251
|
Exosomes induce endolysosomal permeabilization as a gateway by which exosomal tau seeds escape into the cytosol. Acta Neuropathol 2021; 141:235-256. [PMID: 33417012 PMCID: PMC7847444 DOI: 10.1007/s00401-020-02254-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
The microtubule-associated protein tau has a critical role in Alzheimer's disease and other tauopathies. A proposed pathomechanism in the progression of tauopathies is the trans-synaptic spreading of tau seeds, with a role for exosomes which are secretory nanovesicles generated by late endosomes. Our previous work demonstrated that brain-derived exosomes isolated from tau transgenic rTg4510 mice encapsulate tau seeds with the ability to induce tau aggregation in recipient cells. We had also shown that exosomes can hijack the endosomal pathway to spread through interconnected neurons. Here, we reveal how tau seeds contained within internalized exosomes exploit mechanisms of lysosomal degradation to escape the endosome and induce tau aggregation in the cytosol of HEK293T-derived 'tau biosensor cells'. We found that the majority of the exosome-containing endosomes fused with lysosomes to form endolysosomes. Exosomes induced their permeabilization, irrespective of the presence of tau seeds, or whether the exosomal preparations originated from mouse brains or HEK293T cells. We also found that permeabilization is a conserved mechanism, operating in both non-neuronal tau biosensor cells and primary neurons. However, permeabilization of endolysosomes only occurred in a small fraction of cells, which supports the notion that permeabilization occurs by a thresholded mechanism. Interestingly, tau aggregation was only induced in cells that exhibited permeabilization, presenting this as an escape route of exosomal tau seeds into the cytosol. Overexpression of RAB7, which is required for the formation of endolysosomes, strongly increased tau aggregation. Conversely, inhibition of lysosomal function with alkalinizing agents, or by knocking-down RAB7, decreased tau aggregation. Together, we conclude that the enzymatic activities of lysosomes permeabilize exosomal and endosomal membranes, thereby facilitating access of exosomal tau seeds to cytosolic tau to induce its aggregation. Our data underscore the importance of endosomal membrane integrity in mechanisms of cellular invasion by misfolded proteins that are resistant to lysosomal degradation.
Collapse
|
252
|
Johmura Y, Yamanaka T, Omori S, Wang TW, Sugiura Y, Matsumoto M, Suzuki N, Kumamoto S, Yamaguchi K, Hatakeyama S, Takami T, Yamaguchi R, Shimizu E, Ikeda K, Okahashi N, Mikawa R, Suematsu M, Arita M, Sugimoto M, Nakayama KI, Furukawa Y, Imoto S, Nakanishi M. Senolysis by glutaminolysis inhibition ameliorates various age-associated disorders. Science 2021; 371:265-270. [DOI: 10.1126/science.abb5916] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 09/14/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Yoshikazu Johmura
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Takehiro Yamanaka
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Satotaka Omori
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Teh-Wei Wang
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Masaki Matsumoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Narumi Suzuki
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Soichiro Kumamoto
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Kiyoshi Yamaguchi
- Clinical Genome Research, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Seira Hatakeyama
- Clinical Genome Research, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Tomoyo Takami
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Rui Yamaguchi
- Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Eigo Shimizu
- Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Kazutaka Ikeda
- RIKEN Center for Integrative Medical Sciences, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Nobuyuki Okahashi
- RIKEN Center for Integrative Medical Sciences, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Ryuta Mikawa
- Research Institute, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Makoto Arita
- RIKEN Center for Integrative Medical Sciences, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Shibakoen, Minato-ku, Tokyo 105-0011, Japan
| | - Masataka Sugimoto
- Research Institute, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511, Japan
| | - Keiichi I. Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoichi Furukawa
- Clinical Genome Research, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Seiya Imoto
- Health Intelligence Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
253
|
Abstract
The lysosome represents an important regulatory platform within numerous vesicle trafficking pathways including the endocytic, phagocytic, and autophagic pathways. Its ability to fuse with endosomes, phagosomes, and autophagosomes enables the lysosome to break down a wide range of both endogenous and exogenous cargo, including macromolecules, certain pathogens, and old or damaged organelles. Due to its center position in an intricate network of trafficking events, the lysosome has emerged as a central signaling node for sensing and orchestrating the cells metabolism and immune response, for inter-organelle and inter-cellular signaling and in membrane repair. This review highlights the current knowledge of general lysosome function and discusses these findings in their implication for renal glomerular cell types in health and disease including the involvement of glomerular cells in lysosomal storage diseases and the role of lysosomes in nongenetic glomerular injuries.
Collapse
|
254
|
Abstract
Enveloped viruses exit producer cells and acquire their external lipid envelopes by budding through limiting cellular membranes. Most viruses encode multifunctional structural proteins that coordinate the processes of virion assembly, membrane envelopment, budding, and maturation. In many cases, the cellular ESCRT pathway is recruited to facilitate the membrane fission step of budding, but alternative strategies are also employed. Recently, many viruses previously considered to be non-enveloped have been shown to exit cells non-lytically within vesicles, adding further complexity to the intricacies of virus budding and egress.
Collapse
|
255
|
Yabuki A, Miyara M, Umeda-Miyara K, Takao S, Sanoh S, Kotake Y. MiT/TFE family members suppress L-leucyl-L-leucine methyl ester-induced cell death. J Toxicol Sci 2021; 46:143-156. [PMID: 33642520 DOI: 10.2131/jts.46.143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Lysosomes are degradative organelles essential for cell homeostasis. However, various internal and external stimuli, including L-leucyl-L-leucine methyl ester (LLOMe), which is one of the common lysosomotropic agents, permeabilize the lysosomal membrane, leading to lysosome-dependent cell death because of leakage of lysosomal contents to the cytosol. The microphthalmia/transcription factor E (MiT/TFE) family members, which include transcription factor EB (TFEB), transcription factor E3 (TFE3), and microphthalmia-associated transcription factor (MITF), are master regulators of lysosomal biogenesis and are known to be involved in the lysosomal stress response. However, their protective effects against cell death associated with lysosomal-membrane damage are still poorly understood. In this study, we confirmed that LLOMe-induced lysosomal damage triggered nuclear translocation of TFEB/TFE3/MITF and increased the mRNA levels of their target genes encoding lysosomal hydrolases and lysosomal membrane proteins in HeLa cells. Furthermore, we revealed that TFEB/TFE3/MITF knockdown exacerbated LLOMe-induced cell death. However, TFEB overexpression only slightly attenuated LLOMe-induced cell death, despite enhanced LLOMe-induced increase in CTSD mRNA levels, implying that the endogenous levels of MiT/TFE family members might be sufficient to promote lysosomal biogenesis in response to lysosomal-membrane damage. Our results suggest that MiT/TFE family members suppress the cell death associated with lysosomal-membrane damage.
Collapse
Affiliation(s)
- Ayaka Yabuki
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Masatsugu Miyara
- Graduate School of Biomedical and Health Sciences, Hiroshima University
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University
- Research Fellow of Japan Society for the Promotion of Science
| | | | - Saya Takao
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Yaichiro Kotake
- Graduate School of Biomedical and Health Sciences, Hiroshima University
| |
Collapse
|
256
|
Davis OB, Shin HR, Lim CY, Wu EY, Kukurugya M, Maher CF, Perera RM, Ordonez MP, Zoncu R. NPC1-mTORC1 Signaling Couples Cholesterol Sensing to Organelle Homeostasis and Is a Targetable Pathway in Niemann-Pick Type C. Dev Cell 2020; 56:260-276.e7. [PMID: 33308480 DOI: 10.1016/j.devcel.2020.11.016] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/05/2020] [Accepted: 11/17/2020] [Indexed: 01/22/2023]
Abstract
Lysosomes promote cellular homeostasis through macromolecular hydrolysis within their lumen and metabolic signaling by the mTORC1 kinase on their limiting membranes. Both hydrolytic and signaling functions require precise regulation of lysosomal cholesterol content. In Niemann-Pick type C (NPC), loss of the cholesterol exporter, NPC1, causes cholesterol accumulation within lysosomes, leading to mTORC1 hyperactivation, disrupted mitochondrial function, and neurodegeneration. The compositional and functional alterations in NPC lysosomes and nature of aberrant cholesterol-mTORC1 signaling contribution to organelle pathogenesis are not understood. Through proteomic profiling of NPC lysosomes, we find pronounced proteolytic impairment compounded with hydrolase depletion, enhanced membrane damage, and defective mitophagy. Genetic and pharmacologic mTORC1 inhibition restores lysosomal proteolysis without correcting cholesterol storage, implicating aberrant mTORC1 as a pathogenic driver downstream of cholesterol accumulation. Consistently, mTORC1 inhibition ameliorates mitochondrial dysfunction in a neuronal model of NPC. Thus, cholesterol-mTORC1 signaling controls organelle homeostasis and is a targetable pathway in NPC.
Collapse
Affiliation(s)
- Oliver B Davis
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hijai R Shin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chun-Yan Lim
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emma Y Wu
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Pediatric Gastroenterology, Hepatology, and Nutrition, University of California, San Diego, La Jolla, CA 92037, USA
| | - Matthew Kukurugya
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Claire F Maher
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rushika M Perera
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - M Paulina Ordonez
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Pediatric Gastroenterology, Hepatology, and Nutrition, University of California, San Diego, La Jolla, CA 92037, USA.
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
257
|
Wright ZJ, Bartel B. Peroxisomes form intralumenal vesicles with roles in fatty acid catabolism and protein compartmentalization in Arabidopsis. Nat Commun 2020; 11:6221. [PMID: 33277488 PMCID: PMC7718247 DOI: 10.1038/s41467-020-20099-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
Peroxisomes are vital organelles that compartmentalize critical metabolic reactions, such as the breakdown of fats, in eukaryotic cells. Although peroxisomes typically are considered to consist of a single membrane enclosing a protein lumen, more complex peroxisomal membrane structure has occasionally been observed in yeast, mammals, and plants. However, technical challenges have limited the recognition and understanding of this complexity. Here we exploit the unusually large size of Arabidopsis peroxisomes to demonstrate that peroxisomes have extensive internal membranes. These internal vesicles accumulate over time, use ESCRT (endosomal sorting complexes required for transport) machinery for formation, and appear to derive from the outer peroxisomal membrane. Moreover, these vesicles can harbor distinct proteins and do not form normally when fatty acid β-oxidation, a core function of peroxisomes, is impaired. Our findings suggest a mechanism for lipid mobilization that circumvents challenges in processing insoluble metabolites. This revision of the classical view of peroxisomes as single-membrane organelles has implications for all aspects of peroxisome biogenesis and function and may help address fundamental questions in peroxisome evolution.
Collapse
Affiliation(s)
| | - Bonnie Bartel
- Biosciences Department, Rice University, Houston, TX, USA.
| |
Collapse
|
258
|
Liu YJ, Zhang T, Cheng D, Yang J, Chen S, Wang X, Li X, Duan D, Lou H, Zhu L, Luo J, Ho MS, Wang XD, Duan S. Late endosomes promote microglia migration via cytosolic translocation of immature protease cathD. SCIENCE ADVANCES 2020; 6:6/50/eaba5783. [PMID: 33298434 PMCID: PMC7725477 DOI: 10.1126/sciadv.aba5783] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 10/26/2020] [Indexed: 06/12/2023]
Abstract
Organelle transport requires dynamic cytoskeleton remodeling, but whether cytoskeletal dynamics are, in turn, regulated by organelles remains elusive. Here, we demonstrate that late endosomes, a type of prelysosomal organelles, facilitate actin-cytoskeleton remodeling via cytosolic translocation of immature protease cathepsin D (cathD) during microglia migration. After cytosolic translocation, late endosome-derived cathD juxtaposes actin filaments at the leading edge of lamellipodia. Suppressing cathD expression or blocking its cytosolic translocation impairs the maintenance but not the initiation of lamellipodial extension. Moreover, immature cathD balances the activity of the actin-severing protein cofilin to maintain globular-actin (G-actin) monomer pool for local actin recycling. Our study identifies cathD as a key lysosomal molecule that unconventionally contributes to actin cytoskeleton remodeling via cytosolic translocation during adenosine triphosphate-evoked microglia migration.
Collapse
Affiliation(s)
- Yi-Jun Liu
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Ting Zhang
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Daxiao Cheng
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Junhua Yang
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Sicong Chen
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xingyue Wang
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xia Li
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Duo Duan
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Huifang Lou
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liya Zhu
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jianhong Luo
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Margaret S Ho
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Xiao-Dong Wang
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China.
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Shumin Duan
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310058, China.
- Mental Health Center, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
259
|
Ghose P, Wehman AM. The developmental and physiological roles of phagocytosis in Caenorhabditis elegans. Curr Top Dev Biol 2020; 144:409-432. [PMID: 33992160 DOI: 10.1016/bs.ctdb.2020.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Phagocytosis is an essential process by which cellular debris and pathogens are cleared from the environment. Cells extend their plasma membrane to engulf objects and contain them within a limiting membrane for isolation from the cytosol or for intracellular degradation in phagolysosomes. The basic mechanisms of phagocytosis and intracellular clearance are well conserved between animals. Indeed, much of our understanding is derived from studies on the nematode worm, Caenorhabditis elegans. Here, we review the latest progress in understanding the mechanisms and functions of phagocytic clearance from C. elegans studies. In particular, we highlight new insights into phagocytic signaling pathways, phagosome formation and phagolysosome resolution, as well as the challenges in studying these cyclic processes.
Collapse
Affiliation(s)
- Piya Ghose
- Department of Biology, University of Texas, Arlington, TX, United States.
| | - Ann M Wehman
- Department of Biological Sciences, University of Denver, Denver, CO, United States.
| |
Collapse
|
260
|
Krawczyk PA, Laub M, Kozik P. To Kill But Not Be Killed: Controlling the Activity of Mammalian Pore-Forming Proteins. Front Immunol 2020; 11:601405. [PMID: 33281828 PMCID: PMC7691655 DOI: 10.3389/fimmu.2020.601405] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/20/2020] [Indexed: 01/01/2023] Open
Abstract
Pore-forming proteins (PFPs) are present in all domains of life, and play an important role in host-pathogen warfare and in the elimination of cancers. They can be employed to deliver specific effectors across membranes, to disrupt membrane integrity interfering with cell homeostasis, and to lyse membranes either destroying intracellular organelles or entire cells. Considering the destructive potential of PFPs, it is perhaps not surprising that mechanisms controlling their activity are remarkably complex, especially in multicellular organisms. Mammalian PFPs discovered to date include the complement membrane attack complex (MAC), perforins, as well as gasdermins. While the primary function of perforin-1 and gasdermins is to eliminate infected or cancerous host cells, perforin-2 and MAC can target pathogens directly. Yet, all mammalian PFPs are in principle capable of generating pores in membranes of healthy host cells which-if uncontrolled-could have dire, and potentially lethal consequences. In this review, we will highlight the strategies employed to protect the host from destruction by endogenous PFPs, while enabling timely and efficient elimination of target cells.
Collapse
Affiliation(s)
- Patrycja A Krawczyk
- MRC Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Marco Laub
- MRC Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Patrycja Kozik
- MRC Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
261
|
Molecular Mechanisms and Biological Functions of Autophagy for Genetics of Hearing Impairment. Genes (Basel) 2020; 11:genes11111331. [PMID: 33187328 PMCID: PMC7697636 DOI: 10.3390/genes11111331] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 02/08/2023] Open
Abstract
The etiology of hearing impairment following cochlear damage can be caused by many factors, including congenital or acquired onset, ototoxic drugs, noise exposure, and aging. Regardless of the many different etiologies, a common pathologic change is auditory cell death. It may be difficult to explain hearing impairment only from the aspect of cell death including apoptosis, necrosis, or necroptosis because the level of hearing loss varies widely. Therefore, we focused on autophagy as an intracellular phenomenon functionally competing with cell death. Autophagy is a dynamic lysosomal degradation and recycling system in the eukaryotic cell, mandatory for controlling the balance between cell survival and cell death induced by cellular stress, and maintaining homeostasis of postmitotic cells, including hair cells (HCs) and spiral ganglion neurons (SGNs) in the inner ear. Autophagy is considered a candidate for the auditory cell fate decision factor, whereas autophagy deficiency could be one of major causes of hearing impairment. In this paper, we review the molecular mechanisms and biologic functions of autophagy in the auditory system and discuss the latest research concerning autophagy-related genes and sensorineural hearing loss to gain insight into the role of autophagic mechanisms in inner-ear disorders.
Collapse
|
262
|
Weir A, Hughes S, Rashidi M, Hildebrand JM, Vince JE. Necroptotic movers and shakers: cell types, inflammatory drivers and diseases. Curr Opin Immunol 2020; 68:83-97. [PMID: 33160107 DOI: 10.1016/j.coi.2020.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
The necroptotic cell death pathway has received significant attention for its ability to trigger inflammatory responses and its potential involvement in related conditions. Recent insights into the essential membrane damaging necroptotic pseudokinase effector, Mixed lineage kinase domain like (MLKL), have revealed a number of diverse MLKL functions that contribute to the inflammatory nature of necroptosis. Here we review distinct MLKL signalling roles and document the immunogenic molecules released by necroptosis. We discuss specific in vivo MLKL-driven responses, the activation of inflammasome complexes and innate lymphoid cells, which have been documented to drive disease. Finally, we list necroptotic competent cell types and their involvement in MLKL-driven cell death-associated and inflammatory-associated conditions.
Collapse
Affiliation(s)
- Ashley Weir
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sebastian Hughes
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Maryam Rashidi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Joanne M Hildebrand
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia.
| | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia; The Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
263
|
Liu EA, Schultz ML, Mochida C, Chung C, Paulson HL, Lieberman AP. Fbxo2 mediates clearance of damaged lysosomes and modifies neurodegeneration in the Niemann-Pick C brain. JCI Insight 2020; 5:136676. [PMID: 32931479 PMCID: PMC7605537 DOI: 10.1172/jci.insight.136676] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 09/09/2020] [Indexed: 12/24/2022] Open
Abstract
A critical response to lysosomal membrane permeabilization (LMP) is the clearance of damaged lysosomes through a selective form of macroautophagy known as lysophagy. Although regulators of this process are emerging, whether organ- and cell-specific components contribute to the control of lysophagy remains incompletely understood. Here, we examined LMP and lysophagy in Niemann-Pick type C (NPC) disease, an autosomal recessive disorder characterized by the accumulation of unesterified cholesterol within late endosomes and lysosomes, leading to neurodegeneration and early death. We demonstrated that NPC human fibroblasts show enhanced sensitivity to lysosomal damage as a consequence of lipid storage. Moreover, we described a role for the glycan-binding F-box protein 2 (Fbxo2) in CNS lysophagy. Fbxo2 functions as a component of the S phase kinase-associated protein 1–cullin 1–F-box protein (SKP1-CUL1-SCF) ubiquitin ligase complex. Loss of Fbxo2 in mouse primary cortical cultures delayed clearance of damaged lysosomes and decreased viability after lysosomal damage. Moreover, Fbxo2 deficiency in a mouse model of NPC exacerbated deficits in motor function, enhanced neurodegeneration, and reduced survival. Collectively, our data identified a role for Fbxo2 in CNS lysophagy and establish its functional importance in NPC. Glycan binding protein Fbxo2 regulates lysophagy in the brain, and its deficiency exacerbates neuronal deficits in a mouse model of Niemann-Pick type C disease.
Collapse
Affiliation(s)
- Elaine A Liu
- Department of Pathology.,Cellular and Molecular Biology Graduate Program, and.,Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Chisaki Mochida
- Yamaguchi University School of Medicine, Ube, Yamaguchi, Japan
| | | | - Henry L Paulson
- Department of Neurology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | |
Collapse
|
264
|
Saftig P, Puertollano R. How Lysosomes Sense, Integrate, and Cope with Stress. Trends Biochem Sci 2020; 46:97-112. [PMID: 33012625 DOI: 10.1016/j.tibs.2020.09.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
Lysosomes are in the center of the cellular control of catabolic and anabolic processes. These membrane-surrounded acidic organelles contain around 70 hydrolases, 200 membrane proteins, and numerous accessory proteins associated with the cytosolic surface of lysosomes. Accessory and transmembrane proteins assemble in signaling complexes that sense and integrate multiple signals and transmit the information to the nucleus. This communication allows cells to respond to changes in multiple environmental conditions, including nutrient levels, pathogens, energy availability, and lysosomal damage, with the goal of restoring cellular homeostasis. This review summarizes our current understanding of the major molecular players and known pathways that are involved in control of metabolic and stress responses that either originate from lysosomes or regulate lysosomal functions.
Collapse
Affiliation(s)
- Paul Saftig
- Biochemical Institute, Christian-Albrechts-Universität Kiel, Kiel, Germany.
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
265
|
Zhu SY, Yao RQ, Li YX, Zhao PY, Ren C, Du XH, Yao YM. Lysosomal quality control of cell fate: a novel therapeutic target for human diseases. Cell Death Dis 2020; 11:817. [PMID: 32999282 PMCID: PMC7528093 DOI: 10.1038/s41419-020-03032-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/15/2020] [Indexed: 02/08/2023]
Abstract
In eukaryotic cells, lysosomes are digestive centers where biological macromolecules are degraded by phagocytosis and autophagy, thereby maintaining cellular self-renewal capacity and energy supply. Lysosomes also serve as signaling hubs to monitor the intracellular levels of nutrients and energy by acting as platforms for the assembly of multiple signaling pathways, such as mammalian target of rapamycin complex 1 (mTORC1) and adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK). The structural integrity and functional balance of lysosomes are essential for cell function and viability. In fact, lysosomal damage not only disrupts intracellular clearance but also results in the leakage of multiple contents, which pose great threats to the cell by triggering cell death pathways, including apoptosis, necroptosis, pyroptosis, and ferroptosis. The collapse of lysosomal homeostasis is reportedly critical for the pathogenesis and development of various diseases, such as tumors, neurodegenerative diseases, cardiovascular diseases, and inflammatory diseases. Lysosomal quality control (LQC), comprising lysosomal repair, lysophagy, and lysosomal regeneration, is rapidly initiated in response to lysosomal damage to maintain lysosomal structural integrity and functional homeostasis. LQC may be a novel but pivotal target for disease treatment because of its indispensable role in maintaining intracellular homeostasis and cell fate.
Collapse
Affiliation(s)
- Sheng-Yu Zhu
- Trauma Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, 100048, Beijing, People's Republic of China.,Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, 100853, Beijing, People's Republic of China.,School of Medicine, Nankai University, 300071, Tianjin, People's Republic of China
| | - Ren-Qi Yao
- Trauma Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, 100048, Beijing, People's Republic of China.,Department of Burn Surgery, Changhai Hospital, Naval Medical University, 200433, Shanghai, People's Republic of China
| | - Yu-Xuan Li
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, 100853, Beijing, People's Republic of China
| | - Peng-Yue Zhao
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, 100853, Beijing, People's Republic of China
| | - Chao Ren
- Trauma Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, 100048, Beijing, People's Republic of China.
| | - Xiao-Hui Du
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, 100853, Beijing, People's Republic of China.
| | - Yong-Ming Yao
- Trauma Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, 100048, Beijing, People's Republic of China.
| |
Collapse
|
266
|
Nakamura S, Shigeyama S, Minami S, Shima T, Akayama S, Matsuda T, Esposito A, Napolitano G, Kuma A, Namba-Hamano T, Nakamura J, Yamamoto K, Sasai M, Tokumura A, Miyamoto M, Oe Y, Fujita T, Terawaki S, Takahashi A, Hamasaki M, Yamamoto M, Okada Y, Komatsu M, Nagai T, Takabatake Y, Xu H, Isaka Y, Ballabio A, Yoshimori T. LC3 lipidation is essential for TFEB activation during the lysosomal damage response to kidney injury. Nat Cell Biol 2020; 22:1252-1263. [PMID: 32989250 DOI: 10.1038/s41556-020-00583-9] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 08/25/2020] [Indexed: 12/21/2022]
Abstract
Sensing and clearance of dysfunctional lysosomes is critical for cellular homeostasis. Here we show that transcription factor EB (TFEB)-a master transcriptional regulator of lysosomal biogenesis and autophagy-is activated during the lysosomal damage response, and its activation is dependent on the function of the ATG conjugation system, which mediates LC3 lipidation. In addition, lysosomal damage triggers LC3 recruitment on lysosomes, where lipidated LC3 interacts with the lysosomal calcium channel TRPML1, facilitating calcium efflux essential for TFEB activation. Furthermore, we demonstrate the presence and importance of this TFEB activation mechanism in kidneys in a mouse model of oxalate nephropathy accompanying lysosomal damage. A proximal tubule-specific TFEB-knockout mouse exhibited progression of kidney injury induced by oxalate crystals. Together, our results reveal unexpected mechanisms of TFEB activation by LC3 lipidation and their physiological relevance during the lysosomal damage response.
Collapse
Affiliation(s)
- Shuhei Nakamura
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan. .,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan. .,Institute for Advanced Co-Creation Studies, Osaka University, Osaka, Japan.
| | - Saki Shigeyama
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Satoshi Minami
- Department of Nephrology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takayuki Shima
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Shiori Akayama
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Tomoki Matsuda
- Department of Biomolecular Science and Engineering, The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | | | - Gennaro Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.,Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Akiko Kuma
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Tomoko Namba-Hamano
- Department of Nephrology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Jun Nakamura
- Department of Nephrology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Ayaka Tokumura
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Mika Miyamoto
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Yukako Oe
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Toshiharu Fujita
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Seigo Terawaki
- Laboratory of Pathobiochemistry, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Atsushi Takahashi
- Department of Nephrology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Maho Hamasaki
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takeharu Nagai
- Department of Biomolecular Science and Engineering, The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Yoshitsugu Takabatake
- Department of Nephrology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yoshitaka Isaka
- Department of Nephrology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.,Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,SSM School for Advanced Studies, Federico II University, Naples, Italy
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan. .,Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan. .,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.
| |
Collapse
|
267
|
Kondow-McConaghy HM, Muthukrishnan N, Erazo-Oliveras A, Najjar K, Juliano RL, Pellois JP. Impact of the Endosomal Escape Activity of Cell-Penetrating Peptides on the Endocytic Pathway. ACS Chem Biol 2020; 15:2355-2363. [PMID: 32786263 PMCID: PMC7502533 DOI: 10.1021/acschembio.0c00319] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cell-penetrating peptides (CPPs) are routinely used for the delivery of macromolecules into live human cells. To enter the cytosolic space of cells, CPPs typically permeabilize the membrane of endosomes. In turn, several approaches have been developed to increase the endosomal membrane permeation activity of CPPs so as to improve delivery efficiencies. The endocytic pathway is, however, important in maintaining cellular homeostasis, and understanding how endosomal permeation impacts cells is now critical to define the general utility of CPPs. Herein, we investigate how CPP-based delivery protocols affect the endocytic network. We detect that, in some cases, cell penetration induces the activation of Chmp1b, Galectin-3, and TFEB, which are components of endosomal repair, organelle clearance, and biogenesis pathways, respectively. We also detect that cellular delivery modulates endocytosis and endocytic proteolysis. Remarkably, a multimeric analogue of the prototypical CPP TAT permeabilizes endosomes efficiently without inducing membrane damage responses. These results challenge the notion that reagents that make endosomes leaky are generally toxic. Instead, our data indicates that it is possible to enter cells with minimal deleterious effects.
Collapse
Affiliation(s)
- Helena M. Kondow-McConaghy
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Nandhini Muthukrishnan
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Alfredo Erazo-Oliveras
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Kristina Najjar
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Rudolph L. Juliano
- UNC Eshelman School of Pharmacy and UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
268
|
Abstract
The endosomal sorting complexes required for transport (ESCRTs) I, -II and -III, and their associated factors are a collection of ∼20 proteins in yeast and ∼30 in mammals, responsible for severing membrane necks in processes that range from multivesicular body formation, HIV release and cytokinesis, to plasma and lysosomal membrane repair. ESCRTs are best known for 'reverse-topology' membrane scission, where they act on the inner surface of membrane necks, often when membranes are budded away from the cytosol. These events are driven by membrane-associated assemblies of dozens to hundreds of ESCRT molecules. ESCRT-III proteins form filaments with a variety of geometries and ESCRT-I has now been shown to also form helical structures. The complex nature of the system and the unusual topology of its action has made progress challenging, and led to controversies with regard to its underlying mechanism. This Review will focus on recent advances obtained by structural in vitro reconstitution and in silico mechanistic studies, and places them in their biological context. The field is converging towards a consensus on the broad outlines of a mechanism that is driven by a progressive ATP-dependent treadmilling exchange of ESCRT subunits, as well as compositional change and geometric transitions in ESCRT filaments.
Collapse
Affiliation(s)
- Mark Remec Pavlin
- Graduate Group in Biophysics, University of California, Berkeley, Berkeley, CA 94720, USA.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - James H Hurley
- Graduate Group in Biophysics, University of California, Berkeley, Berkeley, CA 94720, USA .,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| |
Collapse
|
269
|
Abstract
Mutations in several genes encoding for lysosomal proteins are involved in Parkinson's disease (PD). In this issue, Herbst et al (2020) show that PD-related leucine-rich repeat kinase 2 (LRRK2) is activated in response to pathogen or membranolytic drug-induced damage of phagolysosomes and lysosomes in macrophages, and regulates endolysosomal homeostasis by controlling the balance between membrane repair and degradation.
Collapse
Affiliation(s)
- Maja Radulovic
- Centre for Cancer Cell ReprogrammingFaculty of MedicineUniversity of OsloOsloNorway
- Department of Molecular Cell BiologyInstitute for Cancer ResearchOslo University HospitalOsloNorway
| | - Harald Stenmark
- Centre for Cancer Cell ReprogrammingFaculty of MedicineUniversity of OsloOsloNorway
- Department of Molecular Cell BiologyInstitute for Cancer ResearchOslo University HospitalOsloNorway
- Centre of Molecular Inflammation ResearchFaculty of MedicineNorwegian University of Science and TechnologyTrondheimNorway
| |
Collapse
|
270
|
Herbst S, Campbell P, Harvey J, Bernard EM, Papayannopoulos V, Wood NW, Morris HR, Gutierrez MG. LRRK2 activation controls the repair of damaged endomembranes in macrophages. EMBO J 2020; 39:e104494. [PMID: 32643832 PMCID: PMC7507578 DOI: 10.15252/embj.2020104494] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 01/15/2023] Open
Abstract
Cells respond to endolysosome damage by either repairing the damage or targeting damaged endolysosomes for degradation via lysophagy. However, the signals regulating the decision for repair or lysophagy are poorly characterised. Here, we show that the Parkinson's disease (PD)-related kinase LRRK2 is activated in macrophages by pathogen- or sterile-induced endomembrane damage. LRRK2 recruits the Rab GTPase Rab8A to damaged endolysosomes as well as the ESCRT-III component CHMP4B, thereby favouring ESCRT-mediated repair. Conversely, in the absence of LRRK2 and Rab8A, damaged endolysosomes are targeted to lysophagy. These observations are recapitulated in macrophages from PD patients where pathogenic LRRK2 gain-of-function mutations result in the accumulation of endolysosomes which are positive for the membrane damage marker Galectin-3. Altogether, this work indicates that LRRK2 regulates endolysosomal homeostasis by controlling the balance between membrane repair and organelle replacement, uncovering an unexpected function for LRRK2, and providing a new link between membrane damage and PD.
Collapse
Affiliation(s)
- Susanne Herbst
- Host‐Pathogen Interactions in Tuberculosis LaboratoryThe Francis Crick InstituteLondonUK
| | - Philip Campbell
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyUCL Movement Disorders CentreUniversity College LondonLondonUK
| | - John Harvey
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyUCL Movement Disorders CentreUniversity College LondonLondonUK
| | - Elliott M Bernard
- Host‐Pathogen Interactions in Tuberculosis LaboratoryThe Francis Crick InstituteLondonUK
| | | | - Nicholas W Wood
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyUCL Movement Disorders CentreUniversity College LondonLondonUK
| | - Huw R Morris
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyUCL Movement Disorders CentreUniversity College LondonLondonUK
| | | |
Collapse
|
271
|
Daussy CF, Wodrich H. "Repair Me if You Can": Membrane Damage, Response, and Control from the Viral Perspective. Cells 2020; 9:cells9092042. [PMID: 32906744 PMCID: PMC7564661 DOI: 10.3390/cells9092042] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Cells are constantly challenged by pathogens (bacteria, virus, and fungi), and protein aggregates or chemicals, which can provoke membrane damage at the plasma membrane or within the endo-lysosomal compartments. Detection of endo-lysosomal rupture depends on a family of sugar-binding lectins, known as galectins, which sense the abnormal exposure of glycans to the cytoplasm upon membrane damage. Galectins in conjunction with other factors orchestrate specific membrane damage responses such as the recruitment of the endosomal sorting complex required for transport (ESCRT) machinery to either repair damaged membranes or the activation of autophagy to remove membrane remnants. If not controlled, membrane damage causes the release of harmful components including protons, reactive oxygen species, or cathepsins that will elicit inflammation. In this review, we provide an overview of current knowledge on membrane damage and cellular responses. In particular, we focus on the endo-lysosomal damage triggered by non-enveloped viruses (such as adenovirus) and discuss viral strategies to control the cellular membrane damage response. Finally, we debate the link between autophagy and inflammation in this context and discuss the possibility that virus induced autophagy upon entry limits inflammation.
Collapse
|
272
|
De La-Rocque S, Moretto E, Butnaru I, Schiavo G. Knockin' on heaven's door: Molecular mechanisms of neuronal tau uptake. J Neurochem 2020; 156:563-588. [PMID: 32770783 PMCID: PMC8432157 DOI: 10.1111/jnc.15144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022]
Abstract
Since aggregates of the microtubule‐binding protein tau were found to be the main component of neurofibrillary tangles more than 30 years ago, their contribution to neurodegeneration in Alzheimer's disease (AD) and tauopathies has become well established. Recent work shows that both tau load and its distribution in the brain of AD patients correlate with cognitive decline more closely compared to amyloid plaque deposition. In addition, the amyloid cascade hypothesis has been recently challenged because of disappointing results of clinical trials designed to treat AD by reducing beta‐amyloid levels, thus fuelling a renewed interest in tau. There is now robust evidence to indicate that tau pathology can spread within the central nervous system via a prion‐like mechanism following a stereotypical pattern, which can be explained by the trans‐synaptic inter‐neuronal transfer of pathological tau. In the receiving neuron, tau has been shown to take multiple routes of internalisation, which are partially dependent on its conformation and aggregation status. Here, we review the emerging mechanisms proposed for the uptake of extracellular tau in neurons and the requirements for the propagation of its pathological conformers, addressing how they gain access to physiological tau monomers in the cytosol. Furthermore, we highlight some of the key mechanistic gaps of the field, which urgently need to be addressed to expand our understanding of tau propagation and lead to the identification of new therapeutic strategies for tauopathies.
Collapse
Affiliation(s)
- Samantha De La-Rocque
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edoardo Moretto
- UK Dementia Research Institute, University College London, London, UK
| | - Ioana Butnaru
- UK Dementia Research Institute, University College London, London, UK
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
273
|
Interaction of Macrophages and Cholesterol-Dependent Cytolysins: The Impact on Immune Response and Cellular Survival. Toxins (Basel) 2020; 12:toxins12090531. [PMID: 32825096 PMCID: PMC7551085 DOI: 10.3390/toxins12090531] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) are key virulence factors involved in many lethal bacterial infections, including pneumonia, necrotizing soft tissue infections, bacterial meningitis, and miscarriage. Host responses to these diseases involve myeloid cells, especially macrophages. Macrophages use several systems to detect and respond to cholesterol-dependent cytolysins, including membrane repair, mitogen-activated protein (MAP) kinase signaling, phagocytosis, cytokine production, and activation of the adaptive immune system. However, CDCs also promote immune evasion by silencing and/or destroying myeloid cells. While there are many common themes between the various CDCs, each CDC also possesses specific features to optimally benefit the pathogen producing it. This review highlights host responses to CDC pathogenesis with a focus on macrophages. Due to their robust plasticity, macrophages play key roles in the outcome of bacterial infections. Understanding the unique features and differences within the common theme of CDCs bolsters new tools for research and therapy.
Collapse
|
274
|
Brock DJ, Kondow-McConaghy H, Allen J, Brkljača Z, Kustigian L, Jiang M, Zhang J, Rye H, Vazdar M, Pellois JP. Mechanism of Cell Penetration by Permeabilization of Late Endosomes: Interplay between a Multivalent TAT Peptide and Bis(monoacylglycero)phosphate. Cell Chem Biol 2020; 27:1296-1307.e5. [PMID: 32783962 DOI: 10.1016/j.chembiol.2020.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 06/06/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023]
Abstract
Many cellular delivery reagents enter the cytosolic space of cells by escaping the lumen of endocytic organelles and, more specifically, late endosomes. The mechanisms involved in endosomal membrane permeation remain largely unresolved, which impedes the improvement of delivery agents. Here, we investigate how 3TAT, a branched analog of the cell-penetrating peptide (CPP) TAT, achieves the permeabilization of bilayers containing bis(monoacylglycero)phosphate (BMP), a lipid found in late endosomes. We establish that the peptide does not induce the leakage of individual lipid bilayers. Instead, leakage requires contact between membranes. Peptide-driven bilayer contacts lead to fusion, lipid mixing, and, critically, peptide encapsulation within proximal bilayers. Notably, this encapsulation is a distinctive property of BMP that explains the specificity of CPP's membrane leakage activity. These results therefore support a model of cell penetration that requires both BMP and the vicinity between bilayers, two features unique to BMP-rich and multivesicular late endosomes.
Collapse
Affiliation(s)
- Dakota J Brock
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Helena Kondow-McConaghy
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Jason Allen
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Zlatko Brkljača
- Division of Organic Chemistry and Biochemistry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Lauren Kustigian
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Mengqiu Jiang
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Junjie Zhang
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Hays Rye
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA
| | - Mario Vazdar
- Division of Organic Chemistry and Biochemistry, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University, Biochemistry and Biophysics Building, Room 430, 300 Olsen Boulevard, College Station, TX 77843-2128, USA; Department of Chemistry, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
275
|
ESCRTs cut some slack. Nat Cell Biol 2020; 22:915-916. [PMID: 32753668 DOI: 10.1038/s41556-020-0557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
276
|
Endosomal membrane tension regulates ESCRT-III-dependent intra-lumenal vesicle formation. Nat Cell Biol 2020; 22:947-959. [PMID: 32753669 PMCID: PMC7612185 DOI: 10.1038/s41556-020-0546-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Abstract
Plasma membrane tension strongly affects cell surface processes, such as migration, endocytosis and signalling. However, it is not known whether membrane tension of organelles regulates their functions, notably intracellular traffic. The ESCRT-III complex is the major membrane remodelling complex that drives Intra-Lumenal Vesicle (ILV) formation on endosomal membranes. Here, we made use of a fluorescent membrane tension probe to show that ESCRT-III subunits are recruited onto endosomal membranes when membrane tension is reduced. We find that tension-dependent recruitment is associated with ESCRT-III polymerization and membrane deformation in vitro, and correlates with increased ILV formation in ESCRT-III decorated endosomes in vivo. Finally, we find that endosomal membrane tension decreases when ILV formation is triggered by EGF under physiological conditions. These results indicate that membrane tension is a major regulator of ILV formation and of endosome trafficking, leading us to conclude that membrane tension can control organelle functions.
Collapse
|
277
|
Bohannon KP, Hanson PI. ESCRT puts its thumb on the nanoscale: Fixing tiny holes in endolysosomes. Curr Opin Cell Biol 2020; 65:122-130. [PMID: 32731154 PMCID: PMC7578027 DOI: 10.1016/j.ceb.2020.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/02/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022]
Abstract
The ESCRT (endosomal complex required for transport) machinery remodels membranes to bud vesicles away from the cytoplasm. In addition to this classic role, ESCRTs are now understood to repair damage in the plasma membrane, nuclear envelope, and throughout the endolysosomal network. Wounds in endolysosomal membranes are caused by pathogens, particulates, and other chemical or metabolic stresses. Nanoscale damage in these membranes promotes activation and engagement of ESCRT proteins. A full understanding of damage signals, molecular sensing, and the mechanism of membrane repair is yet to be developed. Nevertheless, a triggering role for calcium and ESCRT-I in recruiting ESCRT-III machinery for membrane remodeling is a repeated theme in functional studies of this response. In our current understanding of the continuum of cellular responses to lipid bilayer damage, the ESCRT machinery is fast, sensitive, and deployed independently of other systems.
Collapse
Affiliation(s)
- Kevin P Bohannon
- Department of Biological Chemistry, University of Michigan School of Medicine, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109, USA.
| | - Phyllis I Hanson
- Department of Biological Chemistry, University of Michigan School of Medicine, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
278
|
Abstract
Lysosomes are acidic, dynamic organelles that supervise catabolism, integrate signaling cascades, and tune cellular trafficking. Moreover, the loss of their integrity may jeopardize cell viability. In cancer cells, lysosomes are qualitatively and quantitatively modified for the tumor's own benefit. For all these reasons, these organelles emerge as appealing intracellular targets to manipulate non-oncogene addiction. This is of particular interest for brain diseases, including neurodegenerative disorders and cancer, in which stem cells are exhausted and transformed, respectively. Recent publications had demonstrated that stem cells displayed disarmed lysosomes in terms of number and functions during aging and oncogenic progression. Likewise, our laboratory identified that the arginine protease MALT1, normally dedicated to the assembly of proper NF-kB activation and processing a number of substrates, arbitrates lysosome biogenesis and mTOR signaling in glioblastoma stem-like cells. Indeed, blocking either the expression or the activity of this enzyme leads to an aberrant increase of lysosomes, alongside of the down-regulation of the mTOR signaling. This surge of lysosomes eradicates glioblastoma stem-like cells. Targeting lysosomes might thus inspire the design of new strategies to face this devastating human cancer. Here, we provide an overview of the functions of the lysosome as well as its role as a cell death initiator, to highlight the potential of lysosomal drugs for glioblastoma therapy.
Collapse
Affiliation(s)
- Kathryn A Jacobs
- Team SOAP, CRCINA, Inserm, CNRS, Université De Nantes, Université d'Angers , Nantes, France
| | - Clément Maghe
- Team SOAP, CRCINA, Inserm, CNRS, Université De Nantes, Université d'Angers , Nantes, France
| | - Julie Gavard
- Team SOAP, CRCINA, Inserm, CNRS, Université De Nantes, Université d'Angers , Nantes, France.,Integrated Center for Oncology, ICO , St. Herblain, France
| |
Collapse
|
279
|
Albrecht LV, Tejeda-Muñoz N, Bui MH, Cicchetto AC, Di Biagio D, Colozza G, Schmid E, Piccolo S, Christofk HR, De Robertis EM. GSK3 Inhibits Macropinocytosis and Lysosomal Activity through the Wnt Destruction Complex Machinery. Cell Rep 2020; 32:107973. [PMID: 32726636 PMCID: PMC7666578 DOI: 10.1016/j.celrep.2020.107973] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/29/2020] [Accepted: 07/08/2020] [Indexed: 12/17/2022] Open
Abstract
Canonical Wnt signaling is emerging as a major regulator of endocytosis. Here, we report that Wnt-induced macropinocytosis is regulated through glycogen synthase kinase 3 (GSK3) and the β-catenin destruction complex. We find that mutation of Axin1, a tumor suppressor and component of the destruction complex, results in the activation of macropinocytosis. Surprisingly, inhibition of GSK3 by lithium chloride (LiCl), CHIR99021, or dominant-negative GSK3 triggers macropinocytosis. GSK3 inhibition causes a rapid increase in acidic endolysosomes that is independent of new protein synthesis. GSK3 inhibition or Axin1 mutation increases lysosomal activity, which can be followed with tracers of active cathepsin D, β-glucosidase, and ovalbumin degradation. Microinjection of LiCl into the blastula cavity of Xenopus embryos causes a striking increase in dextran macropinocytosis. The effects of GSK3 inhibition on protein degradation in endolysosomes are blocked by the macropinocytosis inhibitors EIPA or IPA-3, suggesting that increases in membrane trafficking drive lysosomal activity.
Collapse
Affiliation(s)
- Lauren V Albrecht
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Maggie H Bui
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Andrew C Cicchetto
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Daniele Di Biagio
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy
| | - Gabriele Colozza
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Ernst Schmid
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy
| | - Heather R Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA
| | - Edward M De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA.
| |
Collapse
|
280
|
Ugbode C, Fort-Aznar L, Sweeney ST. Leaky endosomes push tau over the seed limit. J Biol Chem 2020; 294:18967-18968. [PMID: 31836670 DOI: 10.1074/jbc.h119.011687] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The inter- and intracellular propagation of aggregated proteins like tau is emerging as a central mechanism behind progression of various neurodegenerative diseases. The steps by which tau aggregates and propagates is currently unclear. Chen et al. now combine a cell-based model of tau aggregation with a CRISPR interference (CRISPRi) genetic screen to identify components of the endosomal sorting complex required for transport (ESCRT) machinery as mediators of intracellular propagation of tau aggregates. These findings reveal a role for endolysosomal integrity in blocking tau propagation.
Collapse
Affiliation(s)
- Chris Ugbode
- Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Laura Fort-Aznar
- Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Sean T Sweeney
- Department of Biology, University of York, York YO10 5DD, United Kingdom
| |
Collapse
|
281
|
Lusk CP, Ader NR. CHMPions of repair: Emerging perspectives on sensing and repairing the nuclear envelope barrier. Curr Opin Cell Biol 2020; 64:25-33. [PMID: 32105978 PMCID: PMC7371540 DOI: 10.1016/j.ceb.2020.01.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/20/2022]
Abstract
Understanding how the integrity of the nuclear membranes is protected against internal and external stresses is an emergent challenge. Work reviewed here investigated the mechanisms by which losses of nuclear-cytoplasmic compartmentalization are sensed and ameliorated. Fundamental to these is spatial control over interactions between the endosomal sorting complexes required for transport machinery and LAP2-emerin-MAN1 family inner nuclear membrane proteins, which together promote nuclear envelope sealing in interphase and at the end of mitosis. We suggest that the size of the nuclear envelope hole dictates the mechanism of its repair, with larger holes requiring barrier-to-autointegration factor and the potential triggering of a postmitotic nuclear envelope reassembly pathway in interphase. We also consider why these mechanisms fail at ruptured micronuclei. Together, this work re-emphasizes the need to understand how membrane flow and local lipid metabolism help ensure that the nuclear envelope is refractory to mechanical rupture yet fluid enough to allow its essential dynamics.
Collapse
Affiliation(s)
- C Patrick Lusk
- Department of Cell Biology, Yale School of Medicine, 295 Congress Avenue, New Haven, CT, 06520, USA.
| | - Nicholas R Ader
- Department of Cell Biology, Yale School of Medicine, 295 Congress Avenue, New Haven, CT, 06520, USA
| |
Collapse
|
282
|
Ferling I, Dunn JD, Ferling A, Soldati T, Hillmann F. Conidial Melanin of the Human-Pathogenic Fungus Aspergillus fumigatus Disrupts Cell Autonomous Defenses in Amoebae. mBio 2020; 11:e00862-20. [PMID: 32457245 PMCID: PMC7251208 DOI: 10.1128/mbio.00862-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022] Open
Abstract
The human-pathogenic fungus Aspergillus fumigatus is a ubiquitous saprophyte that causes fatal lung infections in immunocompromised individuals. Following inhalation, conidia are ingested by innate immune cells and can arrest phagolysosome maturation. How this virulence trait could have been selected for in natural environments is unknown. Here, we found that surface exposure of the green pigment 1,8-dihydroxynaphthalene-(DHN)-melanin can protect conidia from phagocytic uptake and intracellular killing by the fungivorous amoeba Protostelium aurantium and delays its exocytosis from the nonfungivorous species Dictyostelium discoideum To elucidate the antiphagocytic properties of the surface pigment, we followed the antagonistic interactions of A. fumigatus conidia with the amoebae in real time. For both amoebae, conidia covered with DHN-melanin were internalized at far lower rates than were seen with conidia lacking the pigment, despite high rates of initial attachment to nonkilling D. discoideum When ingested by D. discoideum, the formation of nascent phagosomes was followed by transient acidification of phagolysosomes, their subsequent neutralization, and, finally, exocytosis of the conidia. While the cycle was completed in less than 1 h for unpigmented conidia, the process was significantly prolonged for conidia covered with DHN-melanin, leading to an extended intracellular residence time. At later stages of this cellular infection, pigmented conidia induced enhanced damage to phagolysosomes and infected amoebae failed to recruit the ESCRT (endosomal sorting complex required for transport) membrane repair machinery or the canonical autophagy pathway to defend against the pathogen, thus promoting prolonged intracellular persistence in the host cell and the establishment of a germination niche in this environmental phagocyte.IMPORTANCE Infections with Aspergillus fumigatus are usually acquired by an inhalation of spores from environmental sources. How spores of a saprophytic fungus have acquired abilities to withstand and escape the phagocytic attacks of innate immune cells is not understood. The fungal surface pigment dihydroxynaphtalene-melanin has been shown to be a crucial factor for the delay in phagosome maturation. Here, we show that this pigment also has a protective function against environmental phagocytes. Pigmented conidia escaped uptake and killing by the fungus-eating amoeba Protostelium aurantium When ingested by the nonfungivorous phagocyte Dictyostelium discoideum, the pigment attenuated the launch of cell autonomous defenses against the fungal invader, such as membrane repair and autophagy, leading to prolonged intracellular retention. Membrane damage and cytoplasmic leakage may result in an influx of nutrients and thus may further promote intracellular germination of the fungus, indicating that A. fumigatus has acquired some of the basic properties of intracellular pathogens.
Collapse
Affiliation(s)
- Iuliia Ferling
- Junior Research Group Evolution of Microbial Interactions, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Joe Dan Dunn
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Alexander Ferling
- Heid-Tech, Technische Schule Heidenheim, Heidenheim an der Brenz, Germany
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Falk Hillmann
- Junior Research Group Evolution of Microbial Interactions, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| |
Collapse
|
283
|
Birgisdottir ÅB, Johansen T. Autophagy and endocytosis – interconnections and interdependencies. J Cell Sci 2020; 133:133/10/jcs228114. [DOI: 10.1242/jcs.228114] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
ABSTRACT
Autophagy and endocytosis are membrane-vesicle-based cellular pathways for degradation and recycling of intracellular and extracellular components, respectively. These pathways have a common endpoint at the lysosome, where their cargo is degraded. In addition, the two pathways intersect at different stages during vesicle formation, fusion and trafficking, and share parts of the molecular machinery. Accumulating evidence shows that autophagy is dependent upon endocytosis and vice versa. The emerging joint network of autophagy and endocytosis is of vital importance for cellular metabolism and signaling, and thus also highly relevant in disease settings. In this Review, we will discuss examples of how the autophagy machinery impacts on endocytosis and cell signaling, and highlight how endocytosis regulates the different steps in autophagy in mammalian cells. Finally, we will focus on the interplay of these pathways in the quality control of their common endpoint, the lysosome.
Collapse
Affiliation(s)
- Åsa B. Birgisdottir
- The Heart and Lung Clinic, University Hospital of North Norway, 9037 Tromsø, Norway
- Clinical Cardiovascular Research Group, Department of Clinical Medicine, University of Tromsø –The Arctic University of Norway, 9037 Tromsø, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø–The Arctic University of Norway, 9037 Tromsø, Norway
| |
Collapse
|
284
|
Eriksson I, Wäster P, Öllinger K. Restoration of lysosomal function after damage is accompanied by recycling of lysosomal membrane proteins. Cell Death Dis 2020; 11:370. [PMID: 32409651 PMCID: PMC7224388 DOI: 10.1038/s41419-020-2527-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022]
Abstract
Lysosomes are central organelles for cellular degradation and energy homeostasis. In addition, lysosomal membrane permeabilization (LMP) and subsequent release of lysosomal content to the cytosol can initiate programmed cell death. The extent of LMP and available repair mechanisms determine the cell fate after lysosomal damage. In this study, we aimed to investigate the premises for lysosomal membrane repair after LMP and found that lysosomal membrane damage initiated by L-leucyl-L-leucine methyl ester (LLOMe) caused caspase-dependent apoptosis in almost 50% of the cells, while the rest recovered. Immediately after LLOMe addition, lysosomal proteases were detected in the cytosol and the ESCRT-components ALIX and CHMP4B were recruited to the lysosomal membrane. Next, lysophagic clearance of damaged lysosomes was evident and a concentration-dependent translocation of several lysosomal membrane proteins, including LAMP2, to the cytosol was found. LAMP2 was present in small vesicles with the N-terminal protein chain facing the lumen of the vesicle. We conclude that lysophagic clearance of damaged lysosomes results in generation of lysosomal membrane protein complexes, which constitute small membrane enclosed units, possibly for recycling of lysosomal membrane proteins. These lysosomal membrane complexes enable an efficient regeneration of lysosomes to regain cell functionality.
Collapse
Affiliation(s)
- Ida Eriksson
- Experimental Pathology, Department of Biomedical and Clinical Sciences, Linköping University, 58185, Linköping, Sweden
| | - Petra Wäster
- Experimental Pathology, Department of Biomedical and Clinical Sciences, Linköping University, 58185, Linköping, Sweden
| | - Karin Öllinger
- Experimental Pathology, Department of Biomedical and Clinical Sciences, Linköping University, 58185, Linköping, Sweden.
| |
Collapse
|
285
|
Rusmini P, Cristofani R, Tedesco B, Ferrari V, Messi E, Piccolella M, Casarotto E, Chierichetti M, Cicardi ME, Galbiati M, Geroni C, Lombardi P, Crippa V, Poletti A. Enhanced Clearance of Neurotoxic Misfolded Proteins by the Natural Compound Berberine and Its Derivatives. Int J Mol Sci 2020; 21:ijms21103443. [PMID: 32414108 PMCID: PMC7279252 DOI: 10.3390/ijms21103443] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Accumulation of misfolded proteins is a common hallmark of several neurodegenerative disorders (NDs) which results from a failure or an impairment of the protein quality control (PQC) system. The PQC system is composed by chaperones and the degradative systems (proteasome and autophagy). Mutant proteins that misfold are potentially neurotoxic, thus strategies aimed at preventing their aggregation or at enhancing their clearance are emerging as interesting therapeutic targets for NDs. Methods: We tested the natural alkaloid berberine (BBR) and some derivatives for their capability to enhance misfolded protein clearance in cell models of NDs, evaluating which degradative pathway mediates their action. Results: We found that both BBR and its semisynthetic derivatives promote degradation of mutant androgen receptor (ARpolyQ) causative of spinal and bulbar muscular atrophy, acting mainly via proteasome and preventing ARpolyQ aggregation. Overlapping effects were observed on other misfolded proteins causative of amyotrophic lateral sclerosis, frontotemporal-lobar degeneration or Huntington disease, but with selective and specific action against each different mutant protein. Conclusions: BBR and its analogues induce the clearance of misfolded proteins responsible for NDs, representing potential therapeutic tools to counteract these fatal disorders.
Collapse
Affiliation(s)
- Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Barbara Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Veronica Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Elio Messi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Margherita Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Elena Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Marta Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Maria Elena Cicardi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Cristina Geroni
- Naxospharma srl, Novate Milanese, 20026 Milan, Italy; (C.G.); (P.L.)
| | - Paolo Lombardi
- Naxospharma srl, Novate Milanese, 20026 Milan, Italy; (C.G.); (P.L.)
| | - Valeria Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
- Correspondence:
| |
Collapse
|
286
|
Kavčič N, Butinar M, Sobotič B, Hafner Česen M, Petelin A, Bojić L, Zavašnik Bergant T, Bratovš A, Reinheckel T, Turk B. Intracellular cathepsin C levels determine sensitivity of cells to leucyl-leucine methyl ester-triggered apoptosis. FEBS J 2020; 287:5148-5166. [PMID: 32319717 DOI: 10.1111/febs.15326] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 02/06/2020] [Accepted: 03/05/2020] [Indexed: 12/20/2022]
Abstract
L-leucyl-leucine methyl ester (LLOMe) is a lysosomotropic detergent, which was evaluated in clinical trials in graft-vs-host disease because it very efficiently killed monocytic cell lines. It was also shown to efficiently trigger apoptosis in cancer cells, suggesting that the drug might have potential in anticancer therapy. Using U-937 and THP-1 promonocytes as models for monocytic cells, U-87-MG and HeLa cells as models for cancer cells, and noncancerous HEK293 cells, we show that the drug triggers rapid cathepsin C-dependent lysosomal membrane permeabilization, followed by the release of other cysteine cathepsins into the cytosol and subsequent apoptosis. However, monocytes were found to be far more sensitive to the drug than the cancer and noncancer cells, which is most likely a consequence of the much higher intracellular levels of cathepsin C-the most upstream molecule in the pathway-in monocytic cell lines as compared to cancer cells. Overexpression of cathepsin C in HEK293 cells substantially enhances their sensitivity to the drug, consistent with the crucial role of cathepsin C. Major involvement of cysteine cathepsins B, S, and L in the downstream signaling pathway to mitochondrial cell death was confirmed in two gene ablation models, including the ablation of the major cytosolic inhibitor of cysteine cathepsins, stefin B, in primary mouse cancer cells, and simultaneous ablation of two major cathepsins, B and L, in mouse embryonic fibroblasts (MEFs). Deletion of stefin B resulted in sensitizing primary murine breast cancer cells to cell death without affecting the release of cathepsins, whereas simultaneous ablation of cathepsins B and L largely protected MEFs against cell death. However, due to the extreme sensitivity of monocytes to LLOMe, it appears that the drug may not be suitable for anticancer therapy due to risk of systemic toxicity.
Collapse
Affiliation(s)
- Nežka Kavčič
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Miha Butinar
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Barbara Sobotič
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Maruša Hafner Česen
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Ana Petelin
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | - Lea Bojić
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Tina Zavašnik Bergant
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Andreja Bratovš
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia.,Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Thomas Reinheckel
- Medical Faculty, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs University, Freiburg, Germany.,German Cancer Consortium (DKTK) partner site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Boris Turk
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, Ljubljana, Slovenia
| |
Collapse
|
287
|
Interactions of the Lysosomotropic Detergent O-Methyl-Serine Dodecylamide Hydrochloride (MSDH) with Lipid Bilayer Membranes-Implications for Cell Toxicity. Int J Mol Sci 2020; 21:ijms21093136. [PMID: 32365555 PMCID: PMC7247706 DOI: 10.3390/ijms21093136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022] Open
Abstract
O-methyl-serine dodecylamine hydrochloride (MSDH) is a detergent that accumulates selectively in lysosomes, a so-called lysosomotropic detergent, with unexpected chemical properties. At physiological pH, it spontaneously forms vesicles, which disassemble into small aggregates (probably micelles) below pH 6.4. In this study, we characterize the interaction between MSDH and liposomes at different pH and correlate the findings to toxicity in human fibroblasts. We find that the effect of MSDH on lipid membranes is highly pH-dependent. At neutral pH, the partitioning of MSDH into the liposome membrane is immediate and causes the leakage of small fluorophores, unless the ratio between MSDH and lipids is kept low. At pH 5, the partitioning of MSDH into the membrane is kinetically impeded since MSDH is charged and a high ratio between MSDH and the lipids is required to permeabilize the membrane. When transferred to cell culture conditions, the ratio between MSDH and plasma membrane lipids must therefore be low, at physiological pH, to maintain plasma membrane integrity. Transmission electron microscopy suggests that MSDH vesicles are taken up by endocytosis. As the pH of the endosomal compartment progressively drops, MSDH vesicles disassemble, leading to a high concentration of increasingly charged MSDH in small aggregates inside the lysosomes. At sufficiently high MSDH concentrations, the lysosome is permeabilized, the proteolytic content released to the cytosol and apoptotic cell death is induced.
Collapse
|
288
|
Nguyen HC, Talledge N, McCullough J, Sharma A, Moss FR, Iwasa JH, Vershinin MD, Sundquist WI, Frost A. Membrane constriction and thinning by sequential ESCRT-III polymerization. Nat Struct Mol Biol 2020; 27:392-399. [PMID: 32251413 PMCID: PMC7343221 DOI: 10.1038/s41594-020-0404-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/05/2020] [Indexed: 01/04/2023]
Abstract
The endosomal sorting complexes required for transport (ESCRTs) mediate diverse membrane remodeling events. These typically require ESCRT-III proteins to stabilize negatively curved membranes; however, recent work has indicated that certain ESCRT-IIIs also participate in positive-curvature membrane-shaping reactions. ESCRT-IIIs polymerize into membrane-binding filaments, but the structural basis for negative versus positive membrane remodeling by these proteins remains poorly understood. To learn how certain ESCRT-IIIs shape positively curved membranes, we determined structures of human membrane-bound CHMP1B-only, membrane-bound CHMP1B + IST1, and IST1-only filaments by cryo-EM. Our structures show how CHMP1B first polymerizes into a single-stranded helical filament, shaping membranes into moderate-curvature tubules. Subsequently, IST1 assembles a second strand on CHMP1B, further constricting the membrane tube and reducing its diameter nearly to the fission point. Each step of constriction thins the underlying bilayer, lowering the barrier to membrane fission. Our structures reveal how a two-component, sequential polymerization mechanism drives membrane tubulation, constriction and bilayer thinning.
Collapse
Affiliation(s)
- Henry C Nguyen
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Nathaniel Talledge
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
- Institute for Molecular Virology, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - John McCullough
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Abhimanyu Sharma
- Department of Physics & Astronomy, University of Utah, Salt Lake City, UT, USA
| | - Frank R Moss
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Janet H Iwasa
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Michael D Vershinin
- Department of Physics & Astronomy, University of Utah, Salt Lake City, UT, USA
- Department of Biology, University of Utah, Salt Lake City, UT, USA
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT, USA
| | - Wesley I Sundquist
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
| | - Adam Frost
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
289
|
Mandal T, Lough W, Spagnolie SE, Audhya A, Cui Q. Molecular Simulation of Mechanical Properties and Membrane Activities of the ESCRT-III Complexes. Biophys J 2020; 118:1333-1343. [PMID: 32078797 PMCID: PMC7091516 DOI: 10.1016/j.bpj.2020.01.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/26/2020] [Accepted: 01/29/2020] [Indexed: 01/16/2023] Open
Abstract
The endosomal sorting complex required for transport (ESCRT) machinery carries out the membrane scission reactions that are required for many biological processes throughout cells. How ESCRTs bind and deform cellular membranes and ultimately produce vesicles has been a matter of active research in recent years. In this study, we use fully atomistic molecular dynamics simulations to scrutinize the structural details of a filament composed of Vps32 protomers, a major component of ESCRT-III complexes. The simulations show that both hydrophobic and electrostatic interactions between monomers help maintain the structural stability of the filament, which exhibits an intrinsic bend and twist. Our findings suggest that the accumulation of bending and twisting stresses as the filament elongates on the membrane surface likely contributes to the driving force for membrane invagination. The filament exposes a large cationic surface that senses the negatively charged lipids in the membrane, and the N-terminal amphipathic helix of the monomers not only acts as a membrane anchor but also generates significant positive membrane curvature. Taking all results together, we discuss a plausible mechanism for membrane invagination driven by ESCRT-III.
Collapse
Affiliation(s)
- Taraknath Mandal
- Department of Chemistry, Boston University, Boston, Massachusetts
| | | | | | - Anjon Audhya
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin
| | - Qiang Cui
- Departments of Chemistry, Physics, and Biomedical Engineering, Boston University, Boston, Massachusetts.
| |
Collapse
|
290
|
Mesquita FS, van der Goot FG, Sergeeva OA. Mammalian membrane trafficking as seen through the lens of bacterial toxins. Cell Microbiol 2020; 22:e13167. [PMID: 32185902 PMCID: PMC7154709 DOI: 10.1111/cmi.13167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/12/2022]
Abstract
A fundamental question of eukaryotic cell biology is how membrane organelles are organised and interact with each other. Cell biologists address these questions by characterising the structural features of membrane compartments and the mechanisms that coordinate their exchange. To do so, they must rely on variety of cargo molecules and treatments that enable targeted perturbation, localisation, and labelling of specific compartments. In this context, bacterial toxins emerged in cell biology as paradigm shifting molecules that enabled scientists to not only study them from the side of bacterial infection but also from the side of the mammalian host. Their selectivity, potency, and versatility made them exquisite tools for uncovering much of our current understanding of membrane trafficking mechanisms. Here, we will follow the steps that lead toxins until their intracellular targets, highlighting how specific events helped us comprehend membrane trafficking and establish the fundamentals of various cellular organelles and processes. Bacterial toxins will continue to guide us in answering crucial questions in cellular biology while also acting as probes for new technologies and applications.
Collapse
Affiliation(s)
| | | | - Oksana A Sergeeva
- Global Health Institute, School of Life Sciences, EPFL, Lausanne, Switzerland
| |
Collapse
|
291
|
Larios J, Mercier V, Roux A, Gruenberg J. ALIX- and ESCRT-III-dependent sorting of tetraspanins to exosomes. J Cell Biol 2020; 219:e201904113. [PMID: 32049272 PMCID: PMC7054990 DOI: 10.1083/jcb.201904113] [Citation(s) in RCA: 259] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 10/31/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
The intraluminal vesicles (ILVs) of endosomes mediate the delivery of activated signaling receptors and other proteins to lysosomes for degradation, but they also modulate intercellular communication when secreted as exosomes. The formation of ILVs requires four complexes, ESCRT-0, -I, -II, and -III, with ESCRT-0, -I, and -II presumably involved in cargo sorting and ESCRT-III in membrane deformation and fission. Here, we report that an active form of the ESCRT-associated protein ALIX efficiently recruits ESCRT-III proteins to endosomes. This recruitment occurs independently of other ESCRTs but requires lysobisphosphatidic acid (LBPA) in vivo, and can be reconstituted on supported bilayers in vitro. Our data indicate that this ALIX- and ESCRT-III-dependent pathway promotes the sorting and delivery of tetraspanins to exosomes. We conclude that ALIX provides an additional pathway of ILV formation, secondary to the canonical pathway, and that this pathway controls the targeting of exosomal proteins.
Collapse
|
292
|
Göser V, Kehl A, Röder J, Hensel M. Role of the ESCRT‐III complex in controlling integrity of the
Salmonella
‐containing vacuole. Cell Microbiol 2020; 22:e13176. [DOI: 10.1111/cmi.13176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/27/2019] [Accepted: 01/20/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Vera Göser
- Abt. MikrobiologieFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
| | - Alexander Kehl
- Abt. MikrobiologieFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
- Institut für HygieneUniversität Münster Münster Germany
- CellNanOs, Center for Cellular NanoanalyticsFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
| | - Jennifer Röder
- Abt. MikrobiologieFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
| | - Michael Hensel
- Abt. MikrobiologieFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
- CellNanOs, Center for Cellular NanoanalyticsFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
| |
Collapse
|
293
|
Meng B, Ip NCY, Abbink TEM, Kenyon JC, Lever AML. ESCRT-II functions by linking to ESCRT-I in human immunodeficiency virus-1 budding. Cell Microbiol 2020; 22:e13161. [PMID: 31922351 PMCID: PMC7187348 DOI: 10.1111/cmi.13161] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 10/29/2019] [Accepted: 11/14/2019] [Indexed: 01/05/2023]
Abstract
Human immunodeficiency virus (HIV) uses the ESCRT (endosomal sorting complexes required for transport) protein pathway to bud from infected cells. Despite the roles of ESCRT-I and -III in HIV budding being firmly established, participation of ESCRT-II in this process has been controversial. EAP45 is a critical component of ESCRT-II. Previously, we utilised a CRISPR-Cas9 EAP45 knockout cell line to assess the involvement of ESCRT-II in HIV replication. We demonstrated that the absence of ESCRT-II impairs HIV budding. Here, we show that virus spread is also defective in physiologically relevant CRISPR/Cas9 EAP45 knockout T cells. We further show reappearance of efficient budding by re-introduction of EAP45 expression into EAP45 knockout cells. Using expression of selected mutants of EAP45, we dissect the domain requirement responsible for this function. Our data show at the steady state that rescue of budding is only observed in the context of a Gag/Pol, but not a Gag expressor, indicating that the size of cargo determines the usage of ESCRT-II. EAP45 acts through the YPXL-ALIX pathway as partial rescue is achieved in a PTAP but not a YPXL mutant virus. Our study clarifies the role of ESCRT-II in the late stages of HIV replication and reinforces the notion that ESCRT-II plays an integral part during this process as it does in sorting ubiquitinated cargos and in cytokinesis.
Collapse
Affiliation(s)
- Bo Meng
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Natasha C Y Ip
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Truus E M Abbink
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Julia C Kenyon
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.,Department of Microbiology and Immunology, National University of Singapore, Singapore.,Homerton College, Cambridge, UK
| | - Andrew M L Lever
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.,Department of Medicine, National University of Singapore, Singapore
| |
Collapse
|
294
|
Yao RQ, Ren C, Xia ZF, Yao YM. Organelle-specific autophagy in inflammatory diseases: a potential therapeutic target underlying the quality control of multiple organelles. Autophagy 2020; 17:385-401. [PMID: 32048886 PMCID: PMC8007140 DOI: 10.1080/15548627.2020.1725377] [Citation(s) in RCA: 269] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The structural integrity and functional stability of organelles are prerequisites for the viability and responsiveness of cells. Dysfunction of multiple organelles is critically involved in the pathogenesis and progression of various diseases, such as chronic obstructive pulmonary disease, cardiovascular diseases, infection, and neurodegenerative diseases. In fact, those organelles synchronously present with evident structural derangement and aberrant function under exposure to different stimuli, which might accelerate the corruption of cells. Therefore, the quality control of multiple organelles is of great importance in maintaining the survival and function of cells and could be a potential therapeutic target for human diseases. Organelle-specific autophagy is one of the major subtypes of autophagy, selectively targeting different organelles for quality control. This type of autophagy includes mitophagy, pexophagy, reticulophagy (endoplasmic reticulum), ribophagy, lysophagy, and nucleophagy. These kinds of organelle-specific autophagy are reported to be beneficial for inflammatory disorders by eliminating damaged organelles and maintaining homeostasis. In this review, we summarized the recent findings and mechanisms covering different kinds of organelle-specific autophagy, as well as their involvement in various diseases, aiming to arouse concern about the significance of the quality control of multiple organelles in the treatment of inflammatory diseases.Abbreviations: ABCD3: ATP binding cassette subfamily D member 3; AD: Alzheimer disease; ALS: amyotrophic lateral sclerosis; AMBRA1: autophagy and beclin 1 regulator 1; AMPK: AMP-activated protein kinase; ARIH1: ariadne RBR E3 ubiquitin protein ligase 1; ATF: activating transcription factor; ATG: autophagy related; ATM: ATM serine/threonine kinase; BCL2: BCL2 apoptosis regulator; BCL2L11/BIM: BCL2 like 11; BCL2L13: BCL2 like 13; BECN1: beclin 1; BNIP3: BCL2 interacting protein 3; BNIP3L/NIX: BCL2 interacting protein 3 like; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CANX: calnexin; CAT: catalase; CCPG1: cell cycle progression 1; CHDH: choline dehydrogenase; COPD: chronic obstructive pulmonary disease; CSE: cigarette smoke exposure; CTSD: cathepsin D; DDIT3/CHOP: DNA-damage inducible transcript 3; DISC1: DISC1 scaffold protein; DNM1L/DRP1: dynamin 1 like; EIF2AK3/PERK: eukaryotic translation initiation factor 2 alpha kinase 3; EIF2S1/eIF2α: eukaryotic translation initiation factor 2 alpha kinase 3; EMD: emerin; EPAS1/HIF-2α: endothelial PAS domain protein 1; ER: endoplasmic reticulum; ERAD: ER-associated degradation; ERN1/IRE1α: endoplasmic reticulum to nucleus signaling 1; FBXO27: F-box protein 27; FKBP8: FKBP prolyl isomerase 8; FTD: frontotemporal dementia; FUNDC1: FUN14 domain containing 1; G3BP1: G3BP stress granule assembly factor 1; GBA: glucocerebrosidase beta; HIF1A/HIF1: hypoxia inducible factor 1 subunit alpha; IMM: inner mitochondrial membrane; LCLAT1/ALCAT1: lysocardiolipin acyltransferase 1; LGALS3/Gal3: galectin 3; LIR: LC3-interacting region; LMNA: lamin A/C; LMNB1: lamin B1; LPS: lipopolysaccharide; MAPK8/JNK: mitogen-activated protein kinase 8; MAMs: mitochondria-associated membranes; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MFN1: mitofusin 1; MOD: multiple organelles dysfunction; MTPAP: mitochondrial poly(A) polymerase; MUL1: mitochondrial E3 ubiquitin protein ligase 1; NBR1: NBR1 autophagy cargo receptor; NLRP3: NLR family pyrin domain containing 3; NUFIP1: nuclear FMR1 interacting protein 1; OMM: outer mitochondrial membrane; OPTN: optineurin; PD: Parkinson disease; PARL: presenilin associated rhomboid like; PEX3: peroxisomal biogenesis factor 3; PGAM5: PGAM family member 5; PHB2: prohibitin 2; PINK1: PTEN induced putative kinase 1; PRKN: parkin RBR E3 ubiquitin protein ligase; RB1CC1/FIP200: RB1 inducible coiled-coil 1; RETREG1/FAM134B: reticulophagy regulator 1; RHOT1/MIRO1: ras homolog family member T1; RIPK3/RIP3: receptor interacting serine/threonine kinase 3; ROS: reactive oxygen species; RTN3: reticulon 3; SEC62: SEC62 homolog, preprotein translocation factor; SESN2: sestrin2; SIAH1: siah E3 ubiquitin protein ligase 1; SNCA: synuclein alpha; SNCAIP: synuclein alpha interacting protein; SQSTM1/p62: sequestosome 1; STING1: stimulator of interferon response cGAMP interactor 1; TAX1BP1: Tax1 binding protein 1; TBK1: TANK binding kinase 1; TFEB: transcription factor EB; TICAM1/TRIF: toll-like receptor adaptor molecule 1; TIMM23: translocase of inner mitochondrial membrane 23; TNKS: tankyrase; TOMM: translocase of the outer mitochondrial membrane; TRIM: tripartite motif containing; UCP2: uncoupling protein 2; ULK1: unc-51 like autophagy activating kinase; UPR: unfolded protein response; USP10: ubiquitin specific peptidase 10; VCP/p97: valosin containing protein; VDAC: voltage dependent anion channels; XIAP: X-linked inhibitor of apoptosis; ZNHIT3: zinc finger HIT-type containing 3.
Collapse
Affiliation(s)
- Ren-Qi Yao
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, People's Republic of China.,Department of Burn Surgery, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Chao Ren
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhao-Fan Xia
- Department of Burn Surgery, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Yong-Ming Yao
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
295
|
Abstract
Autophagy is a major intracellular degradation system that derives its degradative abilities from the lysosome. The most well-studied form of autophagy is macroautophagy, which delivers cytoplasmic material to lysosomes via the double-membraned autophagosome. Other forms of autophagy, namely chaperone-mediated autophagy and microautophagy, occur directly on the lysosome. Besides providing the means for degradation, lysosomes are also involved in autophagy regulation and can become substrates of autophagy when damaged. During autophagy, they exhibit notable changes, including increased acidification, enhanced enzymatic activity, and perinuclear localization. Despite their importance to autophagy, details on autophagy-specific regulation of lysosomes remain relatively scarce. This review aims to provide a summary of current understanding on the behaviour of lysosomes during autophagy and outline unexplored areas of autophagy-specific lysosome research.
Collapse
Affiliation(s)
- Willa Wen-You Yim
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, 113-0033 Japan
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, 113-0033 Japan
| |
Collapse
|
296
|
Cui L, Zhao LP, Ye JY, Yang L, Huang Y, Jiang XP, Zhang Q, Jia JZ, Zhang DX, Huang Y. The Lysosomal Membrane Protein Lamp2 Alleviates Lysosomal Cell Death by Promoting Autophagic Flux in Ischemic Cardiomyocytes. Front Cell Dev Biol 2020; 8:31. [PMID: 32117965 PMCID: PMC7019187 DOI: 10.3389/fcell.2020.00031] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/14/2020] [Indexed: 12/22/2022] Open
Abstract
Lysosomal membrane permeabilization (LMP) has recently been recognized as an important cell death pathway in various cell types. However, studies regarding the correlation between LMP and cardiomyocyte death are scarce. Lysosomal membrane-associated protein 2 (Lamp2) is an important component of lysosomal membranes and is involved in both autophagy and LMP. In the present study, we found that the protein content of Lamp2 gradually decreased in response to oxygen, glucose and serum deprivation (OGD) treatment in vitro. To further elucidate its role in ischemic cardiomyocytes, particularly with respect to autophagy and LMP, we infected cardiomyocytes with adenovirus carrying full-length Lamp2 to restore its protein level in cells. We found that OGD treatment resulted in the occurrence of LMP and a decline in the viability of cardiomyocytes, which were remarkably reversed by Lamp2 restoration. Exogenous expression of Lamp2 also significantly alleviated the autophagic flux blockade induced by OGD treatment by promoting the trafficking of cathepsin B (Cat B) and cathepsin D (Cat D). Through drug intervention and gene regulation to alleviate and exacerbate autophagic flux blockade respectively, we found that impaired autophagic flux in response to ischemic injury contributed to the occurrence of LMP in cardiomyocytes. In conclusion, our present data suggest that Lamp2 overexpression can improve autophagic flux blockade probably by promoting the trafficking of cathepsins and consequently conferring cardiomyocyte resistance against lysosomal cell death (LCD) that is induced by ischemic injury. These results may indicate a new therapeutic target for ischemic heart damage.
Collapse
Affiliation(s)
- Lin Cui
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li-Ping Zhao
- Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, China
| | - Jing-Ying Ye
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lei Yang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yao Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xu-Pin Jiang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qiong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jie-Zhi Jia
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Dong-Xia Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
297
|
Jia J, Bissa B, Brecht L, Allers L, Choi SW, Gu Y, Zbinden M, Burge MR, Timmins G, Hallows K, Behrends C, Deretic V. AMPK, a Regulator of Metabolism and Autophagy, Is Activated by Lysosomal Damage via a Novel Galectin-Directed Ubiquitin Signal Transduction System. Mol Cell 2020; 77:951-969.e9. [PMID: 31995728 DOI: 10.1016/j.molcel.2019.12.028] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/27/2019] [Accepted: 12/24/2019] [Indexed: 12/29/2022]
Abstract
AMPK is a central regulator of metabolism and autophagy. Here we show how lysosomal damage activates AMPK. This occurs via a hitherto unrecognized signal transduction system whereby cytoplasmic sentinel lectins detect membrane damage leading to ubiquitination responses. Absence of Galectin 9 (Gal9) or loss of its capacity to recognize lumenal glycans exposed during lysosomal membrane damage abrogate such ubiquitination responses. Proteomic analyses with APEX2-Gal9 have revealed global changes within the Gal9 interactome during lysosomal damage. Gal9 association with lysosomal glycoproteins increases whereas interactions with a newly identified Gal9 partner, deubiquitinase USP9X, diminishes upon lysosomal injury. In response to damage, Gal9 displaces USP9X from complexes with TAK1 and promotes K63 ubiquitination of TAK1 thus activating AMPK on damaged lysosomes. This triggers autophagy and contributes to autophagic control of membrane-damaging microbe Mycobacterium tuberculosis. Thus, galectin and ubiquitin systems converge to activate AMPK and autophagy during endomembrane homeostasis.
Collapse
Affiliation(s)
- Jingyue Jia
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Bhawana Bissa
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Lukas Brecht
- Munich Cluster of Systems Neurology, Munich, Germany
| | - Lee Allers
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Seong Won Choi
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Yuexi Gu
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Mark Zbinden
- Human Metabolome Technologies America, Boston, MA, USA
| | - Mark R Burge
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Graham Timmins
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; School pf Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Kenneth Hallows
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Vojo Deretic
- Autophagy, Inflammation and Metabolism AIM Center of Biochemical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM, USA.
| |
Collapse
|
298
|
Abstract
Being originally discovered as cellular recycling bins, lysosomes are today recognized as versatile signaling organelles that control a wide range of cellular functions that are essential not only for the well-being of normal cells but also for malignant transformation and cancer progression. In addition to their core functions in waste disposal and recycling of macromolecules and energy, lysosomes serve as an indispensable support system for malignant phenotype by promoting cell growth, cytoprotective autophagy, drug resistance, pH homeostasis, invasion, metastasis, and genomic integrity. On the other hand, malignant transformation reduces the stability of lysosomal membranes rendering cancer cells sensitive to lysosome-dependent cell death. Notably, many clinically approved cationic amphiphilic drugs widely used for the treatment of other diseases accumulate in lysosomes, interfere with their cancer-promoting and cancer-supporting functions and destabilize their membranes thereby opening intriguing possibilities for cancer therapy. Here, we review the emerging evidence that supports the supplementation of current cancer therapies with lysosome-targeting cationic amphiphilic drugs.
Collapse
|
299
|
Zhou K, Guo S, Li F, Sun Q, Liang G. Exosomal PD-L1: New Insights Into Tumor Immune Escape Mechanisms and Therapeutic Strategies. Front Cell Dev Biol 2020; 8:569219. [PMID: 33178688 PMCID: PMC7593554 DOI: 10.3389/fcell.2020.569219] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
As a classical immune checkpoint molecule, PD-L1 on the surface of tumor cells plays a pivotal role in tumor immunosuppression, primarily by inhibiting the antitumor activities of T cells by binding to its receptor PD-1. PD-1/PD-L1 inhibitors have demonstrated unprecedented promise in treating various human cancers with impressive efficacy. However, a significant portion of cancer patients remains less responsive. Therefore, a better understanding of PD-L1-mediated immune escape is imperative. PD-L1 can be expressed on the surface of tumor cells, but it is also found to exist in extracellular forms, such as on exosomes. Recent studies have revealed the importance of exosomal PD-L1 (ExoPD-L1). As an alternative to membrane-bound PD-L1, ExoPD-L1 produced by tumor cells also plays an important regulatory role in the antitumor immune response. We review the recent remarkable findings on the biological functions of ExoPD-L1, including the inhibition of lymphocyte activities, migration to PD-L1-negative tumor cells and immune cells, induction of both local and systemic immunosuppression, and promotion of tumor growth. We also discuss the potential implications of ExoPD-L1 as a predictor for disease progression and treatment response, sensitive methods for detection of circulating ExoPD-L1, and the novel therapeutic strategies combining the inhibition of exosome biogenesis with PD-L1 blockade in the clinic.
Collapse
Affiliation(s)
- Kaijian Zhou
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shu Guo
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Shu Guo,
| | - Fei Li
- Department of Pharmaceutical Science, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Qiang Sun
- Department of Plastic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Guoxin Liang
- Cancer Therapy Research Institute, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
300
|
Allen J, Najjar K, Erazo-Oliveras A, Kondow-McConaghy HM, Brock DJ, Graham K, Hager EC, Marschall ALJ, Dübel S, Juliano RL, Pellois JP. Cytosolic Delivery of Macromolecules in Live Human Cells Using the Combined Endosomal Escape Activities of a Small Molecule and Cell Penetrating Peptides. ACS Chem Biol 2019; 14:2641-2651. [PMID: 31633910 DOI: 10.1021/acschembio.9b00585] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ineffective cellular delivery is a common problem in numerous biological applications. Developing delivery reagents that work robustly in a variety of experimental settings remains a challenge. Herein, we report how peptides derived from the prototypical cell penetrating peptide TAT can be used in combination with a small molecule, UNC7938, to deliver macromolecules into the cytosol of cells by a simple co-incubation protocol. We establish successful delivery of peptides, DNA plasmids, and a single-chain variable fragment antibody. We also demonstrate that delivery works in hard-to-transfect mammalian cells and under conditions typically inhibitory to cell-penetrating peptides. Mechanistically, UNC7938 destabilizes the membrane of endosomes. This, in turn, enhances the endosome-leakage activity of cell-penetrating peptides and facilitates the endosomal escape of macromolecules initially internalized by mammalian cells via endocytosis. This combined selective membrane-destabilization represents a new chemical space for delivery tools and provides a novel solution to the problem of endosomal entrapment that often limits the effectiveness of reagent-based delivery approaches.
Collapse
Affiliation(s)
- Jason Allen
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Kristina Najjar
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Alfredo Erazo-Oliveras
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Helena M. Kondow-McConaghy
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Dakota J. Brock
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Kristin Graham
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Elizabeth C. Hager
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Andrea L. J. Marschall
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Stefan Dübel
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Rudolph L. Juliano
- UNC Eshelman School of Pharmacy and UNC School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|