosine (6mA) in genomic DNA and N6-methyladenosine (m6A) in eukaryotic mRNA. The 6mA modification in genomic DNA adds an extra dimension of epigenetic regulation, potentially impacting the transcriptional dynamics of genes linked to autophagy and, especially, cancer. Conversely, m6A modification, governed by methyltransferases and demethylases, influences mRNA stability, processing, and translation, affecting genes central to autophagic pathways. As we delve deeper into the complexities of autophagy regulation, the importance of these methylation modifications grows more evident. The interplay of 6mA, m6A, and autophagy points to a layered regulatory mechanism, illuminating cellular reactions to a range of conditions. This review delves into the nexus between DNA 6mA and RNA m6A methylation and their influence on autophagy in cancer contexts. By closely examining these epigenetic markers, we underscore their promise as therapeutic avenues, suggesting novel approaches for cancer intervention through autophagy modulation.
Collapse
Affiliation(s)
- Luobin Lin
- Guangdong Province Key Laboratory of Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yuntao Zhao
- Guangdong Province Key Laboratory of Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Qinzhou Zheng
- Guangdong Province Key Laboratory of Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jiayang Zhang
- Guangdong Province Key Laboratory of Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Huaqin Li
- School of Health Sciences, Guangzhou Xinhua University, Guangzhou, Guangdong, China
| | - Wenmei Wu
- Guangdong Province Key Laboratory of Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Hu Q, Lei J, Cheng Z, Xu J, Wang L, Yuan Y, Gan M, Wang Y, Xie Y, Yao L, Wang K, Liu Y, Xun W, Wang JB, Han T. STUB1-mediated ubiquitination regulates the stability of GLUD1 in lung adenocarcinoma. iScience 2023; 26:107151. [PMID: 37416474 PMCID: PMC10319899 DOI: 10.1016/j.isci.2023.107151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 05/03/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
The dysregulation of glutamine metabolism provides survival advantages for tumors by supplementing tricarboxylic acid cycle. Glutamate dehydrogenase 1 (GLUD1) is one of the key enzymes in glutamine catabolism. Here, we found that enhanced protein stability was the key factor for the upregulation of GLUD1 in lung adenocarcinoma. We discovered that GLUD1 showed a high protein expression in lung adenocarcinoma cells or tissues. We elucidated that STIP1 homology and U-box-containing protein 1 (STUB1) was the key E3 ligase responsible for ubiquitin-mediated proteasomal degradation of GLUD1. We further showed that lysine 503 (K503) was the main ubiquitination site of GLUD1, inhibiting the ubiquitination at this site promoted the proliferation and tumor growth of lung adenocarcinoma cells. Taken together, this study clarifies the molecular mechanism of GLUD1 in maintaining protein homeostasis in lung adenocarcinoma, which provides a theoretical basis for the development of anti-cancer drugs targeting GLUD1.
Collapse
Affiliation(s)
- Qifan Hu
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi 330006, China
- School of Basic Medical Sciences, Nanchang University, Nanchang City, Jiangxi 330031, China
- Department of Thoracic Surgery, The First Affifiliated Hospital of Nanchang University, Nanchang City, Jiangxi 330006, China
| | - Jiapeng Lei
- School of Basic Medical Sciences, Nanchang University, Nanchang City, Jiangxi 330031, China
- School of Basic Medical Sciences, Nanchang Medical College, Nanchang City, Jiangxi 330000, China
| | - Zhujun Cheng
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi 330006, China
| | - Jing Xu
- School of Basic Medical Sciences, Nanchang University, Nanchang City, Jiangxi 330031, China
| | - Lei Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang City, Jiangxi 330031, China
| | - Yi Yuan
- School of Huankui Academy, Nanchang University, Nanchang City, Jiangxi 330031, China
| | - Mingxi Gan
- School of Basic Medical Sciences, Nanchang University, Nanchang City, Jiangxi 330031, China
| | - Yanan Wang
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi 330006, China
| | - Yilin Xie
- School of Queen Mary, Nanchang University, Nanchang City, Jiangxi 330031, China
| | - Lu Yao
- School of Huankui Academy, Nanchang University, Nanchang City, Jiangxi 330031, China
| | - Keru Wang
- School of Huankui Academy, Nanchang University, Nanchang City, Jiangxi 330031, China
| | - Yuhan Liu
- School of Basic Medical Sciences, Nanchang University, Nanchang City, Jiangxi 330031, China
| | - Wenze Xun
- School of Basic Medical Sciences, Nanchang University, Nanchang City, Jiangxi 330031, China
| | - Jian-Bin Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang City, Jiangxi 330031, China
- Department of Thoracic Surgery, The First Affifiliated Hospital of Nanchang University, Nanchang City, Jiangxi 330006, China
| | - Tianyu Han
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi 330006, China
- Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City, Jiangxi 330006, China
- China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City, Jiangxi 330200, China
| |
Collapse
|
15
|
Ahn D, Go RE, Choi KC. Oxygen consumption rate to evaluate mitochondrial dysfunction and toxicity in cardiomyocytes. Toxicol Res 2023; 39:333-339. [PMID: 37398565 PMCID: PMC10313613 DOI: 10.1007/s43188-023-00183-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 07/04/2023] Open
Abstract
The increase in the types and complexity of diseases has led to significant advances in diagnostic techniques and the availability of effective therapies. Recent studies have focused on the role of mitochondrial dysfunction in the pathogenesis of cardiovascular diseases (CVDs). Mitochondria are important organelles in cells that generate energy. Besides the production of adenosine triphosphate (ATP), the energy currency of cells, mitochondria are also involved in thermogenesis, control of intracellular calcium ions (Ca2+), apoptosis, regulation of reactive oxygen species (ROS), and inflammation. Mitochondrial dysfunction has been implicated in several diseases including cancer, diabetes, some genetic diseases, and neurogenerative and metabolic diseases. Furthermore, the cardiomyocytes of the heart are rich in mitochondria due to the large energy requirement for optimal cardiac function. One of the main causes of cardiac tissue injuries is believed to be mitochondrial dysfunction, which occurs via complicated pathways which have not yet been completely elucidated. There are various types of mitochondrial dysfunction including mitochondrial morphological change, unbalanced levels of substances to maintain mitochondria, mitochondrial damage by drugs, and mitochondrial deletion and synthesis errors. Most of mitochondrial dysfunctions are linked with symptoms and diseases, thus we focus on parts of mitochondrial dysfunction about fission and fusion in cardiomyocytes, and ways to understand the mechanism of cardiomyocyte damage by detecting oxygen consumption levels in the mitochondria.
Collapse
Affiliation(s)
- Dohee Ahn
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| |
Collapse
|
16
|
Vo MT, Choi CY, Choi YB. The mitophagy receptor NIX induces vIRF-1 oligomerization and interaction with GABARAPL1 for the promotion of HHV-8 reactivation-induced mitophagy. PLoS Pathog 2023; 19:e1011548. [PMID: 37459327 PMCID: PMC10374065 DOI: 10.1371/journal.ppat.1011548] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/27/2023] [Accepted: 07/07/2023] [Indexed: 07/28/2023] Open
Abstract
Recently, viruses have been shown to regulate selective autophagy for productive infections. For instance, human herpesvirus 8 (HHV-8), also known as Kaposi's sarcoma-associated herpesvirus (KSHV), activates selective autophagy of mitochondria, termed mitophagy, thereby inhibiting antiviral innate immune responses during lytic infection in host cells. We previously demonstrated that HHV-8 viral interferon regulatory factor 1 (vIRF-1) plays a crucial role in lytic replication-activated mitophagy by interacting with cellular mitophagic proteins, including NIX and TUFM. However, the precise molecular mechanisms by which these interactions lead to mitophagy activation remain to be determined. Here, we show that vIRF-1 binds directly to mammalian autophagy-related gene 8 (ATG8) proteins, preferentially GABARAPL1 in infected cells, in an LC3-interacting region (LIR)-independent manner. Accordingly, we identified key residues in vIRF-1 and GABARAPL1 required for mutual interaction and demonstrated that the interaction is essential for mitophagy activation and HHV-8 productive replication. Interestingly, the mitophagy receptor NIX promotes vIRF-1-GABARAPL1 interaction, and NIX/vIRF-1-induced mitophagy is significantly inhibited in GABARAPL1-deficient cells. Moreover, a vIRF-1 variant defective in GABARAPL1 binding substantially loses the ability to induce vIRF-1/NIX-induced mitophagy. These results suggest that NIX supports vIRF-1 activity as a mitophagy mediator. In addition, we found that NIX promotes vIRF-1 aggregation and stabilizes aggregated vIRF-1. Together, these findings indicate that vIRF-1 plays a role as a viral mitophagy mediator that can be activated by a cellular mitophagy receptor.
Collapse
Affiliation(s)
- Mai Tram Vo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Chang-Yong Choi
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Young Bong Choi
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
17
|
Shim SM, Choi HR, Kwon SC, Kim HY, Sung KW, Jung EJ, Mun SR, Bae TH, Kim DH, Son YS, Jung CH, Lee J, Lee MJ, Park JW, Kwon YT. The Cys-N-degron pathway modulates pexophagy through the N-terminal oxidation and arginylation of ACAD10. Autophagy 2023; 19:1642-1661. [PMID: 36184612 PMCID: PMC10262816 DOI: 10.1080/15548627.2022.2126617] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 11/02/2022] Open
Abstract
In the N-degron pathway, N-recognins recognize cognate substrates for degradation via the ubiquitin (Ub)-proteasome system (UPS) or the autophagy-lysosome system (hereafter autophagy). We have recently shown that the autophagy receptor SQSTM1/p62 (sequestosome 1) is an N-recognin that binds the N-terminal arginine (Nt-Arg) as an N-degron to modulate autophagic proteolysis. Here, we show that the N-degron pathway mediates pexophagy, in which damaged peroxisomal fragments are degraded by autophagy under normal and oxidative stress conditions. This degradative process initiates when the Nt-Cys of ACAD10 (acyl-CoA dehydrogenase family, member 10), a receptor in pexophagy, is oxidized into Cys sulfinic (CysO2) or sulfonic acid (CysO3) by ADO (2-aminoethanethiol (cysteamine) dioxygenase). Under oxidative stress, the Nt-Cys of ACAD10 is chemically oxidized by reactive oxygen species (ROS). The oxidized Nt-Cys2 is arginylated by ATE1-encoded R-transferases, generating the RCOX N-degron. RCOX-ACAD10 marks the site of pexophagy via the interaction with PEX5 and binds the ZZ domain of SQSTM1/p62, recruiting LC3+-autophagic membranes. In mice, knockout of either Ate1 responsible for Nt-arginylation or Sqstm1/p62 leads to increased levels of peroxisomes. In the cells from patients with peroxisome biogenesis disorders (PBDs), characterized by peroxisomal loss due to uncontrolled pexophagy, inhibition of either ATE1 or SQSTM1/p62 was sufficient to recover the level of peroxisomes. Our results demonstrate that the Cys-N-degron pathway generates an N-degron that regulates the removal of damaged peroxisomal membranes along with their contents. We suggest that tannic acid, a commercially available drug on the market, has a potential to treat PBDs through its activity to inhibit ATE1 R-transferases.Abbreviations: ACAA1, acetyl-Coenzyme A acyltransferase 1; ACAD, acyl-Coenzyme A dehydrogenase; ADO, 2-aminoethanethiol (cysteamine) dioxygenase; ATE1, arginyltransferase 1; CDO1, cysteine dioxygenase type 1; ER, endoplasmic reticulum; LIR, LC3-interacting region; MOXD1, monooxygenase, DBH-like 1; NAC, N-acetyl-cysteine; Nt-Arg, N-terminal arginine; Nt-Cys, N-terminal cysteine; PB1, Phox and Bem1p; PBD, peroxisome biogenesis disorder; PCO, plant cysteine oxidase; PDI, protein disulfide isomerase; PTS, peroxisomal targeting signal; R-COX, Nt-Arg-CysOX; RNS, reactive nitrogen species; ROS, reactive oxygen species; SNP, sodium nitroprusside; UBA, ubiquitin-associated; UPS, ubiquitinproteasome system.
Collapse
Affiliation(s)
- Sang Mi Shim
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ha Rim Choi
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Soon Chul Kwon
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hye Yeon Kim
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ki Woon Sung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
- AUTOTAC Bio Inc., Seoul, Republic of Korea
| | - Eui Jung Jung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Su Ran Mun
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Tae Hyun Bae
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Dong Hyun Kim
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon, Korea
| | - Yeon Sung Son
- Neuroscience Research Institute, Medical Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Chan Hoon Jung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jihoon Lee
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
- AUTOTAC Bio Inc., Seoul, Republic of Korea
| | - Min Jae Lee
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Joo-Won Park
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Yong Tae Kwon
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cellular Degradation Biology Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
- AUTOTAC Bio Inc., Seoul, Republic of Korea
- Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
- SNU Dementia Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
18
|
Xiang J, Gong W, Liu J, Zhang H, Li M, Wang R, Lv Y, Sun P. Identification of DLL3-related genes affecting the prognosis of patients with colon adenocarcinoma. Front Genet 2023; 14:1098190. [PMID: 37274780 PMCID: PMC10233108 DOI: 10.3389/fgene.2023.1098190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/24/2023] [Indexed: 06/07/2023] Open
Abstract
Background: Delta-like ligand 3 (DLL3) is one of the NOTCH family of ligands, which plays a pro- or anti-carcinogenic role in some cancers. But the role of DLL3 in colon adenocarcinoma (COAD) has not been studied in depth. Materials and methods: First, we used Kaplan-Meier (K-M) curve to evaluate the effect of DLL3 on the prognosis of COAD in The Cancer Genome Atlas (TCGA), which was further validated in clinical samples for immunohistochemistry. Then we screened for differentially expressed genes (DEGs) of DLL3 by analyzing datasets of COAD samples from Gene Expression Omnibus (GEO) and TCGA. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, and Gene Set Enrichment Analysis (GSEA) were conducted to explore the underlying mechanisms of DLL3-related in the development and prognosis of COAD. On the basis of DLL3-related signature genes, a prognostic model and a nomogram were constructed. Finally, CIBERSORT was applied to assess the proportion of immune cell types in COAD sample. Results: Survival analysis showed a significant difference in overall survival between high- and low-expression group (p = 0.0092), with COAD patients in the high-group having poorer 5-year survival rate. Gene functional enrichment analysis revealed that DLL3-related DEGs were mainly enriched in tumor- and immunity-related signaling pathways, containing AMPK pathway and mitophagy-animal. The comparison of COAD tumor and normal, DLL3 high- and low-expression groups by GSEA found that AMPK signaling pathway and mitophagy-animal were inhibited. Nomogram constructed from DLL3-related signature genes had a good predictive effect on the prognosis of COAD. We found the highest correlation between DLL3 and interstitial dendritic cell (iDC), natural killer (NK) cell and Interstitial dendritic cell (Tem). DLL3 was also revealed to be diagnostic for COAD. In clinical sample, we identified higher DLL3 expression in colon cancer tissue than in adjacent control (p < 0.0001) and in metastasis than in primary lesion (p = 0.0056). DLL3 expression was associated with stage and high DLL3 expression was observed to predict poorer overall survival (p = 0.004). Conclusion: It suggested that DLL3 may offer prognostic value and therapeutic potential for individualized treatment of COAD, and that it may has a diagnostic role in COAD.
Collapse
Affiliation(s)
- Jinyu Xiang
- Departments of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, Shandong, China
| | - Wenjing Gong
- Departments of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, Shandong, China
| | - Jiannan Liu
- Departments of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, Shandong, China
| | - Huijuan Zhang
- Departments of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, Shandong, China
| | - Ming Li
- Departments of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, Shandong, China
| | - Rujian Wang
- Departments of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, Shandong, China
| | - Yaodong Lv
- Departments of Neurology, Yantai Yuhuangding Hospital, Shandong University, Yantai, Shandong, China
| | - Ping Sun
- Departments of Oncology, Yantai Yuhuangding Hospital, Shandong University, Yantai, Shandong, China
| |
Collapse
|
19
|
Chen L, Zhang Q, Meng Y, Zhao T, Mu C, Fu C, Deng C, Feng J, Du S, Liu W, Geng G, Ma K, Cheng H, Liu Q, Luo Q, Zhang J, Du Z, Cao L, Wang H, Liu Y, Lin J, Chen G, Liu L, Lam SM, Shui G, Zhu Y, Chen Q. Saturated fatty acids increase LPI to reduce FUNDC1 dimerization and stability and mitochondrial function. EMBO Rep 2023; 24:e54731. [PMID: 36847607 PMCID: PMC10074135 DOI: 10.15252/embr.202254731] [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: 01/29/2022] [Revised: 01/19/2023] [Accepted: 02/02/2023] [Indexed: 03/01/2023] Open
Abstract
Ectopic lipid deposition and mitochondrial dysfunction are common etiologies of obesity and metabolic disorders. Excessive dietary uptake of saturated fatty acids (SFAs) causes mitochondrial dysfunction and metabolic disorders, while unsaturated fatty acids (UFAs) counterbalance these detrimental effects. It remains elusive how SFAs and UFAs differentially signal toward mitochondria for mitochondrial performance. We report here that saturated dietary fatty acids such as palmitic acid (PA), but not unsaturated oleic acid (OA), increase lysophosphatidylinositol (LPI) production to impact on the stability of the mitophagy receptor FUNDC1 and on mitochondrial quality. Mechanistically, PA shifts FUNDC1 from dimer to monomer via enhanced production of LPI. Monomeric FUNDC1 shows increased acetylation at K104 due to dissociation of HDAC3 and increased interaction with Tip60. Acetylated FUNDC1 can be further ubiquitinated by MARCH5 for proteasomal degradation. Conversely, OA antagonizes PA-induced accumulation of LPI, and FUNDC1 monomerization and degradation. A fructose-, palmitate-, and cholesterol-enriched (FPC) diet also affects FUNDC1 dimerization and promotes its degradation in a non-alcoholic steatohepatitis (NASH) mouse model. We thus uncover a signaling pathway that orchestrates lipid metabolism with mitochondrial quality.
Collapse
Affiliation(s)
- Linbo Chen
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Qianping Zhang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Yuanyuan Meng
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Tian Zhao
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Chenglong Mu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Changying Fu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Caijuan Deng
- College of Pharmacy, Frontiers Science Center for Cell ResponsesNankai UniversityTianjinChina
| | - Jianyu Feng
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Siling Du
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Wei Liu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Guangfeng Geng
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Kaili Ma
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Hongcheng Cheng
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Qiangqiang Liu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Qian Luo
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Jiaojiao Zhang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Zhanqiang Du
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Lin Cao
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Hui Wang
- Cancer InstituteXuzhou Medical UniversityXuzhouChina
| | - Yong Liu
- Cancer InstituteXuzhou Medical UniversityXuzhouChina
| | - Jianping Lin
- College of Pharmacy, Frontiers Science Center for Cell ResponsesNankai UniversityTianjinChina
| | - Guo Chen
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Lei Liu
- State Key Laboratory of Membrane Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental BiologyChinese Academy of SciencesBeijingChina
- LipidAll Technologies Company LimitedChangzhouChina
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental BiologyChinese Academy of SciencesBeijingChina
| | - Yushan Zhu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| | - Quan Chen
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, Tianjin Key Laboratory of Protein Science, College of Life SciencesNankai UniversityTianjinChina
| |
Collapse
|
20
|
Ke PY. Crosstalk between Autophagy and RLR Signaling. Cells 2023; 12:cells12060956. [PMID: 36980296 PMCID: PMC10047499 DOI: 10.3390/cells12060956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Autophagy plays a homeostatic role in regulating cellular metabolism by degrading unwanted intracellular materials and acts as a host defense mechanism by eliminating infecting pathogens, such as viruses. Upon viral infection, host cells often activate retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) signaling to induce the transcription of type I interferons, thus establishing the first line of the innate antiviral response. In recent years, numerous studies have shown that virus-mediated autophagy activation may benefit viral replication through different actions on host cellular processes, including the modulation of RLR-mediated innate immunity. Here, an overview of the functional molecules and regulatory mechanism of the RLR antiviral immune response as well as autophagy is presented. Moreover, a summary of the current knowledge on the biological role of autophagy in regulating RLR antiviral signaling is provided. The molecular mechanisms underlying the crosstalk between autophagy and RLR innate immunity are also discussed.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry & Molecular Biology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| |
Collapse
|
21
|
Alim Al-Bari A, Ito Y, Thomes PG, Menon MB, García-Macia M, Fadel R, Stadlin A, Peake N, Faris ME, Eid N, Klionsky DJ. Emerging mechanistic insights of selective autophagy in hepatic diseases. Front Pharmacol 2023; 14:1149809. [PMID: 37007026 PMCID: PMC10060854 DOI: 10.3389/fphar.2023.1149809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
Macroautophagy (hereafter referred to as autophagy), a highly conserved metabolic process, regulates cellular homeostasis by degrading dysfunctional cytosolic constituents and invading pathogens via the lysosomal system. In addition, autophagy selectively recycles specific organelles such as damaged mitochondria (via mitophagy), and lipid droplets (LDs; via lipophagy) or eliminates specialized intracellular pathogenic microorganisms such as hepatitis B virus (HBV) and coronaviruses (via virophagy). Selective autophagy, particularly mitophagy, plays a key role in the preservation of healthy liver physiology, and its dysfunction is connected to the pathogenesis of a wide variety of liver diseases. For example, lipophagy has emerged as a defensive mechanism against chronic liver diseases. There is a prominent role for mitophagy and lipophagy in hepatic pathologies including non-alcoholic fatty liver disease (NAFLD), hepatocellular carcinoma (HCC), and drug-induced liver injury. Moreover, these selective autophagy pathways including virophagy are being investigated in the context of viral hepatitis and, more recently, the coronavirus disease 2019 (COVID-19)-associated hepatic pathologies. The interplay between diverse types of selective autophagy and its impact on liver diseases is briefly addressed. Thus, modulating selective autophagy (e.g., mitophagy) would seem to be effective in improving liver diseases. Considering the prominence of selective autophagy in liver physiology, this review summarizes the current understanding of the molecular mechanisms and functions of selective autophagy (mainly mitophagy and lipophagy) in liver physiology and pathophysiology. This may help in finding therapeutic interventions targeting hepatic diseases via manipulation of selective autophagy.
Collapse
Affiliation(s)
- Abdul Alim Al-Bari
- Department of Pharmacy, Faculty of Science, University of Rajshahi, Rajshahi, Bangladesh
| | - Yuko Ito
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Paul G. Thomes
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Manoj B. Menon
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Marina García-Macia
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca-CSIC, Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
| | - Raouf Fadel
- Department of Anatomy, College of Medicine and Medical Sciences, Arabian Gulf University, Al Manama, Bahrain
| | - Alfreda Stadlin
- Basic Medical Sciences Department, College of Medicine, Ajman university, Ajman, United Arab Emirates
| | - Nicholas Peake
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - MoezAlIslam Ezzat Faris
- Department of Clinical Nutrition and Dietetics, College of Health Sciences, University of Sharjah, United Arab Emirates
| | - Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
- *Correspondence: Nabil Eid,
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of MI, Ann Arbor, MI, United States
| |
Collapse
|
22
|
Makarov M, Korkotian E. Differential Role of Active Compounds in Mitophagy and Related Neurodegenerative Diseases. Toxins (Basel) 2023; 15:202. [PMID: 36977093 PMCID: PMC10058020 DOI: 10.3390/toxins15030202] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease or Parkinson's disease, significantly reduce the quality of life of patients and eventually result in complete maladjustment. Disruption of the synapses leads to a deterioration in the communication of nerve cells and decreased plasticity, which is associated with a loss of cognitive functions and neurodegeneration. Maintaining proper synaptic activity depends on the qualitative composition of mitochondria, because synaptic processes require sufficient energy supply and fine calcium regulation. The maintenance of the qualitative composition of mitochondria occurs due to mitophagy. The regulation of mitophagy is usually based on several internal mechanisms, as well as on signals and substances coming from outside the cell. These substances may directly or indirectly enhance or weaken mitophagy. In this review, we have considered the role of some compounds in process of mitophagy and neurodegeneration. Some of them have a beneficial effect on the functions of mitochondria and enhance mitophagy, showing promise as novel drugs for the treatment of neurodegenerative pathologies, while others contribute to a decrease in mitophagy.
Collapse
Affiliation(s)
| | - Eduard Korkotian
- Department of Brain Sciences, The Weizmann Institute of Science, Rehovot 7630031, Israel
| |
Collapse
|
23
|
Puglisi-Allegra S, Lazzeri G, Busceti CL, Giorgi FS, Biagioni F, Fornai F. Lithium engages autophagy for neuroprotection and neuroplasticity: translational evidence for therapy. Neurosci Biobehav Rev 2023; 148:105148. [PMID: 36996994 DOI: 10.1016/j.neubiorev.2023.105148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023]
Abstract
Here an overview is provided on therapeutic/neuroprotective effects of Lithium (Li+) in neurodegenerative and psychiatric disorders focusing on the conspicuous action of Li+ through autophagy. The effects on the autophagy machinery remain the key molecular mechanisms to explain the protective effects of Li+ for neurodegenerative diseases, offering potential therapeutic strategies for the treatment of neuropsychiatric disorders and emphasizes a crossroad linking autophagy, neurodegenerative disorders, and mood stabilization. Sensitization by psychostimulants points to several mechanisms involved in psychopathology, most also crucial in neurodegenerative disorders. Evidence shows the involvement of autophagy and metabotropic Glutamate receptors-5 (mGluR5) in neurodegeneration due to methamphetamine neurotoxicity as well as in neuroprotection, both in vitro and in vivo models. More recently, Li+ was shown to modulate autophagy through its action on mGluR5, thus pointing to an additional way of autophagy engagement by Li+ and to a substantial role of mGluR5 in neuroprotection related to neural e neuropsychiatry diseases. We propose Li+ engagement of autophagy through the canonical mechanisms of autophagy machinery and through the intermediary of mGluR5.
Collapse
|
24
|
Alao JP, Legon L, Dabrowska A, Tricolici AM, Kumar J, Rallis C. Interplays of AMPK and TOR in Autophagy Regulation in Yeast. Cells 2023; 12:cells12040519. [PMID: 36831186 PMCID: PMC9953913 DOI: 10.3390/cells12040519] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Cells survey their environment and need to balance growth and anabolism with stress programmes and catabolism towards maximum cellular bioenergetics economy and survival. Nutrient-responsive pathways, such as the mechanistic target of rapamycin (mTOR) interact and cross-talk, continuously, with stress-responsive hubs such as the AMP-activated protein kinase (AMPK) to regulate fundamental cellular processes such as transcription, protein translation, lipid and carbohydrate homeostasis. Especially in nutrient stresses or deprivations, cells tune their metabolism accordingly and, crucially, recycle materials through autophagy mechanisms. It has now become apparent that autophagy is pivotal in lifespan, health and cell survival as it is a gatekeeper of clearing damaged macromolecules and organelles and serving as quality assurance mechanism within cells. Autophagy is hard-wired with energy and nutrient levels as well as with damage-response, and yeasts have been instrumental in elucidating such connectivities. In this review, we briefly outline cross-talks and feedback loops that link growth and stress, mainly, in the fission yeast Schizosaccharomyces pombe, a favourite model in cell and molecular biology.
Collapse
|
25
|
Alam MB, Park NH, Song BR, Lee SH. Antioxidant Potential-Rich Betel Leaves ( Piper betle L.) Exert Depigmenting Action by Triggering Autophagy and Downregulating MITF/Tyrosinase In Vitro and In Vivo. Antioxidants (Basel) 2023; 12:antiox12020374. [PMID: 36829933 PMCID: PMC9952209 DOI: 10.3390/antiox12020374] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/09/2023] Open
Abstract
Each individual has a unique skin tone based on the types and quantities of melanin pigment, and oxidative stress is a key element in melanogenesis regulation. This research sought to understand the in vitro and in vivo antioxidant and depigmenting properties of betel leaves (Piper betle L.) extract (PBL) and the underlying mechanism. Ethyl acetate fractions of PBL (PBLA) demonstrated excellent phenolic content (342 ± 4.02 mgGAE/g) and strong DPPH, ABTS radicals, and nitric oxide (NO) scavenging activity with an IC50 value of 41.52 ± 1.02 μg/mL, 45.60 ± 0.56 μg/mL, and 51.42 ± 1.25 μg/mL, respectively. Contrarily, ethanolic extract of PBL (PBLE) showed potent mushroom, mice, and human tyrosinase inhibition activity (IC50 = 7.72 ± 0.98 μg/mL, 20.59 ± 0.83 μg/mL and 24.78 ± 0.56 μg/mL, respectively). According to gas chromatography-mass spectrometry, PBL is abundant in caryophyllene, eugenol, O-eugenol, 3-Allyl-6-methoxyphenyl acetate, and chavicol. An in vitro and in vivo investigation showed that PBLE suppressed tyrosinase (Tyr), tyrosinase-related protein-1 and -2 (Trp-1 and Trp-2), and microphthalmia-associated transcription factors (MITF), decreasing the formation of melanin in contrast to the untreated control. PBLE reduced the cyclic adenosine monophosphate (cAMP) response to an element-binding protein (CREB) phosphorylation by preventing the synthesis of cAMP. Additionally, it activates c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinases (p38), destroying Tyr and MITF and avoiding melanin production. Higher levels of microtubule-associated protein-light chain 3 (LC3-II), autophagy-related protein 5 (Atg5), Beclin 1, and lower levels of p62 demonstrate that PBLE exhibits significant anti-melanogenic effects via an autophagy-induction mechanism, both in vitro and in vivo. Additionally, PBLE significantly reduced the amount of lipid peroxidation while increasing the activity of several antioxidant enzymes in vivo, such as catalase, glutathione, superoxide dismutase, and thioredoxin. PBLE can therefore be employed in topical formulations as a potent skin-whitening agent.
Collapse
Affiliation(s)
- Md Badrul Alam
- Department of Food Science and Biotechnology, Graduate School, Kyungpook National University, Daegu 41566, Republic of Korea
- Food and Bio-Industry Research Institute, Inner Beauty/Antiaging Center, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Na Hyun Park
- Department of Food Science and Biotechnology, Graduate School, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Bo-Rim Song
- Department of Food Science and Biotechnology, Graduate School, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Sang-Han Lee
- Department of Food Science and Biotechnology, Graduate School, Kyungpook National University, Daegu 41566, Republic of Korea
- Food and Bio-Industry Research Institute, Inner Beauty/Antiaging Center, Kyungpook National University, Daegu 41566, Republic of Korea
- Correspondence: ; Tel.: +82-053-950-7754
| |
Collapse
|
26
|
Iriondo MN, Etxaniz A, Varela YR, Ballesteros U, Lázaro M, Valle M, Fracchiolla D, Martens S, Montes LR, Goñi FM, Alonso A. Effect of ATG12-ATG5-ATG16L1 autophagy E3-like complex on the ability of LC3/GABARAP proteins to induce vesicle tethering and fusion. Cell Mol Life Sci 2023; 80:56. [PMID: 36729310 PMCID: PMC9894987 DOI: 10.1007/s00018-023-04704-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023]
Abstract
In macroautophagy, the autophagosome (AP) engulfs portions of cytoplasm to allow their lysosomal degradation. AP formation in humans requires the concerted action of the ATG12 and LC3/GABARAP conjugation systems. The ATG12-ATG5-ATG16L1 or E3-like complex (E3 for short) acts as a ubiquitin-like E3 enzyme, promoting LC3/GABARAP proteins anchoring to the AP membrane. Their role in the AP expansion process is still unclear, in part because there are no studies comparing six LC3/GABARAP family member roles under the same conditions, and also because the full human E3 was only recently available. In the present study, the lipidation of six members of the LC3/GABARAP family has been reconstituted in the presence and absence of E3, and the mechanisms by which E3 and LC3/GABARAP proteins participate in vesicle tethering and fusion have been investigated. In the absence of E3, GABARAP and GABARAPL1 showed the highest activities. Differences found within LC3/GABARAP proteins suggest the existence of a lipidation threshold, lower for the GABARAP subfamily, as a requisite for tethering and inter-vesicular lipid mixing. E3 increases and speeds up lipidation and LC3/GABARAP-promoted tethering. However, E3 hampers LC3/GABARAP capacity to induce inter-vesicular lipid mixing or subsequent fusion, presumably through the formation of a rigid scaffold on the vesicle surface. Our results suggest a model of AP expansion in which the growing regions would be areas where the LC3/GABARAP proteins involved should be susceptible to lipidation in the absence of E3, or else a regulatory mechanism would allow vesicle incorporation and phagophore growth when E3 is present.
Collapse
Affiliation(s)
- Marina N Iriondo
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, 48940, Leioa, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country, 48940, Leioa, Spain
| | - Asier Etxaniz
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, 48940, Leioa, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country, 48940, Leioa, Spain
| | - Yaiza R Varela
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, 48940, Leioa, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country, 48940, Leioa, Spain
| | - Uxue Ballesteros
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, 48940, Leioa, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country, 48940, Leioa, Spain
| | - Melisa Lázaro
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160, Derio, Bizkaia, Spain
| | - Mikel Valle
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160, Derio, Bizkaia, Spain
| | - Dorotea Fracchiolla
- Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030, Vienna, Austria
| | - Sascha Martens
- Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030, Vienna, Austria
| | - L Ruth Montes
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, 48940, Leioa, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country, 48940, Leioa, Spain
| | - Félix M Goñi
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, 48940, Leioa, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country, 48940, Leioa, Spain
| | - Alicia Alonso
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, 48940, Leioa, Spain.
- Department of Biochemistry and Molecular Biology, University of the Basque Country, 48940, Leioa, Spain.
| |
Collapse
|
27
|
Takahashi S, Saito C, Koyama-Honda I, Mizushima N. Quantitative 3D correlative light and electron microscopy of organelle association during autophagy. Cell Struct Funct 2022; 47:89-99. [PMID: 36418108 PMCID: PMC10511054 DOI: 10.1247/csf.22071] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022] Open
Abstract
In macroautophagy, disk-shaped double-membrane structures called phagophores elongate to form cup-shaped structures, becoming autophagosomes upon closure. These autophagosomes then fuse with lysosomes to become autolysosomes and degrade engulfed material. Autophagosome formation is reported to involve other organelles, including the endoplasmic reticulum (ER) and mitochondria. Organelles are also taken up by autophagosomes as autophagy cargos. However, few studies have performed systematic spatiotemporal analysis of inter-organelle relationships during macroautophagy. Here, we investigated the organelles in contact with phagophores, autophagosomes, and autolysosomes by using three-dimensional correlative light and electron microscopy with array tomography in cells starved 30 min. As previously reported, all phagophores associate with the ER. The surface area of phagophores in contact with the ER decreases gradually as they mature into autophagosomes and autolysosomes. However, the ER still associates with 92% of autophagosomes and 79% of autolysosomes, suggesting that most autophagosomes remain on the ER after closure and even when they fuse with lysosomes. In addition, we found that phagophores form frequently near other autophagic structures, suggesting the presence of potential hot spots for autophagosome formation. We also analyzed the contents of phagophores and autophagosomes and found that the ER is the most frequently engulfed organelle (detected in 65% of total phagophores and autophagosomes). These quantitative three-dimensional ultrastructural data provide insights into autophagosome-organelle relationships during macroautophagy.Key words: 3D-CLEM, autophagosome, electron microscopy, endoplasmic reticulum, lysosome.
Collapse
Affiliation(s)
- Satoru Takahashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Chieko Saito
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ikuko Koyama-Honda
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
28
|
Lee YJ, Kim JK, Jung CH, Kim YJ, Jung EJ, Lee SH, Choi HR, Son YS, Shim SM, Jeon SM, Choe JH, Lee SH, Whang J, Sohn KC, Hur GM, Kim HT, Yeom J, Jo EK, Kwon YT. Chemical modulation of SQSTM1/p62-mediated xenophagy that targets a broad range of pathogenic bacteria. Autophagy 2022; 18:2926-2945. [PMID: 35316156 PMCID: PMC9673928 DOI: 10.1080/15548627.2022.2054240] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
The N-degron pathway is a proteolytic system in which the N-terminal degrons (N-degrons) of proteins, such as arginine (Nt-Arg), induce the degradation of proteins and subcellular organelles via the ubiquitin-proteasome system (UPS) or macroautophagy/autophagy-lysosome system (hereafter autophagy). Here, we developed the chemical mimics of the N-degron Nt-Arg as a pharmaceutical means to induce targeted degradation of intracellular bacteria via autophagy, such as Salmonella enterica serovar Typhimurium (S. Typhimurium), Escherichia coli, and Streptococcus pyogenes as well as Mycobacterium tuberculosis (Mtb). Upon binding the ZZ domain of the autophagic cargo receptor SQSTM1/p62 (sequestosome 1), these chemicals induced the biogenesis and recruitment of autophagic membranes to intracellular bacteria via SQSTM1, leading to lysosomal degradation. The antimicrobial efficacy was independent of rapamycin-modulated core autophagic pathways and synergistic with the reduced production of inflammatory cytokines. In mice, these drugs exhibited antimicrobial efficacy for S. Typhimurium, Bacillus Calmette-Guérin (BCG), and Mtb as well as multidrug-resistant Mtb and inhibited the production of inflammatory cytokines. This dual mode of action in xenophagy and inflammation significantly protected mice from inflammatory lesions in the lungs and other tissues caused by all the tested bacterial strains. Our results suggest that the N-degron pathway provides a therapeutic target in host-directed therapeutics for a broad range of drug-resistant intracellular pathogens.Abbreviations: ATG: autophagy-related gene; BCG: Bacillus Calmette-Guérin; BMDMs: bone marrow-derived macrophages; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CFUs: colony-forming units; CXCL: C-X-C motif chemokine ligand; EGFP: enhanced green fluorescent protein; IL1B/IL-1β: interleukin 1 beta; IL6: interleukin 6; LIR: MAP1LC3/LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; Mtb: Mycobacterium tuberculosis; MTOR: mechanistic target of rapamycin kinase; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PB1: Phox and Bem1; SQSTM1/p62: sequestosome 1; S. Typhimurium: Salmonella enterica serovar Typhimurium; TAX1BP1: Tax1 binding protein 1; TNF: tumor necrosis factor; UBA: ubiquitin-associated.
Collapse
Affiliation(s)
- Yoon Jee Lee
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Chan Hoon Jung
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Young Jae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Eui Jung Jung
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Su Hyun Lee
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ha Rim Choi
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yeon Sung Son
- Neuroscience Research Institute, Medical Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sang Mi Shim
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sang Min Jeon
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jin Ho Choe
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sang-Hee Lee
- Center for Research Equipment, Korea Basic Science Institute, Cheongju, Korea
| | - Jake Whang
- Korea Mycobacterium Resource Center (KMRC) & Basic Research Section, The Korean Institute of Tuberculosis (KIT), Cheongju, Korea
| | - Kyung-Cheol Sohn
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Department of Pharmacology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Gang Min Hur
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Department of Pharmacology, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyun Tae Kim
- Chemistry R&D Center, AUTOTAC Bio Inc, Seoul, Republic of Korea
| | - Jinki Yeom
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea,Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, Korea,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, Korea,CONTACT Eun-Kyeong Jo Department of Microbiology, and Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon35015, Korea
| | - Yong Tae Kwon
- Cellular Degradation Biology Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea,Chemistry R&D Center, AUTOTAC Bio Inc, Seoul, Republic of Korea,SNU Dementia Research Center, College of Medicine, Seoul National University, Seoul, Republic of Korea,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea,Yong Tae Kwon Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul110-799, Korea
| |
Collapse
|
29
|
Yao Z, Li X, Gao J, Wang Y, Xiao L, Chang X, Liu F, Feng Z, Zhang X. Transcription factor p8 regulates autophagy in response to disulfiram via PI3K/mTOR/p70S6K signaling pathway in pancreatic cancer cells. Hum Cell 2022; 35:1464-1474. [PMID: 35749047 DOI: 10.1007/s13577-022-00731-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/26/2022] [Indexed: 11/25/2022]
Abstract
Disulfiram (DSF), which is an inhibitor of aldehyde dehydrogenase (ALDH) and approved by the FDA for the treatment of alcoholism previously, has been repurposed for use as a cancer treatment because of its potent effect in preclinical studies. In this study, we found that disulfiram forms potent complexes with copper (DSF/Cu) inhibited cell proliferation, induced apoptosis in human pancreatic cancer cells, which was detected by flow cytometry and western blotting. Meanwhile, autophagy and autophagic flux also clearly observed by transmission electron microscopy, confocal microscopy and flow cytometry. Our results also showed that DSF/Cu induced transcription factor p8 upregulation and PI3K/mTOR signaling pathway activation detected by real-time PCR and western blotting. Additionally, suppression of p8 inactivated the mTOR signaling pathway and autophagic flux maintained. Furthermore, mechanism study indicated that autophagy induced by DSF/Cu was regulated by p8 and was related to PI3K/mTOR/p70S6K signaling pathway in pancreatic cancer cells. Our findings provide insights into the role of p8 in regulating autophagy induced by DSF/Cu effects in pancreatic cancer cells.
Collapse
Affiliation(s)
- Zhangyu Yao
- Key Laboratory of Antibody Technology, National Health Commission, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
- Department of Head and Neck Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, 42 Baiziting Street, Nanjing, 210009, Jiangsu, China
| | - Xiang Li
- Key Laboratory of Antibody Technology, National Health Commission, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
| | - Jun Gao
- Key Laboratory of Antibody Technology, National Health Commission, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
| | - Yutao Wang
- Key Laboratory of Antibody Technology, National Health Commission, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
| | - Linmei Xiao
- Key Laboratory of Antibody Technology, National Health Commission, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
| | - Xinxia Chang
- Key Laboratory of Antibody Technology, National Health Commission, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
| | - Fangzhou Liu
- Department of Head and Neck Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, 42 Baiziting Street, Nanjing, 210009, Jiangsu, China
| | - Zhenqing Feng
- Key Laboratory of Antibody Technology, National Health Commission, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China
| | - Xiao Zhang
- Key Laboratory of Antibody Technology, National Health Commission, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China.
- Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, 101 Longmian Road, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
30
|
Yang Y, Gomez M, Marsh T, Poillet-Perez L, Sawant A, Chen L, Park NR, Jackson SR, Hu Z, Alon N, Liu C, Debnath J, Guan JL, Davidson S, Verzi M, White E. Autophagy in PDGFRα+ mesenchymal cells is essential for intestinal stem cell survival. Proc Natl Acad Sci U S A 2022; 119:e2202016119. [PMID: 35537042 PMCID: PMC9173755 DOI: 10.1073/pnas.2202016119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/15/2022] [Indexed: 12/14/2022] Open
Abstract
Autophagy defects are a risk factor for inflammatory bowel diseases (IBDs) through unknown mechanisms. Whole-body conditional deletion of autophagy-related gene (Atg) Atg7 in adult mice (Atg7Δ/Δ) causes tissue damage and death within 3 mo due to neurodegeneration without substantial effect on intestine. In contrast, we report here that whole-body conditional deletion of other essential Atg genes Atg5 or Fip200/Atg17 in adult mice (Atg5Δ/Δ or Fip200Δ/Δ) caused death within 5 d due to rapid autophagy inhibition, elimination of ileum stem cells, and loss of barrier function. Atg5Δ/Δ mice lost PDGFRα+ mesenchymal cells (PMCs) and Wnt signaling essential for stem cell renewal, which were partially rescued by exogenous Wnt. Matrix-assisted laser desorption ionization coupled to mass spectrometry imaging (MALDI-MSI) of Atg5Δ/Δ ileum revealed depletion of aspartate and nucleotides, consistent with metabolic insufficiency underlying PMC loss. The difference in the autophagy gene knockout phenotypes is likely due to distinct kinetics of autophagy loss, as deletion of Atg5 more gradually extended lifespan phenocopying deletion of Atg7 or Atg12. Thus, autophagy is required for PMC metabolism and ileum stem cell and mammalian survival. Failure to maintain PMCs through autophagy may therefore contribute to IBD.
Collapse
Affiliation(s)
- Yang Yang
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903
| | - Maria Gomez
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544
| | - Timothy Marsh
- Department of Pathology, University of California, San Francisco, CA 94143
| | | | - Akshada Sawant
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903
| | - Lei Chen
- Department of Genetics, Rutgers University, Piscataway, NJ, 08854
| | - Noel R. Park
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - S. RaElle Jackson
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Zhixian Hu
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544
| | - Noa Alon
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903
- School of Art Sciences, Rutgers University, New Brunswick, NJ 08901
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson University Hospital, New Brunswick, NJ 08903
| | - Jayanta Debnath
- Department of Pathology, University of California, San Francisco, CA 94143
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Shawn Davidson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544
| | - Michael Verzi
- Department of Genetics, Rutgers University, Piscataway, NJ, 08854
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544
- School of Art Sciences, Rutgers University, New Brunswick, NJ 08901
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| |
Collapse
|
31
|
Ke PY. Autophagy and antiviral defense. IUBMB Life 2022; 74:317-338. [PMID: 34859938 DOI: 10.1002/iub.2582] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/04/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022]
Abstract
Targeting intracellular components for lysosomal degradation by autophagy not only maintains cellular homeostasis but also counteracts the effects of external stimuli, including invading pathogens. Among various kinds of pathogens, viruses have been extensively shown to induce autophagy to benefit viral growth in infected cells and to modulate host defense responses, such as innate antiviral immunity. Recently, numerous lines of evidence have implied that virus-induced autophagy triggers multilayer mechanisms to regulate the innate antiviral response of host cells, thus promoting a balance in virus-host cell interactions. In this review, the detailed mechanisms underlying autophagy and the innate antiviral immune response are first described. Then, I summarize the current information regarding the diverse functional role(s) of autophagy in the control of antiviral defenses against different types of viral infections. Moreover, the physiological significance of autophagy-regulated antiviral responses on the viral life cycle and the potential autophagy alterations induced by virus-associated antiviral signaling is further discussed.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry & Molecular Biology and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| |
Collapse
|
32
|
Ke PY, Chang CW, Hsiao YC. Baicalein Activates Parkin-Dependent Mitophagy through NDP52 and OPTN. Cells 2022; 11:cells11071132. [PMID: 35406696 PMCID: PMC8997844 DOI: 10.3390/cells11071132] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/15/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022] Open
Abstract
The elimination of intracellular components by autophagy maintains metabolic homeostasis and is a quality-control pathway that enables organelle regeneration. Mitophagy is a type of selective autophagy that regulates mitochondrial turnover, and the dysregulation of mitophagy has been implicated in the pathogenesis of liver diseases. However, the detailed molecular mechanism underlying mitophagy regulation in liver cells remains unclear, and the small molecules that may potentially modulate hepatic mitophagy are still unavailable. Here, we report that baicalein, a flavonoid extracted from Scutellaria baicalensis, induces the entire autophagy that proceeds through the autolysosome maturation stage in human hepatoma cells. In addition, baicalein-induced autophagy is demonstrated to target mitochondria for degradation. Further studies show that baicalein triggers the translocation of Parkin and TBK1 to mitochondria to induce mitophagy. Moreover, the phosphorylation of TBK1 at Ser172 and ubiquitin at Ser65 is shown to trigger mitophagy in baicalein-treated cells. Furthermore, two specific autophagy cargo receptors, NDP52 and OPTN, that function in baicalein-activated mitophagy are identified. Taken together, these findings not only delineate the molecular process of Parkin-dependent mitophagy in liver cells, but also reveal baicalein as a novel inducer of hepatic mitophagy.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry & Molecular Biology and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-W.C.); (Y.-C.H.)
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Correspondence: ; Tel.: +886-3-2118800 (ext. 5115); Fax: +886-3-211-8700
| | - Chih-Wei Chang
- Department of Biochemistry & Molecular Biology and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-W.C.); (Y.-C.H.)
| | - Yuan-Chao Hsiao
- Department of Biochemistry & Molecular Biology and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (C.-W.C.); (Y.-C.H.)
| |
Collapse
|
33
|
Chang HY, Yang WY. Golgi quality control and autophagy. IUBMB Life 2022; 74:361-370. [PMID: 35274438 DOI: 10.1002/iub.2611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 02/12/2022] [Indexed: 11/09/2022]
Abstract
Organelles can easily be disrupted by intracellular and extracellular factors. Studies on ER and mitochondria indicate that a wide range of responses are elicited upon organelle disruption. One response thought to be of particular importance is autophagy. Cells can target entire organelles into autophagosomes for removal. This wholesale nature makes autophagy a robust means for eliminating compromised organelles. Recently, it was demonstrated that the Golgi apparatus is a substrate of autophagy. On the other hand, various reports have shown that components traffic away from the Golgi for elimination in an autophagosome-independent manner when the Golgi apparatus is stressed. Future studies will reveal how these different pieces of machinery coordinate to drive Golgi degradation. Quantitative measurements will be needed to determine how much autophagy contributes to the maintenance of the Golgi apparatus.
Collapse
Affiliation(s)
- Hsiang-Yi Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Wei Yuan Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.,Institute of Biochemical Sciences, College of Life Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
34
|
Mitophagy in Traumatic Brain Injury: A New Target for Therapeutic Intervention. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4906434. [PMID: 35126814 PMCID: PMC8813270 DOI: 10.1155/2022/4906434] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/31/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) contributes to death, and disability worldwide more than any other traumatic insult and damage to cellular components including mitochondria leads to the impairment of cellular functions and brain function. In neurons, mitophagy, autophagy-mediated degradation of damaged mitochondria, is a key process in cellular quality control including mitochondrial homeostasis and energy supply and plays a fundamental role in neuronal survival and health. Conversely, defective mitophagy leads to the accumulation of damaged mitochondria and cellular dysfunction, contributing to inflammation, oxidative stress, and neuronal cell death. Therefore, an extensive characterization of mitophagy-related protective mechanisms, taking into account the complex mechanisms by which each molecular player is connected to the others, may provide a rationale for the development of new therapeutic strategies in TBI patients. Here, we discuss the contribution of defective mitophagy in TBI, and the underlying molecular mechanisms of mitophagy in inflammation, oxidative stress, and neuronal cell death highlight novel therapeutics based on newly discovered mitophagy-inducing strategies.
Collapse
|
35
|
Iriondo MN, Etxaniz A, Antón Z, Montes LR, Alonso A. Molecular and mesoscopic geometries in autophagosome generation. A review. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183731. [PMID: 34419487 DOI: 10.1016/j.bbamem.2021.183731] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 01/18/2023]
Abstract
Autophagy is an essential process in cell self-repair and survival. The centre of the autophagic event is the generation of the so-called autophagosome (AP), a vesicle surrounded by a double membrane (two bilayers). The AP delivers its cargo to a lysosome, for degradation and re-use of the hydrolysis products as new building blocks. AP formation is a very complex event, requiring dozens of specific proteins, and involving numerous instances of membrane biogenesis and architecture, including membrane fusion and fission. Many stages of AP generation can be rationalised in terms of curvature, both the molecular geometry of lipids interpreted in terms of 'intrinsic curvature', and the overall mesoscopic curvature of the whole membrane, as observed with microscopy techniques. The present contribution intends to bring together the worlds of biophysics and cell biology of autophagy, in the hope that the resulting cross-pollination will generate abundant fruit.
Collapse
Affiliation(s)
- Marina N Iriondo
- Instituto Biofisika (CSIC, UPV/EHU) and Departamento de Bioquímica y Biología Molecular, Universidad del País Vasco, 48940 Leioa, Spain
| | - Asier Etxaniz
- Instituto Biofisika (CSIC, UPV/EHU) and Departamento de Bioquímica y Biología Molecular, Universidad del País Vasco, 48940 Leioa, Spain
| | - Zuriñe Antón
- Instituto Biofisika (CSIC, UPV/EHU) and Departamento de Bioquímica y Biología Molecular, Universidad del País Vasco, 48940 Leioa, Spain
| | - L Ruth Montes
- Instituto Biofisika (CSIC, UPV/EHU) and Departamento de Bioquímica y Biología Molecular, Universidad del País Vasco, 48940 Leioa, Spain
| | - Alicia Alonso
- Instituto Biofisika (CSIC, UPV/EHU) and Departamento de Bioquímica y Biología Molecular, Universidad del País Vasco, 48940 Leioa, Spain.
| |
Collapse
|
36
|
A Decade of Mighty Lipophagy: What We Know and What Facts We Need to Know? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5539161. [PMID: 34777688 PMCID: PMC8589519 DOI: 10.1155/2021/5539161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/30/2021] [Accepted: 10/15/2021] [Indexed: 12/24/2022]
Abstract
Lipids are integral cellular components that act as substrates for energy provision, signaling molecules, and essential constituents of biological membranes along with a variety of other biological functions. Despite their significance, lipid accumulation may result in lipotoxicity, impair autophagy, and lysosomal function that may lead to certain diseases and metabolic syndromes like obesity and even cell death. Therefore, these lipids are continuously recycled and redistributed by the process of selective autophagy specifically termed as lipophagy. This selective form of autophagy employs lysosomes for the maintenance of cellular lipid homeostasis. In this review, we have reviewed the current literature about how lipid droplets (LDs) are recruited towards lysosomes, cross-talk between a variety of autophagy receptors present on LD surface and lysosomes, and lipid hydrolysis by lysosomal enzymes. In addition to it, we have tried to answer most of the possible questions related to lipophagy regulation at different levels. Moreover, in the last part of this review, we have discussed some of the pathological states due to the accumulation of these LDs and their possible treatments under the light of currently available findings.
Collapse
|
37
|
Macroautophagy and Mitophagy in Neurodegenerative Disorders: Focus on Therapeutic Interventions. Biomedicines 2021; 9:biomedicines9111625. [PMID: 34829854 PMCID: PMC8615936 DOI: 10.3390/biomedicines9111625] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
Macroautophagy, a quality control mechanism, is an evolutionarily conserved pathway of lysosomal degradation of protein aggregates, pathogens, and damaged organelles. As part of its vital homeostatic role, macroautophagy deregulation is associated with various human disorders, including neurodegenerative diseases. There are several lines of evidence that associate protein misfolding and mitochondrial dysfunction in the etiology of Alzheimer’s, Parkinson’s, and Huntington’s diseases. Macroautophagy has been implicated in the degradation of different protein aggregates such as Aβ, tau, alpha-synuclein (α-syn), and mutant huntingtin (mHtt) and in the clearance of dysfunctional mitochondria. Taking these into consideration, targeting autophagy might represent an effective therapeutic strategy to eliminate protein aggregates and to improve mitochondrial function in these disorders. The present review describes our current understanding on the role of macroautophagy in neurodegenerative disorders and focuses on possible strategies for its therapeutic modulation.
Collapse
|
38
|
He L, Qian X, Cui Y. Advances in ER-Phagy and Its Diseases Relevance. Cells 2021; 10:cells10092328. [PMID: 34571977 PMCID: PMC8465915 DOI: 10.3390/cells10092328] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/27/2021] [Accepted: 09/01/2021] [Indexed: 01/18/2023] Open
Abstract
As an important form of selective autophagy in cells, ER-phagy (endoplasmic reticulum-selective autophagy), the autophagic degradation of endoplasmic reticulum (ER), degrades ER membranes and proteins to maintain cellular homeostasis. The relationship between ER-phagy and human diseases, including neurodegenerative disorders, cancer, and other metabolic diseases has been unveiled by extensive research in recent years. Starting with the catabolic process of ER-phagy and key mediators in this pathway, this paper reviews the advances in the mechanism of ER-phagy and its diseases relevance. We hope to provide some enlightenment for further study on ER-phagy and the development of novel therapeutic strategies for related diseases.
Collapse
Affiliation(s)
- Lingang He
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China; (L.H.); (X.Q.)
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Xuehong Qian
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China; (L.H.); (X.Q.)
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Yixian Cui
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China; (L.H.); (X.Q.)
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
- Correspondence: ; Tel.: +86-27-87267099
| |
Collapse
|
39
|
Puglisi-Allegra S, Ruggieri S, Fornai F. Translational evidence for lithium-induced brain plasticity and neuroprotection in the treatment of neuropsychiatric disorders. Transl Psychiatry 2021; 11:366. [PMID: 34226487 PMCID: PMC8257731 DOI: 10.1038/s41398-021-01492-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence indicates lithium (Li+) efficacy in neuropsychiatry, pointing to overlapping mechanisms that occur within distinct neuronal populations. In fact, the same pathway depending on which circuitry operates may fall in the psychiatric and/or neurological domains. Li+ restores both neurotransmission and brain structure unveiling that psychiatric and neurological disorders share common dysfunctional molecular and morphological mechanisms, which may involve distinct brain circuitries. Here an overview is provided concerning the therapeutic/neuroprotective effects of Li+ in different neuropsychiatric disorders to highlight common molecular mechanisms through which Li+ produces its mood-stabilizing effects and to what extent these overlap with plasticity in distinct brain circuitries. Li+ mood-stabilizing effects are evident in typical bipolar disorder (BD) characterized by a cyclic course of mania or hypomania followed by depressive episodes, while its efficacy is weaker in the opposite pattern. We focus here on neural adaptations that may underlie psychostimulant-induced psychotic development and to dissect, through the sensitization process, which features are shared in BD and other psychiatric disorders, including schizophrenia. The multiple functions of Li+ highlighted here prove its exceptional pharmacology, which may help to elucidate its mechanisms of action. These may serve as a guide toward a multi-drug strategy. We propose that the onset of sensitization in a specific BD subtype may predict the therapeutic efficacy of Li+. This model may help to infer in BD which molecular mechanisms are relevant to the therapeutic efficacy of Li+.
Collapse
Affiliation(s)
| | | | - Francesco Fornai
- IRCCS Neuromed, Via Atinense 18, 86077, Pozzilli (IS), Italy.
- Human Anatomy, Department of Translational Research and New technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126, Pisa (PI), Italy.
| |
Collapse
|
40
|
Limanaqi F, Busceti CL, Celli R, Biagioni F, Fornai F. Autophagy as a gateway for the effects of methamphetamine: From neurotransmitter release and synaptic plasticity to psychiatric and neurodegenerative disorders. Prog Neurobiol 2021; 204:102112. [PMID: 34171442 DOI: 10.1016/j.pneurobio.2021.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/27/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
As a major eukaryotic cell clearing machinery, autophagy grants cell proteostasis, which is key for neurotransmitter release, synaptic plasticity, and neuronal survival. In line with this, besides neuropathological events, autophagy dysfunctions are bound to synaptic alterations that occur in mental disorders, and early on, in neurodegenerative diseases. This is also the case of methamphetamine (METH) abuse, which leads to psychiatric disturbances and neurotoxicity. While consistently altering the autophagy machinery, METH produces behavioral and neurotoxic effects through molecular and biochemical events that can be recapitulated by autophagy blockade. These consist of altered physiological dopamine (DA) release, abnormal stimulation of DA and glutamate receptors, as well as oxidative, excitotoxic, and neuroinflammatory events. Recent molecular insights suggest that METH early impairs the autophagy machinery, though its functional significance remains to be investigated. Here we discuss evidence suggesting that alterations of DA transmission and autophagy are intermingled within a chain of events underlying behavioral alterations and neurodegenerative phenomena produced by METH. Understanding how METH alters the autophagy machinery is expected to provide novel insights into the neurobiology of METH addiction sharing some features with psychiatric disorders and parkinsonism.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy
| | | | - Roberta Celli
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56126, Pisa, PI, Italy; IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy.
| |
Collapse
|
41
|
Contextualizing Autophagy during Gametogenesis and Preimplantation Embryonic Development. Int J Mol Sci 2021; 22:ijms22126313. [PMID: 34204653 PMCID: PMC8231133 DOI: 10.3390/ijms22126313] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 01/05/2023] Open
Abstract
Mammals face environmental stressors throughout their lifespan, which may jeopardize cellular homeostasis. Hence, these organisms have acquired mechanisms to cope with stressors by sensing, repairing the damage, and reallocating resources to increase the odds of long-term survival. Autophagy is a pro-survival lysosome-mediated cytoplasm degradation pathway for organelle and macromolecule recycling. Furthermore, autophagy efflux increases, and this pathway becomes idiosyncratic depending upon developmental and environmental contexts. Mammalian germ cells and preimplantation embryos are attractive models for dissecting autophagy due to their metastable phenotypes during differentiation and exposure to varying environmental cues. The aim of this review is to explore autophagy during mammalian gametogenesis, fertilization and preimplantation embryonic development by contemplating its physiological role during development, under key stressors, and within the scope of assisted reproduction technologies.
Collapse
|
42
|
Gong CY, Zhang HH. Autophagy as a potential therapeutic target in intervertebral disc degeneration. Life Sci 2021; 273:119266. [PMID: 33631177 DOI: 10.1016/j.lfs.2021.119266] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/08/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
Autophagy is an evolutionarily conserved intracellular recirculation system that delivers cytoplasmic content to lysosomes for degradation, thereby maintaining metabolism and homeostasis. Recent studies have found that autophagy plays a dual role in intervertebral disc degeneration (IDD). Most studies have shown that inducing autophagy can slow down the process of IDD. A few studies have shown that extensive autophagy activation-mediated apoptosis accelerates IDD. In this review, we describe the pathophysiological characteristics of intervertebral disc (IVD), the mechanism of autophagy and the application of regulating autophagy in the treatment of IDD, hoping to provide a certain theoretical basis for the biotherapy of IDD.
Collapse
Affiliation(s)
- Chao-Yang Gong
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou 730000, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- Lanzhou University Second Hospital, 82 Cuiying Men, Lanzhou 730000, PR China.
| |
Collapse
|
43
|
Nguyen TN, Lazarou M. Plant mitophagy: Beware of Friendly or you might get eaten. Curr Biol 2021; 31:R457-R458. [PMID: 33974877 DOI: 10.1016/j.cub.2021.02.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mitophagy is a selective garbage disposal pathway that rids cells of excess or damaged mitochondria. In this issue, Ma et al. uncover a role for Friendly in driving depolarisation-induced mitophagy in plants and highlight a physiological role for mitophagy during plant development.
Collapse
Affiliation(s)
- Thanh Ngoc Nguyen
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Michael Lazarou
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia.
| |
Collapse
|
44
|
Herpesvirus Regulation of Selective Autophagy. Viruses 2021; 13:v13050820. [PMID: 34062931 PMCID: PMC8147283 DOI: 10.3390/v13050820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/18/2022] Open
Abstract
Selective autophagy has emerged as a key mechanism of quality and quantity control responsible for the autophagic degradation of specific subcellular organelles and materials. In addition, a specific type of selective autophagy (xenophagy) is also activated as a line of defense against invading intracellular pathogens, such as viruses. However, viruses have evolved strategies to counteract the host’s antiviral defense and even to activate some proviral types of selective autophagy, such as mitophagy, for their successful infection and replication. This review discusses the current knowledge on the regulation of selective autophagy by human herpesviruses.
Collapse
|
45
|
Abstract
Synthesis and degradation of cellular constituents must be balanced to maintain cellular homeostasis, especially during adaptation to environmental stress. The role of autophagy in the degradation of proteins and organelles is well-characterized. However, autophagy-mediated RNA degradation in response to stress and the potential preference of specific RNAs to undergo autophagy-mediated degradation have not been examined. In this study, we demonstrate selective mRNA degradation by rapamycin-induced autophagy in yeast. Profiling of mRNAs from the vacuole reveals that subsets of mRNAs, such as those encoding amino acid biosynthesis and ribosomal proteins, are preferentially delivered to the vacuole by autophagy for degradation. We also reveal that autophagy-mediated mRNA degradation is tightly coupled with translation by ribosomes. Genome-wide ribosome profiling suggested a high correspondence between ribosome association and targeting to the vacuole. We propose that autophagy-mediated mRNA degradation is a unique and previously-unappreciated function of autophagy that affords post-transcriptional gene regulation.
Collapse
|
46
|
Onishi M, Okamoto K. Mitochondrial clearance: mechanisms and roles in cellular fitness. FEBS Lett 2021; 595:1239-1263. [PMID: 33615465 DOI: 10.1002/1873-3468.14060] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/29/2021] [Accepted: 02/12/2021] [Indexed: 12/14/2022]
Abstract
Mitophagy is one of the selective autophagy pathways that catabolizes dysfunctional or superfluous mitochondria. Under mitophagy-inducing conditions, mitochondria are labeled with specific molecular landmarks that recruit the autophagy machinery to the surface of mitochondria, enclosed into autophagosomes, and delivered to lysosomes (vacuoles in yeast) for degradation. As damaged mitochondria are the major sources of reactive oxygen species, mitophagy is critical for mitochondrial quality control and cellular health. Moreover, appropriate control of mitochondrial quantity via mitophagy is vital for the energy supply-demand balance in cells and whole organisms, cell differentiation, and developmental programs. Thus, it seems conceivable that defects in mitophagy could elicit pleiotropic pathologies such as excess inflammation, tissue injury, neurodegeneration, and aging. In this review, we will focus on the molecular basis and physiological relevance of mitophagy, and potential of mitophagy as a therapeutic target to overcome such disorders.
Collapse
Affiliation(s)
- Mashun Onishi
- Laboratory of Mitochondrial Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Koji Okamoto
- Laboratory of Mitochondrial Dynamics, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
47
|
Liu L, Li Y, Wang J, Zhang D, Wu H, Li W, Wei H, Ta N, Fan Y, Liu Y, Wang X, Wang J, Pan X, Liao X, Zhu Y, Chen Q. Mitophagy receptor FUNDC1 is regulated by PGC-1α/NRF1 to fine tune mitochondrial homeostasis. EMBO Rep 2021; 22:e50629. [PMID: 33554448 DOI: 10.15252/embr.202050629] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 11/27/2020] [Accepted: 12/15/2020] [Indexed: 12/28/2022] Open
Abstract
Mitophagy is an essential cellular autophagic process that selectively removes superfluous and damaged mitochondria, and it is coordinated with mitochondrial biogenesis to fine tune the quantity and quality of mitochondria. Coordination between these two opposing processes to maintain the functional mitochondrial network is of paramount importance for normal cellular and organismal metabolism. However, the underlying mechanism is not completely understood. Here we report that PGC-1α and nuclear respiratory factor 1 (NRF1), master regulators of mitochondrial biogenesis and metabolic adaptation, also transcriptionally upregulate the gene encoding FUNDC1, a previously characterized mitophagy receptor, in response to cold stress in brown fat tissue. NRF1 binds to the classic consensus site in the promoter of Fundc1 to upregulate its expression and to enhance mitophagy through its interaction with LC3. Specific knockout of Fundc1 in BAT results in reduced mitochondrial turnover and accumulation of functionally compromised mitochondria, leading to impaired adaptive thermogenesis. Our results demonstrate that FUNDC1-dependent mitophagy is directly coupled with mitochondrial biogenesis through the PGC-1α/NRF1 pathway, which dictates mitochondrial quantity, quality, and turnover and contributes to adaptive thermogenesis.
Collapse
Affiliation(s)
- Lei Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yanjun Li
- College of Life Sciences, Nankai University, Tianjin, China
| | - Jianing Wang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Di Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, Nankai University, Tianjin, China
| | - Hao Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wenhui Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Huifang Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Na Ta
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuyuan Fan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yujiao Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaohui Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jun Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xin Pan
- Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Xudong Liao
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yushan Zhu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Quan Chen
- College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
48
|
Onishi M, Yamano K, Sato M, Matsuda N, Okamoto K. Molecular mechanisms and physiological functions of mitophagy. EMBO J 2021; 40:e104705. [PMID: 33438778 PMCID: PMC7849173 DOI: 10.15252/embj.2020104705] [Citation(s) in RCA: 688] [Impact Index Per Article: 172.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 08/03/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
Degradation of mitochondria via a selective form of autophagy, named mitophagy, is a fundamental mechanism conserved from yeast to humans that regulates mitochondrial quality and quantity control. Mitophagy is promoted via specific mitochondrial outer membrane receptors, or ubiquitin molecules conjugated to proteins on the mitochondrial surface leading to the formation of autophagosomes surrounding mitochondria. Mitophagy-mediated elimination of mitochondria plays an important role in many processes including early embryonic development, cell differentiation, inflammation, and apoptosis. Recent advances in analyzing mitophagy in vivo also reveal high rates of steady-state mitochondrial turnover in diverse cell types, highlighting the intracellular housekeeping role of mitophagy. Defects in mitophagy are associated with various pathological conditions such as neurodegeneration, heart failure, cancer, and aging, further underscoring the biological relevance. Here, we review our current molecular understanding of mitophagy, and its physiological implications, and discuss how multiple mitophagy pathways coordinately modulate mitochondrial fitness and populations.
Collapse
Affiliation(s)
- Mashun Onishi
- Laboratory of Mitochondrial DynamicsGraduate School of Frontier BiosciencesOsaka UniversitySuitaJapan
| | - Koji Yamano
- The Ubiquitin ProjectTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Miyuki Sato
- Laboratory of Molecular Membrane BiologyInstitute for Molecular and Cellular RegulationGunma UniversityMaebashiJapan
| | - Noriyuki Matsuda
- The Ubiquitin ProjectTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Koji Okamoto
- Laboratory of Mitochondrial DynamicsGraduate School of Frontier BiosciencesOsaka UniversitySuitaJapan
| |
Collapse
|
49
|
Autophagy status as a gateway for stress-induced catecholamine interplay in neurodegeneration. Neurosci Biobehav Rev 2021; 123:238-256. [PMID: 33497785 DOI: 10.1016/j.neubiorev.2021.01.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 12/13/2022]
Abstract
The catecholamine-containing brainstem nuclei locus coeruleus (LC) and ventral tegmental area (VTA) are critically involved in stress responses. Alterations of catecholamine systems during chronic stress may contribute to neurodegeneration, including cognitive decline. Stress-related catecholamine alterations, while contributing to anxiety and depression, might accelerate neuronal degeneration by increasing the formation of toxic dopamine and norepinephrine by-products. These, in turn, may impair proteostasis within a variety of cortical and subcortical areas. In particular, the molecular events governing neurotransmission, neuroplasticity, and proteostasis within LC and VTA affect a variety of brain areas. Therefore, we focus on alterations of autophagy machinery in these nuclei as a relevant trigger in this chain of events. In fact, these catecholamine-containing areas are mostly prone to autophagy-dependent neurodegeneration. Thus, we propose a dynamic hypothesis according to which stress-induced autophagy alterations within the LC-VTA network foster a cascade towards early neurodegeneration within these nuclei.
Collapse
|
50
|
Zheng Y, Huang C, Lu L, Yu K, Zhao J, Chen M, Liu L, Sun Q, Lin Z, Zheng J, Chen J, Zhang J. STOML2 potentiates metastasis of hepatocellular carcinoma by promoting PINK1-mediated mitophagy and regulates sensitivity to lenvatinib. J Hematol Oncol 2021; 14:16. [PMID: 33446239 PMCID: PMC7807703 DOI: 10.1186/s13045-020-01029-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/25/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dysregulation of both mitochondrial biogenesis and mitophagy is critical to sustain oncogenic signaling pathways. However, the mechanism of mitophagy in promoting hepatocellular carcinoma (HCC) progression remains poorly understood. In this study, we investigated the clinical significance and biological involvement of mitochondrial inner membrane protein STOML2 in HCC. METHODS STOML2 was identified by gene expression profiles of HCC tissues and was measured in tissue microarray and cell lines. Gain/loss-of-function experiment was applied to study the biological function of STOML2 in HCC. Flow cytometry, Western blotting, laser confocal microscopy, transmission electron microscopy, and co-immunoprecipitation were used to detect and analyze mitophagy. ChIP and luciferase reporter assay were conducted to evaluate the relationship between STOML2 and HIF-1α. The sensitivity to lenvatinib was assessed in HCC both in vitro and in vivo. RESULTS Increased expression of STOML2 was found in HCC compared with paired peritumoral tissues. It was more significant in HCC with metastasis and correlated with worse overall survival and higher probability of recurrence after hepatectomy. Upregulation of STOML2 accelerated HCC cells colony formation, migration and invasion. Mechanically, TCGA dataset-based analysis showed enrichment of autophagy-related pathways in STOML2 highly-expressed HCC. Next, STOML2 was demonstrated to interact and stabilize PINK1 under cellular stress, amplify PINK1-Parkin-mediated mitophagy and then promote HCC growth and metastasis. Most interestingly, HIF-1α was upregulated and transcriptionally increased STOML2 expression in HCC cells under the treatment of lenvatinib. Furthermore, higher sensitivity to lenvatinib was found in HCC cells when STOML2 was downregulated. Combination therapy with lenvatinib and mitophagy inhibitor hydroxychloroquine obtained best efficacy. CONCLUSIONS Our findings suggested that STOML2 could amplify mitophagy through interacting and stabilizing PINK1, which promote HCC metastasis and modulate the response of HCC to lenvatinib. Combinations of pharmacologic inhibitors that concurrently block both angiogenesis and mitophagy may serve as an effective treatment for HCC.
Collapse
Affiliation(s)
- Yahui Zheng
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Chong Huang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Lu Lu
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Kangkang Yu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jing Zhao
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Mingquan Chen
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Lu Liu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qingfeng Sun
- Department of Infectious Diseases, Ruian People's Hospital, Ruian, 325200, China
| | - Zhifei Lin
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jianming Zheng
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jinhong Chen
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jubo Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Center of Liver Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|