1
|
Li J, Prange R, Lu M. ESCRT Machinery in HBV Life Cycle: Dual Roles in Autophagy and Membrane Dynamics for Viral Pathogenesis. Cells 2025; 14:603. [PMID: 40277928 PMCID: PMC12025488 DOI: 10.3390/cells14080603] [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: 01/30/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
The endosomal sorting complexes required for transport (ESCRT) comprise a fundamental cellular machinery with remarkable versatility in membrane remodeling. It is multifunctional in the multivesicular body (MVB) biogenesis, exosome formation and secretion, virus budding, cytokinesis, plasma membrane repair, neuron pruning, and autophagy. ESCRT's involvement in cellular mechanisms extends beyond basic membrane trafficking. By directly interacting with autophagy-related (ATG) proteins and facilitating autophagosome-lysosome fusion, ESCRT ensures cellular homeostasis. Dysregulation in ESCRT function has been implicated in cancer, neurodegenerative disorders, and infectious diseases, underscoring its critical role in numerous pathologies. Hepatitis B virus (HBV) is an enveloped virus that exploits ESCRT and autophagy pathways for viral replication, assembly, and secretion. This review synthesizes recent mechanistic insights into ESCRT's multifaceted roles, particularly focusing on its interactions with autophagy formation and the HBV lifecycle.
Collapse
Affiliation(s)
- Jia Li
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany;
| | - Reinhild Prange
- Institute for Virology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany;
| |
Collapse
|
2
|
Chen X, Perry S, Fan Z, Wang B, Loxterkamp E, Wang S, Hu J, Dickman D, Han C. Tissue-specific knockout in the Drosophila neuromuscular system reveals ESCRT's role in formation of synapse-derived extracellular vesicles. PLoS Genet 2024; 20:e1011438. [PMID: 39388480 PMCID: PMC11495600 DOI: 10.1371/journal.pgen.1011438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 10/22/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Tissue-specific gene knockout by CRISPR/Cas9 is a powerful approach for characterizing gene functions during development. However, this approach has not been successfully applied to most Drosophila tissues, including the Drosophila neuromuscular junction (NMJ). To expand tissue-specific CRISPR to this powerful model system, here we present a CRISPR-mediated tissue-restricted mutagenesis (CRISPR-TRiM) toolkit for knocking out genes in motoneurons, muscles, and glial cells. We validated the efficacy of CRISPR-TRiM by knocking out multiple genes in each tissue, demonstrated its orthogonal use with the Gal4/UAS binary expression system, and showed simultaneous knockout of multiple redundant genes. We used CRISPR-TRiM to discover an essential role for SNARE components in NMJ maintenance. Furthermore, we demonstrate that the canonical ESCRT pathway suppresses NMJ bouton growth by downregulating retrograde Gbb signaling. Lastly, we found that axon termini of motoneurons rely on ESCRT-mediated intra-axonal membrane trafficking to release extracellular vesicles at the NMJ. Thus, we have successfully developed an NMJ CRISPR mutagenesis approach which we used to reveal genes important for NMJ structural plasticity.
Collapse
Affiliation(s)
- Xinchen Chen
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, California, United States of America
| | - Ziwei Fan
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Bei Wang
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Elizabeth Loxterkamp
- Department of Neurobiology, University of Southern California, Los Angeles, California, United States of America
| | - Shuran Wang
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Jiayi Hu
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, California, United States of America
| | - Chun Han
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
3
|
Cicchetti R, Basconi M, Litterio G, Mascitti M, Tamborino F, Orsini A, Digiacomo A, Ferro M, Schips L, Marchioni M. Advances in Molecular Mechanisms of Kidney Disease: Integrating Renal Tumorigenesis of Hereditary Cancer Syndrome. Int J Mol Sci 2024; 25:9060. [PMID: 39201746 PMCID: PMC11355026 DOI: 10.3390/ijms25169060] [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: 07/11/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Renal cell carcinoma (RCC) comprises various histologically distinct subtypes, each characterized by specific genetic alterations, necessitating individualized management and treatment strategies for each subtype. An exhaustive search of the PubMed database was conducted without any filters or restrictions. Inclusion criteria encompassed original English articles focusing on molecular mechanisms of kidney cancer. On the other hand, all non-original articles and articles published in any language other than English were excluded. Hereditary kidney cancer represents 5-8% of all kidney cancer cases and is associated with syndromes such as von Hippel-Lindau syndrome, Birt-Hogg-Dubè syndrome, succinate dehydrogenase-deficient renal cell cancer syndrome, tuberous sclerosis complex, hereditary papillary renal cell carcinoma, fumarate hydratase deficiency syndrome, BAP1 tumor predisposition syndrome, and other uncommon hereditary cancer syndromes. These conditions are characterized by distinct genetic mutations and related extra-renal symptoms. The majority of renal cell carcinoma predispositions stem from loss-of-function mutations in tumor suppressor genes. These mutations promote malignant advancement through the somatic inactivation of the remaining allele. This review aims to elucidate the main molecular mechanisms underlying the pathophysiology of major syndromes associated with renal cell carcinoma. By providing a comprehensive overview, it aims to facilitate early diagnosis and to highlight the principal therapeutic options available.
Collapse
Affiliation(s)
- Rossella Cicchetti
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Martina Basconi
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Giulio Litterio
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Marco Mascitti
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Flavia Tamborino
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Angelo Orsini
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Alessio Digiacomo
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 20141 Milan, Italy;
| | - Luigi Schips
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| | - Michele Marchioni
- Department of Medical Oral and Biotechnological Science, Università degli Studi “G. d’Annunzio” of Chieti, 66100 Chieti, Italy; (R.C.); (M.B.); (G.L.); (M.M.); (F.T.); (A.O.); (A.D.); (M.M.)
| |
Collapse
|
4
|
Hermosilla Aguayo V, Martin P, Tian N, Zheng J, Aho R, Losa M, Selleri L. ESCRT-dependent control of craniofacial morphogenesis with concomitant perturbation of NOTCH signaling. Dev Biol 2023; 503:25-42. [PMID: 37573008 DOI: 10.1016/j.ydbio.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Craniofacial development is orchestrated by transcription factor-driven regulatory networks, epigenetic modifications, and signaling pathways. Signaling molecules and their receptors rely on endo-lysosomal trafficking to prevent accumulation on the plasma membrane. ESCRT (Endosomal Sorting Complexes Required for Transport) machinery is recruited to endosomal membranes enabling degradation of such endosomal cargoes. Studies in vitro and in invertebrate models established the requirements of the ESCRT machinery in membrane remodeling, endosomal trafficking, and lysosomal degradation of activated membrane receptors. However, investigations during vertebrate development have been scarce. By ENU-induced mutagenesis, we isolated a mouse line, Vps25ENU/ENU, carrying a hypomorphic allele of the ESCRT-II component Vps25, with craniofacial anomalies resembling features of human congenital syndromes. Here, we assessed the spatiotemporal dynamics of Vps25 and additional ESCRT-encoding genes during murine development. We show that these genes are ubiquitously expressed although enriched in discrete domains of the craniofacial complex, heart, and limbs. ESCRT-encoding genes, including Vps25, are expressed in both cranial neural crest-derived mesenchyme and epithelium. Unlike constitutive ESCRT mutants, Vps25ENU/ENU embryos display late lethality. They exhibit hypoplastic lower jaw, stunted snout, dysmorphic ear pinnae, and secondary palate clefting. Thus, we provide the first evidence for critical roles of ESCRT-II in craniofacial morphogenesis and report perturbation of NOTCH signaling in craniofacial domains of Vps25ENU/ENU embryos. Given the known roles of NOTCH signaling in the developing cranium, and notably the lower jaw, we propose that the NOTCH pathway partly mediates the craniofacial defects of Vps25ENU/ENU mouse embryos.
Collapse
Affiliation(s)
- Viviana Hermosilla Aguayo
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Peter Martin
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nuo Tian
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - James Zheng
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Robert Aho
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marta Losa
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Licia Selleri
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Dept of Orofacial Sciences and Dept of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
5
|
Yang CC, Zheng CC, Luo Y, Guo KW, Gao D, Zhang L, Li L, Zhang L. Cornel Iridoid Glycoside and Its Effective Component Regulate ATPase Vps4A/JNK to Alleviate Autophagy Deficit with Autophagosome Accumulation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1599-1615. [PMID: 35786171 DOI: 10.1142/s0192415x22500677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Improving autophagy-lysosome fusion has been considered a key method in the treatment of Alzheimer's disease (AD). Cornel iridoid glycoside (CIG) is extracted from Cornus officinalis and has been shown to promote the clearance of tau oligomers via the autophagy pathway. However, the mechanisms of CIG on autophagy deficits are not understood. Here, we found autophagy deficit and tau aggregation in the brains of P301S tau transgenic mice and MAPT cells edited using CRISPR-Cas9 technology. CIG decreased tau aggregation and alleviated autophagic markers involving the JNK/Beclin-1 signaling pathway which demonstrated CIG that might enhance lysosome formation by upregulating ATPase Vps4A expression. Knocking down VPS4A increased autophagosome accumulation and attenuated the effect of CIG on p62. In addition, CIG had no effect on tau oligomers but still inhibited the level of tau monomer in VPS4A knockout cells. The effective component (Sweroside, SWE) of CIG attenuated tau oligomers accumulation and increased Vps4A level but not CHMP2B. SWE could not change the level of tau oligomers in VPS4A knockout cells. In conclusion, CIG suppressed autophagosome accumulation by regulating the ATPase Vps4A/JNK. SWE is a core of active factors of CIG in Vps4A regulation. These findings suggest CIG may be a potential drug in AD treatment.
Collapse
Affiliation(s)
- Cui-Cui Yang
- Department of Pharmacy, Xuanwu Hospital of Capital, Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, 45 Changchun St, Xicheng District, Beijing 100053, P. R. China
| | - Ceng-Ceng Zheng
- Department of Pharmacy, Xuanwu Hospital of Capital, Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, 45 Changchun St, Xicheng District, Beijing 100053, P. R. China
| | - Yi Luo
- Department of Pharmacy, Xuanwu Hospital of Capital, Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, 45 Changchun St, Xicheng District, Beijing 100053, P. R. China
| | - Kai-Wen Guo
- Department of Pharmacy, Xuanwu Hospital of Capital, Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, 45 Changchun St, Xicheng District, Beijing 100053, P. R. China
| | - Dan Gao
- Department of Pharmacy, Xuanwu Hospital of Capital, Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, 45 Changchun St, Xicheng District, Beijing 100053, P. R. China
| | - Li Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital, Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, 45 Changchun St, Xicheng District, Beijing 100053, P. R. China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital, Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, 45 Changchun St, Xicheng District, Beijing 100053, P. R. China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital, Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, 45 Changchun St, Xicheng District, Beijing 100053, P. R. China
| |
Collapse
|
6
|
Navarro-Hortal MD, Romero-Márquez JM, Osta S, Jiménez-Trigo V, Muñoz-Ollero P, Varela-López A. Natural Bioactive Products and Alzheimer’s Disease Pathology: Lessons from Caenorhabditis elegans Transgenic Models. Diseases 2022; 10:diseases10020028. [PMID: 35645249 PMCID: PMC9149938 DOI: 10.3390/diseases10020028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is an age-dependent, progressive disorder affecting millions of people. Currently, the therapeutics for AD only treat the symptoms. Although they have been used to discover new products of interest for this disease, mammalian models used to investigate the molecular determinants of this disease are often prohibitively expensive, time-consuming and very complex. On the other hand, cell cultures lack the organism complexity involved in AD. Given the highly conserved neurological pathways between mammals and invertebrates, Caenorhabditis elegans has emerged as a powerful tool for the investigation of the pathophysiology of human AD. Numerous models of both Tau- and Aβ-induced toxicity, the two prime components observed to correlate with AD pathology and the ease of performing RNA interference for any gene in the C. elegans genome, allow for the identification of multiple therapeutic targets. The effects of many natural products in main AD hallmarks using these models suggest promising health-promoting effects. However, the way in which they exert such effects is not entirely clear. One of the reasons is that various possible therapeutic targets have not been evaluated in many studies. The present review aims to explore shared therapeutical targets and the potential of each of them for AD treatment or prevention.
Collapse
|
7
|
Visintin R, Ray SK. Specific microRNAs for Modulation of Autophagy in Spinal Cord Injury. Brain Sci 2022; 12:247. [PMID: 35204010 PMCID: PMC8870708 DOI: 10.3390/brainsci12020247] [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: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/06/2022] [Indexed: 02/05/2023] Open
Abstract
The treatment of spinal cord injury (SCI) is currently a major challenge, with a severe lack of effective therapies for yielding meaningful improvements in function. Therefore, there is a great opportunity for the development of novel treatment strategies for SCI. The modulation of autophagy, a process by which a cell degrades and recycles unnecessary or harmful components (protein aggregates, organelles, etc.) to maintain cellular homeostasis and respond to a changing microenvironment, is thought to have potential for treating many neurodegenerative conditions, including SCI. The discovery of microRNAs (miRNAs), which are short ribonucleotide transcripts for targeting of specific messenger RNAs (mRNAs) for silencing, shows prevention of the translation of mRNAs to the corresponding proteins affecting various cellular processes, including autophagy. The number of known miRNAs and their targets continues to grow rapidly. This review article aims to explore the relationship between autophagy and SCI, specifically with the intent of identifying specific miRNAs that can be useful to modulate autophagy for neuroprotection and the improvement of functional recovery in SCI.
Collapse
Affiliation(s)
- Rhett Visintin
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA;
| | - Swapan K. Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| |
Collapse
|
8
|
Zhang A, Meng Y, Li Q, Liang Y. The endosomal sorting complex required for transport complex negatively regulates Erg6 degradation under specific glucose restriction conditions. Traffic 2021; 21:488-502. [PMID: 32378292 DOI: 10.1111/tra.12732] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022]
Abstract
Lipid droplets (LDs) are cytosolic fat storage organelles that play roles in lipid metabolism, trafficking and signaling. Breakdown of LDs in Saccharomyces cerevisiae is mainly achieved by lipolysis and lipophagy. In this study, we found that the endosomal sorting complex required for transport (ESCRT) in S. cerevisiae negatively regulated the turnover of a LD marker, Erg6, under both simplified glucose restriction (GR) and acute glucose restriction (AGR) conditions by monitoring the localization and degradation of Erg6. Loss of Vps27, Snf7 or Vps4, representative subunits of the ESCRT machinery, facilitated the delivery of Erg6-GFP to vacuoles and its degradation depending on the lipophagy protein Atg15 under simplified GR. Additionally, the lipolysis proteins Tgl3 and Tgl4 were also involved in the enhanced vacuolar localization and degradation of Erg6-GFP in vps4Δ cells. Furthermore, we found that Atg14, which is required for the formation of putatively liquid-ordered (Lo) membrane domains on the vacuole that act as preferential internalization sites for LDs, abundantly localized to vacuolar membranes in ESCRT mutants. Most importantly, the depletion or overexpression of Atg14 correspondingly abolished or promoted the observed Erg6 degradation in ESCRT mutant cells. We propose that Atg14 together with other proteins promotes Erg6 degradation in ESCRT mutant cells under specific glucose restriction conditions. These results shed new light on the regulation of ESCRT on LD turnover.
Collapse
Affiliation(s)
- Ao Zhang
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Ying Meng
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Qunli Li
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Yongheng Liang
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
9
|
Wang N, Zhou Y, Zhao L, Wang C, Ma W, Ge G, Wang Y, Ullah I, Muhammad F, Alwayli D, Zhi D, Li H. Ferulic acid delayed amyloid β-induced pathological symptoms by autophagy pathway via a fasting-like effect in Caenorhabditis elegans. Food Chem Toxicol 2020; 146:111808. [PMID: 33045309 DOI: 10.1016/j.fct.2020.111808] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/27/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022]
Abstract
The amyloid β (Aβ) generation or aggregation plays a crucial role in Alzheimer's disease (AD). Autophagy agonists, which function as the clearance of Aβ, could be the potential drug candidates against AD. In staple food crops, ferulic acid (FA) is an enormously copious and almost ubiquitous phenolic antioxidant. In the present study, FA significantly inhibited Aβ-induced pathological symptoms of paralysis and hypersensitivity to exogenous serotonin, meanwhile restrained Aβ monomers, oligomers, and deposits in AD C. elegans. FA increased the expression of autophagy reporter LGG-1 and enhanced autophagy flux. However, the autophagy inhibitors abolished the restrictive action of FA on the worm paralysis phenotype. According to these results, FA triggered autophagy and ameliorated Aβ-induced pathological symptoms by the autophagy pathway. Moreover, FA activated the HLH-30 transcription factor to nuclear localization, which acts upstream of autophagy in fasted animals, reduced the level of lipids, but affected nor the growth of E. coli OP50, neither animal food intake behavior. These suggest that FA induced a fasting-like effect to activate the autophagy pathway. Additionally, FA ameliorated poly Q aggregations in Huntington's disease worm. Thus, FA could not only affect AD, broadly but also neurodegenerative diseases characterized by misfolded or aggregated proteins.
Collapse
Affiliation(s)
- Ningbo Wang
- School of Life Sciences, Lanzhou University, China.
| | - Yongtao Zhou
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Clinical Center for Parkinson's Disease, Capital Medical University, Beijing, China.
| | - Longhe Zhao
- School of Pharmacy, Lanzhou University, China.
| | - Caiding Wang
- School of Life Sciences, Lanzhou University, China.
| | - Wuli Ma
- School of Life Sciences, Lanzhou University, China.
| | - Guangfei Ge
- School of Life Sciences, Lanzhou University, China.
| | - Yu Wang
- School of Pharmacy, Lanzhou University, China.
| | - Inam Ullah
- School of Life Sciences, Lanzhou University, China.
| | | | | | - Dejuan Zhi
- School of Pharmacy, Lanzhou University, China.
| | - Hongyu Li
- School of Life Sciences, Lanzhou University, China; School of Pharmacy, Lanzhou University, China.
| |
Collapse
|
10
|
Subcellular Localization of ESCRT-II in the Nematode C. elegans by Correlative Light Electron Microscopy. Methods Mol Biol 2020. [PMID: 31250293 DOI: 10.1007/978-1-4939-9492-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
In this chapter, we report a protocol to perform correlative light electron microscopy (CLEM) on adult Caenorhabditis elegans. We use a specific fixation protocol, which preserves both the GFP fluorescence and the structural integrity of the samples. Thin sections are first analyzed by light microscopy to detect GFP-tagged proteins and, subsequently, with transmission electron microscopy (TEM) to characterize the ultrastructural anatomy of cells. The superimposition of light and electron images allows determining the subcellular localization of the fluorescent protein.We used CLEM to characterize the subcellular localization of the C. elegans ESCRT-II component VPS-36. VPS-36 protein localization in C. elegans muscle cell is strongly correlated with the sarcoplasmic reticulum network. Together with genetic evidences, the CLEM data support a role for ESCRT-II proteins in sarcoplasmic reticulum membrane shaping.
Collapse
|
11
|
Haeussler S, Köhler F, Witting M, Premm MF, Rolland SG, Fischer C, Chauve L, Casanueva O, Conradt B. Autophagy compensates for defects in mitochondrial dynamics. PLoS Genet 2020; 16:e1008638. [PMID: 32191694 PMCID: PMC7135339 DOI: 10.1371/journal.pgen.1008638] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 04/06/2020] [Accepted: 01/28/2020] [Indexed: 12/30/2022] Open
Abstract
Compromising mitochondrial fusion or fission disrupts cellular homeostasis; however, the underlying mechanism(s) are not fully understood. The loss of C. elegans fzo-1MFN results in mitochondrial fragmentation, decreased mitochondrial membrane potential and the induction of the mitochondrial unfolded protein response (UPRmt). We performed a genome-wide RNAi screen for genes that when knocked-down suppress fzo-1MFN(lf)-induced UPRmt. Of the 299 genes identified, 143 encode negative regulators of autophagy, many of which have previously not been implicated in this cellular quality control mechanism. We present evidence that increased autophagic flux suppresses fzo-1MFN(lf)-induced UPRmt by increasing mitochondrial membrane potential rather than restoring mitochondrial morphology. Furthermore, we demonstrate that increased autophagic flux also suppresses UPRmt induction in response to a block in mitochondrial fission, but not in response to the loss of spg-7AFG3L2, which encodes a mitochondrial metalloprotease. Finally, we found that blocking mitochondrial fusion or fission leads to increased levels of certain types of triacylglycerols and that this is at least partially reverted by the induction of autophagy. We propose that the breakdown of these triacylglycerols through autophagy leads to elevated metabolic activity, thereby increasing mitochondrial membrane potential and restoring mitochondrial and cellular homeostasis.
Collapse
Affiliation(s)
- Simon Haeussler
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Fabian Köhler
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael Witting
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Analytical Food Chemistry, Technische Universität München, Freising, Germany
| | - Madeleine F. Premm
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Christian Fischer
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
- Center for Integrated Protein Science, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Laetitia Chauve
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Olivia Casanueva
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Barbara Conradt
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
- Center for Integrated Protein Science, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
- Department of Cell and Developmental Biology, Division of Biosciences, University College London, London, United Kingdom
| |
Collapse
|
12
|
Abstract
Cellular membranes can form two principally different involutions, which either exclude or contain cytosol. The 'classical' budding reactions, such as those occurring during endocytosis or formation of exocytic vesicles, involve proteins that assemble on the cytosol-excluding face of the bud neck. Inverse membrane involution occurs in a wide range of cellular processes, supporting cytokinesis, endosome maturation, autophagy, membrane repair and many other processes. Such inverse membrane remodelling is mediated by a heteromultimeric protein machinery known as endosomal sorting complex required for transport (ESCRT). ESCRT proteins assemble on the cytosolic (or nucleoplasmic) face of the neck of the forming involution and cooperate with the ATPase VPS4 to drive membrane scission or sealing. Here, we review similarities and differences of various ESCRT-dependent processes, with special emphasis on mechanisms of ESCRT recruitment.
Collapse
|
13
|
Takahashi Y, Liang X, Hattori T, Tang Z, He H, Chen H, Liu X, Abraham T, Imamura-Kawasawa Y, Buchkovich NJ, Young MM, Wang HG. VPS37A directs ESCRT recruitment for phagophore closure. J Cell Biol 2019; 218:3336-3354. [PMID: 31519728 PMCID: PMC6781443 DOI: 10.1083/jcb.201902170] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/25/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
Takahashi et al. perform a genome-wide CRISPR screen using the HaloTag-LC3 assay to gain insight into the mechanisms of phagophore closure. They identify a role for VPS37A in coordinating the ESCRT assembly on the phagophore for membrane closure. The process of phagophore closure requires the endosomal sorting complex required for transport III (ESCRT-III) subunit CHMP2A and the AAA ATPase VPS4, but their regulatory mechanisms remain unknown. Here, we establish a FACS-based HaloTag-LC3 autophagosome completion assay to screen a genome-wide CRISPR library and identify the ESCRT-I subunit VPS37A as a critical component for phagophore closure. VPS37A localizes on the phagophore through the N-terminal putative ubiquitin E2 variant domain, which is found to be required for autophagosome completion but dispensable for ESCRT-I complex formation and the degradation of epidermal growth factor receptor in the multivesicular body pathway. Notably, loss of VPS37A abrogates the phagophore recruitment of the ESCRT-I subunit VPS28 and CHMP2A, whereas inhibition of membrane closure by CHMP2A depletion or VPS4 inhibition accumulates VPS37A on the phagophore. These observations suggest that VPS37A coordinates the recruitment of a unique set of ESCRT machinery components for phagophore closure in mammalian cells.
Collapse
Affiliation(s)
| | - Xinwen Liang
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Tatsuya Hattori
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Zhenyuan Tang
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA
| | - Haiyan He
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Han Chen
- Microscopy Imaging Facility, Penn State College of Medicine, Hershey, PA
| | - Xiaoming Liu
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Thomas Abraham
- Department of Neural and Behavioral Science, Penn State College of Medicine, Hershey, PA.,Microscopy Imaging Facility, Penn State College of Medicine, Hershey, PA
| | - Yuka Imamura-Kawasawa
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA.,Institute for Personalized Medicine, Penn State College of Medicine, Hershey, PA
| | - Nicholas J Buchkovich
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA
| | - Megan M Young
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA
| | - Hong-Gang Wang
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA .,Department of Pharmacology, Penn State College of Medicine, Hershey, PA
| |
Collapse
|
14
|
Zhen Y, Spangenberg H, Munson MJ, Brech A, Schink KO, Tan KW, Sørensen V, Wenzel EM, Radulovic M, Engedal N, Simonsen A, Raiborg C, Stenmark H. ESCRT-mediated phagophore sealing during mitophagy. Autophagy 2019; 16:826-841. [PMID: 31366282 PMCID: PMC7158923 DOI: 10.1080/15548627.2019.1639301] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Inactivation of the endosomal sorting complex required for transport (ESCRT) machinery has been reported to cause autophagic defects, but the exact functions of ESCRT proteins in macroautophagy/autophagy remain incompletely understood. Using live-cell fluorescence microscopy we found that the filament-forming ESCRT-III subunit CHMP4B was recruited transiently to nascent autophagosomes during starvation-induced autophagy and mitophagy, with residence times of about 1 and 2 min, respectively. Correlative light microscopy and electron tomography revealed CHMP4B recruitment at a late step in mitophagosome formation. The autophagosomal dwell time of CHMP4B was strongly increased by depletion of the regulatory ESCRT-III subunit CHMP2A. Using a novel optogenetic closure assay we observed that depletion of CHMP2A inhibited phagophore sealing during mitophagy. Consistent with this, depletion of CHMP2A and other ESCRT-III subunits inhibited both PRKN/PARKIN-dependent and -independent mitophagy. We conclude that the ESCRT machinery mediates phagophore closure, and that this is essential for mitophagic flux.Abbreviations: BSA: bovine serum albumin; CHMP: chromatin-modifying protein; CLEM: correlative light and electron microscopy; EGFP: enhanced green fluorescent protein; ESCRT: endosomal sorting complex required for transport; HEPES: 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid; HRP: horseradish peroxidase; ILV: intralumenal vesicle; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; LOV2: light oxygen voltage 2; MLS: mitochondrial localization sequence; MT-CO2: mitochondrially encoded cytochrome c oxidase II; O+A: oligomycin and antimycin A; PBS: phosphate-buffered saline; PIPES: piperazine-N,N-bis(2-ethanesulfonic acid); PRKN/PARKIN: parkin RBR E3 ubiquitin protein ligase; RAB: RAS-related in brain; SD: standard deviation; SEM: standard error of the mean; TOMM20: TOMM20: translocase of outer mitochondrial membrane 20; VCL: vinculin; VPS4: vacuolar protein sorting protein 4; Zdk1: Zdark 1; TUBG: Tubulin gamma chain.
Collapse
Affiliation(s)
- Yan Zhen
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| | - Hélène Spangenberg
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| | - Michael J Munson
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Medicine, Institute of Basic Medical Sciences, Oslo, Norway
| | - Andreas Brech
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| | - Kay O Schink
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| | - Kia-Wee Tan
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| | - Vigdis Sørensen
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| | - Eva Maria Wenzel
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| | - Maja Radulovic
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| | - Nikolai Engedal
- Centre for Molecular Medicine Norway (NCMM), University of Oslo, Oslo, Norway
| | - Anne Simonsen
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Medicine, Institute of Basic Medical Sciences, Oslo, Norway
| | - Camilla Raiborg
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| | - Harald Stenmark
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Montebello, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Norway
| |
Collapse
|
15
|
Zhou F, Wu Z, Zhao M, Murtazina R, Cai J, Zhang A, Li R, Sun D, Li W, Zhao L, Li Q, Zhu J, Cong X, Zhou Y, Xie Z, Gyurkovska V, Li L, Huang X, Xue Y, Chen L, Xu H, Xu H, Liang Y, Segev N. Rab5-dependent autophagosome closure by ESCRT. J Cell Biol 2019; 218:1908-1927. [PMID: 31010855 PMCID: PMC6548130 DOI: 10.1083/jcb.201811173] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/09/2019] [Accepted: 03/29/2019] [Indexed: 01/08/2023] Open
Abstract
Zhou et al. identify the mechanism of autophagosome (AP) closure. They show that Rab5 GTPase regulates an interaction between the ESCRT subunit Snf7 and Atg17 to bring ESCRT to APs where it catalyzes AP closure. These findings highlight the convergence of the endocytic and autophagic pathways at this step. In the conserved autophagy pathway, autophagosomes (APs) engulf cellular components and deliver them to the lysosome for degradation. Before fusing with the lysosome, APs have to close via an unknown mechanism. We have previously shown that the endocytic Rab5-GTPase regulates AP closure. Therefore, we asked whether ESCRT, which catalyzes scission of vesicles into late endosomes, mediates the topologically similar process of AP sealing. Here, we show that depletion of representative subunits from all ESCRT complexes causes late autophagy defects and accumulation of APs. Focusing on two subunits, we show that Snf7 and the Vps4 ATPase localize to APs and their depletion results in accumulation of open APs. Moreover, Snf7 and Vps4 proteins complement their corresponding mutant defects in vivo and in vitro. Finally, a Rab5-controlled Atg17–Snf7 interaction is important for Snf7 localization to APs. Thus, we unravel a mechanism in which a Rab5-dependent Atg17–Snf7 interaction leads to recruitment of ESCRT to open APs where ESCRT catalyzes AP closure.
Collapse
Affiliation(s)
- Fan Zhou
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Zulin Wu
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Mengzhu Zhao
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Rakhilya Murtazina
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Juan Cai
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Ao Zhang
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Rui Li
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Dan Sun
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Wenjing Li
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Lei Zhao
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Qunli Li
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Jing Zhu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxia Cong
- Department of Biochemistry and Molecular Biology, Dr. Li Dak Sam and Yap Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiting Zhou
- Department of Biochemistry and Molecular Biology, Dr. Li Dak Sam and Yap Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiping Xie
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Valeriya Gyurkovska
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Liuju Li
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xiaoshuai Huang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Yanhong Xue
- The National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Liangyi Chen
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Hui Xu
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Haiqian Xu
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Yongheng Liang
- College of Life Sciences, Key Laboratory of Agricultural Environmental Microbiology of Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing, China
| | - Nava Segev
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
16
|
Palmisano NJ, Meléndez A. Autophagy in C. elegans development. Dev Biol 2019; 447:103-125. [PMID: 29709599 PMCID: PMC6204124 DOI: 10.1016/j.ydbio.2018.04.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 03/19/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
Abstract
Autophagy involves the sequestration of cytoplasmic contents in a double-membrane structure referred to as the autophagosome and the degradation of its contents upon delivery to lysosomes. Autophagy activity has a role in multiple biological processes during the development of the nematode Caenorhabditis elegans. Basal levels of autophagy are required to remove aggregate prone proteins, paternal mitochondria, and spermatid-specific membranous organelles. During larval development, autophagy is required for the remodeling that occurs during dauer development, and autophagy can selectively degrade components of the miRNA-induced silencing complex, and modulate miRNA-mediated silencing. Basal levels of autophagy are important in synapse formation and in the germ line, to promote the proliferation of proliferating stem cells. Autophagy activity is also required for the efficient removal of apoptotic cell corpses by promoting phagosome maturation. Finally, autophagy is also involved in lipid homeostasis and in the aging process. In this review, we first describe the molecular complexes involved in the process of autophagy, its regulation, and mechanisms for cargo recognition. In the second section, we discuss the developmental contexts where autophagy has been shown to be important. Studies in C. elegans provide valuable insights into the physiological relevance of this process during metazoan development.
Collapse
Affiliation(s)
- Nicholas J Palmisano
- Biology Department, Queens College, CUNY, Flushing, NY, USA; Biology Ph.D. Program, The Graduate Center of the City University of New York, NK, USA
| | - Alicia Meléndez
- Biology Department, Queens College, CUNY, Flushing, NY, USA; Biology Ph.D. Program, The Graduate Center of the City University of New York, NK, USA; Biochemistry Ph.D. Program, The Graduate Center of the City University of New York, NY, USA.
| |
Collapse
|
17
|
Wang H, Wang X, Zhang K, Wang Q, Cao X, Wang Z, Zhang S, Li A, Liu K, Fang Y. Rapid depletion of ESCRT protein Vps4 underlies injury-induced autophagic impediment and Wallerian degeneration. SCIENCE ADVANCES 2019; 5:eaav4971. [PMID: 30788439 PMCID: PMC6374107 DOI: 10.1126/sciadv.aav4971] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/02/2019] [Indexed: 06/01/2023]
Abstract
Injured axons undergo a controlled, self-destruction process, known as Wallerian degeneration. However, the underlying mechanism remains elusive. Using the Drosophila wing nerve as a model, we identify the ESCRT component Vps4 as a previously unidentified essential gene for axonal integrity. Up-regulation of Vps4 remarkably delays degeneration of injured axons. We further reveal that Vps4 is required and sufficient to promote autophagic flux in axons and mammalian cells. Moreover, using both in vitro and in vivo models, we show that the function of Vps4 in maintaining axonal autophagy and suppressing Wallerian degeneration is conserved in mammals. Last, we uncover that Vps4 protein is rapidly depleted in injured mouse axons, which may underlie the injury-induced autophagic impediment and the subsequent axonal degeneration. Together, Vps4 and ESCRT may represent a novel signal transduction mechanism in axon injury and Wallerian degeneration.
Collapse
Affiliation(s)
- Haiqiong Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuejie Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Kai Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingyao Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu Cao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhao Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Shuang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Ang Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Jinan University, Guangzhou 510632, China
| | - Kai Liu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong, China
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Yanshan Fang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
Springhorn A, Hoppe T. Western blot analysis of the autophagosomal membrane protein LGG-1/LC3 in Caenorhabditis elegans. Methods Enzymol 2019; 619:319-336. [DOI: 10.1016/bs.mie.2018.12.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
19
|
Takahashi Y, He H, Tang Z, Hattori T, Liu Y, Young MM, Serfass JM, Chen L, Gebru M, Chen C, Wills CA, Atkinson JM, Chen H, Abraham T, Wang HG. An autophagy assay reveals the ESCRT-III component CHMP2A as a regulator of phagophore closure. Nat Commun 2018; 9:2855. [PMID: 30030437 PMCID: PMC6054611 DOI: 10.1038/s41467-018-05254-w] [Citation(s) in RCA: 252] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 06/23/2018] [Indexed: 01/21/2023] Open
Abstract
The mechanism of phagophore closure remains unclear due to technical limitations in distinguishing unclosed and closed autophagosomal membranes. Here, we report the HaloTag-LC3 autophagosome completion assay that specifically detects phagophores, nascent autophagosomes, and mature autophagic structures. Using this assay, we identify the endosomal sorting complexes required for transport (ESCRT)-III component CHMP2A as a critical regulator of phagophore closure. During autophagy, CHMP2A translocates to the phagophore and regulates the separation of the inner and outer autophagosomal membranes to form double-membrane autophagosomes. Consistently, inhibition of the AAA-ATPase VPS4 activity impairs autophagosome completion. The ESCRT-mediated membrane abscission appears to be a critical step in forming functional autolysosomes by preventing mislocalization of lysosome-associated membrane glycoprotein 1 to the inner autophagosomal membrane. Collectively, our work reveals a function for the ESCRT machinery in the final step of autophagosome formation and provides a useful tool for quantitative analysis of autophagosome biogenesis and maturation.
Collapse
Affiliation(s)
- Yoshinori Takahashi
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA.
| | - Haiyan He
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Zhenyuan Tang
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Tatsuya Hattori
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Ying Liu
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Megan M Young
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Jacob M Serfass
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Longgui Chen
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Melat Gebru
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Chong Chen
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Carson A Wills
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Jennifer M Atkinson
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Han Chen
- Microscopy Imaging Facility, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Thomas Abraham
- Microscopy Imaging Facility, Penn State College of Medicine, Hershey, PA, 17033, USA
- Department of Neural and Behavioral Science, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Hong-Gang Wang
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, 17033, USA.
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
20
|
Kalinowska K, Isono E. All roads lead to the vacuole-autophagic transport as part of the endomembrane trafficking network in plants. JOURNAL OF EXPERIMENTAL BOTANY 2018; 69:1313-1324. [PMID: 29165603 DOI: 10.1093/jxb/erx395] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/14/2017] [Indexed: 05/10/2023]
Abstract
Plants regulate their development and response to the changing environment by sensing and interpreting environmental signals. Intracellular trafficking pathways including endocytic-, vacuolar-, and autophagic trafficking are important for the various aspects of responses in plants. Studies in the last decade have shown that the autophagic transport pathway uses common key components of endomembrane trafficking as well as specific regulators. A number of factors previously described for their function in endosomal trafficking have been discovered to be involved in the regulation of autophagy in plants. These include conserved endocytic machineries, such as the endosomal sorting complex required for transport (ESCRT), subunits of the HOPS and exocyst complexes, SNAREs, and RAB GTPases as well as plant-specific proteins. Defects in these factors have been shown to cause impairment of autophagosome formation, transport, fusion, and degradation, suggesting crosstalk between autophagy and other intracellular trafficking processes. In this review, we focus mainly on possible functions of endosomal trafficking components in autophagy.
Collapse
|
21
|
Synthesis and biological evaluation of N-alkyl-1,4-dihydroquinoline prodrugs of scutellarin methyl ester as neuroprotective agents. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2134-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
22
|
The interactome of EBV LMP1 evaluated by proximity-based BioID approach. Virology 2018; 516:55-70. [PMID: 29329079 DOI: 10.1016/j.virol.2017.12.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/04/2017] [Accepted: 12/28/2017] [Indexed: 12/27/2022]
Abstract
Epstein-Barr virus LMP1 is an oncoprotein required for immortalizing B lymphocytes and also plays important roles in transforming non-lymphoid tissue. The discovery of LMP1 protein interactions will likely generate targets to treat EBV-associated cancers. Here, we define the broader LMP1 interactome using the recently developed BioID method. Combined with mass spectrometry, we identified over 1000 proteins across seven independent experiments with direct or indirect relationships to LMP1. Pathway analysis suggests that a significant number of the proteins identified are involved in signal transduction and protein or vesicle trafficking. Interestingly, a large number of proteins thought to be important in the formation of exosomes and protein targeting were recognized as probable LMP1 interacting partners, including CD63, syntenin-1, ALIX, TSG101, HRS, CHMPs, and sorting nexins. Therefore, it is likely that LMP1 modifies protein trafficking and exosome biogenesis pathways. In support of this, knock-down of syntenin-1 and ALIX resulted in reduced exosomal LMP1.
Collapse
|
23
|
Integration of the Endocytic System into the Network of Cellular Functions. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2018; 57:39-63. [PMID: 30097771 DOI: 10.1007/978-3-319-96704-2_2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Maintenance of physiologic cellular functions and homeostasis requires highly coordinated interactions between different cellular compartments. In this regard, the endocytic system, which plays a key role in cargo internalization and trafficking within the cell, participates in upkeep of intracellular dynamics, while communicating with multiple organelles. This chapter will discuss the function of endosomes from a standpoint of cellular integration. We will present examples of different types of interactions between endosomes and other cellular compartments, such as the endoplasmic reticulum (ER), mitochondria, the plasma membrane (PM), and the nuclear envelope. In addition, we will describe the incorporation of endocytic components, such as endosomal sorting complexes required for transport (ESCRT) proteins and Rab small GTPases, into cellular processes that operate outside of the endolysosomal pathway. The significance of endosomal interactions for processes such as signaling regulation, intracellular trafficking, organelle dynamics, metabolic control, and homeostatic responses will be reviewed. Accumulating data indicate that beyond its involvement in cargo transport, the endocytic pathway is comprehensively integrated into other systems of the cell and plays multiple roles in the complex net of cellular functions.
Collapse
|
24
|
Chemotherapy-Induced Tissue Injury: An Insight into the Role of Extracellular Vesicles-Mediated Oxidative Stress Responses. Antioxidants (Basel) 2017; 6:antiox6040075. [PMID: 28956814 PMCID: PMC5745485 DOI: 10.3390/antiox6040075] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 09/21/2017] [Accepted: 09/22/2017] [Indexed: 12/16/2022] Open
Abstract
The short- and long-term side effects of chemotherapy limit the maximum therapeutic dose and impair quality of life of survivors. Injury to normal tissues, especially chemotherapy-induced cardiomyopathy, is an unintended outcome that presents devastating health impacts. Approximately half of the drugs approved by the Food and Drug Administration for cancer treatment are associated with the generation of reactive oxygen species, and Doxorubicin (Dox) is one of them. Dox undergoes redox cycling by involving its quinone structure in the production of superoxide free radicals, which are thought to be instrumental to the role it plays in cardiomyopathy. Dox-induced protein oxidation changes protein function, translocation, and aggregation that are toxic to cells. To maintain cellular homeostasis, oxidized proteins can be degraded intracellularly by ubiquitin-proteasome pathway or by autophagy, depending on the redox status of the cell. Alternatively, the cell can remove oxidized proteins by releasing extracellular vesicles (EVs), which can be transferred to neighboring or distant cells, thereby instigating an intercellular oxidative stress response. In this article, we discuss the role of EVs in oxidative stress response, the potential of EVs as sensitive biomarkers of oxidative stress, and the role of superoxide dismutase in attenuating EV-associated oxidative stress response resulting from chemotherapy.
Collapse
|
25
|
Abstract
Macroautophagy is an intracellular pathway used for targeting of cellular components to the lysosome for their degradation and involves sequestration of cytoplasmic material into autophagosomes formed from a double membrane structure called the phagophore. The nucleation and elongation of the phagophore is tightly regulated by several autophagy-related (ATG) proteins, but also involves vesicular trafficking from different subcellular compartments to the forming autophagosome. Such trafficking must be tightly regulated by various intra- and extracellular signals to respond to different cellular stressors and metabolic states, as well as the nature of the cargo to become degraded. We are only starting to understand the interconnections between different membrane trafficking pathways and macroautophagy. This review will focus on the membrane trafficking machinery found to be involved in delivery of membrane, lipids, and proteins to the forming autophagosome and in the subsequent autophagosome fusion with endolysosomal membranes. The role of RAB proteins and their regulators, as well as coat proteins, vesicle tethers, and SNARE proteins in autophagosome biogenesis and maturation will be discussed.
Collapse
|
26
|
Palmisano NJ, Rosario N, Wysocki M, Hong M, Grant B, Meléndez A. The recycling endosome protein RAB-10 promotes autophagic flux and localization of the transmembrane protein ATG-9. Autophagy 2017; 13:1742-1753. [PMID: 28872980 DOI: 10.1080/15548627.2017.1356976] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Macroautophagy/autophagy involves the formation of an autophagosome, a double-membrane vesicle that delivers sequestered cytoplasmic cargo to lysosomes for degradation and recycling. Closely related, endocytosis mediates the sorting and transport of cargo throughout the cell, and both processes are important for cellular homeostasis. However, how endocytic proteins functionally intersect with autophagy is not clear. Mutations in the DAF-2/insulin-like IGF-1 (INSR) receptor at the permissive temperature result in a small increase in GFP::LGG-1 foci, i.e. autophagosomes, but a large increase at the nonpermissive temperature, allowing us to control the level of autophagy. In a RNAi screen for endocytic genes that alter the expression of GFP::LGG-1 in daf-2 mutants, we identified RAB-10, a small GTPase that regulates basolateral endocytosis. Loss of rab-10 in daf-2 mutants results in more GFP::LGG-1-positive foci at the permissive, but less GFP::LGG-1 or SQST-1::GFP foci at the nonpermissive temperature. As previously reported, loss of rab-10 alone resulted in an increase of GFP:LGG-1 foci. Exposure of rab-10 mutant animals to chloroquine, a known inhibitor of autophagic flux, failed to increase the number of GFP::LGG-1 foci. Moreover, colocalization between LMP-1::tagRFP and GFP::LGG-1 (the lysosome and autophagosome reporters) was decreased in daf-2; rab-10 dauers at the nonpermissive temperature. Intriguingly, RAB-10 was required to maintain the normal size of GFP::ATG-9-positive structures in daf-2 mutants at both the permissive and nonpermissive temperature. Finally, we found that RAB-10 GTPase cycling was required to control the size of GFP::ATG-9 foci. Collectively, our data support a model where rab-10 controls autophagic flux by regulating autophagosome formation and maturation.
Collapse
Affiliation(s)
- N J Palmisano
- a Biology Department, Queens College, CUNY , Flushing , NY , USA.,b Biology and Biochemistry Ph.D. Programs , The Graduate Center of the City University of New York , NY , USA
| | - N Rosario
- a Biology Department, Queens College, CUNY , Flushing , NY , USA
| | - M Wysocki
- a Biology Department, Queens College, CUNY , Flushing , NY , USA
| | - M Hong
- a Biology Department, Queens College, CUNY , Flushing , NY , USA
| | - B Grant
- c Department of Molecular Biology and Biochemistry , Rutgers University , Piscataway , NJ , USA
| | - A Meléndez
- a Biology Department, Queens College, CUNY , Flushing , NY , USA.,b Biology and Biochemistry Ph.D. Programs , The Graduate Center of the City University of New York , NY , USA
| |
Collapse
|
27
|
Lefebvre C, Legouis R, Culetto E. ESCRT and autophagies: Endosomal functions and beyond. Semin Cell Dev Biol 2017; 74:21-28. [PMID: 28807884 DOI: 10.1016/j.semcdb.2017.08.014] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/27/2017] [Accepted: 08/04/2017] [Indexed: 12/20/2022]
Abstract
ESCRT (endosomal sorting complex required for transport) machinery has been initially identified for its role during endocytosis, which allows membrane proteins and lipids to be degraded in the lysosome. ESCRT function is required to form intraluminal vesicles permitting internalization of cytosolic components or membrane embedded cargoes and promoting endosome maturation. ESCRT machinery also contributes to multiple key cell mechanisms in which it reshapes membranes. In addition, ESCRT actively participates in different types of autophagy processes for degrading cytosolic components, such as endosomal microautophagy and macroautophagy. During macroautophagy, ESCRT promotes formation of multivesicular bodies, which can fuse with autophagosomes to generate amphisomes. This latter fusion probably brings to autophagosomes key membrane molecules necessary for the subsequent fusion with lysosomes. Interestingly, during macroautophagy, ESCRT proteins could be involved in non-canonical functions such as vesicle tethering or phagophore membrane sealing. Additionally, ESCRT subunits could directly interact with key autophagy related proteins to build a closer connection between endocytosis and autophagy pathways.
Collapse
Affiliation(s)
- Christophe Lefebvre
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette Cedex, France
| | - Renaud Legouis
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette Cedex, France.
| | - Emmanuel Culetto
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette Cedex, France.
| |
Collapse
|
28
|
Kim J, You YJ. Regulation of organelle function by metformin. IUBMB Life 2017; 69:459-469. [PMID: 28444922 DOI: 10.1002/iub.1633] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/02/2017] [Indexed: 12/30/2022]
Abstract
Metformin ameliorates hyperglycemia without the side effects of lactic acidosis or hypoglycemia. Metformin lowers the blood glucose level by decreasing hepatic glucose production in the liver and by increasing glucose uptake in the muscle. Recent studies show that metformin induces cell death in certain cancer cell lines by interfering with the metabolism of the cancer cells. Therefore, understanding the mechanisms of action for metformin will provide insights into how to better treat diabetes and other metabolic disorders and also into the development of new therapeutic drugs. One of the best understood molecular targets of metformin is the mitochondrial complex I. However, given metformin's broad effects on metabolism, it could act on multiple targets. In this review, we summarize current findings in metformin's mechanisms of action regarding its known targets in mitochondria and known effects in cancer cell lines. Then, we introduce endosomal Na+ /H+ exchangers and the V-ATPase as new potential targets of metformin's action. Finally, we will discuss the hypothesis that metformin directly acts on endosome/lysosome regulation so as to regulate metabolism and ultimately alleviate type 2 diabetes. © 2017 IUBMB Life, 69(7):459-469, 2017.
Collapse
Affiliation(s)
- Jeongho Kim
- Department of Biological Sciences, Inha University, Incheon, South Korea
| | - Young-Jai You
- Nagoya Research Center for Brain and Neural Circuits, Graduate School of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
29
|
Rödelsperger C, Menden K, Serobyan V, Witte H, Baskaran P. First insights into the nature and evolution of antisense transcription in nematodes. BMC Evol Biol 2016; 16:165. [PMID: 27549405 PMCID: PMC4994411 DOI: 10.1186/s12862-016-0740-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/11/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The development of multicellular organisms is coordinated by various gene regulatory mechanisms that ensure correct spatio-temporal patterns of gene expression. Recently, the role of antisense transcription in gene regulation has moved into focus of research. To characterize genome-wide patterns of antisense transcription and to study their evolutionary conservation, we sequenced a strand-specific RNA-seq library of the nematode Pristionchus pacificus. RESULTS We identified 1112 antisense configurations of which the largest group represents 465 antisense transcripts (ASTs) that are fully embedded in introns of their host genes. We find that most ASTs show homology to protein-coding genes and are overrepresented in proteomic data. Together with the finding, that expression levels of ASTs and host genes are uncorrelated, this indicates that most ASTs in P. pacificus do not represent non-coding RNAs and do not exhibit regulatory functions on their host genes. We studied the evolution of antisense gene pairs across 20 nematode genomes, showing that the majority of pairs is lineage-specific and even the highly conserved vps-4, ddx-27, and sel-2 loci show abundant structural changes including duplications, deletions, intron gains and loss of antisense transcription. In contrast, host genes in general, are remarkably conserved and encode exceptionally long introns leading to unusually large blocks of conserved synteny. CONCLUSIONS Our study has shown that in P. pacificus antisense transcription as such does not define non-coding RNAs but is rather a feature of highly conserved genes with long introns. We hypothesize that the presence of regulatory elements imposes evolutionary constraint on the intron length, but simultaneously, their large size makes them a likely target for translocation of genomic elements including protein-coding genes that eventually end up as ASTs.
Collapse
Affiliation(s)
- Christian Rödelsperger
- Department for Evolutionary Biology, Max Planck Institute for Developmental Biology, Spemannstr. 35, Tübingen, 72076, Germany.
| | - Kevin Menden
- Department for Evolutionary Biology, Max Planck Institute for Developmental Biology, Spemannstr. 35, Tübingen, 72076, Germany.,Eberhard Karls University, Tübingen, Germany
| | - Vahan Serobyan
- Department for Evolutionary Biology, Max Planck Institute for Developmental Biology, Spemannstr. 35, Tübingen, 72076, Germany
| | - Hanh Witte
- Department for Evolutionary Biology, Max Planck Institute for Developmental Biology, Spemannstr. 35, Tübingen, 72076, Germany
| | - Praveen Baskaran
- Department for Evolutionary Biology, Max Planck Institute for Developmental Biology, Spemannstr. 35, Tübingen, 72076, Germany
| |
Collapse
|
30
|
Kim J, Lee HY, Ahn J, Hyun M, Lee I, Min KJ, You YJ. NHX-5, an Endosomal Na+/H+ Exchanger, Is Associated with Metformin Action. J Biol Chem 2016; 291:18591-9. [PMID: 27435670 DOI: 10.1074/jbc.c116.744037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Indexed: 01/18/2023] Open
Abstract
Diabetes is one of the most impactful diseases worldwide. The most commonly prescribed anti-diabetic drug is metformin. In this study, we identified an endosomal Na(+)/H(+) exchanger (NHE) as a new potential target of metformin from an unbiased screen in Caenorhabditis elegans The same NHE homolog also exists in flies, where it too mediates the effects of metformin. Our results suggest that endosomal NHEs could be a metformin target and provide an insight into a novel mechanism of action of metformin on regulating the endocytic cycle.
Collapse
Affiliation(s)
- Jeongho Kim
- From the Department of Biological Sciences, Inha University, Incheon 22212, South Korea
| | - Hye-Yeon Lee
- From the Department of Biological Sciences, Inha University, Incheon 22212, South Korea
| | - Jheesoo Ahn
- the Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia 23298, and
| | - Moonjung Hyun
- the Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia 23298, and
| | - Inhwan Lee
- the Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia 23298, and
| | - Kyung-Jin Min
- From the Department of Biological Sciences, Inha University, Incheon 22212, South Korea
| | - Young-Jai You
- the Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia 23298, and the Nagoya Research Center for Brain & Neural Circuits, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| |
Collapse
|
31
|
Chapin HC, Okada M, Merz AJ, Miller DL. Tissue-specific autophagy responses to aging and stress in C. elegans. Aging (Albany NY) 2016; 7:419-34. [PMID: 26142908 PMCID: PMC4505168 DOI: 10.18632/aging.100765] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cellular function relies on a balance between protein synthesis and breakdown. Macromolecular breakdown through autophagy is broadly required for cellular and tissue development, function, and recovery from stress. While Caenorhabditis elegans is frequently used to explore cellular responses to development and stress, the most common assays for autophagy in this system lack tissue-level resolution. Different tissues within an organism have unique functional characteristics and likely vary in their reliance on autophagy under different conditions. To generate a tissue-specific map of autophagy in C. elegans we used a dual fluorescent protein (dFP) tag that releases monomeric fluorescent protein (mFP) upon arrival at the lysosome. Tissue-specific expression of dFP::LGG-1 revealed autophagic flux in all tissues, but mFP accumulation was most dramatic in the intestine. We also observed variable responses to stress: starvation increased autophagic mFP release in all tissues, whereas anoxia primarily increased intestinal autophagic flux. We observed autophagic flux with tagged LGG-1, LGG-2, and two autophagic cargo reporters: a soluble cytoplasmic protein, and mitochondrial TOMM-7. Finally, an increase in mFP in older worms was consistent with an age-dependent shift in proteostasis. These novel measures of autophagic flux in C. elegans reveal heterogeneity in autophagic response across tissues during stress and aging.
Collapse
Affiliation(s)
- Hannah C Chapin
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Megan Okada
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alexey J Merz
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Dana L Miller
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
32
|
|
33
|
Lefebvre C, Largeau C, Michelet X, Fourrage C, Maniere X, Matic I, Legouis R, Culetto E. The ESCRT-II proteins are involved in shaping the sarcoplasmic reticulum in C. elegans. J Cell Sci 2016; 129:1490-9. [PMID: 26906413 DOI: 10.1242/jcs.178467] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 02/15/2016] [Indexed: 12/22/2022] Open
Abstract
The sarcoplasmic reticulum is a network of tubules and cisternae localized in close association with the contractile apparatus, and regulates Ca(2+)dynamics within striated muscle cell. The sarcoplasmic reticulum maintains its shape and organization despite repeated muscle cell contractions, through mechanisms which are still under investigation. The ESCRT complexes are essential to organize membrane subdomains and modify membrane topology in multiple cellular processes. Here, we report for the first time that ESCRT-II proteins play a role in the maintenance of sarcoplasmic reticulum integrity inC. elegans ESCRT-II proteins colocalize with the sarcoplasmic reticulum marker ryanodine receptor UNC-68. The localization at the sarcoplasmic reticulum of ESCRT-II and UNC-68 are mutually dependent. Furthermore, the characterization of ESCRT-II mutants revealed a fragmentation of the sarcoplasmic reticulum network, associated with an alteration of Ca(2+)dynamics. Our data provide evidence that ESCRT-II proteins are involved in sarcoplasmic reticulum shaping.
Collapse
Affiliation(s)
- Christophe Lefebvre
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France
| | - Céline Largeau
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France
| | - Xavier Michelet
- Brigham and Women's Hospital, 1 Jimmy Fund Way, Boston, MA 02115, USA
| | - Cécile Fourrage
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France
| | - Xavier Maniere
- Faculté de médecine Paris Descartes, Inserm U1001 - 24, rue du Faubourg St-Jacques, Paris 75014, France
| | - Ivan Matic
- Faculté de médecine Paris Descartes, Inserm U1001 - 24, rue du Faubourg St-Jacques, Paris 75014, France
| | - Renaud Legouis
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France
| | - Emmanuel Culetto
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette cedex 91198, France
| |
Collapse
|
34
|
Palmisano NJ, Meléndez A. Detection of Autophagy in Caenorhabditis elegans by Western Blotting Analysis of LGG-1. Cold Spring Harb Protoc 2016; 2016:pdb.prot086512. [PMID: 26832685 PMCID: PMC8112127 DOI: 10.1101/pdb.prot086512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A common way to measure the induction of autophagy in yeast and mammalian cells is to compare the amount of Atg8/LC3-I with that of Atg8-PE/LC3-II by using western blot analysis. This is because changes in the amount of LC3-II correlate closely with changes in the number of autophagosomes present in cells. Atg8/LC3 is initially synthesized as an unprocessed form, which is proteolytically processed to form Atg8/LC3-I, and then this is modified into the phosphatidylethanolamine (PE)-conjugated Atg8-PE/LC3-II form. Atg8/LC3-II is membrane bound, whereas Atg8-PE/LC3-I is cytosolic. By associating with both the inner and outer membranes of the autophagosome, Atg8-PE/LC3-II is the only autophagy reporter that is reliably associated with completed autophagosomes. In the nematode Caenorhabditis elegans, the ortholog of Atg8/LC3 is LGG-1. Here, we discuss how changes in the levels of LGG-1-II (and the paralog LGG-2) protein can, with appropriate controls, be used to monitor autophagy activity in nematodes and present a protocol for monitoring changes in the protein levels of different forms of LGG-1 by western blotting.
Collapse
Affiliation(s)
- Nicholas J. Palmisano
- Queens College-CUNY, Department of Biology, Flushing, NY, USA
- The Graduate Center, The City University of New York, New York, USA
| | - Alicia Meléndez
- Queens College-CUNY, Department of Biology, Flushing, NY, USA
- The Graduate Center, The City University of New York, New York, USA
| |
Collapse
|
35
|
Juan T, Fürthauer M. [The ESCRT complex: from endosomal transport to the development of multicellular organisms]. Biol Aujourdhui 2015; 209:111-124. [PMID: 26115716 DOI: 10.1051/jbio/2015009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Indexed: 06/04/2023]
Abstract
Since its discovery more than 50 years ago, the endo-lysosomal system has emerged as a central integrator of different cellular activities. This vesicular trafficking apparatus governs processes as diverse as the transduction of stimuli by growth factor receptors, the recycling and secretion of signaling molecules and the regulation of cellular homeostasis through autophagy. Accordingly, dysfunctions of the vesicular transport machinery have been linked to a growing number of pathologies. In this review we take the "Endosomal Sorting Complex Required for Transport" (ESCRT) as an example to illustrate the multiple functions of an evolutionarily conserved endosomal transport machinery. We describe the major concepts that have emerged from the study of this machinery at the level of the development and the physiology of multi-cellular organisms. In particular, we highlight the essential contributions of ESCRT proteins on the regulation of three biological processes: the endocytic regulation of cell signaling, autophagy and its role in neuronal morphogenesis and finally the biogenesis and function of extracellular vesicles.
Collapse
|
36
|
Müller M, Schmidt O, Angelova M, Faserl K, Weys S, Kremser L, Pfaffenwimmer T, Dalik T, Kraft C, Trajanoski Z, Lindner H, Teis D. The coordinated action of the MVB pathway and autophagy ensures cell survival during starvation. eLife 2015; 4:e07736. [PMID: 25902403 PMCID: PMC4424281 DOI: 10.7554/elife.07736] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/19/2015] [Indexed: 01/19/2023] Open
Abstract
The degradation and recycling of cellular components is essential for cell growth and survival. Here we show how selective and non-selective lysosomal protein degradation pathways cooperate to ensure cell survival upon nutrient limitation. A quantitative analysis of starvation-induced proteome remodeling in yeast reveals comprehensive changes already in the first three hours. In this period, many different integral plasma membrane proteins undergo endocytosis and degradation in vacuoles via the multivesicular body (MVB) pathway. Their degradation becomes essential to maintain critical amino acids levels that uphold protein synthesis early during starvation. This promotes cellular adaptation, including the de novo synthesis of vacuolar hydrolases to boost the vacuolar catabolic activity. This order of events primes vacuoles for the efficient degradation of bulk cytoplasm via autophagy. Hence, a catabolic cascade including the coordinated action of the MVB pathway and autophagy is essential to enter quiescence to survive extended periods of nutrient limitation.
Collapse
Affiliation(s)
- Martin Müller
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Oliver Schmidt
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Mihaela Angelova
- Division of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus Faserl
- Division of Clinical Biochemistry, ProteinMicroAnalysis Facility, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Sabine Weys
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Leopold Kremser
- Division of Clinical Biochemistry, ProteinMicroAnalysis Facility, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Thomas Dalik
- Department of Chemistry, University of Natural Resources and Applied Biosciences, Vienna, Austria
| | - Claudine Kraft
- Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Zlatko Trajanoski
- Division of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Lindner
- Division of Clinical Biochemistry, ProteinMicroAnalysis Facility, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - David Teis
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
- Austrian Drug Screening Institute, Innsbruck, Austria
| |
Collapse
|
37
|
Dual roles of an Arabidopsis ESCRT component FREE1 in regulating vacuolar protein transport and autophagic degradation. Proc Natl Acad Sci U S A 2015; 112:1886-91. [PMID: 25624505 DOI: 10.1073/pnas.1421271112] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Protein turnover can be achieved via the lysosome/vacuole and the autophagic degradation pathways. Evidence has accumulated revealing that efficient autophagic degradation requires functional endosomal sorting complex required for transport (ESCRT) machinery. However, the interplay between the ESCRT machinery and the autophagy regulator remains unclear. Here, we show that FYVE domain protein required for endosomal sorting 1 (FREE1), a recently identified plant-specific ESCRT component essential for multivesicular body (MVB) biogenesis and plant growth, plays roles both in vacuolar protein transport and autophagic degradation. FREE1 also regulates vacuole biogenesis in both seeds and vegetative cells of Arabidopsis. Additionally, FREE1 interacts directly with a unique plant autophagy regulator SH3 domain-containing protein2 and associates with the PI3K complex, to regulate the autophagic degradation in plants. Thus, FREE1 plays multiple functional roles in vacuolar protein trafficking and organelle biogenesis as well as in autophagic degradation via a previously unidentified regulatory mechanism of cross-talk between the ESCRT machinery and autophagy process.
Collapse
|
38
|
Jenzer C, Simionato E, Legouis R. Tools and methods to analyze autophagy in C. elegans. Methods 2014; 75:162-71. [PMID: 25484340 DOI: 10.1016/j.ymeth.2014.11.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 11/27/2022] Open
Abstract
For a long time, autophagy has been mainly studied in yeast or mammalian cell lines, and assays for analyzing autophagy in these models have been well described. More recently, the involvement of autophagy in various physiological functions has been investigated in multicellular organisms. Modification of autophagy flux is involved in developmental processes, resistance to stress conditions, aging, cell death and multiple pathologies. So, the use of animal models is essential to understand these processes in the context of different cell types and during the whole life. For ten years, the nematode Caenorhabditis elegans has emerged as a powerful model to analyze autophagy in physiological or pathological contexts. In this article, we present some of the established approaches and the emerging tools available to monitor and manipulate autophagy in C. elegans, and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Céline Jenzer
- Centre de Génétique Moléculaire, CNRS UPR3404, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France; Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris Sud, 1 Avenue de la Terrasse, 91198 Gif sur Yvette, France
| | - Elena Simionato
- Centre de Génétique Moléculaire, CNRS UPR3404, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France; Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris Sud, 1 Avenue de la Terrasse, 91198 Gif sur Yvette, France
| | - Renaud Legouis
- Centre de Génétique Moléculaire, CNRS UPR3404, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France; Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris Sud, 1 Avenue de la Terrasse, 91198 Gif sur Yvette, France.
| |
Collapse
|
39
|
Baixauli F, López-Otín C, Mittelbrunn M. Exosomes and autophagy: coordinated mechanisms for the maintenance of cellular fitness. Front Immunol 2014; 5:403. [PMID: 25191326 PMCID: PMC4138502 DOI: 10.3389/fimmu.2014.00403] [Citation(s) in RCA: 342] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 08/07/2014] [Indexed: 12/19/2022] Open
Abstract
Conditions resulting from loss of cellular homeostasis, including oxidative stress, inflammation, protein aggregation, endoplasmic reticulum stress, metabolic stress, and perturbation of mitochondrial function, are common to many pathological disorders and contribute to aging. Cells face these stress situations by engaging quality control mechanisms aimed to restore cellular homeostasis and preserve cell viability. Among them, the autophagy-lysosomal pathway mediates the specific degradation of damaged proteins and organelles, and its proper function is related to cellular protection and increased life span in many model organisms. Besides autophagy, increasing evidence underscores a role for exosomes in the selective secretion of harmful/damaged proteins and RNAs and thus in the maintenance of cellular fitness. In this perspective article, we discuss the emerging function of exosomes as a means of alleviating intracellular stress conditions, and how secretion of harmful or unwanted material in exosomes, in coordination with the autophagy-lysosomal pathway, is essential to preserve intracellular protein and RNA homeostasis. Finally, we provide an overview about the consequences of the spreading of the exosome content in physiological and pathological situations, and suggest putative therapeutic strategies for these exosome-mediated alterations.
Collapse
Affiliation(s)
- Francesc Baixauli
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares , Madrid , Spain
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo , Oviedo , Spain
| | - Maria Mittelbrunn
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares , Madrid , Spain ; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo , Oviedo , Spain
| |
Collapse
|
40
|
Jenzer C, Manil-Ségalen M, Lefebvre C, Largeau C, Glatigny A, Legouis R. Human GABARAP can restore autophagosome biogenesis in a C. elegans lgg-1 mutant. Autophagy 2014; 10:1868-72. [PMID: 25126728 DOI: 10.4161/auto.29745] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We recently described in C. elegans embryos, the acquisition of specialized functions for orthologs of yeast Atg8 (e.g., mammalian MAP1LC3/LC3) in allophagy, a selective and developmentally regulated autophagic process. During the formation of double-membrane autophagosomes, the ubiquitin-like Atg8/LC3 proteins are recruited to the membrane through a lipidation process. While at least 6 orthologs and paralogs are present in mammals, C. elegans only possesses 2 orthologs, LGG-1 and LGG-2, corresponding to the GABARAP-GABARAPL2/GATE-16 and the MAP1LC3 families, respectively. During allophagy, LGG-1 acts upstream of LGG-2 and is essential for autophagosome biogenesis, whereas LGG-2 facilitates their maturation. We demonstrated that LGG-2 directly interacts with the HOPS complex subunit VPS-39, and mediates the tethering between autophagosomes and lysosomes, which also requires RAB-7. In the present addendum, we compared the localization of autophagosomes, endosomes, amphisomes, and lysosomes in vps-39, rab-7, and lgg-2 depleted embryos. Our results suggest that lysosomes interact with autophagosomes or endosomes through a similar mechanism. We also performed a functional complementation of an lgg-1 null mutant with human GABARAP, its closer homolog, and showed that it localizes to autophagosomes and can rescue LGG-1 functions in the early embryo.
Collapse
Affiliation(s)
- Céline Jenzer
- Centre de Génétique Moléculaire; CNRS UPR3404;Université Paris Sud; Gif-sur-Yvette, France
| | - Marion Manil-Ségalen
- Centre de Génétique Moléculaire; CNRS UPR3404;Université Paris Sud; Gif-sur-Yvette, France
| | - Christophe Lefebvre
- Centre de Génétique Moléculaire; CNRS UPR3404;Université Paris Sud; Gif-sur-Yvette, France
| | - Céline Largeau
- Centre de Génétique Moléculaire; CNRS UPR3404;Université Paris Sud; Gif-sur-Yvette, France
| | - Annie Glatigny
- Centre de Génétique Moléculaire; CNRS UPR3404;Université Paris Sud; Gif-sur-Yvette, France
| | - Renaud Legouis
- Centre de Génétique Moléculaire; CNRS UPR3404;Université Paris Sud; Gif-sur-Yvette, France
| |
Collapse
|
41
|
Possik E, Jalali Z, Nouët Y, Yan M, Gingras MC, Schmeisser K, Panaite L, Dupuy F, Kharitidi D, Chotard L, Jones RG, Hall DH, Pause A. Folliculin regulates ampk-dependent autophagy and metabolic stress survival. PLoS Genet 2014; 10:e1004273. [PMID: 24763318 PMCID: PMC3998892 DOI: 10.1371/journal.pgen.1004273] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 02/14/2014] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of AMPK signaling has been implicated in many human diseases, which emphasizes the importance of characterizing AMPK regulators. The tumor suppressor FLCN, responsible for the Birt-Hogg Dubé renal neoplasia syndrome (BHD), is an AMPK-binding partner but the genetic and functional links between FLCN and AMPK have not been established. Strikingly, the majority of naturally occurring FLCN mutations predisposing to BHD are predicted to produce truncated proteins unable to bind AMPK, pointing to the critical role of this interaction in the tumor suppression mechanism. Here, we demonstrate that FLCN is an evolutionarily conserved negative regulator of AMPK. Using Caenorhabditis elegans and mammalian cells, we show that loss of FLCN results in constitutive activation of AMPK which induces autophagy, inhibits apoptosis, improves cellular bioenergetics, and confers resistance to energy-depleting stresses including oxidative stress, heat, anoxia, and serum deprivation. We further show that AMPK activation conferred by FLCN loss is independent of the cellular energy state suggesting that FLCN controls the AMPK energy sensing ability. Together, our data suggest that FLCN is an evolutionarily conserved regulator of AMPK signaling that may act as a tumor suppressor by negatively regulating AMPK function. The FLCN gene is responsible for the hereditary human tumor disease called Birt-Hogg-Dube syndrome (BHD). Patients that inherit an inactivating mutation in the FLCN gene develop lung collapse as well as tumors in the kidney, colon, and skin. It is not clear yet what the exact function of this protein is in the cell or an organism. In this study, we used a simple model organism (the round worm C. elegans) to study the function of FLCN. We found that it is involved in the regulation of energy metabolism in the cell. FLCN normally binds and blocks the action of another protein (AMPK), which is involved in the maintenance of energy levels. When energy levels fall, AMPK is activated and drives a recycling pathway called autophagy, where cellular components are recycled producing energy. In the absence of FLCN in worms and mammalian cells, like in tumors of BHD patients, AMPK and autophagy are chronically activated leading to an increased energy level, which makes the cells/organism very resistant to many stresses that would normally kill them, which in the end could lead to progression of tumorigenesis.
Collapse
Affiliation(s)
- Elite Possik
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Zahra Jalali
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Yann Nouët
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Ming Yan
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Marie-Claude Gingras
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Kathrin Schmeisser
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Lorena Panaite
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Fanny Dupuy
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Physiology, McGill University, Montréal, Québec, Canada
| | - Dmitri Kharitidi
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Laëtitia Chotard
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Russell G. Jones
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Physiology, McGill University, Montréal, Québec, Canada
| | - David H. Hall
- Department of Neuroscience, Albert Einstein College of Medicine, New York, New York, United States of America
| | - Arnim Pause
- Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
42
|
Manil-Ségalen M, Culetto E, Legouis R, Lefebvre C. Interactions between endosomal maturation and autophagy: analysis of ESCRT machinery during Caenorhabditis elegans development. Methods Enzymol 2014; 534:93-118. [PMID: 24359950 DOI: 10.1016/b978-0-12-397926-1.00006-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Endocytosis and autophagy are key vesicular pathways involved in degradation and recycling of cellular material. Both degradative pathways finally fuse with lysosome but are indeed interconnected at several levels. In particular, the fusion between endosomes and autophagosomes can generate intermediate vesicles named amphisomes. We analyzed the physiological and developmental roles of the ESCRT machinery in a model organism, the nematode Caenorhabditis elegans and showed that the blockage of the endosomal maturation triggers the induction of autophagic activity. This chapter describes several methods for studying endocytosis, autophagy, and their interconnection in C. elegans. A series of genetic, biochemical, and microscopy analyses has been used to study at the cellular and developmental levels, the cross talks between autophagy and endocytosis.
Collapse
Affiliation(s)
- Marion Manil-Ségalen
- Centre de Génétique Moléculaire, CNRS UPR3404, Université Paris-Sud, 91198 Gif-sur-Yvette Cedex, France
| | - Emmanuel Culetto
- Centre de Génétique Moléculaire, CNRS UPR3404, Université Paris-Sud, 91198 Gif-sur-Yvette Cedex, France
| | - Renaud Legouis
- Centre de Génétique Moléculaire, CNRS UPR3404, Université Paris-Sud, 91198 Gif-sur-Yvette Cedex, France.
| | - Christophe Lefebvre
- Centre de Génétique Moléculaire, CNRS UPR3404, Université Paris-Sud, 91198 Gif-sur-Yvette Cedex, France.
| |
Collapse
|
43
|
Manil-Ségalen M, Lefebvre C, Jenzer C, Trichet M, Boulogne C, Satiat-Jeunemaitre B, Legouis R. The C. elegans LC3 acts downstream of GABARAP to degrade autophagosomes by interacting with the HOPS subunit VPS39. Dev Cell 2013; 28:43-55. [PMID: 24374177 DOI: 10.1016/j.devcel.2013.11.022] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 10/17/2013] [Accepted: 11/26/2013] [Indexed: 01/07/2023]
Abstract
The formation of the autophagic vesicles requires the recruitment of ubiquitin-like Atg8 proteins to the membrane of nascent autophagosomes. Seven Atg8 homologs are present in mammals, split into the LC3 and the GABARAP/GATE-16 families, whose respective functions are unknown. Using Caenorhabditis elegans, we investigated the functions of the GABARAP and the LC3 homologs, LGG-1 and LGG-2, in autophagosome biogenesis. Both LGG-1 and LGG-2 localize to the autophagosomes but display partially overlapping patterns. During allophagy, a developmentally stereotyped autophagic flux, LGG-1 acts upstream of LGG-2 to allow its localization to autophagosomes. LGG-2 controls the maturation of LGG-1-positive autophagosomes and facilitates the tethering with the lysosomes through a direct interaction with the VPS-39 HOPS complex subunit. Genetic analyses sustain a sequential implication of LGG-1, LGG-2, RAB-7, and HOPS complex to generate autolysosomes. The duplications of Atg8 in metazoans thus allowed the acquisition of specialized functions for autophagosome maturation.
Collapse
Affiliation(s)
- Marion Manil-Ségalen
- Centre de Génétique Moléculaire, CNRS UPR3404 associée à l'Université Paris Sud, Avenue de la terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Christophe Lefebvre
- Centre de Génétique Moléculaire, CNRS UPR3404 associée à l'Université Paris Sud, Avenue de la terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Céline Jenzer
- Centre de Génétique Moléculaire, CNRS UPR3404 associée à l'Université Paris Sud, Avenue de la terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Michael Trichet
- Institut des Sciences du Végetal, CNRS UPR2355, Avenue de la terrasse, 91198 Gif-sur-Yvette Cedex, France; Imagif FRC 3115, Avenue de la terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Claire Boulogne
- Imagif FRC 3115, Avenue de la terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Béatrice Satiat-Jeunemaitre
- Institut des Sciences du Végetal, CNRS UPR2355, Avenue de la terrasse, 91198 Gif-sur-Yvette Cedex, France; Imagif FRC 3115, Avenue de la terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Renaud Legouis
- Centre de Génétique Moléculaire, CNRS UPR3404 associée à l'Université Paris Sud, Avenue de la terrasse, 91198 Gif-sur-Yvette Cedex, France.
| |
Collapse
|
44
|
Spallek T, Beck M, Ben Khaled S, Salomon S, Bourdais G, Schellmann S, Robatzek S. ESCRT-I mediates FLS2 endosomal sorting and plant immunity. PLoS Genet 2013; 9:e1004035. [PMID: 24385929 PMCID: PMC3873229 DOI: 10.1371/journal.pgen.1004035] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 11/01/2013] [Indexed: 11/19/2022] Open
Abstract
The plant immune receptor FLAGELLIN SENSING 2 (FLS2) is present at the plasma membrane and is internalized following activation of its ligand flagellin (flg22). We show that ENDOSOMAL SORTING COMPLEX REQUIRED FOR TRANSPORT (ESCRT)-I subunits play roles in FLS2 endocytosis in Arabidopsis. VPS37-1 co-localizes with FLS2 at endosomes and immunoprecipitates with the receptor upon flg22 elicitation. Vps37-1 mutants are reduced in flg22-induced FLS2 endosomes but not in endosomes labeled by Rab5 GTPases suggesting a defect in FLS2 trafficking rather than formation of endosomes. FLS2 localizes to the lumen of multivesicular bodies, but this is altered in vps37-1 mutants indicating compromised endosomal sorting of FLS2 by ESCRT-I loss-of-function. VPS37-1 and VPS28-2 are critical for immunity against bacterial infection through a role in stomatal closure. Our findings identify that VPS37-1, and likewise VPS28-2, regulate late FLS2 endosomal sorting and reveals that ESCRT-I is critical for flg22-activated stomatal defenses involved in plant immunity. Plants deploy plasma membrane immune receptors to survey their environment for potential threats. One of these receptors, FLAGELIN SENSING 2 (FLS2) recognizes bacterial flagellin (flg22) and thereby triggers a multitude of defense responses, enhancing immunity against infectious pathogens. Regulation of the subcellular localization of FLS2 is therefore an important aspect in plant disease resistance. FLS2 is known to shuttle between the plasma membrane and endosomal compartments but enters the late endosomal trafficking pathway upon ligand-dependent activation. A key question is the regulation of activated FLS2 in late endosomal trafficking. Here, we show that FLS2 is internalized into the lumen of multivesicular bodies and discovered by genetic inhibition that this step is regulated by components of the ENDOSOMAL SORTING COMPLEXES REQUIRED FOR TRANSPORT-I (ESCRT-I). Furthermore, we reveal that these ESCRT-I components play crucial roles in plant immunity impacting the flg22-triggered closure of stomata, prominent entry points of pathogenic bacteria, which occurred downstream of the known flg22 responses. These findings highlight the roles of endosomal trafficking in regulating FLS2 subcellular localization and plant immunity.
Collapse
Affiliation(s)
- Thomas Spallek
- The Sainsbury Laboratory, Norwich Research Park, Norwich, United Kingdom
| | - Martina Beck
- The Sainsbury Laboratory, Norwich Research Park, Norwich, United Kingdom
| | - Sara Ben Khaled
- The Sainsbury Laboratory, Norwich Research Park, Norwich, United Kingdom
| | - Susanne Salomon
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Gildas Bourdais
- The Sainsbury Laboratory, Norwich Research Park, Norwich, United Kingdom
| | | | - Silke Robatzek
- The Sainsbury Laboratory, Norwich Research Park, Norwich, United Kingdom
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
- * E-mail:
| |
Collapse
|
45
|
Michelet X, Legouis R. Autophagy in endosomal mutants: Desperately seeking to survive. WORM 2013; 1:216-20. [PMID: 24058852 PMCID: PMC3670222 DOI: 10.4161/worm.20848] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 05/16/2012] [Accepted: 05/22/2012] [Indexed: 12/19/2022]
Abstract
The endosomal and autophagic pathways are essential for the degradation and renewal of cellular components. After a complex maturation process, both pathways converge to their final destination, the lysosome. A close link between these two pathways was described along the last decade, notably through the analysis of ESCRT mutants. Although in mammals ESCRT mutants are unable to complete autophagic maturation due to the lack of fusion with the endolysosomal system, the role of ESCRT in the autophagic process still remains an open issue. Using C. elegans, we recently showed that blockage of the endosomal maturation triggers the induction of autophagic activity in ESCRT mutant.1 This increase of autophagic flux is an attempt to correct cellular defects and promote the survival of mutant animals.
Collapse
Affiliation(s)
- Xavier Michelet
- Centre de Génétique Moléculaire; Université Paris Sud; Gif-sur-Yvette, France
| | | |
Collapse
|
46
|
Katsiarimpa A, Kalinowska K, Anzenberger F, Weis C, Ostertag M, Tsutsumi C, Schwechheimer C, Brunner F, Hückelhoven R, Isono E. The deubiquitinating enzyme AMSH1 and the ESCRT-III subunit VPS2.1 are required for autophagic degradation in Arabidopsis. THE PLANT CELL 2013; 25:2236-52. [PMID: 23800962 PMCID: PMC3723623 DOI: 10.1105/tpc.113.113399] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
In eukaryotes, posttranslational modification by ubiquitin regulates the activity and stability of many proteins and thus influences a variety of developmental processes as well as environmental responses. Ubiquitination also plays a critical role in intracellular trafficking by serving as a signal for endocytosis. We have previously shown that the Arabidopsis thaliana associated molecule with the SH3 domain of STAM3 (AMSH3) is a deubiquitinating enzyme (DUB) that interacts with endosomal complex required for transport-III (ESCRT-III) and is essential for intracellular transport and vacuole biogenesis. However, physiological functions of AMSH3 in the context of its ESCRT-III interaction are not well understood due to the severe seedling lethal phenotype of its null mutant. In this article, we show that Arabidopsis AMSH1, an AMSH3-related DUB, interacts with the ESCRT-III subunit vacuolar protein sorting2.1 (VPS2.1) and that impairment of both AMSH1 and VPS2.1 causes early senescence and hypersensitivity to artificial carbon starvation in the dark similar to previously reported autophagy mutants. Consistent with this, both mutants accumulate autophagosome markers and accumulate less autophagic bodies in the vacuole. Taken together, our results demonstrate that AMSH1 and the ESCRT-III-subunit VPS2.1 are important for autophagic degradation and autophagy-mediated physiological processes.
Collapse
Affiliation(s)
- Anthi Katsiarimpa
- Department of Plant Systems Biology, Technische Universität München, 85354 Freising, Germany
| | - Kamila Kalinowska
- Department of Plant Systems Biology, Technische Universität München, 85354 Freising, Germany
| | - Franziska Anzenberger
- Department of Plant Systems Biology, Technische Universität München, 85354 Freising, Germany
| | - Corina Weis
- Department of Phytopathology, Technische Universität München, 85354 Freising, Germany
| | - Maya Ostertag
- Department of Phytopathology, Technische Universität München, 85354 Freising, Germany
| | - Chie Tsutsumi
- Department of Botany, National Museum of Nature and Science, Tsukuba 305-0005, Japan
| | - Claus Schwechheimer
- Department of Plant Systems Biology, Technische Universität München, 85354 Freising, Germany
| | - Frédéric Brunner
- Department of Plant Biochemistry, Center for Plant Molecular Biology, Tübingen University, 72076 Tuebingen, Germany
| | - Ralph Hückelhoven
- Department of Phytopathology, Technische Universität München, 85354 Freising, Germany
| | - Erika Isono
- Department of Plant Systems Biology, Technische Universität München, 85354 Freising, Germany
- Address correspondence to
| |
Collapse
|
47
|
Manil-Segalén M, Lefebvre C, Culetto E, Legouis R. Need an ESCRT for autophagosomal maturation? Commun Integr Biol 2013; 5:566-71. [PMID: 23336026 PMCID: PMC3541323 DOI: 10.4161/cib.21522] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Several reports in fly, nematode and mammalian cells have revealed that the inactivation of endosomal sorting complexes required for transport (ESCRT) blocks the endosomal maturation but also leads to the increased number of autophagosomal structures. In this review we compare these data and conclude that the way ESCRT mutations affect the relationships between autophagosomes and endosomes cannot be generalized but depends on the studied species. We propose that the effect of ESCRT mutations on autophagy is directly dependent of the level of interaction between autophagosomes and endosomes. In particular, the formation of amphisomes during autophagosomal maturation could be the key point to explain the differences observed between species. These observations highlight the importance of multiple model organisms to decipher the complexity of relationships between such dynamic vesicles.
Collapse
Affiliation(s)
- Marion Manil-Segalén
- Centre de Génétique Moléculaire; Université Paris-Sud; CNRS UPR3404; Gif-sur-Yvette Cedex, France
| | | | | | | |
Collapse
|