51
|
Collins MP, Stransky LA, Forgac M. AKT Ser/Thr kinase increases V-ATPase-dependent lysosomal acidification in response to amino acid starvation in mammalian cells. J Biol Chem 2020; 295:9433-9444. [PMID: 32409581 PMCID: PMC7363138 DOI: 10.1074/jbc.ra120.013223] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
The vacuolar H+-ATPase (V-ATPase) is an ATP-dependent proton pump that is essential for cellular homeostasis. V-ATPase activity is controlled by the regulated assembly of the enzyme from its component V1 and V0 domains. We previously reported that amino acid starvation rapidly increases V-ATPase assembly and activity in mammalian lysosomes, but the signaling pathways controlling this effect are unknown. In testing inhibitors of pathways important for controlling cellular metabolism, we found here that the cAMP-dependent protein kinase (PKA) inhibitor H89 increases lysosomal V-ATPase activity and blocks any further change upon starvation. The AMP-activated protein kinase (AMPK) inhibitor dorsomorphin decreased lysosomal V-ATPase activity and also blocked any increase upon starvation. However, CRISPR-mediated gene editing revealed that PKA and AMPK are not required for the starvation-dependent increase in lysosomal V-ATPase activity, indicating that H89 and dorsomorphin modify V-ATPase activity through other cellular targets. We next found that the AKT Ser/Thr kinase (AKT) inhibitor MK2206 blocks the starvation-dependent increase in lysosomal V-ATPase activity without altering basal activity. Expression of AKT1 or AKT3, but not AKT2, was required for increased lysosomal V-ATPase activity in response to amino acid starvation in mouse fibroblasts. Finally, HEK293T cells expressing only AKT1 responded normally to starvation, whereas cells expressing only AKT2 displayed a significantly reduced increase in V-ATPase activity and assembly upon starvation. These results show that AKT is required for controlling the rapid response of lysosomal V-ATPase activity to changes in amino acid availability and that this response depends on specific AKT isoforms.
Collapse
Affiliation(s)
- Michael P Collins
- Program in Cell, Molecular and Developmental Biology, Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
| | - Laura A Stransky
- Program in Cellular and Molecular Physiology, Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
| | - Michael Forgac
- Program in Cell, Molecular and Developmental Biology, Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
- Program in Cellular and Molecular Physiology, Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
52
|
Banerjee S, Kane PM. Regulation of V-ATPase Activity and Organelle pH by Phosphatidylinositol Phosphate Lipids. Front Cell Dev Biol 2020; 8:510. [PMID: 32656214 PMCID: PMC7324685 DOI: 10.3389/fcell.2020.00510] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Luminal pH and the distinctive distribution of phosphatidylinositol phosphate (PIP) lipids are central identifying features of organelles in all eukaryotic cells that are also critical for organelle function. V-ATPases are conserved proton pumps that populate and acidify multiple organelles of the secretory and the endocytic pathway. Complete loss of V-ATPase activity causes embryonic lethality in higher animals and conditional lethality in yeast, while partial loss of V-ATPase function is associated with multiple disease states. On the other hand, many cancer cells increase their virulence by upregulating V-ATPase expression and activity. The pH of individual organelles is tightly controlled and essential for function, but the mechanisms for compartment-specific pH regulation are not completely understood. There is substantial evidence indicating that the PIP content of membranes influences organelle pH. We present recent evidence that PIPs interact directly with subunit isoforms of the V-ATPase to dictate localization of V-ATPase subpopulations and participate in their regulation. In yeast cells, which have only one set of organelle-specific V-ATPase subunit isoforms, the Golgi-enriched lipid PI(4)P binds to the cytosolic domain of the Golgi-enriched a-subunit isoform Stv1, and loss of PI(4)P binding results in mislocalization of Stv1-containing V-ATPases from the Golgi to the vacuole/lysosome. In contrast, levels of the vacuole/lysosome-enriched signaling lipid PI(3,5)P2 affect assembly and activity of V-ATPases containing the Vph1 a-subunit isoform. Mutations in the Vph1 isoform that disrupt the lipid interaction increase sensitivity to stress. These studies have decoded “zip codes” for PIP lipids in the cytosolic N-terminal domain of the a-subunit isoforms of the yeast V-ATPase, and similar interactions between PIP lipids and the V-ATPase subunit isoforms are emerging in higher eukaryotes. In addition to direct effects on the V-ATPase, PIP lipids are also likely to affect organelle pH indirectly, through interactions with other membrane transporters. We discuss direct and indirect effects of PIP lipids on organelle pH, and the functional consequences of the interplay between PIP lipid content and organelle pH.
Collapse
Affiliation(s)
- Subhrajit Banerjee
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Patricia M Kane
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
53
|
Yue S, Wang Z, Wang L, Peng Q, Xue B. Transcriptome Functional Analysis of Mammary Gland of Cows in Heat Stress and Thermoneutral Condition. Animals (Basel) 2020; 10:ani10061015. [PMID: 32532099 PMCID: PMC7341491 DOI: 10.3390/ani10061015] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The current study employed RNA-seq technology to analyze the impact of heat stress on the whole transcript sequencing profile in the mammary glands of lactating Holstein dairy cows. In the findings of the current study, heat stress downregulated the expression of casein genes, which resulted in a decrease in milk production. Moreover, heat stress upregulated the gene expression of HSPA1A and HSP90B1, while it downregulated the expression of immune response-related genes that resulted in a reduction in milk yield. Furthermore, there was an increased synthesis of heat shock proteins and unfolded proteins that could reduce the availability of circulating amino acids for milk protein synthesis. The findings of the current experiment may help to explore the impact of heat stress on immune function, milk production, and milk protein synthesis in cows. Abstract Heat stress (HS) exerts significant effects on the production of dairy animals through impairing health and biological functions. However, the molecular mechanisms related to the effect of HS on dairy cow milk production are still largely unknown. The present study employed an RNA-sequencing approach to explore the molecular mechanisms associated with a decline in milk production by the functional analysis of differentially expressed genes (DEGs) in mammary glands of cows exposed to HS and non-heat-stressed cows. The results of the current study reveal that HS increases the rectal temperature and respiratory rate. Cows under HS result in decreased bodyweight, dry matter intake (DMI), and milk yield. In the current study, a total of 213 genes in experimental cow mammary glands was identified as being differentially expressed by DEGs analysis. Among identified genes, 89 were upregulated, and 124 were downregulated. Gene Ontology functional analysis found that biological processes, such as immune response, chaperone-dependent refolding of protein, and heat shock protein binding activity, were notably affected by HS. The Kyoto Encyclopedia of Genes and Genomes enrichment analysis found that almost all of the top-affected pathways were related to immune response. Under HS, the expression of heat shock protein 90 kDa beta I (HSP90B1) and heat shock 70 kDa protein 1A was upregulated, while the expression of bovine lymphocyte antigen (BoLA) and histocompatibility complex, class II, DRB3 (BoLA-DRB3) was downregulated. We further explored the effects of HS on lactation-related genes and pathways and found that HS significantly downregulated the casein genes. Furthermore, HS increased the expression of phosphorylation of mammalian target of rapamycin, cytosolic arginine sensor for mTORC1 subunit 2 (CASTOR2), and cytosolic arginine sensor for mTORC1 subunit 1 (CASTOR1), but decreased the phosphorylation of Janus kinase-2, a signal transducer and activator of transcription factor-5. Based on the findings of DMI, milk yield, casein gene expression, and the genes and pathways identified by functional annotation analysis, it is concluded that HS adversely affects the immune function of dairy cows. These results will be beneficial to understand the underlying mechanism of reduced milk yield in HS cows.
Collapse
|
54
|
Hayashi H, Wang T, Tanaka M, Ogiwara S, Okada C, Ito M, Fukunishi N, Iida Y, Nakamura A, Sasaki A, Amano S, Yoshida K, Otomo A, Ohtsuka M, Hadano S. Monitoring the autophagy-endolysosomal system using monomeric Keima-fused MAP1LC3B. PLoS One 2020; 15:e0234180. [PMID: 32511278 PMCID: PMC7279612 DOI: 10.1371/journal.pone.0234180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 05/20/2020] [Indexed: 12/20/2022] Open
Abstract
The autophagy-endolysosomal pathway is an evolutionally conserved degradation system that is tightly linked to a wide variety of physiological processes. Dysfunction of this system is associated with many pathological conditions such as cancer, inflammation and neurodegenerative diseases. Therefore, monitoring the cellular autophagy-endolysosomal activity is crucial for studies on the pathogenesis as well as therapeutics of such disorders. To this end, we here sought to create a novel means exploiting Keima, an acid-stable fluorescent protein possessing pH-dependent fluorescence excitation spectra, for precisely monitoring the autophagy-endolysosomal system. First, we generated three lines of transgenic (tg) mouse expressing monomeric Keima-fused MAP1LC3B (mKeima-LC3B). Then, these tg mice were subjected to starvation by food-restriction, and also challenged to neurodegeneration by genetically crossing with a mouse model of amyotrophic lateral sclerosis; i.e., SOD1H46R transgenic mouse. Unexpectedly, despite that a lipidated-form of endogenous LC3 (LC3-II) was significantly increased, those of mKeima-LC3B (mKeima-LC3B-II) were not changed under both stressed conditions. It was also noted that mKeima-LC3B-positive aggregates were progressively accumulated in the spinal cord of SOD1H46R;mKeima-LC3B double-tg mice, suggestive of acid-resistance and aggregate-prone natures of long-term overexpressed mKeima-LC3B in vivo. Next, we characterized mouse embryonic fibroblasts (MEFs) derived from mKeima-LC3B-tg mice. In contrast with in vivo, levels of mKeima-LC3B-I were decreased under starved conditions. Furthermore, when starved MEFs were treated with chloroquine (CQ), the abundance of mKeima-LC3B-II was significantly increased. Remarkably, when cultured medium was repeatedly changed between DMEM (nutrient-rich) and EBSS (starvation), acidic/neutral signal ratios of mKeima-LC3B-positive compartments were rapidly and reversibly shifted, which were suppressed by the CQ treatment, indicating that intraluminal pH of mKeima-LC3B-positive vesicles was changeable upon nutritional conditions of culture media. Taken together, although mKeima-LC3B-tg mice may not be an appropriate tool to monitor the autophagy-endolysosomal system in vivo, mKeima-LC3B must be one of the most sensitive reporter molecules for monitoring this system under in vitro cultured conditions.
Collapse
Affiliation(s)
- Hideki Hayashi
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Ting Wang
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Masayuki Tanaka
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Sanae Ogiwara
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Chisa Okada
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Masatoshi Ito
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Nahoko Fukunishi
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Yumi Iida
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Ayaka Nakamura
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Ayumi Sasaki
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Shunji Amano
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Kazuhiro Yoshida
- Support Center for Medical Research and Education, Isehara Research Promotion Division, Tokai University, Isehara, Kanagawa, Japan
| | - Asako Otomo
- Molecular Neuropathobiology Laboratory, Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- The Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
- Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, Japan
| | - Masato Ohtsuka
- Genetic Engineering and Genome Editing Laboratory, Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Shinji Hadano
- Molecular Neuropathobiology Laboratory, Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- The Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
- Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, Japan
- Research Center for Brain and Nervous Diseases, Tokai University Graduate School of Medicine, Isehara, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
55
|
Collins MP, Forgac M. Regulation and function of V-ATPases in physiology and disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183341. [PMID: 32422136 DOI: 10.1016/j.bbamem.2020.183341] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 02/07/2023]
Abstract
The vacuolar H+-ATPases (V-ATPases) are essential, ATP-dependent proton pumps present in a variety of eukaryotic cellular membranes. Intracellularly, V-ATPase-dependent acidification functions in such processes as membrane traffic, protein degradation, autophagy and the coupled transport of small molecules. V-ATPases at the plasma membrane of certain specialized cells function in such processes as bone resorption, sperm maturation and urinary acidification. V-ATPases also function in disease processes such as pathogen entry and cancer cell invasiveness, while defects in V-ATPase genes are associated with disorders such as osteopetrosis, renal tubular acidosis and neurodegenerative diseases. This review highlights recent advances in our understanding of V-ATPase structure, mechanism, function and regulation, with an emphasis on the signaling pathways controlling V-ATPase assembly in mammalian cells. The role of V-ATPases in cancer and other human pathologies, and the prospects for therapeutic intervention, are also discussed.
Collapse
Affiliation(s)
- Michael P Collins
- Cell, Molecular and Developmental Biology, Tufts University Graduate School of Biomedical Sciences, United States of America
| | - Michael Forgac
- Cell, Molecular and Developmental Biology, Tufts University Graduate School of Biomedical Sciences, United States of America; Dept. of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, United States of America.
| |
Collapse
|
56
|
Song Q, Meng B, Xu H, Mao Z. The emerging roles of vacuolar-type ATPase-dependent Lysosomal acidification in neurodegenerative diseases. Transl Neurodegener 2020; 9:17. [PMID: 32393395 PMCID: PMC7212675 DOI: 10.1186/s40035-020-00196-0] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
Background Lysosomes digest extracellular material from the endocytic pathway and intracellular material from the autophagic pathway. This process is performed by the resident hydrolytic enzymes activated by the highly acidic pH within the lysosomal lumen. Lysosome pH gradients are mainly maintained by the vacuolar (H+) ATPase (or V-ATPase), which pumps protons into lysosomal lumen by consuming ATP. Dysfunction of V-ATPase affects lysosomal acidification, which disrupts the clearance of substrates and leads to many disorders, including neurodegenerative diseases. Main body As a large multi-subunit complex, the V-ATPase is composed of an integral membrane V0 domain involved in proton translocation and a peripheral V1 domain catalyzing ATP hydrolysis. The canonical functions of V-ATPase rely on its H+-pumping ability in multiple vesicle organelles to regulate endocytic traffic, protein processing and degradation, synaptic vesicle loading, and coupled transport. The other non-canonical effects of the V-ATPase that are not readily attributable to its proton-pumping activity include membrane fusion, pH sensing, amino-acid-induced activation of mTORC1, and scaffolding for protein-protein interaction. In response to various stimuli, V-ATPase complex can reversibly dissociate into V1 and V0 domains and thus close ATP-dependent proton transport. Dysregulation of pH and lysosomal dysfunction have been linked to many human diseases, including neurodegenerative disorders such as Alzheimer disease, Parkinson’s disease, amyotrophic lateral sclerosis as well as neurodegenerative lysosomal storage disorders. Conclusion V-ATPase complex is a universal proton pump and plays an important role in lysosome acidification in all types of cells. Since V-ATPase dysfunction contributes to the pathogenesis of multiple neurodegenerative diseases, further understanding the mechanisms that regulate the canonical and non-canonical functions of V-ATPase will reveal molecular details of disease process and help assess V-ATPase or molecules related to its regulation as therapeutic targets.
Collapse
Affiliation(s)
- Qiaoyun Song
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang, Hebei Province, 050051, People's Republic of China.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Bo Meng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Haidong Xu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zixu Mao
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
57
|
Rivera OC, Geddes DT, Barber-Zucker S, Zarivach R, Gagnon A, Soybel DI, Kelleher SL. A common genetic variant in zinc transporter ZnT2 (Thr288Ser) is present in women with low milk volume and alters lysosome function and cell energetics. Am J Physiol Cell Physiol 2020; 318:C1166-C1177. [PMID: 32320289 DOI: 10.1152/ajpcell.00383.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Suboptimal lactation is a common, yet underappreciated cause for early cessation of breastfeeding. Molecular regulation of mammary gland function is critical to the process lactation; however, physiological factors underlying insufficient milk production are poorly understood. The zinc (Zn) transporter ZnT2 is critical for regulation of mammary gland development and maturation during puberty, lactation, and postlactation gland remodeling. Numerous genetic variants in the gene encoding ZnT2 (SLC30A2) are associated with low milk Zn concentration and result in severe Zn deficiency in exclusively breastfed infants. However, the functional impacts of genetic variation in ZnT2 on key mammary epithelial cell functions have not yet been systematically explored at the cellular level. Here we determined a common mutation in SLC30A2/ZnT2 substituting serine for threonine at amino acid 288 (Thr288Ser) was found in 20% of women producing low milk volume (n = 2/10) but was not identified in women producing normal volume. Exploration of cellular consequences in vitro using phosphomimetics showed the serine substitution promoted preferential phosphorylation of ZnT2, driving localization to the lysosome and increasing lysosome biogenesis and acidification. While the substitution did not initiate lysosome-mediated cell death, cellular ATP levels were significantly reduced. Our findings demonstrate the Thr288Ser mutation in SLC30A2/ZnT2 impairs critical functions of mammary epithelial cells and suggest a role for genetic variation in the regulation of milk production and lactation performance.
Collapse
Affiliation(s)
- Olivia C Rivera
- Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, Pennsylvania.,Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania
| | - Donna T Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Shiran Barber-Zucker
- Department of Life Sciences, The National Institute for Biotechnology in the Negev and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Raz Zarivach
- Department of Life Sciences, The National Institute for Biotechnology in the Negev and Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Annie Gagnon
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts
| | - David I Soybel
- Surgery, Penn State Hershey College of Medicine, Hershey, Pennsylvania
| | - Shannon L Kelleher
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts
| |
Collapse
|
58
|
Aasebø E, Berven FS, Bartaula-Brevik S, Stokowy T, Hovland R, Vaudel M, Døskeland SO, McCormack E, Batth TS, Olsen JV, Bruserud Ø, Selheim F, Hernandez-Valladares M. Proteome and Phosphoproteome Changes Associated with Prognosis in Acute Myeloid Leukemia. Cancers (Basel) 2020; 12:cancers12030709. [PMID: 32192169 PMCID: PMC7140113 DOI: 10.3390/cancers12030709] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/05/2020] [Accepted: 03/13/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological cancer that mainly affects the elderly. Although complete remission (CR) is achieved for the majority of the patients after induction and consolidation therapies, nearly two-thirds relapse within a short interval. Understanding biological factors that determine relapse has become of major clinical interest in AML. We utilized liquid chromatography tandem mass spectrometry (LC-MS/MS) to identify the protein changes and protein phosphorylation events associated with AML relapse in primary cells from 41 AML patients at time of diagnosis. Patients were defined as relapse-free if they had not relapsed within a five-year clinical follow-up after AML diagnosis. Relapse was associated with increased expression of RNA processing proteins and decreased expression of V-ATPase proteins. We also observed an increase in phosphorylation events catalyzed by cyclin-dependent kinases (CDKs) and casein kinase 2 (CSK2). The biological relevance of the proteome findings was supported by cell proliferation assays using inhibitors of V-ATPase (bafilomycin), CSK2 (CX-4945), CDK4/6 (abemaciclib) and CDK2/7/9 (SNS-032). While bafilomycin preferentially inhibited the cells from relapse patients, the kinase inhibitors were less efficient in these cells. This suggests that therapy against the upregulated kinases could also target the factors inducing their upregulation rather than their activity. This study, therefore, presents markers that could help predict AML relapse and direct therapeutic strategies.
Collapse
Affiliation(s)
- Elise Aasebø
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (S.B.-B.); (T.S.); (M.V.); (Ø.B.)
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
| | - Frode S. Berven
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
- The Department of Biomedicine, University of Bergen, 5009 Bergen, Norway;
| | - Sushma Bartaula-Brevik
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (S.B.-B.); (T.S.); (M.V.); (Ø.B.)
| | - Tomasz Stokowy
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (S.B.-B.); (T.S.); (M.V.); (Ø.B.)
- Department for Medical Genetics, Haukeland University Hospital, 5021 Bergen, Norway;
| | - Randi Hovland
- Department for Medical Genetics, Haukeland University Hospital, 5021 Bergen, Norway;
- Department of Biological Sciences, University of Bergen, 5006 Bergen, Norway
| | - Marc Vaudel
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (S.B.-B.); (T.S.); (M.V.); (Ø.B.)
| | | | - Emmet McCormack
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway;
| | - Tanveer S. Batth
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200 Copenhagen, Denmark; (T.S.B.); (J.V.O.)
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200 Copenhagen, Denmark; (T.S.B.); (J.V.O.)
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (S.B.-B.); (T.S.); (M.V.); (Ø.B.)
| | - Frode Selheim
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
- The Department of Biomedicine, University of Bergen, 5009 Bergen, Norway;
| | - Maria Hernandez-Valladares
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (E.A.); (S.B.-B.); (T.S.); (M.V.); (Ø.B.)
- The Proteomics Facility of the University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway; (F.S.B.); (F.S.)
- Correspondence: ; Tel.: +47-5558-6368
| |
Collapse
|
59
|
Abstract
Autophagy is a major intracellular degradation system that derives its degradative abilities from the lysosome. The most well-studied form of autophagy is macroautophagy, which delivers cytoplasmic material to lysosomes via the double-membraned autophagosome. Other forms of autophagy, namely chaperone-mediated autophagy and microautophagy, occur directly on the lysosome. Besides providing the means for degradation, lysosomes are also involved in autophagy regulation and can become substrates of autophagy when damaged. During autophagy, they exhibit notable changes, including increased acidification, enhanced enzymatic activity, and perinuclear localization. Despite their importance to autophagy, details on autophagy-specific regulation of lysosomes remain relatively scarce. This review aims to provide a summary of current understanding on the behaviour of lysosomes during autophagy and outline unexplored areas of autophagy-specific lysosome research.
Collapse
Affiliation(s)
- Willa Wen-You Yim
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, 113-0033 Japan
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, 113-0033 Japan
| |
Collapse
|
60
|
Vasanthakumar T, Rubinstein JL. Structure and Roles of V-type ATPases. Trends Biochem Sci 2020; 45:295-307. [PMID: 32001091 DOI: 10.1016/j.tibs.2019.12.007] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/05/2019] [Accepted: 12/31/2019] [Indexed: 12/19/2022]
Abstract
V-ATPases are membrane-embedded protein complexes that function as ATP hydrolysis-driven proton pumps. V-ATPases are the primary source of organellar acidification in all eukaryotes, making them essential for many fundamental cellular processes. Enzymatic activity can be modulated by regulated and reversible disassembly of the complex, and several subunits of mammalian V-ATPase have multiple isoforms that are differentially localized. Although the biochemical properties of the different isoforms are currently unknown, mutations in specific subunit isoforms have been associated with various diseases, making V-ATPases potential drug targets. V-ATPase structure and activity have been best characterized in Saccharomyces cerevisiae, where recent structures have revealed details about the dynamics of the enzyme, the proton translocation pathway, and conformational changes associated with regulated disassembly and autoinhibition.
Collapse
Affiliation(s)
- Thamiya Vasanthakumar
- The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, The University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - John L Rubinstein
- The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, The University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medical Biophysics, The University of Toronto, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
61
|
Zhong B, Liu M, Bai C, Ruan Y, Wang Y, Qiu L, Hong Y, Wang X, Li L, Li B. Caspase-8 Induces Lysosome-Associated Cell Death in Cancer Cells. Mol Ther 2020; 28:1078-1091. [PMID: 32053770 DOI: 10.1016/j.ymthe.2020.01.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/10/2020] [Accepted: 01/10/2020] [Indexed: 12/31/2022] Open
Abstract
Caspase-8, a well-characterized initiator of apoptosis, has also been found to play non-apoptotic roles in cells. In this study, we reveal that caspase-8 can induce cell death in a special way, which does not depend on activation of caspases and mitochondrial initiation. Instead, we prove that caspase-8 can cause lysosomal deacidification and thus lysosomal membrane permeabilization. V-ATPase is a multi-subunit proton pump that acidifies the lumen of lysosome. Our results demonstrate that caspase-8 can bind to the V0 domain of lysosomal Vacuolar H+-ATPase (V-ATPase), but not the V1 domain, to block the assembly of functional V-ATPase and alkalinize lysosomes. We further demonstrate that the C-terminal of caspase-8 is mainly responsible for the interaction with V-ATPase and can suffice to inhibit survival of cancer cells. Interestingly, regardless of the protein level, it is the expression rate of caspase-8 that is the major cause of cell death. Taken together, we identify a previously unrevealed caspase-8-mediated cell death pathway different form typical apoptosis, which could render caspase-8 a particular physiological function and may be potentially applied in treatments for apoptosis-resistant cancers.
Collapse
Affiliation(s)
- Benfu Zhong
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China; Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, P.R. China
| | - Miao Liu
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Changsen Bai
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Yuxia Ruan
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Yuanyuan Wang
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Li Qiu
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Yang Hong
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Xin Wang
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Lifang Li
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China.
| | - Binghui Li
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China; Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, P.R. China.
| |
Collapse
|
62
|
Jaskolka MC, Kane PM. Interaction between the yeast RAVE complex and Vph1-containing V o sectors is a central glucose-sensitive interaction required for V-ATPase reassembly. J Biol Chem 2020; 295:2259-2269. [PMID: 31941791 DOI: 10.1074/jbc.ra119.011522] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/02/2020] [Indexed: 12/12/2022] Open
Abstract
The yeast vacuolar H+-ATPase (V-ATPase) of budding yeast (Saccharomyces cerevisiae) is regulated by reversible disassembly. Disassembly inhibits V-ATPase activity under low-glucose conditions by releasing peripheral V1 subcomplexes from membrane-bound Vo subcomplexes. V-ATPase reassembly and reactivation requires intervention of the conserved regulator of H+-ATPase of vacuoles and endosomes (RAVE) complex, which binds to cytosolic V1 subcomplexes and assists reassembly with integral membrane Vo complexes. Consistent with its role, the RAVE complex itself is reversibly recruited to the vacuolar membrane by glucose, but the requirements for its recruitment are not understood. We demonstrate here that RAVE recruitment to the membrane does not require an interaction with V1 Glucose-dependent RAVE localization to the vacuolar membrane required only intact Vo complexes containing the Vph1 subunit, suggesting that the RAVE-Vo interaction is glucose-dependent. We identified a short conserved sequence in the center of the RAVE subunit Rav1 that is essential for the interaction with Vph1 in vivo and in vitro Mutations in this region resulted in the temperature- and pH-dependent growth phenotype characteristic of ravΔ mutants. However, this region did not account for glucose sensitivity of the Rav1-Vph1 interaction. We quantitated glucose-dependent localization of a GFP-tagged RAVE subunit to the vacuolar membrane in several mutants previously implicated in altering V-ATPase assembly state or glucose-induced assembly. RAVE localization did not correlate with V-ATPase assembly levels reported previously in these mutants, highlighting both the catalytic nature of RAVE's role in V-ATPase assembly and the likelihood of glucose signaling to RAVE independently of V1.
Collapse
Affiliation(s)
- Michael C Jaskolka
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| | - Patricia M Kane
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210.
| |
Collapse
|
63
|
Elingaard-Larsen LO, Rolver MG, Sørensen EE, Pedersen SF. How Reciprocal Interactions Between the Tumor Microenvironment and Ion Transport Proteins Drive Cancer Progression. Rev Physiol Biochem Pharmacol 2020; 182:1-38. [PMID: 32737753 DOI: 10.1007/112_2020_23] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Solid tumors comprise two major components: the cancer cells and the tumor stroma. The stroma is a mixture of cellular and acellular components including fibroblasts, mesenchymal and cancer stem cells, endothelial cells, immune cells, extracellular matrix, and tumor interstitial fluid. The insufficient tumor perfusion and the highly proliferative state and dysregulated metabolism of the cancer cells collectively create a physicochemical microenvironment characterized by altered nutrient concentrations and varying degrees of hypoxia and acidosis. Furthermore, both cancer and stromal cells secrete numerous growth factors, cytokines, and extracellular matrix proteins which further shape the tumor microenvironment (TME), favoring cancer progression.Transport proteins expressed by cancer and stromal cells localize at the interface between the cells and the TME and are in a reciprocal relationship with it, as both sensors and modulators of TME properties. It has been amply demonstrated how acid-base and nutrient transporters of cancer cells enable their growth, presumably by contributing both to the extracellular acidosis and the exchange of metabolic substrates and waste products between cells and TME. However, the TME also impacts other transport proteins important for cancer progression, such as multidrug resistance proteins. In this review, we summarize current knowledge of the cellular and acellular components of solid tumors and their interrelationship with key ion transport proteins. We focus in particular on acid-base transport proteins with known or proposed roles in cancer development, and we discuss their relevance for novel therapeutic strategies.
Collapse
Affiliation(s)
- Line O Elingaard-Larsen
- Translational Type 2 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Michala G Rolver
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Ester E Sørensen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Stine F Pedersen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
64
|
Cui X, Feng R, Wang J, Du C, Pi X, Chen D, Li J, Li H, Zhang J, Zhang J, Mu H, Zhang F, Liu M, Hu Y. Heat shock factor 4 regulates lysosome activity by modulating the αB-crystallin-ATP6V1A-mTOR complex in ocular lens. Biochim Biophys Acta Gen Subj 2019; 1864:129496. [PMID: 31786107 DOI: 10.1016/j.bbagen.2019.129496] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/28/2019] [Accepted: 11/13/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Germline mutations in heat shock factor 4 (HSF4) cause congenital cataracts. Previously, we have shown that HSF4 is involved in regulating lysosomal pH in mouse lens epithelial cell in vitro. However, the underlying mechanism remains unclear. METHODS HSF4-deficient mouse lens epithelial cell lines and zebrafish were used in this study. Immunoblotting and quantitative RT-PCR were used for expression analysis. The protein-protein interactions were tested with GST-pull downs. The lysosomes were fractioned by ultracentrifugation. RESULTS HSF4 deficiency or knock down of αB-crystallin elevates lysosomal pH and increases the ubiquitination and degradation of ATP6V1A by the proteasome. αB-crystallin localizes partially in the lysosome and interacts solely with the ATP6V1A protein of the V1 complex of V-ATPase. Furthermore, αB-crystallin can co-precipitate with mTORC1 and ATP6V1A in GST pull down assays. Inhibition of mTORC1 by rapamycin or siRNA can lead to dissociation of αB-crystallin from the ATP6V1A and mTORC1complex, shortening the half-life of ATP6V1A and increasing the lysosomal pH. Mutation of ATP6V1A/S441A (the predicted mTOR phosphorylation site) reduces its association with αB-crystallin. In the zebrafish model, HSF4 deficiency reduces αB-crystallin expression and elevates the lysosomal pH in lens tissues. CONCLUSION HSF4 regulates lysosomal acidification by controlling the association of αB-crystallin with ATP6V1A and mTOR and regulating ATP6V1A protein stabilization. GENERAL SIGNIFICANCE This study uncovers a novel function of αB-crystallin, demonstrating that αB-crystallin can regulate lysosomal ATP6V1A protein stabilization by complexing to ATP6V1A and mTOR. This highlights a novel mechanism by which HSF4 regulates the proteolytic process of organelles during lens development.
Collapse
Affiliation(s)
- Xiukun Cui
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Ruiping Feng
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Jungai Wang
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Chunxiao Du
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Xiahui Pi
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Danling Chen
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Jing Li
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Hui Li
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Jun Zhang
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Jing Zhang
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China
| | - Hongmei Mu
- Kaifeng Key Lab of Myopia and Cataract, Institute of Eye Disease, Kaifeng Central Hospital, Kaifeng, China
| | - Fengyan Zhang
- Department of Ophthalmology, the First Affiliate Hospital of Zhengzhou University, China
| | - Mugen Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yanzhong Hu
- Joint National Laboratory for Antibody Drug Engineering, Henan International Union Lab of Antibody Medicine, Henan University School of Medicine, Kaifeng, China; Department of Ophthalmology, the First Affiliate Hospital of Zhengzhou University, China; Kaifeng Key Lab of Myopia and Cataract, Institute of Eye Disease, Kaifeng Central Hospital, Kaifeng, China.
| |
Collapse
|
65
|
Peng W, Minakaki G, Nguyen M, Krainc D. Preserving Lysosomal Function in the Aging Brain: Insights from Neurodegeneration. Neurotherapeutics 2019; 16:611-634. [PMID: 31183763 PMCID: PMC6694346 DOI: 10.1007/s13311-019-00742-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lysosomes are acidic, membrane-bound organelles that serve as the primary catabolic compartment of the cell. They are crucial to a variety of cellular processes from nutrient storage to autophagy. Given the diversity of lysosomal functions, it is unsurprising that lysosomes are also emerging as important players in aging. Lysosomal dysfunction is implicated in several aging-related neurodegenerative diseases including Alzheimer's, Parkinson's, amyotrophic lateral sclerosis/frontotemporal dementia, and Huntington's. Although the precise role of lysosomes in the aging brain is not well-elucidated, some insight into their function has been gained from our understanding of the pathophysiology of age-dependent neurodegenerative diseases. Therapeutic strategies targeting lysosomes and autophagic machinery have already been tested in several of these diseases with promising results, suggesting that improving lysosomal function could be similarly beneficial in preserving function in the aging brain.
Collapse
Affiliation(s)
- Wesley Peng
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA
| | - Georgia Minakaki
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA
| | - Maria Nguyen
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA
| | - Dimitri Krainc
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA.
| |
Collapse
|
66
|
Latifkar A, Ling L, Hingorani A, Johansen E, Clement A, Zhang X, Hartman J, Fischbach C, Lin H, Cerione RA, Antonyak MA. Loss of Sirtuin 1 Alters the Secretome of Breast Cancer Cells by Impairing Lysosomal Integrity. Dev Cell 2019; 49:393-408.e7. [PMID: 30982660 DOI: 10.1016/j.devcel.2019.03.011] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/09/2019] [Accepted: 03/13/2019] [Indexed: 12/23/2022]
Abstract
The NAD+-dependent deacetylase Sirtuin 1 (SIRT1) is down-regulated in triple-negative breast cancer. To determine the mechanistic basis by which reduced SIRT1 expression influences processes related to certain aggressive cancers, we examined the consequences of depleting breast cancer cells of SIRT1. We discovered that reducing SIRT1 levels decreased the expression of one particular subunit of the vacuolar-type H+ ATPase (V-ATPase), which is responsible for proper lysosomal acidification and protein degradation. This impairment in lysosomal function caused a reduction in the number of multi-vesicular bodies (MVBs) targeted for lysosomal degradation and resulted in larger MVBs prior to their fusing with the plasma membrane to release their contents. Collectively, these findings help explain how reduced SIRT1 expression, by disrupting lysosomal function and generating a secretome comprising exosomes with unique cargo and soluble hydrolases that degrade the extracellular matrix, can promote processes that increase breast-cancer-cell survival and invasion.
Collapse
Affiliation(s)
- Arash Latifkar
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Lu Ling
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, NY 14853, USA; Kavli Institute at Cornell for Nanoscale Science Cornell University, Ithaca, NY 14853, USA
| | - Amrit Hingorani
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Eric Johansen
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Amdiel Clement
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Xiaoyu Zhang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - John Hartman
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, NY 14853, USA; Kavli Institute at Cornell for Nanoscale Science Cornell University, Ithaca, NY 14853, USA
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA; Howard Hughes Medical Institute, Cornell University, Ithaca, NY 14853, USA
| | - Richard A Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA.
| | - Marc A Antonyak
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
67
|
Sharma S, Oot RA, Khan MM, Wilkens S. Functional reconstitution of vacuolar H +-ATPase from V o proton channel and mutant V 1-ATPase provides insight into the mechanism of reversible disassembly. J Biol Chem 2019; 294:6439-6449. [PMID: 30792311 DOI: 10.1074/jbc.ra119.007577] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/10/2019] [Indexed: 12/12/2022] Open
Abstract
The vacuolar H+-ATPase (V-ATPase; V1Vo-ATPase) is an ATP-dependent proton pump that acidifies subcellular compartments in all eukaryotic organisms. V-ATPase activity is regulated by reversible disassembly into autoinhibited V1-ATPase and Vo proton channel subcomplexes, a process that is poorly understood on the molecular level. V-ATPase is a rotary motor, and recent structural analyses have revealed different rotary states for disassembled V1 and Vo, a mismatch that is likely responsible for their inability to reconstitute into holo V-ATPase in vitro Here, using the model organism Saccharomyces cerevisiae, we show that a key impediment for binding of V1 to Vo is the conformation of the inhibitory C-terminal domain of subunit H (HCT). Using biolayer interferometry and biochemical analyses of purified mutant V1-ATPase and Vo proton channel reconstituted into vacuolar lipid-containing nanodiscs, we further demonstrate that disruption of HCT's V1-binding site facilitates assembly of a functionally coupled and stable V1Vo-ATPase. Unlike WT, this mutant enzyme was resistant to MgATP hydrolysis-induced dissociation, further highlighting HCT's role in the mechanism of V-ATPase regulation. Our findings provide key insight into the molecular events underlying regulation of V-ATPase activity by reversible disassembly.
Collapse
Affiliation(s)
- Stuti Sharma
- From the Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| | - Rebecca A Oot
- From the Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| | - Md Murad Khan
- From the Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| | - Stephan Wilkens
- From the Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| |
Collapse
|
68
|
Hayek SR, Rane HS, Parra KJ. Reciprocal Regulation of V-ATPase and Glycolytic Pathway Elements in Health and Disease. Front Physiol 2019; 10:127. [PMID: 30828305 PMCID: PMC6384264 DOI: 10.3389/fphys.2019.00127] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/31/2019] [Indexed: 12/24/2022] Open
Abstract
The ability of cells to adapt to fluctuations in glucose availability is crucial for their survival and involves the vacuolar proton-translocating ATPase (V-ATPase), a proton pump found in all eukaryotes. V-ATPase hydrolyzes ATP via its V1 domain and uses the energy released to transport protons across membranes via its Vo domain. This activity is critical for pH homeostasis and generation of a membrane potential that drives cellular metabolism. A number of stimuli have been reported to alter V-ATPase assembly in yeast and higher eukaryotes. Glucose flux is one of the strongest and best-characterized regulators of V-ATPase; this review highlights current models explaining how glycolysis and V-ATPase are coordinated in both the Saccharomyces cerevisiae model fungus and in mammalian systems. Glucose-dependent assembly and trafficking of V-ATPase, V-ATPase-dependent modulations in glycolysis, and the recent discovery that glucose signaling through V-ATPase acts as a molecular switch to dictate anabolic versus catabolic metabolism are discussed. Notably, metabolic plasticity and altered glycolytic flux are critical drivers of numerous human pathologies, and the expression and activity of V-ATPase is often altered in disease states or can be pharmacologically manipulated as treatment. This overview will specifically discuss connections between V-ATPase and glycolysis in cancer.
Collapse
Affiliation(s)
- Summer R Hayek
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Hallie S Rane
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Karlett J Parra
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
69
|
Onishi K, Shibutani S, Goto N, Maeda Y, Iwata H. Amino acid starvation accelerates replication of Ibaraki virus. Virus Res 2018; 260:94-101. [PMID: 30503718 DOI: 10.1016/j.virusres.2018.10.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/07/2018] [Accepted: 10/12/2018] [Indexed: 01/01/2023]
Abstract
Ibaraki virus (IBAV) is a strain of epizootic hemorrhagic disease virus 2 that belongs to the genus Orbivirus of the family Reoviridae. IBAV replication is suppressed by the inhibition of autophagy, and since mechanistic target of rapamycin complex 1 (mTORC1) is a key regulator of autophagy, we examined if mTORC1 inhibition by amino acid starvation or mTOR inhibitors (Torin 1 and rapamycin) affects IBAV replication. We found that IBAV replication is significantly enhanced after amino acid starvation of host cells, but not after treatment with mTOR inhibitors, during early stages of viral infection (0-1 hpi). Notably, inhibition of mTORC1 by amino acid starvation was reversible and thus restricted to 0-1 hpi, whereas mTOR inhibitors sustainably suppressed mTORC1 even after the 1-h treatment, suggesting that mTORC1 suppression itself does not affect IBAV replication. To investigate the mechanism of enhanced IBAV replication by amino acid starvation, we examined the endocytic pathway, since IBAV utilizes acidification of endosomes as a trigger for viral replication. Accordingly, we found that amino acid starvation, but not mTOR inhibitors, strongly induced acidification of endosomes/lysosomes and that inhibition of endosomal acidification by bafilomycin A1 effectively blocked enhancement of IBAV replication. Altogether, the inactivation of mTORC1 by amino acid starvation during early stages of infection enhances acidification of endosomes, which in turn enhances IBAV replication.
Collapse
Affiliation(s)
- Keiko Onishi
- Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi, 753-8515, Japan
| | - Shusaku Shibutani
- Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi, 753-8515, Japan
| | - Nanami Goto
- Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi, 753-8515, Japan
| | - Yuki Maeda
- Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi, 753-8515, Japan
| | - Hiroyuki Iwata
- Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi, 753-8515, Japan.
| |
Collapse
|
70
|
Duan X, Yang S, Zhang L, Yang T. V-ATPases and osteoclasts: ambiguous future of V-ATPases inhibitors in osteoporosis. Theranostics 2018; 8:5379-5399. [PMID: 30555553 PMCID: PMC6276090 DOI: 10.7150/thno.28391] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022] Open
Abstract
Vacuolar ATPases (V-ATPases) play a critical role in regulating extracellular acidification of osteoclasts and bone resorption. The deficiencies of subunit a3 and d2 of V-ATPases result in increased bone density in humans and mice. One of the traditional drug design strategies in treating osteoporosis is the use of subunit a3 inhibitor. Recent findings connect subunits H and G1 with decreased bone density. Given the controversial effects of ATPase subunits on bone density, there is a critical need to review the subunits of V-ATPase in osteoclasts and their functions in regulating osteoclasts and bone remodeling. In this review, we comprehensively address the following areas: information about all V-ATPase subunits and their isoforms; summary of V-ATPase subunits associated with human genetic diseases; V-ATPase subunits and osteopetrosis/osteoporosis; screening of all V-ATPase subunits variants in GEFOS data and in-house data; spectrum of V-ATPase subunits during osteoclastogenesis; direct and indirect roles of subunits of V-ATPases in osteoclasts; V-ATPase-associated signaling pathways in osteoclasts; interactions among V-ATPase subunits in osteoclasts; osteoclast-specific V-ATPase inhibitors; perspective of future inhibitors or activators targeting V-ATPase subunits in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Xiaohong Duan
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral Biology, Clinic of Oral Rare and Genetic Diseases, School of Stomatology, the Fourth Military Medical University, 145 West Changle Road, Xi'an 710032, P. R. China
| | - Shaoqing Yang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral Biology, Clinic of Oral Rare and Genetic Diseases, School of Stomatology, the Fourth Military Medical University, 145 West Changle Road, Xi'an 710032, P. R. China
| | - Lei Zhang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, P. R. China
| | - Tielin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, People's Republic of China
| |
Collapse
|
71
|
Rao VK, Zavala G, Deb Roy A, Mains RE, Eipper BA. A pH-sensitive luminal His-cluster promotes interaction of PAM with V-ATPase along the secretory and endocytic pathways of peptidergic cells. J Cell Physiol 2018; 234:8683-8697. [PMID: 30317586 DOI: 10.1002/jcp.27528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/11/2018] [Indexed: 01/18/2023]
Abstract
The biosynthetic and endocytic pathways of secretory cells are characterized by progressive luminal acidification, a process which is crucial for posttranslational modifications and membrane trafficking. This progressive fall in luminal pH is mainly achieved by the vacuolar-type-H+ ATPase (V-ATPase). V-ATPases are large, evolutionarily ancient rotary proton pumps that consist of a peripheral V1 complex, which hydrolyzes ATP, and an integral membrane V0 complex, which transports protons from the cytosol into the lumen. Upon sensing the desired luminal pH, V-ATPase activity is regulated by reversible dissociation of the complex into its V1 and V0 components. Molecular details of how intraluminal pH is sensed and transmitted to the cytosol are not fully understood. Peptidylglycine α-amidating mono-oxygenase (PAM; EC 1.14.17.3), a secretory pathway membrane enzyme which shares similar topology with two V-ATPase accessory proteins (Ac45 and prorenin receptor), has a pH-sensitive luminal linker region. Immunofluorescence and sucrose gradient analysis of peptidergic cells (AtT-20) identified distinct subcellular compartments exhibiting spatial co-occurrence of PAM and V-ATPase. In vitro binding assays demonstrated direct binding of the cytosolic domain of PAM to V1H. Blue native PAGE identified heterogeneous high-molecular weight complexes of PAM and V-ATPase. A PAM-1 mutant (PAM-1/H3A) with altered pH sensitivity had diminished ability to form high-molecular weight complexes. In addition, V-ATPase assembly status was altered in PAM-1/H3A expressing cells. Our analysis of the secretory and endocytic pathways of peptidergic cells supports the hypothesis that PAM serves as a luminal pH-sensor, regulating V-ATPase action by altering its assembly status.
Collapse
Affiliation(s)
- Vishwanatha K Rao
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut
| | - Gerardo Zavala
- Department of Chemistry, University of Texas at El Paso, El Paso, Texas
| | - Abhijit Deb Roy
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, Connecticut
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut
| | - Betty A Eipper
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut.,Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
72
|
Marrone G, De Chiara F, Böttcher K, Levi A, Dhar D, Longato L, Mazza G, Zhang Z, Marrali M, Fernández-Iglesias A, Hall A, Luong TV, Viollet B, Pinzani M, Rombouts K. The adenosine monophosphate-activated protein kinase-vacuolar adenosine triphosphatase-pH axis: A key regulator of the profibrogenic phenotype of human hepatic stellate cells. Hepatology 2018; 68:1140-1153. [PMID: 29663481 DOI: 10.1002/hep.30029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/06/2018] [Accepted: 04/10/2018] [Indexed: 12/18/2022]
Abstract
UNLABELLED Liver fibrosis and cirrhosis are characterized by activation of hepatic stellate cells (HSCs), which is associated with higher intracellular pH (pHi). The vacuolar H+ adenosine-triphosphatase (v-ATPase) multisubunit complex is a key regulator of pHi homeostasis. The present work investigated the functional role of v-ATPase in primary human HSC (hHSC) activation and its modulation by specific adenosine monophosphate-activated protein kinase (AMPK) subunits. We demonstrate that the expression of different v-ATPase subunits was increased in in vivo and in vitro activated hHSCs compared to nonactivated hHSCs. Specific inhibition of v-ATPase with bafilomycin and KM91104 induced a down-regulation of the HSC fibrogenic gene profile, which coincided with increased lysosomal pH, decreased pHi, activation of AMPK, reduced proliferation, and lower metabolic activity. Similarly, pharmacological activation of AMPK by treatment with diflunisal, A769662, and ZLN024 reduced the expression of v-ATPase subunits and profibrogenic markers. v-ATPase expression was differently regulated by the AMPK α1 subunit (AMPKα1) and AMPKα2, as demonstrated in mouse embryo fibroblasts specifically deficient for AMPK α subunits. In addition, activation of v-ATPase in hHSCs was shown to be AMPKα1-dependent. Accordingly, pharmacological activation of AMPK in AMPKα1-depleted hHSCs prevented v-ATPase down-regulation. Finally, we showed that v-ATPase expression was increased in fibrotic livers from bile duct-ligated mice and in human cirrhotic livers. CONCLUSION The down-regulation of v-ATPase might represent a promising target for the development of antifibrotic strategies. (Hepatology 2018).
Collapse
Affiliation(s)
- Giusi Marrone
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| | - Francesco De Chiara
- Liver Failure Group, Institute for Liver & Digestive Health, University College London, Royal Free Hospital, London, UK
| | - Katrin Böttcher
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| | - Ana Levi
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| | - Dipok Dhar
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| | - Lisa Longato
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| | - Giuseppe Mazza
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| | - Zhenzhen Zhang
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| | - Martina Marrali
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| | - Anabel Fernández-Iglesias
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Laboratory, IDIBAPS Biomedical Research Institute-CIBEREHD, Barcelona, Spain
| | - Andrew Hall
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| | - Tu Vinh Luong
- Department of Cellular Pathology, Royal Free Hospital, London, UK
| | - Benoit Viollet
- INSERM, Institut Cochin.,CNRS UMR 8104, Sorbonne Paris cité, Paris, France.,Université Paris Descartes, Sorbonne Paris cité, Paris, France
| | - Massimo Pinzani
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| | - Krista Rombouts
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, London, UK
| |
Collapse
|
73
|
STAT3 associates with vacuolar H +-ATPase and regulates cytosolic and lysosomal pH. Cell Res 2018; 28:996-1012. [PMID: 30127373 PMCID: PMC6170402 DOI: 10.1038/s41422-018-0080-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/28/2018] [Accepted: 07/16/2018] [Indexed: 12/02/2022] Open
Abstract
Dysregulated intracellular pH is emerging as a hallmark of cancer. In spite of their acidic environment and increased acid production, cancer cells maintain alkaline intracellular pH that promotes cancer progression by inhibiting apoptosis and increasing glycolysis, cell growth, migration, and invasion. Here we identify signal transducer and activator of transcription-3 (STAT3) as a key factor in the preservation of alkaline cytosol. STAT3 associates with the vacuolar H+-ATPase in a coiled-coil domain-dependent manner and increases its activity in living cells and in vitro. Accordingly, STAT3 depletion disrupts intracellular proton equilibrium by decreasing cytosolic pH and increasing lysosomal pH, respectively. This dysregulation can be reverted by reconstitution with wild-type STAT3 or STAT3 mutants unable to activate target genes (Tyr705Phe and DNA-binding mutant) or to regulate mitochondrial respiration (Ser727Ala). Upon cytosolic acidification, STAT3 is transcriptionally inactivated and further recruited to lysosomal membranes to reestablish intracellular proton equilibrium. These data reveal STAT3 as a regulator of intracellular pH and, vice versa, intracellular pH as a regulator of STAT3 localization and activity.
Collapse
|
74
|
Whitton B, Okamoto H, Packham G, Crabb SJ. Vacuolar ATPase as a potential therapeutic target and mediator of treatment resistance in cancer. Cancer Med 2018; 7:3800-3811. [PMID: 29926527 PMCID: PMC6089187 DOI: 10.1002/cam4.1594] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 01/10/2023] Open
Abstract
Vacuolar ATPase (V-ATPase) is an ATP-dependent H+ -transporter that pumps protons across intracellular and plasma membranes. It consists of a large multi-subunit protein complex and influences a wide range of cellular processes. This review focuses on emerging evidence for the roles for V-ATPase in cancer. This includes how V-ATPase dysregulation contributes to cancer growth, metastasis, invasion and proliferation, and the potential link between V-ATPase and the development of drug resistance.
Collapse
Affiliation(s)
- Bradleigh Whitton
- Southampton Cancer Research UK CentreUniversity of SouthamptonSouthamptonUK
- Biological SciencesFaculty of Natural and Environmental SciencesUniversity of SouthamptonSouthamptonUK
| | - Haruko Okamoto
- Biological SciencesFaculty of Natural and Environmental SciencesUniversity of SouthamptonSouthamptonUK
| | - Graham Packham
- Southampton Cancer Research UK CentreUniversity of SouthamptonSouthamptonUK
| | - Simon J. Crabb
- Southampton Cancer Research UK CentreUniversity of SouthamptonSouthamptonUK
| |
Collapse
|
75
|
Collins MP, Forgac M. Regulation of V-ATPase Assembly in Nutrient Sensing and Function of V-ATPases in Breast Cancer Metastasis. Front Physiol 2018; 9:902. [PMID: 30057555 PMCID: PMC6053528 DOI: 10.3389/fphys.2018.00902] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 06/21/2018] [Indexed: 12/20/2022] Open
Abstract
V-ATPases are proton pumps that function to acidify intracellular compartments in all eukaryotic cells, and to transport protons across the plasma membrane of certain specialized cells. V-ATPases function in many normal and disease processes, including membrane traffic, protein degradation, pathogen entry, and cancer cell invasion. An important mechanism of regulating V-ATPase activity in vivo is regulated assembly, which is the reversible dissociation of the ATP-hydrolytic V1 domain from the proton-conducting V0 domain. Regulated assembly is highly conserved and occurs in response to various nutrient cues, suggesting that it plays an important role in cellular homeostasis. We have recently found that starvation of mammalian cells for either amino acids or glucose increases V-ATPase assembly on lysosomes, possibly to increase protein degradation (for amino acid homeostasis) or for the utilization of alternative energy sources (during glucose starvation). While regulation of assembly in response to amino acid starvation does not involve PI3K or mTORC1, glucose-regulated assembly involves both PI3K and AMPK. Another important form of V-ATPase regulation is the targeting of the enzyme to different cellular membranes, which is controlled by isoforms of subunit a. We have shown that V-ATPases are localized to the plasma membrane of highly invasive breast cancer cells, where they promote cell migration and invasion. Furthermore, overexpression of the a3 isoform is responsible for plasma membrane targeting of V-ATPases in breast tumor cells leading to their increased invasiveness.
Collapse
Affiliation(s)
- Michael P Collins
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, United States
| | - Michael Forgac
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, United States.,Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, Boston, MA, United States
| |
Collapse
|
76
|
Sharma S, Oot RA, Wilkens S. MgATP hydrolysis destabilizes the interaction between subunit H and yeast V 1-ATPase, highlighting H's role in V-ATPase regulation by reversible disassembly. J Biol Chem 2018; 293:10718-10730. [PMID: 29754144 DOI: 10.1074/jbc.ra118.002951] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/22/2018] [Indexed: 01/01/2023] Open
Abstract
Vacuolar H+-ATPases (V-ATPases; V1Vo-ATPases) are rotary-motor proton pumps that acidify intracellular compartments and, in some tissues, the extracellular space. V-ATPase is regulated by reversible disassembly into autoinhibited V1-ATPase and Vo proton channel sectors. An important player in V-ATPase regulation is subunit H, which binds at the interface of V1 and Vo H is required for MgATPase activity in holo-V-ATPase but also for stabilizing the MgADP-inhibited state in membrane-detached V1 However, how H fulfills these two functions is poorly understood. To characterize the H-V1 interaction and its role in reversible disassembly, we determined binding affinities of full-length H and its N-terminal domain (HNT) for an isolated heterodimer of subunits E and G (EG), the N-terminal domain of subunit a (aNT), and V1 lacking subunit H (V1ΔH). Using isothermal titration calorimetry (ITC) and biolayer interferometry (BLI), we show that HNT binds EG with moderate affinity, that full-length H binds aNT weakly, and that both H and HNT bind V1ΔH with high affinity. We also found that only one molecule of HNT binds V1ΔH with high affinity, suggesting conformational asymmetry of the three EG heterodimers in V1ΔH. Moreover, MgATP hydrolysis-driven conformational changes in V1 destabilized the interaction of H or HNT with V1ΔH, suggesting an interplay between MgADP inhibition and subunit H. Our observation that H binding is affected by MgATP hydrolysis in V1 points to H's role in the mechanism of reversible disassembly.
Collapse
Affiliation(s)
- Stuti Sharma
- From the Department of Biochemistry and Molecular Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York 13210
| | - Rebecca A Oot
- From the Department of Biochemistry and Molecular Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York 13210
| | - Stephan Wilkens
- From the Department of Biochemistry and Molecular Biology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York 13210
| |
Collapse
|
77
|
AMPK Inhibits ULK1-Dependent Autophagosome Formation and Lysosomal Acidification via Distinct Mechanisms. Mol Cell Biol 2018; 38:MCB.00023-18. [PMID: 29507183 PMCID: PMC5954193 DOI: 10.1128/mcb.00023-18] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/16/2018] [Indexed: 11/20/2022] Open
Abstract
Autophagy maintains metabolism in response to starvation, but each nutrient is sensed distinctly. Amino acid deficiency suppresses mechanistic target of rapamycin complex 1 (MTORC1), while glucose deficiency promotes AMP-activated protein kinase (AMPK). The MTORC1 and AMPK signaling pathways converge onto the ULK1/2 autophagy initiation complex. Here, we show that amino acid starvation promoted formation of ULK1- and sequestosome 1/p62-positive early autophagosomes. Autophagosome initiation was controlled by MTORC1 sensing glutamine, leucine, and arginine levels together. In contrast, glucose starvation promoted AMPK activity, phosphorylation of ULK1 Ser555, and LC3-II accumulation, but with dynamics consistent with a block in autophagy flux. We studied the flux pathway and found that starvation of amino acid but not of glucose activated lysosomal acidification, which occurred independently of autophagy and ULK1. In addition to lack of activation, glucose starvation inhibited the ability of amino acid starvation to activate both autophagosome formation and the lysosome. Activation of AMPK and phosphorylation of ULK1 were determined to specifically inhibit autophagosome formation. AMPK activation also was sufficient to prevent lysosome acidification. These results indicate concerted but distinct AMPK-dependent mechanisms to suppress early and late phases of autophagy.
Collapse
|
78
|
McGuire CM, Forgac M. Glucose starvation increases V-ATPase assembly and activity in mammalian cells through AMP kinase and phosphatidylinositide 3-kinase/Akt signaling. J Biol Chem 2018. [PMID: 29540478 DOI: 10.1074/jbc.ra117.001327] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The vacuolar H+-ATPase (V-ATPase) is an ATP-driven proton pump involved in many cellular processes. An important mechanism by which V-ATPase activity is controlled is the reversible assembly of its two domains, namely the peripheral V1 domain and the integral V0 domain. Although reversible assembly is conserved across all eukaryotic organisms, the signaling pathways controlling it have not been fully characterized. Here, we identify glucose starvation as a novel regulator of V-ATPase assembly in mammalian cells. During acute glucose starvation, the V-ATPase undergoes a rapid and reversible increase in assembly and activity as measured by lysosomal acidification. Because the V-ATPase has recently been implicated in the activation of AMP kinase (AMPK), a critical cellular energy sensor that is also activated upon glucose starvation, we compared the time course of AMPK activation and V-ATPase assembly upon glucose starvation. We observe that AMPK activation precedes increased V-ATPase activity. Moreover, the starvation-induced increase in V-ATPase activity and assembly are prevented by the AMPK inhibitor dorsomorphin. These results suggest that increased assembly and activity of the V-ATPase upon glucose starvation are dependent upon AMPK. We also find that the PI3K/Akt pathway, which has previously been implicated in controlling V-ATPase assembly in mammalian cells, also plays a role in the starvation-induced increase in V-ATPase assembly and activity. These studies thus identify a novel stimulus of V-ATPase assembly and a novel signaling pathway involved in regulating this process. The possible function of starvation-induced increase in lysosomal V-ATPase activity is discussed.
Collapse
Affiliation(s)
- Christina M McGuire
- From the Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine and the Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| | - Michael Forgac
- From the Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine and the Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts 02111
| |
Collapse
|
79
|
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) coordinates cellular growth and metabolism with environmental inputs to ensure that cells grow only under favourable conditions. When active, mTORC1 stimulates biosynthetic pathways including protein, lipid and nucleotide synthesis and inhibits cellular catabolism through repression of the autophagic pathway, thereby promoting cell growth and proliferation. The recruitment of mTORC1 to the lysosomal surface has been shown to be essential for its activation. This finding has significantly enhanced our knowledge of mTORC1 regulation and has focused the attention of the field on the lysosome as a signalling hub which coordinates several homeostatic pathways. The intriguing localisation of mTORC1 to the cellular organelle that plays a crucial role in catabolism enables mTORC1 to feedback to autophagy and lysosomal biogenesis, thus leading mTORC1 to enact precise spatial and temporal control of cell growth. This review will cover the signalling interactions which take place on the surface of lysosomes and the cross-talk which exists between mTORC1 activity and lysosomal function.
Collapse
Affiliation(s)
- Yoana Rabanal-Ruiz
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| | - Viktor I Korolchuk
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| |
Collapse
|
80
|
Roh SH, Stam NJ, Hryc CF, Couoh-Cardel S, Pintilie G, Chiu W, Wilkens S. The 3.5-Å CryoEM Structure of Nanodisc-Reconstituted Yeast Vacuolar ATPase V o Proton Channel. Mol Cell 2018. [PMID: 29526695 DOI: 10.1016/j.molcel.2018.02.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The molecular mechanism of transmembrane proton translocation in rotary motor ATPases is not fully understood. Here, we report the 3.5-Å resolution cryoEM structure of the lipid nanodisc-reconstituted Vo proton channel of the yeast vacuolar H+-ATPase, captured in a physiologically relevant, autoinhibited state. The resulting atomic model provides structural detail for the amino acids that constitute the proton pathway at the interface of the proteolipid ring and subunit a. Based on the structure and previous mutagenesis studies, we propose the chemical basis of transmembrane proton transport. Moreover, we discovered that the C terminus of the assembly factor Voa1 is an integral component of mature Vo. Voa1's C-terminal transmembrane α helix is bound inside the proteolipid ring, where it contributes to the stability of the complex. Our structure rationalizes possible mechanisms by which mutations in human Vo can result in disease phenotypes and may thus provide new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Soung-Hun Roh
- Department of Bioengineering and James H. Clark Center, Stanford University, Stanford, CA 94305, USA; Biosciences Division, SLAC National Accelerator Laboratory, Menlo Park, CA 94025, USA
| | - Nicholas J Stam
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Corey F Hryc
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX 77030, USA; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sergio Couoh-Cardel
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Grigore Pintilie
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Wah Chiu
- Department of Bioengineering and James H. Clark Center, Stanford University, Stanford, CA 94305, USA; Biosciences Division, SLAC National Accelerator Laboratory, Menlo Park, CA 94025, USA; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Stephan Wilkens
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
81
|
Paquette M, El-Houjeiri L, Pause A. mTOR Pathways in Cancer and Autophagy. Cancers (Basel) 2018; 10:cancers10010018. [PMID: 29329237 PMCID: PMC5789368 DOI: 10.3390/cancers10010018] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/22/2017] [Accepted: 01/09/2018] [Indexed: 12/11/2022] Open
Abstract
TOR (target of rapamycin), an evolutionarily-conserved serine/threonine kinase, acts as a central regulator of cell growth, proliferation and survival in response to nutritional status, growth factor, and stress signals. It plays a crucial role in coordinating the balance between cell growth and cell death, depending on cellular conditions and needs. As such, TOR has been identified as a key modulator of autophagy for more than a decade, and several deregulations of this pathway have been implicated in a variety of pathological disorders, including cancer. At the molecular level, autophagy regulates several survival or death signaling pathways that may decide the fate of cancer cells; however, the relationship between autophagy pathways and cancer are still nascent. In this review, we discuss the recent cellular signaling pathways regulated by TOR, their interconnections to autophagy, and the clinical implications of TOR inhibitors in cancer.
Collapse
Affiliation(s)
- Mathieu Paquette
- Goodman Cancer Research Center, McGill University, Montréal, QC H3A 1A3, Canada.
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada.
| | - Leeanna El-Houjeiri
- Goodman Cancer Research Center, McGill University, Montréal, QC H3A 1A3, Canada.
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada.
| | - Arnim Pause
- Goodman Cancer Research Center, McGill University, Montréal, QC H3A 1A3, Canada.
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada.
| |
Collapse
|
82
|
Kuchitsu Y, Homma Y, Fujita N, Fukuda M. Rab7 knockout unveiled regulated autolysosome maturation induced by glutamine starvation. J Cell Sci 2018. [DOI: 10.1242/jcs.215442] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Macroautophagy (simply called autophagy hereafter) is an intracellular degradation mechanism that is activated by nutrient starvation. Although it is well-known that starvation induces autophagosome formation in an mTORC1-dependent manner, whether starvation also regulates autophagosome/autolysosome maturation was unclear. In the present study we succeeded in demonstrating that starvation activates autolysosome maturation in mammalian cells. We found that knockout (KO) of Rab7 caused accumulation of a massive number of LC3-positive autolysosomes under nutrient-rich conditions, indicating that Rab7 is dispensable for autophagosome-lysosome fusion. Intriguingly, the autolysosomes that had accumulated in Rab7-KO cells matured and disappeared by starvation for a brief period (∼10 min), and we identified glutamine as an essential nutrient for autolysosome maturation. In contrast, forced mTORC1 inactivation by Torin2 failed to induce autolysosome maturation, suggesting that the process is controlled by an mTORC1-independent mechanism. Since starvation-induced autolysosome maturation was also observed in wild-type cells, the nutrient-starvation-induced maturation of autolysosomes is likely to be a generalized mechanism, the same as starvation-induced autophagosome formation is. Such multistep regulatory mechanisms would enable efficient autophagic flux during starvation.
Collapse
Affiliation(s)
- Yoshihiko Kuchitsu
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Yuta Homma
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Naonobu Fujita
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
83
|
Podinovskaia M, Spang A. The Endosomal Network: Mediators and Regulators of Endosome Maturation. ENDOCYTOSIS AND SIGNALING 2018; 57:1-38. [DOI: 10.1007/978-3-319-96704-2_1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
84
|
mTORC1 as the main gateway to autophagy. Essays Biochem 2017; 61:565-584. [PMID: 29233869 PMCID: PMC5869864 DOI: 10.1042/ebc20170027] [Citation(s) in RCA: 356] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 12/16/2022]
Abstract
Cells and organisms must coordinate their metabolic activity with changes in their environment to ensure their growth only when conditions are favourable. In order to maintain cellular homoeostasis, a tight regulation between the synthesis and degradation of cellular components is essential. At the epicentre of the cellular nutrient sensing is the mechanistic target of rapamycin complex 1 (mTORC1) which connects environmental cues, including nutrient and growth factor availability as well as stress, to metabolic processes in order to preserve cellular homoeostasis. Under nutrient-rich conditions mTORC1 promotes cell growth by stimulating biosynthetic pathways, including synthesis of proteins, lipids and nucleotides, and by inhibiting cellular catabolism through repression of the autophagic pathway. Its close signalling interplay with the energy sensor AMP-activated protein kinase (AMPK) dictates whether the cell actively favours anabolic or catabolic processes. Underlining the role of mTORC1 in the coordination of cellular metabolism, its deregulation is linked to numerous human diseases ranging from metabolic disorders to many cancers. Although mTORC1 can be modulated by a number of different inputs, amino acids represent primordial cues that cannot be compensated for by any other stimuli. The understanding of how amino acids signal to mTORC1 has increased considerably in the last years; however this area of research remains a hot topic in biomedical sciences. The current ideas and models proposed to explain the interrelationship between amino acid sensing, mTORC1 signalling and autophagy is the subject of the present review.
Collapse
|
85
|
Aasebø E, Bartaula-Brevik S, Hernandez-Valladares M, Bruserud Ø. Vacuolar ATPase as a possible therapeutic target in human acute myeloid leukemia. Expert Rev Hematol 2017; 11:13-24. [PMID: 29168399 DOI: 10.1080/17474086.2018.1407239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION V-ATPase is a proton pump expressed both in the membrane of intracellular organelles (e.g. endosomes, lysosomes, Golgi structures) and the plasma membrane. It is an important regulator of organellar functions, intracellular molecular trafficking, intercellular communication and intracellular signaling. It is therefore considered as a possible therapeutic target in the treatment of human malignancies. Areas covered: Relevant publications were identified through literature searches in the PubMed database. We searched for original articles and reviews describing the possible importance of V-ATPase for leukemogenesis and chemosensitivity in human myeloid cells, especially acute myeloid leukemia (AML) cells. Expert commentary: The expression of V-ATPase in the primary human AML cells varies between patients, and high levels are associated with high constitutive release of a wide range of soluble mediators. Several of the molecules included in the V-ATPase interactome may also be important in leukemogenesis and/or development of chemoresistance in human AML. Therapeutic targeting of V-ATPase should therefore be regarded as a possible therapeutic strategy in human AML, but the efficiency of such targeting will probably differ between patients. The possibility of toxicity, especially hematological toxicity and immunosuppression, also has to be clarified.
Collapse
Affiliation(s)
- Elise Aasebø
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,b Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , Bergen , Norway
| | - Sushma Bartaula-Brevik
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway
| | - Maria Hernandez-Valladares
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,b Proteomics Unit (PROBE), Department of Biomedicine , University of Bergen , Bergen , Norway
| | - Øystein Bruserud
- a Section for Hematology, Department of Clinical Science , University of Bergen , Bergen , Norway.,c Department of Medicine , Haukeland University Hospital , Bergen , Norway
| |
Collapse
|
86
|
Naufer A, Hipolito VEB, Ganesan S, Prashar A, Zaremberg V, Botelho RJ, Terebiznik MR. pH of endophagosomes controls association of their membranes with Vps34 and PtdIns(3)P levels. J Cell Biol 2017; 217:329-346. [PMID: 29089378 PMCID: PMC5748975 DOI: 10.1083/jcb.201702179] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 09/03/2017] [Accepted: 09/26/2017] [Indexed: 12/19/2022] Open
Abstract
Specific changes in phospholipid content are a hallmark of the membranes of maturing endosomes and phagosomes, but is it unclear how this is controlled. Naufer et al. now show that acidification of the lumen of endosomes and phagosomes triggers dissociation of the Vps34 lipid kinase from these organelles, which terminates PtdIns(3)P synthesis and signaling. Phagocytosis of filamentous bacteria occurs through tubular phagocytic cups (tPCs) and takes many minutes to engulf these filaments into phagosomes. Contravening the canonical phagocytic pathway, tPCs mature by fusing with endosomes. Using this model, we observed the sequential recruitment of early and late endolysosomal markers to the elongating tPCs. Surprisingly, the regulatory early endosomal lipid phosphatidylinositol-3-phosphate (PtdIns(3)P) persists on tPCs as long as their luminal pH remains neutral. Interestingly, by manipulating cellular pH, we determined that PtdIns(3)P behaves similarly in canonical phagosomes as well as endosomes. We found that this is the product of a pH-based mechanism that induces the dissociation of the Vps34 class III phosphatidylinositol-3-kinase from these organelles as they acidify. The detachment of Vps34 stops the production of PtdIns(3)P, allowing for the turnover of this lipid by PIKfyve. Given that PtdIns(3)P-dependent signaling is important for multiple cellular pathways, this mechanism for pH-dependent regulation of Vps34 could be at the center of many PtdIns(3)P-dependent cellular processes.
Collapse
Affiliation(s)
- Amriya Naufer
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Canada.,Department of Cell and System Biology, University of Toronto Scarborough, Toronto, Canada
| | - Victoria E B Hipolito
- Molecular Science Graduate Program, Ryerson University, Toronto, Canada.,Department of Chemistry and Biology, Ryerson University, Toronto, Canada
| | | | - Akriti Prashar
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Canada.,Department of Cell and System Biology, University of Toronto Scarborough, Toronto, Canada
| | - Vanina Zaremberg
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Roberto J Botelho
- Molecular Science Graduate Program, Ryerson University, Toronto, Canada .,Department of Chemistry and Biology, Ryerson University, Toronto, Canada
| | - Mauricio R Terebiznik
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Canada .,Department of Cell and System Biology, University of Toronto Scarborough, Toronto, Canada
| |
Collapse
|
87
|
Liu J, Wang Y, Li D, Wang Y, Li M, Chen C, Fang X, Chen H, Zhang C. Milk protein synthesis is regulated by T1R1/T1R3, a G protein-coupled taste receptor, through the mTOR pathway in the mouse mammary gland. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201601017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/01/2017] [Accepted: 04/18/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Junqiang Liu
- Institute of Cellular and Molecular Biology; School of Life Sciences; Jiangsu Normal University; Xuzhou Jiangsu China
| | - Yanhong Wang
- Institute of Cellular and Molecular Biology; School of Life Sciences; Jiangsu Normal University; Xuzhou Jiangsu China
| | - Dewei Li
- Institute of Cellular and Molecular Biology; School of Life Sciences; Jiangsu Normal University; Xuzhou Jiangsu China
| | - Yanhuan Wang
- Institute of Cellular and Molecular Biology; School of Life Sciences; Jiangsu Normal University; Xuzhou Jiangsu China
| | - Menglu Li
- Institute of Cellular and Molecular Biology; School of Life Sciences; Jiangsu Normal University; Xuzhou Jiangsu China
| | - Caifa Chen
- The Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province; Jiangsu Normal University; Xuzhou Jiangsu China
| | - Xingtang Fang
- Institute of Cellular and Molecular Biology; School of Life Sciences; Jiangsu Normal University; Xuzhou Jiangsu China
| | - Hong Chen
- Institute of Cellular and Molecular Biology; School of Life Sciences; Jiangsu Normal University; Xuzhou Jiangsu China
| | - Chunlei Zhang
- Institute of Cellular and Molecular Biology; School of Life Sciences; Jiangsu Normal University; Xuzhou Jiangsu China
| |
Collapse
|
88
|
The yeast protein kinase Sch9 adjusts V-ATPase assembly/disassembly to control pH homeostasis and longevity in response to glucose availability. PLoS Genet 2017; 13:e1006835. [PMID: 28604780 PMCID: PMC5484544 DOI: 10.1371/journal.pgen.1006835] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 06/26/2017] [Accepted: 05/23/2017] [Indexed: 11/19/2022] Open
Abstract
The conserved protein kinase Sch9 is a central player in the nutrient-induced signaling network in yeast, although only few of its direct substrates are known. We now provide evidence that Sch9 controls the vacuolar proton pump (V-ATPase) to maintain cellular pH homeostasis and ageing. A synthetic sick phenotype arises when deletion of SCH9 is combined with a dysfunctional V-ATPase, and the lack of Sch9 has a significant impact on cytosolic pH (pHc) homeostasis. Sch9 physically interacts with, and influences glucose-dependent assembly/disassembly of the V-ATPase, thereby integrating input from TORC1. Moreover, we show that the role of Sch9 in regulating ageing is tightly connected with V-ATPase activity and vacuolar acidity. As both Sch9 and the V-ATPase are highly conserved in higher eukaryotes, it will be interesting to further clarify their cooperative action on the cellular processes that influence growth and ageing. The evolutionary conserved TOR complex 1 controls growth in response to the quality and quantity of nutrients such as carbon and amino acids. The protein kinase Sch9 is the main TORC1 effector in yeast. However, only few of its direct targets are known. In this study, we performed a genome-wide screening looking for mutants which require Sch9 function for their survival and growth. In this way, we identified multiple components of the highly conserved vacuolar proton pump (V-ATPase) which mediates the luminal acidification of multiple biosynthetic and endocytic organelles. Besides a genetic interaction, we found Sch9 also physically interacts with the V-ATPase to regulate its assembly state in response to glucose availability and TORC1 activity. Moreover, the interaction with the V-ATPase has consequences for ageing as it allowed Sch9 to control vacuolar pH and thereby trigger either lifespan extension or lifespan shortening. Hence, our results provide insights into the signaling mechanism coupling glucose availability, TORC1 signaling, pH homeostasis and longevity. As both Sch9 and the V-ATPase are highly conserved and implicated in various pathologies, these results offer fertile ground for further research in higher eukaryotes.
Collapse
|
89
|
Rotating with the brakes on and other unresolved features of the vacuolar ATPase. Biochem Soc Trans 2017; 44:851-5. [PMID: 27284051 PMCID: PMC4900747 DOI: 10.1042/bst20160043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Indexed: 12/31/2022]
Abstract
The rotary ATPase family comprises the ATP synthase (F-ATPase), vacuolar ATPase (V-ATPase) and archaeal ATPase (A-ATPase). These either predominantly utilize a proton gradient for ATP synthesis or use ATP to produce a proton gradient, driving secondary transport and acidifying organelles. With advances in EM has come a significant increase in our understanding of the rotary ATPase family. Following the sub nm resolution reconstructions of both the F- and V-ATPases, the secondary structure organization of the elusive subunit a has now been resolved, revealing a novel helical arrangement. Despite these significant developments in our understanding of the rotary ATPases, there are still a number of unresolved questions about the mechanism, regulation and overall architecture, which this mini-review aims to highlight and discuss.
Collapse
|
90
|
Abstract
The vacuolar ATPases (V-ATPases) are a family of proton pumps that couple ATP hydrolysis to proton transport into intracellular compartments and across the plasma membrane. They function in a wide array of normal cellular processes, including membrane traffic, protein processing and degradation, and the coupled transport of small molecules, as well as such physiological processes as urinary acidification and bone resorption. The V-ATPases have also been implicated in a number of disease processes, including viral infection, renal disease, and bone resorption defects. This review is focused on the growing evidence for the important role of V-ATPases in cancer. This includes functions in cellular signaling (particularly Wnt, Notch, and mTOR signaling), cancer cell survival in the highly acidic environment of tumors, aiding the development of drug resistance, as well as crucial roles in tumor cell invasion, migration, and metastasis. Of greatest excitement is evidence that at least some tumors express isoforms of V-ATPase subunits whose disruption is not lethal, leading to the possibility of developing anti-cancer therapeutics that selectively target V-ATPases that function in cancer cells.
Collapse
Affiliation(s)
- Laura Stransky
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Program in Cellular and Molecular Physiology, Program in Biochemistry, and Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts
| | - Kristina Cotter
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Program in Cellular and Molecular Physiology, Program in Biochemistry, and Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts
| | - Michael Forgac
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Program in Cellular and Molecular Physiology, Program in Biochemistry, and Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts
| |
Collapse
|
91
|
Abstract
Beyond protein synthesis and autophagy, emerging evidence has implicated mTORC1 in regulating protein folding and proteasomal degradation as well, highlighting its prominent role in cellular proteome homeostasis or proteostasis. In addition to growth signals, mTORC1 senses and responds to a wide array of stresses, including energetic/metabolic stress, genotoxic stress, oxidative stress, osmotic stress, ER stress, proteotoxic stress, and psychological stress. Whereas growth signals unanimously stimulate mTORC1, stresses exert complex impacts on mTORC1, most of which are repressive. mTORC1 suppression, as a generic adaptive strategy, empowers cell survival under various stressful conditions. In this essay, we provide an overview of the emerging role of mTORC1 in proteostasis, the distinct molecular mechanisms through which mTORC1 reacts to diverse stresses, and the schemes exploited by cancer cells to circumvent stress-induced mTORC1 suppression. Hence, acting as a stress sensor, mTORC1 intimately couples stresses to cellular proteostasis.
Collapse
Affiliation(s)
- Kuo-Hui Su
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Chengkai Dai
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA,Corresponding author: Chengkai Dai,
| |
Collapse
|
92
|
Abstract
Beyond protein synthesis and autophagy, emerging evidence has implicated mTORC1 in regulating protein folding and proteasomal degradation as well, highlighting its prominent role in cellular proteome homeostasis or proteostasis. In addition to growth signals, mTORC1 senses and responds to a wide array of stresses, including energetic/metabolic stress, genotoxic stress, oxidative stress, osmotic stress, ER stress, proteotoxic stress, and psychological stress. Whereas growth signals unanimously stimulate mTORC1, stresses exert complex impacts on mTORC1, most of which are repressive. mTORC1 suppression, as a generic adaptive strategy, empowers cell survival under various stressful conditions. In this essay, we provide an overview of the emerging role of mTORC1 in proteostasis, the distinct molecular mechanisms through which mTORC1 reacts to diverse stresses, and the schemes exploited by cancer cells to circumvent stress-induced mTORC1 suppression. Hence, acting as a stress sensor, mTORC1 intimately couples stresses to cellular proteostasis.
Collapse
Affiliation(s)
- Kuo-Hui Su
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Chengkai Dai
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| |
Collapse
|
93
|
The lysosome: a crucial hub for AMPK and mTORC1 signalling. Biochem J 2017; 474:1453-1466. [PMID: 28408430 DOI: 10.1042/bcj20160780] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 12/18/2022]
Abstract
Much attention has recently been focussed on the lysosome as a signalling hub. Following the initial discovery that localisation of the nutrient-sensitive kinase, mammalian target of rapamycin complex 1 (mTORC1), to the lysosome was essential for mTORC1 activation, the field has rapidly expanded to reveal the role of the lysosome as a platform permitting the co-ordination of several homeostatic signalling pathways. Much is now understood about how the lysosome contributes to amino acid sensing by mTORC1, the involvement of the energy-sensing kinase, AMP-activated protein kinase (AMPK), at the lysosome and how both AMPK and mTORC1 signalling pathways feedback to lysosomal biogenesis and regeneration following autophagy. This review will cover the classical role of the lysosome in autophagy, the dynamic signalling interactions which take place on the lysosomal surface and the multiple levels of cross-talk which exist between lysosomes, AMPK and mTORC1.
Collapse
|
94
|
Oot RA, Couoh-Cardel S, Sharma S, Stam NJ, Wilkens S. Breaking up and making up: The secret life of the vacuolar H + -ATPase. Protein Sci 2017; 26:896-909. [PMID: 28247968 DOI: 10.1002/pro.3147] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 02/21/2017] [Indexed: 01/24/2023]
Abstract
The vacuolar ATPase (V-ATPase; V1 Vo -ATPase) is a large multisubunit proton pump found in the endomembrane system of all eukaryotic cells where it acidifies the lumen of subcellular organelles including lysosomes, endosomes, the Golgi apparatus, and clathrin-coated vesicles. V-ATPase function is essential for pH and ion homeostasis, protein trafficking, endocytosis, mechanistic target of rapamycin (mTOR), and Notch signaling, as well as hormone secretion and neurotransmitter release. V-ATPase can also be found in the plasma membrane of polarized animal cells where its proton pumping function is involved in bone remodeling, urine acidification, and sperm maturation. Aberrant (hypo or hyper) activity has been associated with numerous human diseases and the V-ATPase has therefore been recognized as a potential drug target. Recent progress with moderate to high-resolution structure determination by cryo electron microscopy and X-ray crystallography together with sophisticated single-molecule and biochemical experiments have provided a detailed picture of the structure and unique mode of regulation of the V-ATPase. This review summarizes the recent advances, focusing on the structural and biophysical aspects of the field.
Collapse
Affiliation(s)
- Rebecca A Oot
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, 13210
| | - Sergio Couoh-Cardel
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, 13210
| | - Stuti Sharma
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, 13210
| | - Nicholas J Stam
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, 13210
| | - Stephan Wilkens
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, 13210
| |
Collapse
|
95
|
Sharma S, Wilkens S. Biolayer interferometry of lipid nanodisc-reconstituted yeast vacuolar H + -ATPase. Protein Sci 2017; 26:1070-1079. [PMID: 28241399 DOI: 10.1002/pro.3143] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 01/24/2023]
Abstract
Vacuolar H+ -ATPase (V-ATPase) is a large, multisubunit membrane protein complex responsible for the acidification of subcellular compartments and the extracellular space. V-ATPase activity is regulated by reversible disassembly, resulting in cytosolic V1 -ATPase and membrane-integral V0 proton channel sectors. Reversible disassembly is accompanied by transient interaction with cellular factors and assembly chaperones. Quantifying protein-protein interactions involving membrane proteins, however, is challenging. Here we present a novel method to determine kinetic constants of membrane protein-protein interactions using biolayer interferometry (BLI). Yeast vacuoles are solubilized, vacuolar proteins are reconstituted into lipid nanodiscs with native vacuolar lipids and biotinylated membrane scaffold protein (MSP) followed by affinity purification of nanodisc-reconstituted V-ATPase (V1 V0 ND). We show that V1 V0 ND can be immobilized on streptavidin-coated BLI sensors to quantitate binding of a pathogen derived inhibitor and to measure the kinetics of nucleotide dependent enzyme dissociation.
Collapse
Affiliation(s)
- Stuti Sharma
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University 750 East Adams Street, Syracuse, New York, 13210
| | - Stephan Wilkens
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University 750 East Adams Street, Syracuse, New York, 13210
| |
Collapse
|
96
|
Abstract
For almost all cells, nutrient availability, from glucose to amino acids, dictates their growth or developmental programs. This nutrient availability is closely coupled to the overall intracellular metabolic state of the cell. Therefore, cells have evolved diverse, robust and versatile modules to sense intracellular metabolic states, activate signaling outputs and regulate outcomes to these states. Yet, signaling and metabolism have been viewed as important but separate. This short review attempts to position aspects of intracellular signaling from a metabolic perspective, highlighting how conserved, core principles of metabolic sensing and signaling can emerge from an understanding of metabolic regulation. I briefly explain the nature of metabolic sensors, using the example of the AMP activated protein kinase (AMPK) as an "energy sensing" hub. Subsequently, I explore how specific central metabolites, particularly acetyl-CoA, but also S -adenosyl methionine and SAICAR, can act as signaling molecules. I extensively illustrate the nature of a metabolic signaling hub using the specific example of the Target of Rapamycin Complex 1 (TORC1), and amino acid sensing. A highlight is the emergence of the lysosome/vacuole as a metabolic and signaling hub. Finally, the need to expand our understanding of the intracellular dynamics (in concentration and localization) of several metabolites, and their signaling hubs is emphasized.
Collapse
Affiliation(s)
- Sunil Laxman
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), NCBS Campus, GKVK, Bellary Road, Bangalore 560065, India
| |
Collapse
|
97
|
Eriksson I, Öllinger K, Appelqvist H. Analysis of Lysosomal pH by Flow Cytometry Using FITC-Dextran Loaded Cells. Methods Mol Biol 2017; 1594:179-189. [PMID: 28456983 DOI: 10.1007/978-1-4939-6934-0_11] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The acidic environment of the lysosomal lumen provides an optimal milieu for the acid hydrolases and is also essential for fusion/fission of endo-lysosomal compartments and sorting of cargo. Evidence suggests that maintaining lysosomal acidity is essential to avoid disease. In this chapter, we describe a protocol for analyzing the lysosomal pH in cultured cells using the fluorescent probe fluorescein isothiocyanate (FITC)-dextran together with a dual-emission ratiometric technique suitable for flow cytometry. Fluorescence-labeled dextran is endocytosed and accumulated in the lysosomal compartment. FITC shows a pH-dependent variation in fluorescence when analyzed at maximum emission wavelength and no variation when analyzing at the isosbestic point, thereby the ratio can be used to determine the lysosomal pH. A standard curve is obtained by equilibrating intralysosomal pH with extracellular pH using the ionophore nigericin. The protocol also includes information regarding procedures to induce lysosomal alkalinization and lysosomal membrane permeabilization.
Collapse
Affiliation(s)
- Ida Eriksson
- Experimental Pathology, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden
| | - Karin Öllinger
- Experimental Pathology, Department of Clinical and Experimental Medicine, Linköping University, SE-58185, Linköping, Sweden.
| | - Hanna Appelqvist
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| |
Collapse
|
98
|
Subcellular Trafficking of Mammalian Lysosomal Proteins: An Extended View. Int J Mol Sci 2016; 18:ijms18010047. [PMID: 28036022 PMCID: PMC5297682 DOI: 10.3390/ijms18010047] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/15/2016] [Accepted: 12/18/2016] [Indexed: 01/02/2023] Open
Abstract
Lysosomes clear macromolecules, maintain nutrient and cholesterol homeostasis, participate in tissue repair, and in many other cellular functions. To assume these tasks, lysosomes rely on their large arsenal of acid hydrolases, transmembrane proteins and membrane-associated proteins. It is therefore imperative that, post-synthesis, these proteins are specifically recognized as lysosomal components and are correctly sorted to this organelle through the endosomes. Lysosomal transmembrane proteins contain consensus motifs in their cytosolic regions (tyrosine- or dileucine-based) that serve as sorting signals to the endosomes, whereas most lysosomal acid hydrolases acquire mannose 6-phosphate (Man-6-P) moieties that mediate binding to two membrane receptors with endosomal sorting motifs in their cytosolic tails. These tyrosine- and dileucine-based motifs are tickets for boarding in clathrin-coated carriers that transport their cargo from the trans-Golgi network and plasma membrane to the endosomes. However, increasing evidence points to additional mechanisms participating in the biogenesis of lysosomes. In some cell types, for example, there are alternatives to the Man-6-P receptors for the transport of some acid hydrolases. In addition, several “non-consensus” sorting motifs have been identified, and atypical transport routes to endolysosomes have been brought to light. These “unconventional” or “less known” transport mechanisms are the focus of this review.
Collapse
|
99
|
Abstract
Lysosomes are highly acidic cellular organelles traditionally viewed as sacs of enzymes involved in digesting extracellular or intracellular macromolecules for the regeneration of basic building blocks, cellular housekeeping, or pathogen degradation. Bound by a single lipid bilayer, lysosomes receive their substrates by fusing with endosomes or autophagosomes, or through specialized translocation mechanisms such as chaperone-mediated autophagy or microautophagy. Lysosomes degrade their substrates using up to 60 different soluble hydrolases and release their products either to the cytosol through poorly defined exporting and efflux mechanisms or to the extracellular space by fusing with the plasma membrane. However, it is becoming evident that the role of the lysosome in nutrient homeostasis goes beyond the disposal of waste or the recycling of building blocks. The lysosome is emerging as a signaling hub that can integrate and relay external and internal nutritional information to promote cellular and organismal homeostasis, as well as a major contributor to the processing of energy-dense molecules like glycogen and triglycerides. Here we describe the current knowledge of the nutrient signaling pathways governing lysosomal function, the role of the lysosome in nutrient mobilization, and how lysosomes signal other organelles, distant tissues, and even themselves to ensure energy homeostasis in spite of fluctuations in energy intake. At the same time, we highlight the value of genomics approaches to the past and future discoveries of how the lysosome simultaneously executes and controls cellular homeostasis.
Collapse
Affiliation(s)
- Vinod K Mony
- a Department of Biology , College of Arts and Sciences, University of Virginia , Charlottesville , VA , USA
| | - Shawna Benjamin
- a Department of Biology , College of Arts and Sciences, University of Virginia , Charlottesville , VA , USA.,b Department of Cell Biology , School of Medicine, University of Virginia , Charlottesville , VA , USA
| | - Eyleen J O'Rourke
- a Department of Biology , College of Arts and Sciences, University of Virginia , Charlottesville , VA , USA.,b Department of Cell Biology , School of Medicine, University of Virginia , Charlottesville , VA , USA.,c Robert M. Berne Cardiovascular Research Center, University of Virginia , Charlottesville , VA , USA
| |
Collapse
|
100
|
Colacurcio DJ, Nixon RA. Disorders of lysosomal acidification-The emerging role of v-ATPase in aging and neurodegenerative disease. Ageing Res Rev 2016; 32:75-88. [PMID: 27197071 DOI: 10.1016/j.arr.2016.05.004] [Citation(s) in RCA: 322] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/02/2016] [Accepted: 05/13/2016] [Indexed: 12/21/2022]
Abstract
Autophagy and endocytosis deliver unneeded cellular materials to lysosomes for degradation. Beyond processing cellular waste, lysosomes release metabolites and ions that serve signaling and nutrient sensing roles, linking the functions of the lysosome to various pathways for intracellular metabolism and nutrient homeostasis. Each of these lysosomal behaviors is influenced by the intraluminal pH of the lysosome, which is maintained in the low acidic range by a proton pump, the vacuolar ATPase (v-ATPase). New reports implicate altered v-ATPase activity and lysosomal pH dysregulation in cellular aging, longevity, and adult-onset neurodegenerative diseases, including forms of Parkinson disease and Alzheimer disease. Genetic defects of subunits composing the v-ATPase or v-ATPase-related proteins occur in an increasingly recognized group of familial neurodegenerative diseases. Here, we review the expanding roles of the v-ATPase complex as a platform regulating lysosomal hydrolysis and cellular homeostasis. We discuss the unique vulnerability of neurons to persistent low level lysosomal dysfunction and review recent clinical and experimental studies that link dysfunction of the v-ATPase complex to neurodegenerative diseases across the age spectrum.
Collapse
|