1
|
Misaghi E, Kannu P, MacDonald IM, Benson MD. Genetic variants in PIKFYVE: A review of ocular phenotypes. Exp Eye Res 2025; 251:110211. [PMID: 39694407 DOI: 10.1016/j.exer.2024.110211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/25/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Many studies have identified disease-causing variants of PIKFYVE in ocular tissues; however, a comprehensive review of these variants and their ocular phenotypes is lacking. The phosphoinositide kinase PIKFYVE plays crucial roles in the endolysosomal pathway in autophagy and phagocytosis, both essential for cellular homeostasis. In this review, we evaluate the reported disease-causing PIKFYVE variants and their associated phenotypes in humans to identify potential genotype-phenotype correlations. Variants in PIKFYVE have been associated with corneal fleck dystrophy, congenital cataracts and possibly keratoconus. There are unvalidated associations of variants in PIKFYVE with autism spectrum disorder and congenital heart disease. We show that variants causing corneal fleck dystrophy exist in the chaperonin-like domain of PIKFYVE as well as the region between the chaperonin-like and the kinase domains. Similarly, congenital cataract variants appear to be specific to the kinase domain of the protein. This review consolidates existing knowledge on PIKFYVE variants in ocular disease and bridges fundamental science and clinical manifestations, potentially informing future diagnostic and treatment strategies for PIKFYVE-associated ocular disorders.
Collapse
Affiliation(s)
- Ehsan Misaghi
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, Canada; Department of Medical Genetics, University of Alberta, Edmonton, Canada
| | - Peter Kannu
- Department of Medical Genetics, University of Alberta, Edmonton, Canada
| | - Ian M MacDonald
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, Canada; Department of Medical Genetics, University of Alberta, Edmonton, Canada; Department of Ophthalmology, University of Montreal, Montreal, Canada
| | - Matthew D Benson
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, Canada.
| |
Collapse
|
2
|
Uwada J, Nakazawa H, Kiyoi T, Yazawa T, Muramatsu I, Masuoka T. PIKFYVE inhibition induces endosome- and lysosome-derived vacuole enlargement via ammonium accumulation. J Cell Sci 2025; 138:jcs262236. [PMID: 39588583 DOI: 10.1242/jcs.262236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024] Open
Abstract
FYVE-type zinc finger-containing phosphoinositide kinase (PIKFYVE), which is essential for phosphatidylinositol 3,5-bisphosphate [PtdIns(3,5)P2] production, is an important regulator of lysosomal homeostasis. PIKFYVE dysfunction leads to cytoplasmic vacuolization; however, the underlying mechanism remains unknown. In this study, we explored the cause of vacuole enlargement upon PIKFYVE inhibition in DU145 prostate cancer cells. Enlargement of vacuoles upon PIKFYVE inhibition required glutamine and its metabolism by glutaminases. Addition of ammonia, a metabolite of glutamine, was sufficient to enlarge vacuoles via PIKFYVE inhibition. Moreover, PIKFYVE inhibition led to intracellular ammonium accumulation. Endosome-lysosome permeabilization resulted in ammonium leakage from the cells, indicating ammonium accumulation in the endosomes and lysosomes. Ammonium accumulation and vacuole expansion were suppressed by the lysosomal lumen neutralization. It is therefore assumed that PIKFYVE inhibition interferes with the efflux of NH4+, which formed through protonation of NH3 in the lysosomal lumen, leading to osmotic swelling of vacuoles. Notably, glutamine or ammonium is required for PIKFYVE inhibition-induced suppression of lysosomal function and autophagic flux. In conclusion, this study shows that PIKFYVE inhibition disrupts lysosomal homeostasis via ammonium accumulation.
Collapse
Affiliation(s)
- Junsuke Uwada
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Hitomi Nakazawa
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Takeshi Kiyoi
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Ikunobu Muramatsu
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Takayoshi Masuoka
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, Ishikawa 920-0293, Japan
| |
Collapse
|
3
|
de Calbiac H, Renault S, Haouy G, Jung V, Roger K, Zhou Q, Campanari ML, Chentout L, Demy DL, Marian A, Goudin N, Edbauer D, Guerrera C, Ciura S, Kabashi E. Poly-GP accumulation due to C9orf72 loss of function induces motor neuron apoptosis through autophagy and mitophagy defects. Autophagy 2024; 20:2164-2185. [PMID: 39316747 PMCID: PMC11423671 DOI: 10.1080/15548627.2024.2358736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 09/26/2024] Open
Abstract
The GGGGCC hexanucleotide repeat expansion (HRE) of the C9orf72 gene is the most frequent cause of amyotrophic lateral sclerosis (ALS), a devastative neurodegenerative disease characterized by motor neuron degeneration. C9orf72 HRE is associated with lowered levels of C9orf72 expression and its translation results in the production of dipeptide-repeats (DPRs). To recapitulate C9orf72-related ALS disease in vivo, we developed a zebrafish model where we expressed glycine-proline (GP) DPR in a c9orf72 knockdown context. We report that C9orf72 gain- and loss-of-function properties act synergistically to induce motor neuron degeneration and paralysis with poly(GP) accumulating preferentially within motor neurons along with Sqstm1/p62 aggregation indicating macroautophagy/autophagy deficits. Poly(GP) levels were shown to accumulate upon c9orf72 downregulation and were comparable to levels assessed in autopsy samples of patients carrying C9orf72 HRE. Chemical boosting of autophagy using rapamycin or apilimod, is able to rescue motor deficits. Proteomics analysis of zebrafish-purified motor neurons unravels mitochondria dysfunction confirmed through a comparative analysis of previously published C9orf72 iPSC-derived motor neurons. Consistently, 3D-reconstructions of motor neuron demonstrate that poly(GP) aggregates colocalize to mitochondria, thus inducing their elongation and swelling and the failure of their processing by mitophagy, with mitophagy activation through urolithin A preventing locomotor deficits. Finally, we report apoptotic-related increased amounts of cleaved Casp3 (caspase 3, apoptosis-related cysteine peptidase) and rescue of motor neuron degeneration by constitutive inhibition of Casp9 or treatment with decylubiquinone. Here we provide evidence of key pathogenic steps in C9ALS-FTD that can be targeted through pharmacological avenues, thus raising new therapeutic perspectives for ALS patients.
Collapse
Affiliation(s)
- Hortense de Calbiac
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Solène Renault
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Grégoire Haouy
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Vincent Jung
- Proteomics Platform 3P5Necker, INSERM US24/CNRS UMS, Paris Descartes University, Structure Fédérative de Recherche Necker, Paris, France
| | - Kevin Roger
- Proteomics Platform 3P5Necker, INSERM US24/CNRS UMS, Paris Descartes University, Structure Fédérative de Recherche Necker, Paris, France
| | - Qihui Zhou
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Maria-Letizia Campanari
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Loïc Chentout
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Doris Lou Demy
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Anca Marian
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Nicolas Goudin
- Imaging Core Facility, INSERM US24/CNRS UMS3633, Paris, France
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Chiara Guerrera
- Proteomics Platform 3P5Necker, INSERM US24/CNRS UMS, Paris Descartes University, Structure Fédérative de Recherche Necker, Paris, France
| | - Sorana Ciura
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Edor Kabashi
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| |
Collapse
|
4
|
Hou WC, Massey LA, Rhoades D, Wu Y, Ren W, Frank C, Overkleeft HS, Kelly JW. A PIKfyve modulator combined with an integrated stress response inhibitor to treat lysosomal storage diseases. Proc Natl Acad Sci U S A 2024; 121:e2320257121. [PMID: 39150784 PMCID: PMC11348278 DOI: 10.1073/pnas.2320257121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/19/2024] [Indexed: 08/18/2024] Open
Abstract
Lysosomal degradation pathways coordinate the clearance of superfluous and damaged cellular components. Compromised lysosomal degradation is a hallmark of many degenerative diseases, including lysosomal storage diseases (LSDs), which are caused by loss-of-function mutations within both alleles of a lysosomal hydrolase, leading to lysosomal substrate accumulation. Gaucher's disease, characterized by <15% of normal glucocerebrosidase function, is the most common LSD and is a prominent risk factor for developing Parkinson's disease. Here, we show that either of two structurally distinct small molecules that modulate PIKfyve activity, identified in a high-throughput cellular lipid droplet clearance screen, can improve glucocerebrosidase function in Gaucher patient-derived fibroblasts through an MiT/TFE transcription factor that promotes lysosomal gene translation. An integrated stress response (ISR) antagonist used in combination with a PIKfyve modulator further improves cellular glucocerebrosidase activity, likely because ISR signaling appears to also be slightly activated by treatment by either small molecule at the higher doses employed. This strategy of combining a PIKfyve modulator with an ISR inhibitor improves mutant lysosomal hydrolase function in cellular models of additional LSD.
Collapse
Affiliation(s)
- William C. Hou
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92122
| | - Lynée A. Massey
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92122
| | - Derek Rhoades
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92122
| | - Yin Wu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA92122
| | - Wen Ren
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92122
| | - Chiara Frank
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92122
| | - Herman S. Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden2333 CC, The Netherlands
| | - Jeffrey W. Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA92122
| |
Collapse
|
5
|
Roy A, DePamphilis ML. Selective Termination of Autophagy-Dependent Cancers. Cells 2024; 13:1096. [PMID: 38994949 PMCID: PMC11240546 DOI: 10.3390/cells13131096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024] Open
Abstract
The goal of cancer research is to identify characteristics of cancer cells that allow them to be selectively eliminated without harming the host. One such characteristic is autophagy dependence. Cancer cells survive, proliferate, and metastasize under conditions where normal cells do not. Thus, the requirement in cancer cells for more energy and macromolecular biosynthesis can evolve into a dependence on autophagy for recycling cellular components. Recent studies have revealed that autophagy, as well as different forms of cellular trafficking, is regulated by five phosphoinositides associated with eukaryotic cellular membranes and that the enzymes that synthesize them are prime targets for cancer therapy. For example, PIKFYVE inhibitors rapidly disrupt lysosome homeostasis and suppress proliferation in all cells. However, these inhibitors selectively terminate PIKFYVE-dependent cancer cells and cancer stem cells with not having adverse effect on normal cells. Here, we describe the biochemical distinctions between PIKFYVE-sensitive and -insensitive cells, categorize PIKFYVE inhibitors into four groups that differ in chemical structure, target specificity and efficacy on cancer cells and normal cells, identify the mechanisms by which they selectively terminate autophagy-dependent cancer cells, note their paradoxical effects in cancer immunotherapy, and describe their therapeutic applications against cancers.
Collapse
Affiliation(s)
- Ajit Roy
- National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Room 6N105, 10 Center Dr., Bethesda, MD 20892-0001, USA;
| | - Melvin L. DePamphilis
- National Institute of Child Health and Human Development, National Institutes of Health, 9000 Rockville Pike, Room 4B413, 6 Center Dr., Bethesda, MD 20892-2790, USA
| |
Collapse
|
6
|
Elmansy MF, Reidl CT, Rahaman M, Özdinler PH, Silverman RB. Small molecules targeting different cellular pathologies for the treatment of amyotrophic lateral sclerosis. Med Res Rev 2023; 43:2260-2302. [PMID: 37243319 PMCID: PMC10592673 DOI: 10.1002/med.21974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 02/28/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease in which the motor neuron circuitry displays progressive degeneration, affecting mostly the motor neurons in the brain and in the spinal cord. There are no effective cures, albeit three drugs, riluzole, edaravone, and AMX0035 (a combination of sodium phenylbutyrate and taurursodiol), have been approved by the Food and Drug Administration, with limited improvement in patients. There is an urgent need to build better and more effective treatment strategies for ALS. Since the disease is very heterogenous, numerous approaches have been explored, such as targeting genetic mutations, decreasing oxidative stress and excitotoxicity, enhancing mitochondrial function and protein degradation mechanisms, and inhibiting neuroinflammation. In addition, various chemical libraries or previously identified drugs have been screened for potential repurposing in the treatment of ALS. Here, we review previous drug discovery efforts targeting a variety of cellular pathologies that occur from genetic mutations that cause ALS, such as mutations in SOD1, C9orf72, FUS, and TARDP-43 genes. These mutations result in protein aggregation, which causes neuronal degeneration. Compounds used to target cellular pathologies that stem from these mutations are discussed and comparisons among different preclinical models are presented. Because the drug discovery landscape for ALS and other motor neuron diseases is changing rapidly, we also offer recommendations for a novel, more effective, direction in ALS drug discovery that could accelerate translation of effective compounds from animals to patients.
Collapse
Affiliation(s)
- Mohamed F. Elmansy
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois, USA
- Department of Organometallic and Organometalloid Chemistry, National Research Centre, Cairo, Egypt
| | - Cory T. Reidl
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois, USA
| | - Mizzanoor Rahaman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois, USA
| | - P. Hande Özdinler
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Richard B. Silverman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
7
|
Li C, Qiao Y, Jiang X, Liu L, Zheng Y, Qiu Y, Cheng C, Zhou F, Zhou Y, Huang W, Ren X, Wang Y, Wang Z, Chinnaiyan AM, Ding K. Discovery of a First-in-Class Degrader for the Lipid Kinase PIKfyve. J Med Chem 2023; 66:12432-12445. [PMID: 37605297 PMCID: PMC10510382 DOI: 10.1021/acs.jmedchem.3c00912] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Indexed: 08/23/2023]
Abstract
The phosphoinositide kinase PIKfyve has emerged as a new potential therapeutic target in various cancers. However, limited clinical progress has been achieved with PIKfyve inhibitors. Here, we report the discovery of a first-in-class PIKfyve degrader 12d (PIK5-12d) by employing the proteolysis-targeting chimera approach. PIK5-12d potently degraded PIKfyve protein with a DC50 value of 1.48 nM and a Dmax value of 97.7% in prostate cancer VCaP cells. Mechanistic studies revealed that it selectively induced PIKfyve degradation in a VHL- and proteasome-dependent manner. PIKfyve degradation by PIK5-12d caused massive cytoplasmic vacuolization and blocked autophagic flux in multiple prostate cancer cell lines. Importantly, PIK5-12d was more effective in suppressing the growth of prostate cancer cells than the parent inhibitor and exerted prolonged inhibition of downstream signaling. Further, intraperitoneal administration of PIK5-12d exhibited potent PIKfyve degradation and suppressed tumor proliferation in vivo. Overall, PIK5-12d is a valuable chemical tool for exploring PIKfyve-based targeted therapy.
Collapse
Affiliation(s)
- Chungen Li
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, #345 Lingling Roadd, Shanghai 200032, People’s Republic of China
| | - Yuanyuan Qiao
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Xia Jiang
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lianchao Liu
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, #345 Lingling Roadd, Shanghai 200032, People’s Republic of China
| | - Yang Zheng
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yudi Qiu
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, #345 Lingling Roadd, Shanghai 200032, People’s Republic of China
| | - Caleb Cheng
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Fengtao Zhou
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
and Innovative Drug Discovery of Chinese Ministry of Education (MOE),
Guangzhou City Key Laboratory of Precision Chemical Drug Development,
College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, People’s Republic of China
| | - Yang Zhou
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
and Innovative Drug Discovery of Chinese Ministry of Education (MOE),
Guangzhou City Key Laboratory of Precision Chemical Drug Development,
College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, People’s Republic of China
| | - Weixue Huang
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, #345 Lingling Roadd, Shanghai 200032, People’s Republic of China
| | - Xiaomei Ren
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, #345 Lingling Roadd, Shanghai 200032, People’s Republic of China
| | - Yuzhuo Wang
- The
Vancouver Prostate Centre, Vancouver General Hospital and Department
of Urologic Sciences, The University of
British Columbia, Vancouver, British Columbia V6H 3Z6, Canada
| | - Zhen Wang
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, #345 Lingling Roadd, Shanghai 200032, People’s Republic of China
| | - Arul M. Chinnaiyan
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Howard
Hughes Medical Institute, University of
Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Urology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ke Ding
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, #345 Lingling Roadd, Shanghai 200032, People’s Republic of China
- Institute
of Basic Medicine and Cancer (IBMC), Chinese
Academy of Sciences, Hangzhou, Zhejiang 310022, People’s Republic of China
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
and Innovative Drug Discovery of Chinese Ministry of Education (MOE),
Guangzhou City Key Laboratory of Precision Chemical Drug Development,
College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, People’s Republic of China
| |
Collapse
|
8
|
Wang H, Zhu Y, Liu H, Liang T, Wei Y. Advances in Drug Discovery Targeting Lysosomal Membrane Proteins. Pharmaceuticals (Basel) 2023; 16:ph16040601. [PMID: 37111358 PMCID: PMC10145713 DOI: 10.3390/ph16040601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 04/29/2023] Open
Abstract
Lysosomes are essential organelles of eukaryotic cells and are responsible for various cellular functions, including endocytic degradation, extracellular secretion, and signal transduction. There are dozens of proteins localized to the lysosomal membrane that control the transport of ions and substances across the membrane and are integral to lysosomal function. Mutations or aberrant expression of these proteins trigger a variety of disorders, making them attractive targets for drug development for lysosomal disorder-related diseases. However, breakthroughs in R&D still await a deeper understanding of the underlying mechanisms and processes of how abnormalities in these membrane proteins induce related diseases. In this article, we summarize the current progress, challenges, and prospects for developing therapeutics targeting lysosomal membrane proteins for the treatment of lysosomal-associated diseases.
Collapse
Affiliation(s)
- Hongna Wang
- Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangzhou 510095, China
- Key Laboratory for Cell Homeostasis, Cancer Research of Guangdong Higher Education Institutes, Guangzhou 510095, China
| | - Yidong Zhu
- Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangzhou 510095, China
- Key Laboratory for Cell Homeostasis, Cancer Research of Guangdong Higher Education Institutes, Guangzhou 510095, China
| | - Huiyan Liu
- Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangzhou 510095, China
- Key Laboratory for Cell Homeostasis, Cancer Research of Guangdong Higher Education Institutes, Guangzhou 510095, China
| | - Tianxiang Liang
- Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangzhou 510095, China
- Key Laboratory for Cell Homeostasis, Cancer Research of Guangdong Higher Education Institutes, Guangzhou 510095, China
| | - Yongjie Wei
- Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangzhou 510095, China
- Key Laboratory for Cell Homeostasis, Cancer Research of Guangdong Higher Education Institutes, Guangzhou 510095, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou 510095, China
| |
Collapse
|
9
|
Wang H, Zhang J, Liu H, Wang M, Dong Y, Zhou Y, Wong SM, Xu K, Xu Q. A plant virus hijacks phosphatidylinositol-3,5-bisphosphate to escape autophagic degradation in its insect vector. Autophagy 2023; 19:1128-1143. [PMID: 36093594 PMCID: PMC10012956 DOI: 10.1080/15548627.2022.2116676] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 06/29/2022] [Accepted: 08/20/2022] [Indexed: 02/07/2023] Open
Abstract
Hosts can initiate macroautophagy/autophagy as an antiviral defense response, while viruses have developed multiple ways to evade the host autophagic degradation. However, little is known as to whether viruses can target lipids to subvert autophagic degradation. Here, we show that a low abundant signaling lipid, phosphatidylinositol 3,5-bisphosphate (PtdIns(3,5)P2), is required for rice black-streaked dwarf virus (RBSDV) to evade the autophagic degradation in the insect vector Laodelphax striatellus. RBSDV binds to PtdIns(3,5)P2 and elevates its level through its main capsid protein P10, leading to inhibited autophagy and promoted virus propagation. Furthermore, we show that PtdIns(3,5)P2 inhibits the autophagy pathway by preventing the fusion of autophagosomes and lysosomes through activation of Trpml (transient receptor potential cation channel, mucolipin), an effector of PtdIns(3,5)P2. These findings uncover a strategy whereby a plant virus hijacks PtdIns(3,5)P2 via its viral capsid protein to evade autophagic degradation and promote its survival in insects.
Collapse
Affiliation(s)
- Haitao Wang
- Institute of Plant Protection, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Jianhua Zhang
- Institute of Plant Protection, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Institute of Industrial Crops, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Haoqiu Liu
- Institute of Plant Protection, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- National University of Singapore Research Institute, Suzhou, China
| | - Man Wang
- Institute of Plant Protection, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Yan Dong
- Institute of Plant Protection, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Yijun Zhou
- Institute of Plant Protection, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Sek-Man Wong
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- National University of Singapore Research Institute, Suzhou, China
| | - Kai Xu
- Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Qiufang Xu
- Institute of Plant Protection, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- College of Life Sciences, Anhui Normal University, Wuhu, China
| |
Collapse
|
10
|
Somogyi A, Kirkham ED, Lloyd-Evans E, Winston J, Allen ND, Mackrill JJ, Anderson KE, Hawkins PT, Gardiner SE, Waller-Evans H, Sims R, Boland B, O'Neill C. The synthetic TRPML1 agonist ML-SA1 rescues Alzheimer-related alterations of the endosomal-autophagic-lysosomal system. J Cell Sci 2023; 136:jcs259875. [PMID: 36825945 PMCID: PMC10112969 DOI: 10.1242/jcs.259875] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Abnormalities in the endosomal-autophagic-lysosomal (EAL) system are an early event in Alzheimer's disease (AD) pathogenesis. However, the mechanisms underlying these abnormalities are unclear. The transient receptor potential channel mucolipin 1(TRPML1, also known as MCOLN1), a vital endosomal-lysosomal Ca2+ channel whose loss of function leads to neurodegeneration, has not been investigated with respect to EAL pathogenesis in late-onset AD (LOAD). Here, we identify pathological hallmarks of TRPML1 dysregulation in LOAD neurons, including increased perinuclear clustering and vacuolation of endolysosomes. We reveal that induced pluripotent stem cell (iPSC)-derived human cortical neurons expressing APOE ε4, the strongest genetic risk factor for LOAD, have significantly diminished TRPML1-induced endolysosomal Ca2+ release. Furthermore, we found that blocking TRPML1 function in primary neurons by depleting the TRPML1 agonist PI(3,5)P2 via PIKfyve inhibition, recreated multiple features of EAL neuropathology evident in LOAD. This included increased endolysosomal Ca2+ content, enlargement and perinuclear clustering of endolysosomes, autophagic vesicle accumulation and early endosomal enlargement. Strikingly, these AD-like neuronal EAL defects were rescued by TRPML1 reactivation using its synthetic agonist ML-SA1. These findings implicate defects in TRPML1 in LOAD EAL pathogenesis and present TRPML1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Aleksandra Somogyi
- School of Biochemistry and Cell Biology, BioSciences Institute, University College Cork, T12 YT20 Cork, Ireland
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
- Cork Neuroscience Centre (CNSC), University College Cork, T12 YT20 Cork, Ireland
| | - Emily D Kirkham
- School of Biosciences, Sir Martin Evans building, Cardiff University, CF10 3AX Cardiff, UK
| | - Emyr Lloyd-Evans
- School of Biosciences, Sir Martin Evans building, Cardiff University, CF10 3AX Cardiff, UK
| | - Jincy Winston
- UK Dementia Research Institute, Hadyn Ellis Building, Cardiff University, CF24 4HQ Cardiff, UK
| | - Nicholas D Allen
- School of Biosciences, Sir Martin Evans building, Cardiff University, CF10 3AX Cardiff, UK
| | - John J Mackrill
- Department of Physiology, School of Medicine, University College Cork, T12 YT20 Cork, Ireland
| | - Karen E Anderson
- The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - Phillip T Hawkins
- The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - Sian E Gardiner
- Medicines Discovery Institute, Main Building, Cardiff University, CF10 3AT Cardiff, UK
| | - Helen Waller-Evans
- Medicines Discovery Institute, Main Building, Cardiff University, CF10 3AT Cardiff, UK
| | - Rebecca Sims
- Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, C14 4XN Cardiff, UK
| | - Barry Boland
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
- Cork Neuroscience Centre (CNSC), University College Cork, T12 YT20 Cork, Ireland
| | - Cora O'Neill
- School of Biochemistry and Cell Biology, BioSciences Institute, University College Cork, T12 YT20 Cork, Ireland
- Cork Neuroscience Centre (CNSC), University College Cork, T12 YT20 Cork, Ireland
| |
Collapse
|
11
|
Lu W, Liu Y, Gao Y, Geng Q, Gurbani D, Li L, Ficarro SB, Meyer CJ, Sinha D, You I, Tse J, He Z, Ji W, Che J, Kim AY, Yu T, Wen K, Anderson KC, Marto JA, Westover KD, Zhang T, Gray NS. Development of a Covalent Inhibitor of c-Jun N-Terminal Protein Kinase (JNK) 2/3 with Selectivity over JNK1. J Med Chem 2023; 66:3356-3371. [PMID: 36826833 PMCID: PMC11190964 DOI: 10.1021/acs.jmedchem.2c01834] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
The c-Jun N-terminal kinases (JNKs) are members of the mitogen-activated protein kinase (MAPK) family, which includes JNK1-JNK3. Interestingly, JNK1 and JNK2 show opposing functions, with JNK2 activity favoring cell survival and JNK1 stimulating apoptosis. Isoform-selective small molecule inhibitors of JNK1 or JNK2 would be useful as pharmacological probes but have been difficult to develop due to the similarity of their ATP binding pockets. Here, we describe the discovery of a covalent inhibitor YL5084, the first such inhibitor that displays selectivity for JNK2 over JNK1. We demonstrated that YL5084 forms a covalent bond with Cys116 of JNK2, exhibits a 20-fold higher Kinact/KI compared to that of JNK1, and engages JNK2 in cells. However, YL5084 exhibited JNK2-independent antiproliferative effects in multiple myeloma cells, suggesting the existence of additional targets relevant in this context. Thus, although not fully optimized, YL5084 represents a useful chemical starting point for the future development of JNK2-selective chemical probes.
Collapse
Affiliation(s)
- Wenchao Lu
- Department of Chemical and Systems Biology, Chem-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, United States
- Lingang Laboratory, Shanghai 200031, China
| | - Yao Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Yang Gao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Qixiang Geng
- Department of Chemical and Systems Biology, Chem-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Deepak Gurbani
- Department of Radiation Oncology, Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Lianbo Li
- Department of Radiation Oncology, Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Scott B Ficarro
- Department of Cancer Biology, Blais Proteomics Center, Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Cynthia J Meyer
- Department of Radiation Oncology, Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Dhiraj Sinha
- Department of Radiation Oncology, Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Inchul You
- Department of Chemical and Systems Biology, Chem-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Jason Tse
- Department of Chemical and Systems Biology, Chem-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Zhixiang He
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Wenzhi Ji
- Department of Chemical and Systems Biology, Chem-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Jianwei Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Audrey Y Kim
- Department of Chemical and Systems Biology, Chem-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Tengteng Yu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Kenneth Wen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Kenneth C Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Jarrod A Marto
- Department of Cancer Biology, Blais Proteomics Center, Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Kenneth D Westover
- Department of Radiation Oncology, Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Tinghu Zhang
- Department of Chemical and Systems Biology, Chem-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, Chem-H, and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
12
|
Chen X, Ma L, Zhao J, Pan X, Chen S. Effect of empagliflozin on cytoskeletal repair in the hippocampus of obese mice. Front Neurosci 2022; 16:1000839. [PMID: 36408417 PMCID: PMC9667058 DOI: 10.3389/fnins.2022.1000839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/22/2022] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE We aimed to investigate the effect of empagliflozin on hippocampal phosphorylated protein levels in obese mice. MATERIALS AND METHODS Sixteen obese mice successfully modeled on high-fat diet were randomly divided into high-fat feeding group (group H) and empagliflozin group (group H + empagliflozin, group E), eight mice in each group, and eight C57BL/6J male normal mice were selected as the control group (normal control, group C). Group E was treated with empagliflozin 10 mg/kg/d for 12 weeks, while mice in groups H and C were treated with equal amounts of saline. The spatial learning memory ability of the mice was determined by the Morris water maze experiment. Further, their body weights and serological indices were measured. Finally, total proteins were extracted from hippocampal tissues for functional analysis by the phosphorylated proteomics method. RESULTS The results showed that escape latency was prolonged, retention time in the target quadrant was shortened, and the number of loop penetrations was reduced in the obese mice induced by a high-calorie diet compared with normal controls, whereas escape latency was shortened, retention time in the target quadrant was increased, and the number of loop penetrations was increased after empagliflozin treatment. Phosphoproteomics in the high-fat/control (H/C), empagliflozin/high-fat (E/H), and E/C groups showed 844, 1,552, and 1,512 differentially significant phosphorylation sites, respectively. The proteins corresponding to these differentially phosphorylated sites were mainly involved in neurodegenerative pathways and actin cytoskeleton regulation. Notably, myosin heavy chain 10 (MYH10), p21 protein-activated kinase 4 (PAK4), phosphatidylinositol 3 -phosphate 5-kinase (PIKfyve), and other differentially phosphorylated proteins were involved in actin cytoskeleton regulation. CONCLUSION We concluded that empagliflozin protects cognitive functions by inducing serine phosphorylation in MYH10, PAK4, and PIKfyve in the hippocampal tissue of obese mice.
Collapse
Affiliation(s)
- Xiaoyi Chen
- Graduate School of Hebei North University, Zhangjiakou, China,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Liang Ma
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Jingyu Zhao
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China,Graduate School of North China University of Science and Technology, Tangshan, China
| | - Xiaoyu Pan
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Shuchun Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China,*Correspondence: Shuchun Chen,
| |
Collapse
|
13
|
Drewry D, Potjewyd FM, Bayati A, Smith JL, Dickmander RJ, Howell S, Taft-Benz S, Min SM, Hossain MA, Heise M, McPherson PS, Moorman NJ, Axtman AD. Identification and Utilization of a Chemical Probe to Interrogate the Roles of PIKfyve in the Lifecycle of β-Coronaviruses. J Med Chem 2022; 65:12860-12882. [PMID: 36111834 PMCID: PMC9574855 DOI: 10.1021/acs.jmedchem.2c00697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Indexed: 11/29/2022]
Abstract
From a designed library of indolyl pyrimidinamines, we identified a highly potent and cell-active chemical probe (17) that inhibits phosphatidylinositol-3-phosphate 5-kinase (PIKfyve). Comprehensive evaluation of inhibitor selectivity confirmed that this PIKfyve probe demonstrates excellent kinome-wide selectivity. A structurally related indolyl pyrimidinamine (30) was characterized as a negative control that lacks PIKfyve inhibitory activity and exhibits exquisite selectivity when profiled broadly. Chemical probe 17 disrupts multiple phases of the lifecycle of β-coronaviruses: viral replication and viral entry. The diverse antiviral roles of PIKfyve have not been previously probed comprehensively in a single study or using the same compound set. Our scaffold is a distinct chemotype that lacks the canonical morpholine hinge-binder of classical lipid kinase inhibitors and has a non-overlapping kinase off-target profile with known PIKfyve inhibitors. Our chemical probe set can be used by the community to further characterize the role of PIKfyve in virology.
Collapse
Affiliation(s)
- David
H. Drewry
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- UNC
Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Frances M. Potjewyd
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Armin Bayati
- Structural
Genomics Consortium, Department of Neurology and Neurosurgery, Montreal
Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Jeffery L. Smith
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rebekah J. Dickmander
- UNC
Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Rapidly
Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department
of Microbiology & Immunology, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department
of Chemistry, University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Stefanie Howell
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Sharon Taft-Benz
- Rapidly
Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department
of Genetics, University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Sophia M. Min
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mohammad Anwar Hossain
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mark Heise
- Rapidly
Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department
of Genetics, University of North Carolina
at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Peter S. McPherson
- Structural
Genomics Consortium, Department of Neurology and Neurosurgery, Montreal
Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Nathaniel J. Moorman
- UNC
Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Rapidly
Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department
of Microbiology & Immunology, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Alison D. Axtman
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
14
|
Rodgers SJ, Jones EI, Arumugam S, Hamila SA, Danne J, Gurung R, Eramo MJ, Nanayakkara R, Ramm G, McGrath MJ, Mitchell CA. Endosome maturation links PI3Kα signaling to lysosome repopulation during basal autophagy. EMBO J 2022; 41:e110398. [PMID: 35968799 PMCID: PMC9531306 DOI: 10.15252/embj.2021110398] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022] Open
Abstract
Autophagy depends on the repopulation of lysosomes to degrade intracellular components and recycle nutrients. How cells co‐ordinate lysosome repopulation during basal autophagy, which occurs constitutively under nutrient‐rich conditions, is unknown. Here, we identify an endosome‐dependent phosphoinositide pathway that links PI3Kα signaling to lysosome repopulation during basal autophagy. We show that PI3Kα‐derived PI(3)P generated by INPP4B on late endosomes was required for basal but not starvation‐induced autophagic degradation. PI(3)P signals were maintained as late endosomes matured into endolysosomes, and served as the substrate for the 5‐kinase, PIKfyve, to generate PI(3,5)P2. The SNX‐BAR protein, SNX2, was recruited to endolysosomes by PI(3,5)P2 and promoted lysosome reformation. Inhibition of INPP4B/PIKfyve‐dependent lysosome reformation reduced autophagic clearance of protein aggregates during proteotoxic stress leading to increased cytotoxicity. Therefore under nutrient‐rich conditions, PI3Kα, INPP4B, and PIKfyve sequentially contribute to basal autophagic degradation and protection from proteotoxic stress via PI(3,5)P2‐dependent lysosome reformation from endolysosomes. These findings reveal that endosome maturation couples PI3Kα signaling to lysosome reformation during basal autophagy.
Collapse
Affiliation(s)
- Samuel J Rodgers
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Emily I Jones
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Senthil Arumugam
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,European Molecular Biological Laboratory Australia, Monash University, Clayton, VIC, Australia
| | - Sabryn A Hamila
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jill Danne
- Monash Ramaciotti Centre for Cryo Electron Microscopy, A Node of Microscopy Australia, Monash University, Clayton, VIC, Australia
| | - Rajendra Gurung
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Matthew J Eramo
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Randini Nanayakkara
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Monash Ramaciotti Centre for Cryo Electron Microscopy, A Node of Microscopy Australia, Monash University, Clayton, VIC, Australia
| | - Georg Ramm
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Monash Ramaciotti Centre for Cryo Electron Microscopy, A Node of Microscopy Australia, Monash University, Clayton, VIC, Australia
| | - Meagan J McGrath
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Christina A Mitchell
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
15
|
Caux M, Mansour R, Xuereb JM, Chicanne G, Viaud J, Vauclard A, Boal F, Payrastre B, Tronchère H, Severin S. PIKfyve-Dependent Phosphoinositide Dynamics in Megakaryocyte/Platelet Granule Integrity and Platelet Functions. Arterioscler Thromb Vasc Biol 2022; 42:987-1004. [PMID: 35708031 DOI: 10.1161/atvbaha.122.317559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Secretory granules are key elements for platelet functions. Their biogenesis and integrity are regulated by fine-tuned mechanisms that need to be fully characterized. Here, we investigated the role of the phosphoinositide 5-kinase PIKfyve and its lipid products, PtdIns5P (phosphatidylinositol 5 monophosphate) and PtdIns(3,5)P2 (phosphatidylinositol (3,5) bisphosphate) in granule homeostasis in megakaryocytes and platelets. METHODS For that, we invalidated PIKfyve by pharmacological inhibition or gene silencing in megakaryocytic cell models (human MEG-01 cell line, human imMKCLs, mouse primary megakaryocytes) and in human platelets. RESULTS We unveiled that PIKfyve expression and its lipid product levels increased with megakaryocytic maturation. In megakaryocytes, PtdIns5P and PtdIns(3,5)P2 were found in alpha and dense granule membranes with higher levels in dense granules. Pharmacological inhibition or knock-down of PIKfyve in megakaryocytes decreased PtdIns5P and PtdIns(3,5)P2 synthesis and induced a vacuolar phenotype with a loss of alpha and dense granule identity. Permeant PtdIns5P and PtdIns(3,5)P2 and the cation channel TRPML1 (transient receptor potential mucolipins) and TPC2 activation were able to accelerate alpha and dense granule integrity recovery following release of PIKfyve pharmacological inhibition. In platelets, PIKfyve inhibition specifically impaired the integrity of dense granules culminating in defects in their secretion, platelet aggregation, and thrombus formation. CONCLUSIONS These data demonstrated that PIKfyve and its lipid products PtdIns5P and PtdIns(3,5)P2 control granule integrity both in megakaryocytes and platelets.
Collapse
Affiliation(s)
- Manuella Caux
- INSERM U1297, I2MC and Université Paul Sabatier, Toulouse, France (M.C., R.M., J.-M.X., G.C., J.V., A.V., F.B., B.P., H.T., S.S.)
| | - Rana Mansour
- INSERM U1297, I2MC and Université Paul Sabatier, Toulouse, France (M.C., R.M., J.-M.X., G.C., J.V., A.V., F.B., B.P., H.T., S.S.)
| | - Jean-Marie Xuereb
- INSERM U1297, I2MC and Université Paul Sabatier, Toulouse, France (M.C., R.M., J.-M.X., G.C., J.V., A.V., F.B., B.P., H.T., S.S.)
| | - Gaëtan Chicanne
- INSERM U1297, I2MC and Université Paul Sabatier, Toulouse, France (M.C., R.M., J.-M.X., G.C., J.V., A.V., F.B., B.P., H.T., S.S.)
| | - Julien Viaud
- INSERM U1297, I2MC and Université Paul Sabatier, Toulouse, France (M.C., R.M., J.-M.X., G.C., J.V., A.V., F.B., B.P., H.T., S.S.)
| | - Alicia Vauclard
- INSERM U1297, I2MC and Université Paul Sabatier, Toulouse, France (M.C., R.M., J.-M.X., G.C., J.V., A.V., F.B., B.P., H.T., S.S.)
| | - Frédéric Boal
- INSERM U1297, I2MC and Université Paul Sabatier, Toulouse, France (M.C., R.M., J.-M.X., G.C., J.V., A.V., F.B., B.P., H.T., S.S.)
| | - Bernard Payrastre
- INSERM U1297, I2MC and Université Paul Sabatier, Toulouse, France (M.C., R.M., J.-M.X., G.C., J.V., A.V., F.B., B.P., H.T., S.S.).,CHU de Toulouse, Laboratoire d'Hématologie, Toulouse, France (B.P.)
| | - Hélène Tronchère
- INSERM U1297, I2MC and Université Paul Sabatier, Toulouse, France (M.C., R.M., J.-M.X., G.C., J.V., A.V., F.B., B.P., H.T., S.S.)
| | - Sonia Severin
- INSERM U1297, I2MC and Université Paul Sabatier, Toulouse, France (M.C., R.M., J.-M.X., G.C., J.V., A.V., F.B., B.P., H.T., S.S.)
| |
Collapse
|
16
|
Jain V, Bose S, Arya AK, Arif T. Lysosomes in Stem Cell Quiescence: A Potential Therapeutic Target in Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:1618. [PMID: 35406389 PMCID: PMC8996909 DOI: 10.3390/cancers14071618] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Lysosomes are cellular organelles that regulate essential biological processes such as cellular homeostasis, development, and aging. They are primarily connected to the degradation/recycling of cellular macromolecules and participate in cellular trafficking, nutritional signaling, energy metabolism, and immune regulation. Therefore, lysosomes connect cellular metabolism and signaling pathways. Lysosome's involvement in the critical biological processes has rekindled clinical interest towards this organelle for treating various diseases, including cancer. Recent research advancements have demonstrated that lysosomes also regulate the maintenance and hemostasis of hematopoietic stem cells (HSCs), which play a critical role in the progression of acute myeloid leukemia (AML) and other types of cancer. Lysosomes regulate both HSCs' metabolic networks and identity transition. AML is a lethal type of blood cancer with a poor prognosis that is particularly associated with aging. Although the genetic landscape of AML has been extensively described, only a few targeted therapies have been produced, warranting the need for further research. This review summarizes the functions and importance of targeting lysosomes in AML, while highlighting the significance of lysosomes in HSCs maintenance.
Collapse
Affiliation(s)
- Vaibhav Jain
- Abramson Cancer Center, Department of Medicine, 421 Curie Blvd., Philadelphia, PA 19104, USA;
| | - Swaroop Bose
- Department of Dermatology, Mount Sinai Icahn School of Medicine, New York, NY 10029, USA;
| | - Awadhesh K. Arya
- Department of Anesthesiology, Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Tasleem Arif
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai Icahn School of Medicine, New York, NY 10029, USA
| |
Collapse
|
17
|
Saffi GT, Tang E, Mamand S, Inpanathan S, Fountain A, Salmena L, Botelho RJ. Reactive oxygen species prevent lysosome coalescence during PIKfyve inhibition. PLoS One 2021; 16:e0259313. [PMID: 34813622 PMCID: PMC8610251 DOI: 10.1371/journal.pone.0259313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/16/2021] [Indexed: 11/19/2022] Open
Abstract
Lysosomes are terminal, degradative organelles of the endosomal pathway that undergo repeated fusion-fission cycles with themselves, endosomes, phagosomes, and autophagosomes. Lysosome number and size depends on balanced fusion and fission rates. Thus, conditions that favour fusion over fission can reduce lysosome numbers while enlarging their size. Conversely, favouring fission over fusion may cause lysosome fragmentation and increase their numbers. PIKfyve is a phosphoinositide kinase that generates phosphatidylinositol-3,5-bisphosphate to modulate lysosomal functions. PIKfyve inhibition causes an increase in lysosome size and reduction in lysosome number, consistent with lysosome coalescence. This is thought to proceed through reduced lysosome reformation and/or fission after fusion with endosomes or other lysosomes. Previously, we observed that photo-damage during live-cell imaging prevented lysosome coalescence during PIKfyve inhibition. Thus, we postulated that lysosome fusion and/or fission dynamics are affected by reactive oxygen species (ROS). Here, we show that ROS generated by various independent mechanisms all impaired lysosome coalescence during PIKfyve inhibition and promoted lysosome fragmentation during PIKfyve re-activation. However, depending on the ROS species or mode of production, lysosome dynamics were affected distinctly. H2O2 impaired lysosome motility and reduced lysosome fusion with phagosomes, suggesting that H2O2 reduces lysosome fusogenecity. In comparison, inhibitors of oxidative phosphorylation, thiol groups, glutathione, or thioredoxin, did not impair lysosome motility but instead promoted clearance of actin puncta on lysosomes formed during PIKfyve inhibition. Additionally, actin depolymerizing agents prevented lysosome coalescence during PIKfyve inhibition. Thus, we discovered that ROS can generally prevent lysosome coalescence during PIKfyve inhibition using distinct mechanisms depending on the type of ROS.
Collapse
Affiliation(s)
- Golam T. Saffi
- Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Evan Tang
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Sami Mamand
- Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- Polytechnic Research Center, Erbil Polytechnic University, Kurdistan Regional Government, Erbil, Kurdistan
| | - Subothan Inpanathan
- Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Aaron Fountain
- Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Leonardo Salmena
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Roberto J. Botelho
- Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Qiu S, Lavallée-Adam M, Côté M. Proximity Interactome Map of the Vac14-Fig4 Complex Using BioID. J Proteome Res 2021; 20:4959-4973. [PMID: 34554760 DOI: 10.1021/acs.jproteome.1c00408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Conversion between phosphatidylinositol-3-phosphate and phosphatidylinositol-3,5-bisphosphate on endosomal membranes is critical for the maturation of early endosomes to late endosomes/lysosomes and is regulated by the PIKfyve-Vac14-Fig4 complex. Despite the importance of this complex for endosomal homeostasis and vesicular trafficking, there is little known about how its activity is regulated or how it interacts with other cellular proteins. Here, we screened for the cellular interactome of Vac14 and Fig4 using proximity-dependent biotin labeling (BioID). After independently screening the interactomes of Vac14 and Fig4, we identified 89 high-confidence protein hits shared by both proteins. Network analysis of these hits revealed pathways with known involvement of the PIKfyve-Vac14-Fig4 complex, including vesicular organization and PI3K/Akt signaling, as well as novel pathways including cell cycle and mitochondrial regulation. We also identified subunits of coatomer complex I (COPI), a Golgi-associated complex with an emerging role in endosomal dynamics. Using proximity ligation assays, we validated the interaction between Vac14 and COPI subunit COPB1 and between Vac14 and Arf1, a GTPase required for COPI assembly. In summary, this study used BioID to comprehensively map the Vac14-Fig4 interactome, revealing potential roles for these proteins in diverse cellular processes and pathways, including preliminary evidence of an interaction between Vac14 and COPI. Data are available via ProteomeXchange with the identifier PXD027917.
Collapse
Affiliation(s)
- Shirley Qiu
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada.,Ottawa Institute of Systems Biology, University of Ottawa, Ottawa K1H 8M5, Canada.,Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Mathieu Lavallée-Adam
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada.,Ottawa Institute of Systems Biology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada.,Ottawa Institute of Systems Biology, University of Ottawa, Ottawa K1H 8M5, Canada.,Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa K1H 8M5, Canada
| |
Collapse
|
19
|
Welcome MO, Mastorakis NE. Neuropathophysiology of coronavirus disease 2019: neuroinflammation and blood brain barrier disruption are critical pathophysiological processes that contribute to the clinical symptoms of SARS-CoV-2 infection. Inflammopharmacology 2021; 29:939-963. [PMID: 33822324 PMCID: PMC8021940 DOI: 10.1007/s10787-021-00806-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 03/22/2021] [Indexed: 12/17/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the novel SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) first discovered in Wuhan, Hubei province, China in December 2019. SARS-CoV-2 has infected several millions of people, resulting in a huge socioeconomic cost and over 2.5 million deaths worldwide. Though the pathogenesis of COVID-19 is not fully understood, data have consistently shown that SARS-CoV-2 mainly affects the respiratory and gastrointestinal tracts. Nevertheless, accumulating evidence has implicated the central nervous system in the pathogenesis of SARS-CoV-2 infection. Unfortunately, however, the mechanisms of SARS-CoV-2 induced impairment of the central nervous system are not completely known. Here, we review the literature on possible neuropathogenic mechanisms of SARS-CoV-2 induced cerebral damage. The results suggest that downregulation of angiotensin converting enzyme 2 (ACE2) with increased activity of the transmembrane protease serine 2 (TMPRSS2) and cathepsin L in SARS-CoV-2 neuroinvasion may result in upregulation of proinflammatory mediators and reactive species that trigger neuroinflammatory response and blood brain barrier disruption. Furthermore, dysregulation of hormone and neurotransmitter signalling may constitute a fundamental mechanism involved in the neuropathogenic sequelae of SARS-CoV-2 infection. The viral RNA or antigenic peptides also activate or interact with molecular signalling pathways mediated by pattern recognition receptors (e.g., toll-like receptors), nuclear factor kappa B, Janus kinase/signal transducer and activator of transcription, complement cascades, and cell suicide molecules. Potential molecular targets and therapeutics of SARS-CoV-2 induced neurologic damage are also discussed.
Collapse
Affiliation(s)
- Menizibeya O Welcome
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, Plot 681 Cadastral Zone, C-00 Research and Institution Area, Jabi Airport Road Bypass, FCT, Abuja, Nigeria.
| | - Nikos E Mastorakis
- Technical University of Sofia, Klement Ohridksi 8, 1000, Sofia, Bulgaria
| |
Collapse
|
20
|
Huang P, Wu SP, Wang N, Seto S, Chang D. Hydroxysafflor yellow A alleviates cerebral ischemia reperfusion injury by suppressing apoptosis via mitochondrial permeability transition pore. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 85:153532. [PMID: 33735723 DOI: 10.1016/j.phymed.2021.153532] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/15/2021] [Accepted: 02/25/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Mitochondria are key cellular organelles that are essential for cell fate decisions. Hydroxysafflor yellow A (HSYA) has displayed an impressively essential role in protection of cerebral ischemia/reperfusion (I/R). However, the mitochondrial effect of HSYA on Brain Microvascular Endothelial Cells (BMECs) under I/R remains to be largely unclear. PURPOSE To evaluate the protective effects of HSYA-mediated mitochondrial permeability transition pore (mPTP) on cerebral I/R injury and its mechanism. METHODS Cerebral I/R injury was established by the model of Middle cerebral artery occlusion (MCAO) in rats. Furthermore, to further clarify the relevant mechanism of HSYA's effects on mPTP, inhibition of extracellular regulated protein kinases (ERK) with U0126 and transfect with Cyclophilin D (CypD) SiRNA to reversely verified whether the protective effects of HSYA were exerted by regulating the Mitogen-activated protein kinase kinase (MEK)/ERK/CypD pathway. RESULTS HSYA treatment significantly increased BMECs viability, decreased the generation of ROS, opening of mPTP and translocation of cytochrome c after OGD/R. In addition to inhibited CypD, HSYA potentiated MEK and increased phosphorylation of ERK expression in BMECs, inhibited apoptosis mediated by mitochondrial. Notably, HSYA also significantly ameliorated neurological deficits and decreased the infarct volume in rats. CONCLUSION HSYA reduced the CytC export from mitochondrial by inhibited the open of mPTP via MEK/ERK/CypD pathway, contributing to the protection of I/R. Thus, our study not only revealed novel mechanisms of HSYA for its anti-I/R function, but also provided a template for the design of novel mPTP inhibitor for the treatment of various mPTP-related diseases.
Collapse
Affiliation(s)
- Ping Huang
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Si-Peng Wu
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei 230012, China; State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China.
| | - Ning Wang
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Saiwang Seto
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Dennis Chang
- National Institute of Complementary Medicine, Western Sydney University; Penrith, NSW 2751, Australia
| |
Collapse
|
21
|
DoĞan E, DÜzgÜn Z, Yildirim Z, Özdİl B, AktuĞ H, Bozok ÇetİntaŞ V. The effects of PIKfyve inhibitor YM201636 on claudins and malignancy potential of nonsmall cell cancer cells. ACTA ACUST UNITED AC 2021; 45:26-34. [PMID: 33597819 PMCID: PMC7877718 DOI: 10.3906/biy-2010-32] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/22/2020] [Indexed: 11/15/2022]
Abstract
PIKfyve is an evolutionarily conserved lipid and protein kinase enzyme that has pleiotropic cellular functions. The aim of the present study was to investigate the effects of phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) inhibitor, YM201636, on nonsmall cell lung cancer (NSCLC) cells growth, tumorigenicity, and claudin (CLDN) expressions. Three NSCLC cell lines (Calu-1, H1299 and HCC827) were used to compare the effects of YM201636. Cytotoxic effects of YM201636 were analysed using XTT assay. Malignancy potential of cells assesses with wound healing and soft agar colony-forming assays. mRNA and protein expressions of claudins were analysed by qRT-PCR and immunofluorescence staining. Our results revealed that YM201636 inhibited the proliferation and malignancy potential of Calu-1, H1299, and HCC827 cells in a dose-dependent manner. After YM201636 treatment CLDN1, -3 and -5 expressions increased significantly in HCC827 cells. CLDN3 and -5 expressions also significantly increased in Calu1 cell line. YM201636 treatment significantly reduced the CLDN1 and increased the CLDN5 expression in H1299 cells. Immunofluorescence staining of CLDN1, -3 and -5 proteins showed a significant increase after YM201636 treatment. Besides, YM201636 induced EGFR mRNA expression in all NSCLC cell lines. Our results have shown that YM201636 inhibits tumorigenicity of NSCLC cells. Furthermore, estimated glomerular filtration rate (EGFR) pathway is important signalling involved in the regulation of claudins. Understanding the mechanisms of PIKfyve inhibitors may improve cancer treatment particularly for EGFR overactivated NSCLC.
Collapse
Affiliation(s)
- Eda DoĞan
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir Turkey
| | - Zekeriya DÜzgÜn
- Department of Medical Biology, Faculty of Medicine, Giresun University, Giresun Turkey
| | - Zafer Yildirim
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir Turkey
| | - Berrin Özdİl
- Department of Histology and Embryology, Faculty of Medicine, Ege University, İzmir Turkey
| | - Hüseyin AktuĞ
- Department of Histology and Embryology, Faculty of Medicine, Ege University, İzmir Turkey
| | | |
Collapse
|
22
|
Casari I, Manfredi M, Metharom P, Falasca M. Dissecting lipid metabolism alterations in SARS-CoV-2. Prog Lipid Res 2021; 82:101092. [PMID: 33571544 PMCID: PMC7869689 DOI: 10.1016/j.plipres.2021.101092] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the COVID-19 pandemic that has infected over a hundred million people globally. There have been more than two million deaths recorded worldwide, with no end in sight until a widespread vaccination will be achieved. Current research has centred on different aspects of the virus interaction with cell surface receptors, but more needs to be done to further understand its mechanism of action in order to develop a targeted therapy and a method to control the spread of the virus. Lipids play a crucial role throughout the viral life cycle, and viruses are known to exploit lipid signalling and synthesis to affect host cell lipidome. Emerging studies using untargeted metabolomic and lipidomic approaches are providing new insight into the host response to COVID-19 infection. Indeed, metabolomic and lipidomic approaches have identified numerous circulating lipids that directly correlate to the severity of the disease, making lipid metabolism a potential therapeutic target. Circulating lipids play a key function in the pathogenesis of the virus and exert an inflammatory response. A better knowledge of lipid metabolism in the host-pathogen interaction will provide valuable insights into viral pathogenesis and to the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Ilaria Casari
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Pat Metharom
- Platelet Research Group, Perth Blood Institute, West Perth, WA 6005, Australia; Western Australian Centre for Thrombosis and Haemostasis, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; Curtin Medical School, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
| | - Marco Falasca
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia.
| |
Collapse
|
23
|
Saric A, Freeman SA. Endomembrane Tension and Trafficking. Front Cell Dev Biol 2021; 8:611326. [PMID: 33490077 PMCID: PMC7820182 DOI: 10.3389/fcell.2020.611326] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
Eukaryotic cells employ diverse uptake mechanisms depending on their specialized functions. While such mechanisms vary widely in their defining criteria: scale, molecular machinery utilized, cargo selection, and cargo destination, to name a few, they all result in the internalization of extracellular solutes and fluid into membrane-bound endosomes. Upon scission from the plasma membrane, this compartment is immediately subjected to extensive remodeling which involves tubulation and vesiculation/budding of the limiting endomembrane. This is followed by a maturation process involving concomitant retrograde transport by microtubule-based motors and graded fusion with late endosomes and lysosomes, organelles that support the degradation of the internalized content. Here we review an important determinant for sorting and trafficking in early endosomes and in lysosomes; the control of tension on the endomembrane. Remodeling of endomembranes is opposed by high tension (caused by high hydrostatic pressure) and supported by the relief of tension. We describe how the timely and coordinated efflux of major solutes along the endocytic pathway affords the cell control over such tension. The channels and transporters that expel the smallest components of the ingested medium from the early endocytic fluid are described in detail as these systems are thought to enable endomembrane deformation by curvature-sensing/generating coat proteins. We also review similar considerations for the lysosome where resident hydrolases liberate building blocks from luminal macromolecules and transporters flux these organic solutes to orchestrate trafficking events. How the cell directs organellar trafficking based on the luminal contents of organelles of the endocytic pathway is not well-understood, however, we propose that the control over membrane tension by solute transport constitutes one means for this to ensue.
Collapse
Affiliation(s)
- Amra Saric
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Spencer A Freeman
- Program in Cell Biology, Peter Gilgan Center for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Abstract
Lysosomes are membrane-bound organelles with roles in processes involved in degrading and recycling cellular waste, cellular signalling and energy metabolism. Defects in genes encoding lysosomal proteins cause lysosomal storage disorders, in which enzyme replacement therapy has proved successful. Growing evidence also implicates roles for lysosomal dysfunction in more common diseases including inflammatory and autoimmune disorders, neurodegenerative diseases, cancer and metabolic disorders. With a focus on lysosomal dysfunction in autoimmune disorders and neurodegenerative diseases - including lupus, rheumatoid arthritis, multiple sclerosis, Alzheimer disease and Parkinson disease - this Review critically analyses progress and opportunities for therapeutically targeting lysosomal proteins and processes, particularly with small molecules and peptide drugs.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France
| | - Fengjuan Wang
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell Signalling, Illkirch, France.
- Laboratory of Excellence Medalis, Team Neuroimmunology and Peptide Therapy, Institut de Science et d'Ingénierie Supramoléculaire (ISIS), Strasbourg, France.
- University of Strasbourg Institute for Advanced Study, Strasbourg, France.
- Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University, Strasbourg, France.
| |
Collapse
|
25
|
Min SH, Suzuki A, Weaver L, Guzman J, Chung Y, Jin H, Gonzalez F, Trasorras C, Zhao L, Spruce LA, Seeholzer SH, Behrens EM, Abrams CS. PIKfyve Deficiency in Myeloid Cells Impairs Lysosomal Homeostasis in Macrophages and Promotes Systemic Inflammation in Mice. Mol Cell Biol 2019; 39:e00158-19. [PMID: 31427458 PMCID: PMC6791654 DOI: 10.1128/mcb.00158-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/29/2019] [Accepted: 08/12/2019] [Indexed: 01/15/2023] Open
Abstract
Macrophages are professional phagocytes that are essential for host defense and tissue homeostasis. Proper membrane trafficking and degradative functions of the endolysosomal system are known to be critical for the function of these cells. We have found that PIKfyve, the kinase that synthesizes the endosomal phosphoinositide phosphatidylinositol-3,5-bisphosphate, is an essential regulator of lysosomal biogenesis and degradative functions in macrophages. Genetically engineered mice lacking PIKfyve in their myeloid cells (PIKfyvefl/fl LysM-Cre) develop diffuse tissue infiltration of foamy macrophages, hepatosplenomegaly, and systemic inflammation. PIKfyve loss in macrophages causes enlarged endolysosomal compartments and impairs the lysosomal degradative function. Moreover, PIKfyve deficiency increases the cellular levels of lysosomal proteins. Although PIKfyve deficiency reduced the activation of mTORC1 pathway and was associated with increased cleavage of TFEB proteins, this does not translate into transcriptional activation of lysosomal genes, suggesting that PIKfyve modulates the abundance of lysosomal proteins by affecting the degradation of these proteins. Our study shows that PIKfyve modulation of lysosomal degradative activity and protein expression is essential to maintain lysosomal homeostasis in macrophages.
Collapse
Affiliation(s)
- Sang Hee Min
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Aae Suzuki
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lehn Weaver
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jessica Guzman
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yutein Chung
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Huiyan Jin
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Francina Gonzalez
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Claire Trasorras
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Liang Zhao
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lynn A Spruce
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Edward M Behrens
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Charles S Abrams
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
26
|
Staiano L, Zappa F. Hijacking intracellular membranes to feed autophagosomal growth. FEBS Lett 2019; 593:3120-3134. [PMID: 31603532 DOI: 10.1002/1873-3468.13637] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 12/21/2022]
Abstract
Autophagy is widely considered as a housekeeping mechanism that enables cells to survive stress conditions and, in particular, nutrient deprivation. Autophagy begins with the formation of the phagophore that expands and closes around cytosolic material and damaged organelles destined for degradation. The execution of this complex machinery is guaranteed by the coordinated action of more than 40 ATG (autophagy-related) proteins that control the entire process at different stages from the biogenesis of the autophagosome to cargo sequestration and fusion with lysosomes. Autophagosome biogenesis occurs at multiple intracellular sites, such as the endoplasmic reticulum (ER) and the plasma membrane. Soon after the formation of the phagophore, the nascent autophagosome progressively grows in size and ultimately closes by recruiting intracellular membranes. In this review, we focus on the contribution of three membrane sources - the ER, the ER-Golgi intermediate compartment, and the Golgi complex - to autophagosome biogenesis and expansion. We also highlight the interplay between the secretory pathway and autophagy in cells when nutrients are scarce.
Collapse
Affiliation(s)
- Leopoldo Staiano
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Francesca Zappa
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
27
|
Sharma G, Guardia CM, Roy A, Vassilev A, Saric A, Griner LN, Marugan J, Ferrer M, Bonifacino JS, DePamphilis ML. A family of PIKFYVE inhibitors with therapeutic potential against autophagy-dependent cancer cells disrupt multiple events in lysosome homeostasis. Autophagy 2019; 15:1694-1718. [PMID: 30806145 DOI: 10.1080/15548627.2019.1586257] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
High-throughput screening identified 5 chemical analogs (termed the WX8-family) that disrupted 3 events in lysosome homeostasis: (1) lysosome fission via tubulation without preventing homotypic lysosome fusion; (2) trafficking of molecules into lysosomes without altering lysosomal acidity, and (3) heterotypic fusion between lysosomes and autophagosomes. Remarkably, these compounds did not prevent homotypic fusion between lysosomes, despite the fact that homotypic fusion required some of the same machinery essential for heterotypic fusion. These effects varied 400-fold among WX8-family members, were time and concentration dependent, reversible, and resulted primarily from their ability to bind specifically to the PIKFYVE phosphoinositide kinase. The ability of the WX8-family to prevent lysosomes from participating in macroautophagy/autophagy suggested they have therapeutic potential in treating autophagy-dependent diseases. In fact, the most potent family member (WX8) was 100-times more lethal to 'autophagy-addicted' melanoma A375 cells than the lysosomal inhibitors hydroxychloroquine and chloroquine. In contrast, cells that were insensitive to hydroxychloroquine and chloroquine were also insensitive to WX8. Therefore, the WX8-family of PIKFYVE inhibitors provides a basis for developing drugs that could selectively kill autophagy-dependent cancer cells, as well as increasing the effectiveness of established anti-cancer therapies through combinatorial treatments. Abbreviations: ACTB: actin beta; Baf: bafilomycin A1; BECN1: beclin 1; BODIPY: boron-dipyrromethene; BORC: BLOC-1 related complex; BRAF: B-Raf proto-oncogene, serine/threonine kinase; BSA: bovine serum albumin; CTSD: cathepsin D; CQ: chloroquine; DNA: deoxyribonucleic acid; EC50: half maximal effective concentration; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; HCQ: hydroxychloroquine; HOPS complex: homotypic fusion and protein sorting complex; Kd: equilibrium binding constant; IC50: half maximal inhibitory concentration; KO: knockout; LAMP1: lysosomal associated membrane protein 1; MAP1LC3A: microtubule associated protein 1 light chain 3 alpha; MES: 2-(N-morpholino)ethanesulphonic acid; MTOR: mechanistic target of rapamycin kinase; μM: micromolar; NDF: 3-methylbenzaldehyde (2,6-dimorpholin-4-ylpyrimidin-4-yl)hydrazine;NEM: N-ethylmaleimide; NSF: N-ethylmaleimide sensitive factor; PBS: phosphate-buffered saline; PIKFYVE: phosphoinositide kinase, FYVE-type zinc finger containing; PIP4K2C: phosphatidylinositol-5-phosphate 4-kinase type 2 gamma; PtdIns3P: phosphatidylinositol 3-phosphate; PtdIns(3,5)P2: phosphatidylinositol 3,5-biphosphate; RFP: red fluorescent protein; RPS6: ribosomal protein S6; RPS6KB1: ribosomal protein S6 kinase B1; SQSTM1: sequestosome 1; TWEEN 20: polysorbate 20; V-ATPase: vacuolar-type H+-translocating ATPase; VPS39: VPS39 subunit of HOPS complex; VPS41: VPS41 subunit of HOPS complex; WWL: benzaldehyde [2,6-di(4-morpholinyl)-4-pyrimidinyl]hydrazone; WX8: 1H-indole-3-carbaldehyde [4-anilino-6-(4-morpholinyl)-1,3,5-triazin-2-yl]hydrazine; XBA: N-(3-chloro-4-fluorophenyl)-4,6-dimorpholino-1,3,5-triazin-2-amine hydrochloride; XB6: N-(4-ethylphenyl)-4,6-dimorpholino-1,3,5-triazin-2-amine hydrochloride.
Collapse
Affiliation(s)
- Gaurav Sharma
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Carlos M Guardia
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Ajit Roy
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Alex Vassilev
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Amra Saric
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Lori N Griner
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Juan Marugan
- Division of Pre-Clinical Innovation, NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health , Rockville , MD , USA
| | - Marc Ferrer
- Division of Pre-Clinical Innovation, NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health , Rockville , MD , USA
| | - Juan S Bonifacino
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Melvin L DePamphilis
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
28
|
Buckley CM, Heath VL, Guého A, Bosmani C, Knobloch P, Sikakana P, Personnic N, Dove SK, Michell RH, Meier R, Hilbi H, Soldati T, Insall RH, King JS. PIKfyve/Fab1 is required for efficient V-ATPase and hydrolase delivery to phagosomes, phagosomal killing, and restriction of Legionella infection. PLoS Pathog 2019; 15:e1007551. [PMID: 30730983 PMCID: PMC6382210 DOI: 10.1371/journal.ppat.1007551] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 02/20/2019] [Accepted: 01/03/2019] [Indexed: 12/11/2022] Open
Abstract
By engulfing potentially harmful microbes, professional phagocytes are continually at risk from intracellular pathogens. To avoid becoming infected, the host must kill pathogens in the phagosome before they can escape or establish a survival niche. Here, we analyse the role of the phosphoinositide (PI) 5-kinase PIKfyve in phagosome maturation and killing, using the amoeba and model phagocyte Dictyostelium discoideum. PIKfyve plays important but poorly understood roles in vesicular trafficking by catalysing formation of the lipids phosphatidylinositol (3,5)-bisphosphate (PI(3,5)2) and phosphatidylinositol-5-phosphate (PI(5)P). Here we show that its activity is essential during early phagosome maturation in Dictyostelium. Disruption of PIKfyve inhibited delivery of both the vacuolar V-ATPase and proteases, dramatically reducing the ability of cells to acidify newly formed phagosomes and digest their contents. Consequently, PIKfyve- cells were unable to generate an effective antimicrobial environment and efficiently kill captured bacteria. Moreover, we demonstrate that cells lacking PIKfyve are more susceptible to infection by the intracellular pathogen Legionella pneumophila. We conclude that PIKfyve-catalysed phosphoinositide production plays a crucial and general role in ensuring early phagosomal maturation, protecting host cells from diverse pathogenic microbes.
Collapse
Affiliation(s)
- Catherine M. Buckley
- Centre for Membrane Interactions and Dynamics, Department of Biomedical Sciences, University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom
| | - Victoria L. Heath
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Aurélie Guého
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Cristina Bosmani
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Paulina Knobloch
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Phumzile Sikakana
- Centre for Membrane Interactions and Dynamics, Department of Biomedical Sciences, University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom
| | - Nicolas Personnic
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Stephen K. Dove
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Robert H. Michell
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Roger Meier
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zürich, Zürich, Switzerland
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Robert H. Insall
- CRUK Beatson Institute, Switchback Road, Bearsden, Glasgow, United Kingdom
| | - Jason S. King
- Centre for Membrane Interactions and Dynamics, Department of Biomedical Sciences, University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, United Kingdom
| |
Collapse
|
29
|
Li Z, Mbah NE, Overmeyer JH, Sarver JG, George S, Trabbic CJ, Erhardt PW, Maltese WA. The JNK signaling pathway plays a key role in methuosis (non-apoptotic cell death) induced by MOMIPP in glioblastoma. BMC Cancer 2019; 19:77. [PMID: 30651087 PMCID: PMC6335761 DOI: 10.1186/s12885-019-5288-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/07/2019] [Indexed: 12/12/2022] Open
Abstract
Background Synthetic indolyl- pyridinyl- propenones (IPPs) induce methuosis, a form of non-apoptotic cell death, in glioblastoma and other cancer cell lines. Methuosis is characterized by accumulation of cytoplasmic vacuoles derived from macropinosomes and late endosomes, followed by metabolic failure and rupture of the plasma membrane. However, not all IPPs that cause vacuolization are cytotoxic. The main goals of the present study were to identify key signaling pathways that contribute to methuosis induced by cytotoxic IPPs and to evaluate the anti-tumor potential of a prototype IPP in vivo. Methods We utilized metabolic flux analysis, glucose uptake, immunoblotting, and selective pharmacological inhibitors to compare the effects of closely related cytotoxic and non-cytotoxic IPPs in cultured glioblastoma cells. To determine whether the use of methuosis-inducing IPPs might be feasible in a therapeutic context, we quantified the distribution of our lead IPP compound, MOMIPP, in mouse plasma and brain, and tested its ability to inhibit tumor growth in an intracerebral glioblastoma xenograft model. Results The cytotoxic IPP compound, MOMIPP, causes early disruptions of glucose uptake and glycolytic metabolism. Coincident with these metabolic changes, MOMIPP selectively activates the JNK1/2 stress kinase pathway, resulting in phosphorylation of c-Jun, Bcl-2 and Bcl-xL. At the same concentration, the non-cytotoxic analog, MOPIPP, does not activate these pathways. Pharmacologic inhibition of JNK activity promotes survival, even when cells are extensively vacuolated, but suppression of c-Jun transcriptional activity offers no protection. MOMIPP readily penetrates the blood-brain barrier and is moderately effective in suppressing progression of intracerebral glioblastoma xenografts. Conclusions The results suggest that interference with glucose uptake and induction of JNK-mediated phosphorylation of pro-survival members of the Bcl-2 family represent key events in the methuosis death process. In addition to providing new insights into the underlying molecular mechanism of methuosis, the results indicate that compounds of the cytotoxic IPP class may have potential for further development as therapeutic agents for brain tumors. Electronic supplementary material The online version of this article (10.1186/s12885-019-5288-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zehui Li
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, 43614, United States
| | - Nneka E Mbah
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, 43614, United States
| | - Jean H Overmeyer
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, 43614, United States
| | - Jeffrey G Sarver
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43606, USA
| | - Sage George
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, 43614, United States
| | - Christopher J Trabbic
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43606, USA
| | - Paul W Erhardt
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43606, USA
| | - William A Maltese
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, 43614, United States.
| |
Collapse
|
30
|
Martinez E, Siadous FA, Bonazzi M. Tiny architects: biogenesis of intracellular replicative niches by bacterial pathogens. FEMS Microbiol Rev 2018; 42:425-447. [PMID: 29596635 DOI: 10.1093/femsre/fuy013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/26/2018] [Indexed: 11/13/2022] Open
Abstract
Co-evolution of bacterial pathogens with their hosts led to the emergence of a stunning variety of strategies aiming at the evasion of host defences, colonisation of host cells and tissues and, ultimately, the establishment of a successful infection. Pathogenic bacteria are typically classified as extracellular and intracellular; however, intracellular lifestyle comes in many different flavours: some microbes rapidly escape to the cytosol whereas other microbes remain within vacuolar compartments and harness membrane trafficking pathways to generate their host-derived, pathogen-specific replicative niche. Here we review the current knowledge on a variety of vacuolar lifestyles, the effector proteins used by bacteria as tools to take control of the host cell and the main membrane trafficking signalling pathways targeted by vacuolar pathogens as source of membranes and nutrients. Finally, we will also discuss how host cells have developed countermeasures to sense the biogenesis of the aberrant organelles harbouring bacteria. Understanding the dialogue between bacterial and eukaryotic proteins is the key to unravel the molecular mechanisms of infection and in turn, this may lead to the identification of new targets for the development of new antimicrobials.
Collapse
Affiliation(s)
- Eric Martinez
- IRIM, University of Montpellier, CNRS, 34293 Montpellier, France
| | | | - Matteo Bonazzi
- IRIM, University of Montpellier, CNRS, 34293 Montpellier, France
| |
Collapse
|
31
|
Chaytow H, Huang YT, Gillingwater TH, Faller KME. The role of survival motor neuron protein (SMN) in protein homeostasis. Cell Mol Life Sci 2018; 75:3877-3894. [PMID: 29872871 PMCID: PMC6182345 DOI: 10.1007/s00018-018-2849-1] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 12/11/2022]
Abstract
Ever since loss of survival motor neuron (SMN) protein was identified as the direct cause of the childhood inherited neurodegenerative disorder spinal muscular atrophy, significant efforts have been made to reveal the molecular functions of this ubiquitously expressed protein. Resulting research demonstrated that SMN plays important roles in multiple fundamental cellular homeostatic pathways, including a well-characterised role in the assembly of the spliceosome and biogenesis of ribonucleoproteins. More recent studies have shown that SMN is also involved in other housekeeping processes, including mRNA trafficking and local translation, cytoskeletal dynamics, endocytosis and autophagy. Moreover, SMN has been shown to influence mitochondria and bioenergetic pathways as well as regulate function of the ubiquitin-proteasome system. In this review, we summarise these diverse functions of SMN, confirming its key role in maintenance of the homeostatic environment of the cell.
Collapse
Affiliation(s)
- Helena Chaytow
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Yu-Ting Huang
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Thomas H Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK.
| | - Kiterie M E Faller
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
32
|
Isobe Y, Nigorikawa K, Tsurumi G, Takemasu S, Takasuga S, Kofuji S, Hazeki K. PIKfyve accelerates phagosome acidification through activation of TRPML1 while arrests aberrant vacuolation independent of the Ca2+ channel. J Biochem 2018; 165:75-84. [DOI: 10.1093/jb/mvy084] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/05/2018] [Indexed: 01/17/2023] Open
Affiliation(s)
- Yuri Isobe
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Minamiku Kasumi 1-2-3, Hiroshima, Japan
| | - Kiyomi Nigorikawa
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Minamiku Kasumi 1-2-3, Hiroshima, Japan
| | - Go Tsurumi
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Minamiku Kasumi 1-2-3, Hiroshima, Japan
| | - Shinya Takemasu
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Minamiku Kasumi 1-2-3, Hiroshima, Japan
| | - Shunsuke Takasuga
- Department of Pathology and Immunology, Akita University School of Medicine, Akita, Japan
| | - Satoshi Kofuji
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Minamiku Kasumi 1-2-3, Hiroshima, Japan
| | - Kaoru Hazeki
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Minamiku Kasumi 1-2-3, Hiroshima, Japan
| |
Collapse
|
33
|
PIKfyve inhibitor cytotoxicity requires AKT suppression and excessive cytoplasmic vacuolation. Toxicol Appl Pharmacol 2018; 356:151-158. [DOI: 10.1016/j.taap.2018.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/16/2018] [Accepted: 08/02/2018] [Indexed: 11/22/2022]
|
34
|
Choy CH, Saffi G, Gray MA, Wallace C, Dayam RM, Ou ZYA, Lenk G, Puertollano R, Watkins SC, Botelho RJ. Lysosome enlargement during inhibition of the lipid kinase PIKfyve proceeds through lysosome coalescence. J Cell Sci 2018; 131:jcs.213587. [PMID: 29661845 DOI: 10.1242/jcs.213587] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/10/2018] [Indexed: 01/07/2023] Open
Abstract
Lysosomes receive and degrade cargo from endocytosis, phagocytosis and autophagy. They also play an important role in sensing and instructing cells on their metabolic state. The lipid kinase PIKfyve generates phosphatidylinositol-3,5-bisphosphate to modulate lysosome function. PIKfyve inhibition leads to impaired degradative capacity, ion dysregulation, abated autophagic flux and a massive enlargement of lysosomes. Collectively, this leads to various physiological defects, including embryonic lethality, neurodegeneration and overt inflammation. The reasons for such drastic lysosome enlargement remain unclear. Here, we examined whether biosynthesis and/or fusion-fission dynamics contribute to swelling. First, we show that PIKfyve inhibition activates TFEB, TFE3 and MITF, enhancing lysosome gene expression. However, this did not augment lysosomal protein levels during acute PIKfyve inhibition, and deletion of TFEB and/or related proteins did not impair lysosome swelling. Instead, PIKfyve inhibition led to fewer but enlarged lysosomes, suggesting that an imbalance favouring lysosome fusion over fission causes lysosome enlargement. Indeed, conditions that abated fusion curtailed lysosome swelling in PIKfyve-inhibited cells.
Collapse
Affiliation(s)
- Christopher H Choy
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3.,The Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada, M5B2K3
| | - Golam Saffi
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3.,The Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada, M5B2K3
| | - Matthew A Gray
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3
| | - Callen Wallace
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Roya M Dayam
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3.,The Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada, M5B2K3
| | - Zhen-Yi A Ou
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3
| | - Guy Lenk
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Room 3537, Bethesda, MD 20892, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Roberto J Botelho
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3 .,The Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada, M5B2K3
| |
Collapse
|
35
|
Fogarty K, Kashem M, Bauer A, Bernardino A, Brennan D, Cook B, Farrow N, Molinaro T, Nelson R. Development of Three Orthogonal Assays Suitable for the Identification and Qualification of PIKfyve Inhibitors. Assay Drug Dev Technol 2018; 15:210-219. [PMID: 28723271 DOI: 10.1089/adt.2017.790] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
FYVE-type zinc finger-containing phosphoinositide kinase (PIKfyve) catalyzes the formation of phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2) from phosphatidylinositol 3-phosphate (PI(3)P). PIKfyve has been implicated in multiple cellular processes, and its role in the regulation of toll-like receptor (TLR) pathways and the production of proinflammatory cytokines has sparked interest in developing small-molecule PIKfyve inhibitors as potential therapeutics to treat autoimmune and inflammatory diseases. We developed three orthogonal assays to identify and qualify small-molecule inhibitors of PIKfyve: (1) a purified component microfluidic enzyme assay that measures the conversion of fluorescently labeled PI(3)P to PI(3,5)P2 by purified recombinant full-length human 6His-PIKfyve (rPIKfyve); (2) an intracellular protein stabilization assay using the kinase domain of PIKfyve expressed in HEK293 cells; and (3) a cell-based functional assay that measures the production of interleukin (IL)-12p70 in human peripheral blood mononuclear cells stimulated with TLR agonists lipopolysaccharide and R848. We determined apparent Km values for both ATP and labeled PI(3)P in the rPIKfyve enzyme assay and evaluated the enzyme's ability to use phosphatidylinositol as a substrate. We also tested four reference compounds in the three assays and showed that together these assays provide a platform that is suitable to select promising inhibitors having appropriate functional activity and confirmed cellular target engagement to advance into preclinical models of inflammation.
Collapse
Affiliation(s)
- Kylie Fogarty
- 1 Department of Small Molecule Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield, Connecticut
| | - Mohammed Kashem
- 1 Department of Small Molecule Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield, Connecticut
| | - Andras Bauer
- 2 Department of Immunology and Inflammation, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield, Connecticut
| | - Alexandra Bernardino
- 2 Department of Immunology and Inflammation, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield, Connecticut
| | - Debra Brennan
- 1 Department of Small Molecule Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield, Connecticut
| | - Brian Cook
- 1 Department of Small Molecule Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield, Connecticut
| | - Neil Farrow
- 1 Department of Small Molecule Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield, Connecticut
| | - Teresa Molinaro
- 1 Department of Small Molecule Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield, Connecticut
| | - Richard Nelson
- 1 Department of Small Molecule Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc. , Ridgefield, Connecticut
| |
Collapse
|
36
|
Liggins MC, Flesher JL, Jahid S, Vasudeva P, Eby V, Takasuga S, Sasaki J, Sasaki T, Boissy RE, Ganesan AK. PIKfyve regulates melanosome biogenesis. PLoS Genet 2018; 14:e1007290. [PMID: 29584722 PMCID: PMC5889185 DOI: 10.1371/journal.pgen.1007290] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/06/2018] [Accepted: 03/05/2018] [Indexed: 12/18/2022] Open
Abstract
PIKfyve, VAC14, and FIG4 form a complex that catalyzes the production of PI(3,5)P2, a signaling lipid implicated in process ranging from lysosome maturation to neurodegeneration. While previous studies have identified VAC14 and FIG4 mutations that lead to both neurodegeneration and coat color defects, how PIKfyve regulates melanogenesis is unknown. In this study, we sought to better understand the role of PIKfyve in melanosome biogenesis. Melanocyte-specific PIKfyve knockout mice exhibit greying of the mouse coat and the accumulation of single membrane vesicle structures in melanocytes resembling multivesicular endosomes. PIKfyve inhibition blocks melanosome maturation, the processing of the melanosome protein PMEL, and the trafficking of the melanosome protein TYRP1. Taken together, these studies identify a novel role for PIKfyve in controlling the delivery of proteins from the endosomal compartment to the melanosome, a role that is distinct from the role of PIKfyve in the reformation of lysosomes from endolysosomes.
Collapse
Affiliation(s)
- Marc C. Liggins
- Department of Dermatology, University of California, San Diego, San Diego, CA, United States of America
| | - Jessica L. Flesher
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States of America
| | - Sohail Jahid
- Department of Dermatology, University of California, Irvine, Irvine, CA, United States of America
| | - Priya Vasudeva
- Department of Dermatology, University of California, Irvine, Irvine, CA, United States of America
| | - Victoria Eby
- Department of Dermatology, University of Cincinnati, Cincinnati, OH, United States of America
| | - Shunsuke Takasuga
- Department of Medical Biology, Akita University School of Medicine, Akita, Japan
| | - Junko Sasaki
- Department of Medical Biology, Akita University School of Medicine, Akita, Japan
| | - Takehiko Sasaki
- Department of Medical Biology, Akita University School of Medicine, Akita, Japan
| | - Raymond E. Boissy
- Department of Dermatology, University of Cincinnati, Cincinnati, OH, United States of America
| | - Anand K. Ganesan
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, United States of America
- Department of Dermatology, University of California, Irvine, Irvine, CA, United States of America
- * E-mail:
| |
Collapse
|
37
|
Shi Y, Lin S, Staats KA, Li Y, Chang WH, Hung ST, Hendricks E, Linares GR, Wang Y, Son EY, Wen X, Kisler K, Wilkinson B, Menendez L, Sugawara T, Woolwine P, Huang M, Cowan MJ, Ge B, Koutsodendris N, Sandor KP, Komberg J, Vangoor VR, Senthilkumar K, Hennes V, Seah C, Nelson AR, Cheng TY, Lee SJJ, August PR, Chen JA, Wisniewski N, Victor HS, Belgard TG, Zhang A, Coba M, Grunseich C, Ward ME, van den Berg LH, Pasterkamp RJ, Trotti D, Zlokovic BV, Ichida JK. Haploinsufficiency leads to neurodegeneration in C9ORF72 ALS/FTD human induced motor neurons. Nat Med 2018; 24:313-325. [PMID: 29400714 PMCID: PMC6112156 DOI: 10.1038/nm.4490] [Citation(s) in RCA: 435] [Impact Index Per Article: 62.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 01/01/2018] [Indexed: 12/13/2022]
Abstract
An intronic GGGGCC repeat expansion in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), but the pathogenic mechanism of this repeat remains unclear. Using human induced motor neurons (iMNs), we found that repeat-expanded C9ORF72 was haploinsufficient in ALS. We found that C9ORF72 interacted with endosomes and was required for normal vesicle trafficking and lysosomal biogenesis in motor neurons. Repeat expansion reduced C9ORF72 expression, triggering neurodegeneration through two mechanisms: accumulation of glutamate receptors, leading to excitotoxicity, and impaired clearance of neurotoxic dipeptide repeat proteins derived from the repeat expansion. Thus, cooperativity between gain- and loss-of-function mechanisms led to neurodegeneration. Restoring C9ORF72 levels or augmenting its function with constitutively active RAB5 or chemical modulators of RAB5 effectors rescued patient neuron survival and ameliorated neurodegenerative processes in both gain- and loss-of-function C9ORF72 mouse models. Thus, modulating vesicle trafficking was able to rescue neurodegeneration caused by the C9ORF72 repeat expansion. Coupled with rare mutations in ALS2, FIG4, CHMP2B, OPTN and SQSTM1, our results reveal mechanistic convergence on vesicle trafficking in ALS and FTD.
Collapse
Affiliation(s)
- Yingxiao Shi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Shaoyu Lin
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Kim A. Staats
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Yichen Li
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Wen-Hsuan Chang
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Shu-Ting Hung
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Eric Hendricks
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Gabriel R. Linares
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Yaoming Wang
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Esther Y. Son
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinmei Wen
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Kassandra Kisler
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Brent Wilkinson
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Louise Menendez
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Tohru Sugawara
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Phillip Woolwine
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Mickey Huang
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Michael J. Cowan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Brandon Ge
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Nicole Koutsodendris
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Kaitlin P. Sandor
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Jacob Komberg
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Vamshidhar R. Vangoor
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Ketharini Senthilkumar
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Valerie Hennes
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Carina Seah
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Amy R. Nelson
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | | | | | | - Marcelo Coba
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Chris Grunseich
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael E. Ward
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Berislav V. Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Justin K. Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
38
|
Minakaki G, Menges S, Kittel A, Emmanouilidou E, Schaeffner I, Barkovits K, Bergmann A, Rockenstein E, Adame A, Marxreiter F, Mollenhauer B, Galasko D, Buzás EI, Schlötzer-Schrehardt U, Marcus K, Xiang W, Lie DC, Vekrellis K, Masliah E, Winkler J, Klucken J. Autophagy inhibition promotes SNCA/alpha-synuclein release and transfer via extracellular vesicles with a hybrid autophagosome-exosome-like phenotype. Autophagy 2018; 14:98-119. [PMID: 29198173 DOI: 10.1080/15548627.2017.1395992] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The autophagy-lysosome pathway (ALP) regulates intracellular homeostasis of the cytosolic protein SNCA/alpha-synuclein and is impaired in synucleinopathies, including Parkinson disease and dementia with Lewy bodies (DLB). Emerging evidence suggests that ALP influences SNCA release, but the underlying cellular mechanisms are not well understood. Several studies identified SNCA in exosome/extracellular vesicle (EV) fractions. EVs are generated in the multivesicular body compartment and either released upon its fusion with the plasma membrane, or cleared via the ALP. We therefore hypothesized that inhibiting ALP clearance 1) enhances SNCA release via EVs by increasing extracellular shuttling of multivesicular body contents, 2) alters EV biochemical profile, and 3) promotes SNCA cell-to-cell transfer. Indeed, ALP inhibition increased the ratio of extra- to intracellular SNCA and upregulated SNCA association with EVs in neuronal cells. Ultrastructural analysis revealed a widespread, fused multivesicular body-autophagosome compartment. Biochemical characterization revealed the presence of autophagosome-related proteins, such as LC3-II and SQSTM1. This distinct "autophagosome-exosome-like" profile was also identified in human cerebrospinal fluid (CSF) EVs. After a single intracortical injection of SNCA-containing EVs derived from CSF into mice, human SNCA colocalized with endosome and neuronal markers. Prominent SNCA immunoreactivity and a higher number of neuronal SNCA inclusions were observed after DLB patient CSF EV injections. In summary, this study provides compelling evidence that a) ALP inhibition increases SNCA in neuronal EVs, b) distinct ALP components are present in EVs, and c) CSF EVs transfer SNCA from cell to cell in vivo. Thus, macroautophagy/autophagy may regulate EV protein composition and consequently progression in synucleinopathies.
Collapse
Affiliation(s)
- Georgia Minakaki
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Stefanie Menges
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Agnes Kittel
- b Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences , Semmelweis University , Budapest , Hungary
| | - Evangelia Emmanouilidou
- c Department of Neuroscience, Center for Basic Research , Biomedical Research Foundation of the Academy of Athens , Athens , Greece
| | | | - Katalin Barkovits
- e Medizinisches Proteom-Center, Medical Faculty , Ruhr University Bochum , Bochum , Germany
| | - Anna Bergmann
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Edward Rockenstein
- f Department of Neurosciences , University of California , San Diego , CA USA
| | - Anthony Adame
- f Department of Neurosciences , University of California , San Diego , CA USA
| | - Franz Marxreiter
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Brit Mollenhauer
- g Paracelsus-Elena-Klinik, Kassel and Departments of Neuropathology & Neurosurgery , & University Medical Center , Göttingen
| | - Douglas Galasko
- f Department of Neurosciences , University of California , San Diego , CA USA
| | - Edit Irén Buzás
- h Department of Genetics, Cell and Immunobiology , Semmelweis University , Budapest , Hungary
| | | | - Katrin Marcus
- e Medizinisches Proteom-Center, Medical Faculty , Ruhr University Bochum , Bochum , Germany
| | - Wei Xiang
- d Institute of Biochemistry , FAU , Erlangen , Germany
| | | | - Kostas Vekrellis
- c Department of Neuroscience, Center for Basic Research , Biomedical Research Foundation of the Academy of Athens , Athens , Greece
| | - Eliezer Masliah
- f Department of Neurosciences , University of California , San Diego , CA USA.,j Department of Pathology , University of California , San Diego , CA USA
| | - Jürgen Winkler
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| | - Jochen Klucken
- a Department of Molecular Neurology , University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen , Germany
| |
Collapse
|
39
|
Zhu D, Yan H, Zhou Z, Tang J, Liu X, Hartmann R, Parak WJ, Feliu N, Shen Y. Detailed investigation on how the protein corona modulates the physicochemical properties and gene delivery of polyethylenimine (PEI) polyplexes. Biomater Sci 2018; 6:1800-1817. [DOI: 10.1039/c8bm00128f] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Given the various cationic polymers developed as non-viral gene delivery vectors, polyethylenimine (PEI) has been/is frequently used in in vitro transfection.
Collapse
Affiliation(s)
- Dingcheng Zhu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
- Fachbereich Physik
| | - Huijie Yan
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
- Fachbereich Physik
| | - Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
| | - Jianbin Tang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
| | - Xiangrui Liu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
| | | | - Wolfgang J. Parak
- Fachbereich Physik
- Philipps Universität Marburg
- Germany
- Fachbereich Physik und Chemie and CHyN
- Universität Hamburg
| | - Neus Feliu
- Fachbereich Physik
- Philipps Universität Marburg
- Germany
- Fachbereich Physik und Chemie and CHyN
- Universität Hamburg
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education
- College of Chemical and Biological Engineering
- Zhejiang University
- China
| |
Collapse
|
40
|
Al-Ramahi I, Giridharan SSP, Chen YC, Patnaik S, Safren N, Hasegawa J, de Haro M, Wagner Gee AK, Titus SA, Jeong H, Clarke J, Krainc D, Zheng W, Irvine RF, Barmada S, Ferrer M, Southall N, Weisman LS, Botas J, Marugan JJ. Inhibition of PIP4Kγ ameliorates the pathological effects of mutant huntingtin protein. eLife 2017; 6:29123. [PMID: 29256861 PMCID: PMC5743427 DOI: 10.7554/elife.29123] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 11/13/2017] [Indexed: 12/15/2022] Open
Abstract
The discovery of the causative gene for Huntington’s disease (HD) has promoted numerous efforts to uncover cellular pathways that lower levels of mutant huntingtin protein (mHtt) and potentially forestall the appearance of HD-related neurological defects. Using a cell-based model of pathogenic huntingtin expression, we identified a class of compounds that protect cells through selective inhibition of a lipid kinase, PIP4Kγ. Pharmacological inhibition or knock-down of PIP4Kγ modulates the equilibrium between phosphatidylinositide (PI) species within the cell and increases basal autophagy, reducing the total amount of mHtt protein in human patient fibroblasts and aggregates in neurons. In two Drosophila models of Huntington’s disease, genetic knockdown of PIP4K ameliorated neuronal dysfunction and degeneration as assessed using motor performance and retinal degeneration assays respectively. Together, these results suggest that PIP4Kγ is a druggable target whose inhibition enhances productive autophagy and mHtt proteolysis, revealing a useful pharmacological point of intervention for the treatment of Huntington’s disease, and potentially for other neurodegenerative disorders.
Collapse
Affiliation(s)
- Ismael Al-Ramahi
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Baylor College of Medicine, Texas Medical Center, Houston, United States
| | | | - Yu-Chi Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, United States
| | - Samarjit Patnaik
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, United States
| | - Nathaniel Safren
- Department of Neurology, University of Michigan, Ann Arbor, United States
| | - Junya Hasegawa
- Department of Cell and Developmental Biology, Life Sciences Institute, University of Michigan, Ann Arbor, United States
| | - Maria de Haro
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Baylor College of Medicine, Texas Medical Center, Houston, United States
| | - Amanda K Wagner Gee
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, United States
| | - Steven A Titus
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, United States
| | - Hyunkyung Jeong
- The Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Jonathan Clarke
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Dimitri Krainc
- The Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Wei Zheng
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, United States
| | - Robin F Irvine
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Sami Barmada
- Department of Neurology, University of Michigan, Ann Arbor, United States
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, United States
| | - Noel Southall
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, United States
| | - Lois S Weisman
- Department of Cell and Developmental Biology, Life Sciences Institute, University of Michigan, Ann Arbor, United States
| | - Juan Botas
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Baylor College of Medicine, Texas Medical Center, Houston, United States
| | - Juan Jose Marugan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, Rockville, United States
| |
Collapse
|
41
|
Bissig C, Hurbain I, Raposo G, van Niel G. PIKfyve activity regulates reformation of terminal storage lysosomes from endolysosomes. Traffic 2017; 18:747-757. [DOI: 10.1111/tra.12525] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 08/25/2017] [Accepted: 08/25/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Christin Bissig
- Institut Curie; PSL Research University, CNRS, UMR144; Paris France
- Sorbonne Universités; UPMC Univ Paris 06, CNRS, UMR 144; Paris France
| | - Ilse Hurbain
- Institut Curie; PSL Research University, CNRS, UMR144; Paris France
- Sorbonne Universités; UPMC Univ Paris 06, CNRS, UMR 144; Paris France
- Cell and Tissue Imaging Core Facility PICT-IBiSA; Institut Curie; Paris France
| | - Graça Raposo
- Institut Curie; PSL Research University, CNRS, UMR144; Paris France
- Sorbonne Universités; UPMC Univ Paris 06, CNRS, UMR 144; Paris France
- Cell and Tissue Imaging Core Facility PICT-IBiSA; Institut Curie; Paris France
| | - Guillaume van Niel
- Institut Curie; PSL Research University, CNRS, UMR144; Paris France
- Sorbonne Universités; UPMC Univ Paris 06, CNRS, UMR 144; Paris France
- Cell and Tissue Imaging Core Facility PICT-IBiSA; Institut Curie; Paris France
| |
Collapse
|
42
|
Qiu S, Leung A, Bo Y, Kozak RA, Anand SP, Warkentin C, Salambanga FDR, Cui J, Kobinger G, Kobasa D, Côté M. Ebola virus requires phosphatidylinositol (3,5) bisphosphate production for efficient viral entry. Virology 2017; 513:17-28. [PMID: 29031163 DOI: 10.1016/j.virol.2017.09.028] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/28/2017] [Accepted: 09/30/2017] [Indexed: 12/11/2022]
Abstract
For entry, Ebola virus (EBOV) requires the interaction of its viral glycoprotein with the cellular protein Niemann-Pick C1 (NPC1) which resides in late endosomes and lysosomes. How EBOV is trafficked and delivered to NPC1 and whether this is positively regulated during entry remain unclear. Here, we show that the PIKfyve-ArPIKfyve-Sac3 cellular complex, which is involved in the metabolism of phosphatidylinositol (3,5) bisphosphate (PtdIns(3,5)P2), is critical for EBOV infection. Although the expression of all subunits of the complex was required for efficient entry, PIKfyve kinase activity was specifically critical for entry by all pathogenic filoviruses. Inhibition of PIKfyve prevented colocalization of EBOV with NPC1 and led to virus accumulation in intracellular vesicles with characteristics of early endosomes. Importantly, genetically-encoded phosphoinositide probes revealed an increase in PtdIns(3,5)P2-positive vesicles in cells during EBOV entry. Taken together, our studies suggest that EBOV requires PtdIns(3,5)P2 production in cells to promote efficient delivery to NPC1.
Collapse
Affiliation(s)
- Shirley Qiu
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Canada
| | - Anders Leung
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Yuxia Bo
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Canada
| | - Robert A Kozak
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Sai Priya Anand
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Canada
| | - Corina Warkentin
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Canada
| | - Fabiola D R Salambanga
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Canada
| | - Jennifer Cui
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Canada
| | - Gary Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
43
|
Dayam RM, Sun CX, Choy CH, Mancuso G, Glogauer M, Botelho RJ. The Lipid Kinase PIKfyve Coordinates the Neutrophil Immune Response through the Activation of the Rac GTPase. THE JOURNAL OF IMMUNOLOGY 2017; 199:2096-2105. [DOI: 10.4049/jimmunol.1601466] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 07/11/2017] [Indexed: 11/19/2022]
|
44
|
Demirsoy S, Martin S, Motamedi S, van Veen S, Holemans T, Van den Haute C, Jordanova A, Baekelandt V, Vangheluwe P, Agostinis P. ATP13A2/PARK9 regulates endo-/lysosomal cargo sorting and proteostasis through a novel PI(3, 5)P2-mediated scaffolding function. Hum Mol Genet 2017; 26:1656-1669. [PMID: 28334751 DOI: 10.1093/hmg/ddx070] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/20/2017] [Indexed: 12/20/2022] Open
Abstract
ATP13A2 (also called PARK9), is a transmembrane endo-/lysosomal-associated P5 type transport ATPase. Loss-of-function mutations in ATP13A2 result in the Kufor-Rakeb Syndrome (KRS), a form of autosomal Parkinson's disease (PD). In spite of a growing interest in ATP13A2, very little is known about its physiological role in stressed cells. Recent studies suggest that the N-terminal domain of ATP13A2 may hold key regulatory functions, but their nature remains incompletely understood. To this end, we generated a set of melanoma and neuroblastoma cell lines stably overexpressing wild-type (WT), catalytically inactive (D508N) and N-terminal mutants, or shRNA against ATP13A2. We found that under proteotoxic stress conditions, evoked by the proteasome inhibitor Bortezomib, endo-/lysosomal associated full-length ATP13A2 WT, catalytically-inactive or N-terminal fragment mutants, reduced the intracellular accumulation of ubiquitin-conjugated (Ub) proteins, independent of autophagic degradation. In contrast, ATP13A2 silencing increased the intracellular accumulation of Ub-proteins, a pattern also observed in patient-derived fibroblasts harbouring ATP13A2 loss-of function mutations. In treated cells, ATP13A2 evoked endocytic vesicle relocation and increased cargo export through nanovesicles. Expression of an ATP13A2 mutant abrogating PI(3,5)P2 binding or chemical inhibition of the PI(3,5)P2-generating enzyme PIKfyve, compromised vesicular trafficking/nanovesicles export and rescued intracellular accumulation of Ub-proteins in response to proteasomal inhibition. Hence, our study unravels a novel activity-independent scaffolding role of ATP13A2 in trafficking/export of intracellular cargo in response to proteotoxic stress.
Collapse
Affiliation(s)
- S Demirsoy
- Laboratory for Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven)
| | - S Martin
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Campus Gasthuisberg, O&N1, Herestraat 49, Box 802, B-3000 Leuven, Belgium
| | - S Motamedi
- Laboratory for Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven)
| | - S van Veen
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Campus Gasthuisberg, O&N1, Herestraat 49, Box 802, B-3000 Leuven, Belgium
| | - T Holemans
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Campus Gasthuisberg, O&N1, Herestraat 49, Box 802, B-3000 Leuven, Belgium
| | - C Van den Haute
- Research Group for Neurobiology and Gene Therapy, Department of Neurosciences, University of Leuven (KU Leuven), B3000 Leuven, Belgium
| | - A Jordanova
- Molecular Neurogenomics Group, VIB Center for Molecular Neurology, University of Antwerp, 2610 Antwerpen, Belgium
- Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical University-Sofia, 1431 Sofia, Bulgaria
| | - V Baekelandt
- Research Group for Neurobiology and Gene Therapy, Department of Neurosciences, University of Leuven (KU Leuven), B3000 Leuven, Belgium
| | - P Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Campus Gasthuisberg, O&N1, Herestraat 49, Box 802, B-3000 Leuven, Belgium
| | - P Agostinis
- Laboratory for Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven)
| |
Collapse
|
45
|
Hasegawa J, Strunk BS, Weisman LS. PI5P and PI(3,5)P 2: Minor, but Essential Phosphoinositides. Cell Struct Funct 2017; 42:49-60. [PMID: 28302928 DOI: 10.1247/csf.17003] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In most eukaryotes, phosphoinositides (PIs) have crucial roles in multiple cellular functions. Although the cellular levels of phosphatidylinositol 5-phosphate (PI5P) and phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2) are extremely low relative to some other PIs, emerging evidence demonstrates that both lipids are crucial for the endocytic pathway, intracellular signaling, and adaptation to stress. Mutations that causes defects in the biosynthesis of PI5P and PI(3,5)P2 are linked to human diseases including neurodegenerative disorders. Here, we review recent findings on cellular roles of PI5P and PI(3,5)P2, as well as the pathophysiological importance of these lipids.Key words: Phosphoinositides, Membrane trafficking, Endocytosis, Vacuoles/Lysosomes, Fab1/PIKfyve.
Collapse
|
46
|
Hessvik NP, Øverbye A, Brech A, Torgersen ML, Jakobsen IS, Sandvig K, Llorente A. PIKfyve inhibition increases exosome release and induces secretory autophagy. Cell Mol Life Sci 2016; 73:4717-4737. [PMID: 27438886 PMCID: PMC11108566 DOI: 10.1007/s00018-016-2309-8] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 01/08/2023]
Abstract
Exosomes are vesicles released from cells by fusion of multivesicular bodies (MVBs) with the plasma membrane. This study aimed to investigate whether the phosphoinositide kinase PIKfyve affects this process. Our results show that in PC-3 cells inhibition of PIKfyve by apilimod or depletion by siRNA increased the secretion of the exosomal fraction. Moreover, quantitative electron microscopy analysis showed that cells treated with apilimod contained more MVBs per cell and more intraluminal vesicles per MVB. Interestingly, mass spectrometry analysis revealed a considerable enrichment of autophagy-related proteins (NBR1, p62, LC3, WIPI2) in exosomal fractions released by apilimod-treated cells, a result that was confirmed by immunoblotting. When the exosome preparations were investigated by electron microscopy a small population of p62-labelled electron dense structures was observed together with CD63-containing exosomes. The p62-positive structures were found in less dense fractions than exosomes in density gradients. Inside the cells, p62 and CD63 were found in the same MVB-like organelles. Finally, both the degradation of EGF and long-lived proteins were shown to be reduced by apilimod. In conclusion, inhibition of PIKfyve increases secretion of exosomes and induces secretory autophagy, showing that these pathways are closely linked. We suggest this is due to impaired fusion of lysosomes with both MVBs and autophagosomes, and possibly increased fusion of MVBs with autophagosomes, and that the cells respond by secreting the content of these organelles to maintain cellular homeostasis.
Collapse
Affiliation(s)
- Nina Pettersen Hessvik
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
| | - Anders Øverbye
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
| | - Andreas Brech
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
- Department of Biosciences, University of Oslo, 0316, Oslo, Norway
| | - Maria Lyngaas Torgersen
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
| | - Ida Seim Jakobsen
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
- Department of Biosciences, University of Oslo, 0316, Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway.
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway.
| |
Collapse
|
47
|
Phosphatidylinositol 3,5-bisphosphate: regulation of cellular events in space and time. Biochem Soc Trans 2016; 44:177-84. [PMID: 26862203 DOI: 10.1042/bst20150174] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Phosphorylated phosphatidylinositol lipids are crucial for most eukaryotes and have diverse cellular functions. The low-abundance signalling lipid phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2] is critical for cellular homoeostasis and adaptation to stimuli. A large complex of proteins that includes the lipid kinase Fab1-PIKfyve, dynamically regulates the levels of PI(3,5)P2. Deficiencies in PI(3,5)P2 are linked to some human diseases, especially those of the nervous system. Future studies will probably determine new, undiscovered regulatory roles of PI(3,5)P2, as well as uncover mechanistic insights into how PI(3,5)P2 contributes to normal human physiology.
Collapse
|
48
|
Hasegawa J, Iwamoto R, Otomo T, Nezu A, Hamasaki M, Yoshimori T. Autophagosome-lysosome fusion in neurons requires INPP5E, a protein associated with Joubert syndrome. EMBO J 2016; 35:1853-67. [PMID: 27340123 DOI: 10.15252/embj.201593148] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 05/27/2016] [Indexed: 12/20/2022] Open
Abstract
Autophagy is a multistep membrane traffic pathway. In contrast to autophagosome formation, the mechanisms underlying autophagosome-lysosome fusion remain largely unknown. Here, we describe a novel autophagy regulator, inositol polyphosphate-5-phosphatase E (INPP5E), involved in autophagosome-lysosome fusion process. In neuronal cells, INPP5E knockdown strongly inhibited autophagy by impairing the fusion step. A fraction of INPP5E is localized to lysosomes, and its membrane anchoring and enzymatic activity are necessary for autophagy. INPP5E decreases lysosomal phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), one of the substrates of the phosphatase, that counteracts cortactin-mediated actin filament stabilization on lysosomes. Lysosomes require actin filaments on their surface for fusing with autophagosomes. INPP5E is one of the genes responsible for Joubert syndrome, a rare brain abnormality, and mutations found in patients with this disease caused defects in autophagy. Taken together, our data reveal a novel role of phosphoinositide on lysosomes and an association between autophagy and neuronal disease.
Collapse
Affiliation(s)
- Junya Hasegawa
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan Department of Genetics, Graduate School of Medicine Osaka University, Osaka, Japan
| | - Ryo Iwamoto
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan
| | - Takanobu Otomo
- Department of Genetics, Graduate School of Medicine Osaka University, Osaka, Japan
| | - Akiko Nezu
- Department of Genetics, Graduate School of Medicine Osaka University, Osaka, Japan
| | - Maho Hamasaki
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan Department of Genetics, Graduate School of Medicine Osaka University, Osaka, Japan
| | - Tamotsu Yoshimori
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan Department of Genetics, Graduate School of Medicine Osaka University, Osaka, Japan
| |
Collapse
|
49
|
Coxiella burnetii effector CvpB modulates phosphoinositide metabolism for optimal vacuole development. Proc Natl Acad Sci U S A 2016; 113:E3260-9. [PMID: 27226300 DOI: 10.1073/pnas.1522811113] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The Q fever bacterium Coxiella burnetii replicates inside host cells within a large Coxiella-containing vacuole (CCV) whose biogenesis relies on the Dot/Icm-dependent secretion of bacterial effectors. Several membrane trafficking pathways contribute membranes, proteins, and lipids for CCV biogenesis. These include the endocytic and autophagy pathways, which are characterized by phosphatidylinositol 3-phosphate [PI(3)P]-positive membranes. Here we show that the C. burnetii secreted effector Coxiella vacuolar protein B (CvpB) binds PI(3)P and phosphatidylserine (PS) on CCVs and early endosomal compartments and perturbs the activity of the phosphatidylinositol 5-kinase PIKfyve to manipulate PI(3)P metabolism. CvpB association to early endosome triggers vacuolation and clustering, leading to the channeling of large PI(3)P-positive membranes to CCVs for vacuole expansion. At CCVs, CvpB binding to early endosome- and autophagy-derived PI(3)P and the concomitant inhibition of PIKfyve favor the association of the autophagosomal machinery to CCVs for optimal homotypic fusion of the Coxiella-containing compartments. The importance of manipulating PI(3)P metabolism is highlighted by mutations in cvpB resulting in a multivacuolar phenotype, rescuable by gene complementation, indicative of a defect in CCV biogenesis. Using the insect model Galleria mellonella, we demonstrate the in vivo relevance of defective CCV biogenesis by highlighting an attenuated virulence phenotype associated with cvpB mutations.
Collapse
|
50
|
Vacuolin-1 inhibits autophagy by impairing lysosomal maturation via PIKfyve inhibition. FEBS Lett 2016; 590:1576-85. [DOI: 10.1002/1873-3468.12195] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/07/2016] [Accepted: 04/14/2016] [Indexed: 12/29/2022]
|