1
|
Sharma S, Chaudhary V. Dissociation of Drosophila Evi-Wg Complex Occurs Post Apical Internalization in the Maturing Acidic Endosomes. Traffic 2024; 25:e12955. [PMID: 39313313 DOI: 10.1111/tra.12955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 06/27/2024] [Accepted: 07/30/2024] [Indexed: 09/25/2024]
Abstract
Signaling pathways activated by secreted Wnt ligands play an essential role in tissue development and the progression of diseases, like cancer. Secretion of the lipid-modified Wnt proteins is tightly regulated by a repertoire of intracellular factors. For instance, a membrane protein, Evi, interacts with the Wnt ligand in the ER, and it is essential for its further trafficking and release in the extracellular space. After dissociating from the Wnt, the Wnt-unbound Evi is recycled back to the ER via Golgi. However, where in this trafficking path Wnt proteins dissociate from Evi remains unclear. Here, we have used the Drosophila wing epithelium to trace the route of the Evi-Wg (Wnt homolog) complex leading up to their separation. In these polarized cells, Wg is first trafficked to the apical surface; however, the secretion of Wg is believed to occurs post-internalization via recycling. Our results show that the Evi-Wg complex is internalized from the apical surface and transported to the retromer-positive endosomes. Furthermore, using antibodies that specifically label the Wnt-unbound Evi, we show that Evi and Wg separation occurs post-internalization in the acidic endosomes. These results refine our understanding of the polarized trafficking of Wg and highlight the importance of Wg endocytosis in its secondary secretion.
Collapse
Affiliation(s)
- Satyam Sharma
- Cell and Developmental Signaling Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh, India
| | - Varun Chaudhary
- Cell and Developmental Signaling Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh, India
| |
Collapse
|
2
|
Jia M, Yue X, Sun W, Zhou Q, Chang C, Gong W, Feng J, Li X, Zhan R, Mo K, Zhang L, Qian Y, Sun Y, Wang A, Zou Y, Chen W, Li Y, Huang L, Yang Y, Zhao Y, Cheng X. ULK1-mediated metabolic reprogramming regulates Vps34 lipid kinase activity by its lactylation. SCIENCE ADVANCES 2023; 9:eadg4993. [PMID: 37267363 PMCID: PMC10413652 DOI: 10.1126/sciadv.adg4993] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 05/01/2023] [Indexed: 06/04/2023]
Abstract
Autophagy and glycolysis are highly conserved biological processes involved in both physiological and pathological cellular programs, but the interplay between these processes is poorly understood. Here, we show that the glycolytic enzyme lactate dehydrogenase A (LDHA) is activated upon UNC-51-like kinase 1 (ULK1) activation under nutrient deprivation. Specifically, ULK1 directly interacts with LDHA, phosphorylates serine-196 when nutrients are scarce and promotes lactate production. Lactate connects autophagy and glycolysis through Vps34 lactylation (at lysine-356 and lysine-781), which is mediated by the acyltransferase KAT5/TIP60. Vps34 lactylation enhances the association of Vps34 with Beclin1, Atg14L, and UVRAG, and then increases Vps34 lipid kinase activity. Vps34 lactylation promotes autophagic flux and endolysosomal trafficking. Vps34 lactylation in skeletal muscle during intense exercise maintains muscle cell homeostasis and correlates with cancer progress by inducing cell autophagy. Together, our findings describe autophagy regulation mechanism and then integrate cell autophagy and glycolysis.
Collapse
Affiliation(s)
- Mengshu Jia
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Xiao Yue
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Weixia Sun
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Qianjun Zhou
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Cheng Chang
- Department of Nuclear Medicine, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Weihua Gong
- Department of Surgery of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310012, China
| | - Jian Feng
- Department of Thoracic Surgery, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xie Li
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Ruonan Zhan
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Kemin Mo
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Lijuan Zhang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Yajie Qian
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Yuying Sun
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Aoxue Wang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yejun Zou
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Weicai Chen
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Yan Li
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Li Huang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Yi Yang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| | - Yuzheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiawei Cheng
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
3
|
Postoak JL, Song W, Yang G, Guo X, Xiao S, Saffold CE, Zhang J, Joyce S, Manley NR, Wu L, Van Kaer L. Thymic epithelial cells require lipid kinase Vps34 for CD4 but not CD8 T cell selection. J Exp Med 2022; 219:e20212554. [PMID: 35997680 PMCID: PMC9402993 DOI: 10.1084/jem.20212554] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 06/22/2022] [Accepted: 08/03/2022] [Indexed: 11/04/2022] Open
Abstract
The generation of a functional, self-tolerant T cell receptor (TCR) repertoire depends on interactions between developing thymocytes and antigen-presenting thymic epithelial cells (TECs). Cortical TECs (cTECs) rely on unique antigen-processing machinery to generate self-peptides specialized for T cell positive selection. In our current study, we focus on the lipid kinase Vps34, which has been implicated in autophagy and endocytic vesicle trafficking. We show that loss of Vps34 in TECs causes profound defects in the positive selection of the CD4 T cell lineage but not the CD8 T cell lineage. Utilizing TCR sequencing, we show that T cell selection in conditional mutants causes altered repertoire properties including reduced clonal sharing. cTECs from mutant mice display an increased abundance of invariant chain intermediates bound to surface MHC class II molecules, indicating altered antigen processing. Collectively, these studies identify lipid kinase Vps34 as an important contributor to the repertoire of selecting ligands processed and presented by TECs to developing CD4 T cells.
Collapse
Affiliation(s)
- J. Luke Postoak
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Wenqiang Song
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Guan Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Xingyi Guo
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN
| | - Shiyun Xiao
- Department of Genetics, University of Georgia, Athens, GA
| | - Cherie E. Saffold
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
- Birmingham Veterans Affairs Medical Center, Birmingham, AL
| | - Sebastian Joyce
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
| | | | - Lan Wu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
4
|
Early Endosomal Vps34-Derived Phosphatidylinositol-3-Phosphate Is Indispensable for the Biogenesis of the Endosomal Recycling Compartment. Cells 2022; 11:cells11060962. [PMID: 35326413 PMCID: PMC8946653 DOI: 10.3390/cells11060962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/17/2022] [Accepted: 03/09/2022] [Indexed: 12/29/2022] Open
Abstract
Phosphatidylinositol-3-phosphate (PI3P), a major identity tag of early endosomes (EEs), provides a platform for the recruitment of numerous cellular proteins containing an FYVE or PX domain that is required for PI3P-dependent maturation of EEs. Most of the PI3P in EEs is generated by the activity of Vps34, a catalytic component of class III phosphatidylinositol-3-phosphate kinase (PI3Ks) complex. In this study, we analyzed the role of Vps34-derived PI3P in the EE recycling circuit of unperturbed cells using VPS34-IN1 (IN1), a highly specific inhibitor of Vps34. IN1-mediated PI3P depletion resulted in the rapid dissociation of recombinant FYVE- and PX-containing PI3P-binding modules and endogenous PI3P-binding proteins, including EEA1 and EE sorting nexins. IN1 treatment triggered the rapid restructuring of EEs into a PI3P-independent functional configuration, and after IN1 washout, EEs were rapidly restored to a PI3P-dependent functional configuration. Analysis of the PI3P-independent configuration showed that the Vps34-derived PI3P is not essential for the pre-EE-associated functions and the fast recycling loop of the EE recycling circuit but contributes to EE maturation toward the degradation circuit, as previously shown in Vps34 knockout and knockdown studies. However, our study shows that Vps34-derived PI3P is also essential for the establishment of the Rab11a-dependent pathway, including recycling cargo sorting in this pathway and membrane flux from EEs to the pericentriolar endosomal recycling compartment (ERC). Rab11a endosomes of PI3P-depleted cells expanded and vacuolized outside the pericentriolar area without the acquisition of internalized transferrin (Tf). These endosomes had high levels of FIP5 and low levels of FIP3, suggesting that their maturation was arrested before the acquisition of FIP3. Consequently, Tf-loaded-, Rab11a/FIP5-, and Rab8a-positive endosomes disappeared from the pericentriolar area, implying that PI3P-associated functions are essential for ERC biogenesis. ERC loss was rapidly reversed after IN1 washout, which coincided with the restoration of FIP3 recruitment to Rab11a-positive endosomes and their dynein-dependent migration to the cell center. Thus, our study shows that Vps34-derived PI3P is indispensable in the recycling circuit to maintain the slow recycling pathway and biogenesis of the ERC.
Collapse
|
5
|
Caglioti C, Palazzetti F, Monarca L, Lobello R, Ceccarini MR, Iannitti RG, Russo R, Ragonese F, Pennetta C, De Luca A, Codini M, Fioretti B. LY294002 Inhibits Intermediate Conductance Calcium-Activated Potassium (KCa3.1) Current in Human Glioblastoma Cells. Front Physiol 2022; 12:790922. [PMID: 35069252 PMCID: PMC8782274 DOI: 10.3389/fphys.2021.790922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Glioblastomas (GBs) are among the most common tumors with high malignancy and invasiveness of the central nervous system. Several alterations in protein kinase and ion channel activity are involved to maintain the malignancy. Among them, phosphatidylinositol 3-kinase (PI3K) activity and intermediate conductance calcium-activated potassium (KCa3.1) current are involved in several aspects of GB biology. By using the electrophysiological approach and noise analysis, we observed that KCa3.1 channel activity is LY294002-sensitive and Wortmannin-resistant in accordance with the involvement of PI3K class IIβ (PI3KC2β). This modulation was observed also during the endogenous activation of KCa3.1 current with histamine. The principal action of PI3KC2β regulation was the reduction of open probability in intracellular free calcium saturating concentration. An explanation based on the “three-gate” model of the KCa3.1 channel by PI3KC2β was proposed. Based on the roles of KCa3.1 and PI3KC2β in GB biology, a therapeutic implication was suggested to prevent chemo- and radioresistance mechanisms.
Collapse
Affiliation(s)
- Concetta Caglioti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy.,Department of Medicine, Perugia Medical School, University of Perugia, Perugia, Italy
| | - Federico Palazzetti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Lorenzo Monarca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy.,Department of Medicine, Perugia Medical School, University of Perugia, Perugia, Italy
| | | | | | | | - Roberta Russo
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Chiara Pennetta
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Antonella De Luca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Michela Codini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| |
Collapse
|
6
|
TFEB phosphorylation on Serine 211 is induced by autophagy in human synovial fibroblasts and by p62/SQSTM1 overexpression in HEK293 cells. Biochem J 2021; 478:3145-3155. [PMID: 34405859 PMCID: PMC8421036 DOI: 10.1042/bcj20210174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 11/17/2022]
Abstract
Autophagy receptor p62/SQSTM1 signals a complex network that links autophagy-lysosomal system to proteasome. Phosphorylation of p62 on Serine 349 (P-Ser349 p62) is involved in a cell protective, antioxidant pathway. We have shown previously that P-Ser349 p62 occurs and is rapidly degraded during human synovial fibroblasts autophagy. In this work we observed that fingolimod (FTY720), used as a medication for multiple sclerosis, induced coordinated expression of p62, P-Ser349 p62 and inhibitory TFEB form, phosphorylated on Serine 211 (P-Ser211 TFEB), in human synovial fibroblasts. These effects were mimicked and potentiated by proteasome inhibitor MG132. In addition, FTY720 induced autophagic flux, LC3B-II up-regulation, Akt phosphorylation inhibition on Serine 473 but down-regulated TFEB, suggesting stalled autophagy. FTY720 decreased cytoplasmic fraction contained TFEB but induced TFEB in nuclear fraction. FTY720-induced P-Ser211 TFEB was mainly found in membrane fraction. Autophagy and VPS34 kinase inhibitor, autophinib, further increased FTY720-induced P-Ser349 p62 but inhibited concomitant expression of P-Ser211 TFEB. These results suggested that P-Ser211 TFEB expression depends on autophagy. Overexpression of GFP tagged TFEB in HEK293 cells showed concomitant expression of its phosphorylated form on Serine 211, that was down-regulated by autophinib. These results suggested that autophagy might be autoregulated through P-Ser211 TFEB as a negative feedback loop. Of interest, overexpression of p62, p62 phosphorylation mimetic (S349E) mutant and phosphorylation deficient mutant (S349A) in HEK293 cells markedly induced P-Ser211 TFEB. These results showed that p62 is involved in regulation of TFEB phosphorylation on Serine 211 but that this involvement does not depend on p62 phosphorylation on Serine 349.
Collapse
|
7
|
Mercer TJ, Ohashi Y, Boeing S, Jefferies HBJ, De Tito S, Flynn H, Tremel S, Zhang W, Wirth M, Frith D, Snijders AP, Williams RL, Tooze SA. Phosphoproteomic identification of ULK substrates reveals VPS15-dependent ULK/VPS34 interplay in the regulation of autophagy. EMBO J 2021; 40:e105985. [PMID: 34121209 PMCID: PMC8280838 DOI: 10.15252/embj.2020105985] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 03/29/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a process through which intracellular cargoes are catabolised inside lysosomes. It involves the formation of autophagosomes initiated by the serine/threonine kinase ULK and class III PI3 kinase VPS34 complexes. Here, unbiased phosphoproteomics screens in mouse embryonic fibroblasts deleted for Ulk1/2 reveal that ULK loss significantly alters the phosphoproteome, with novel high confidence substrates identified including VPS34 complex member VPS15 and AMPK complex subunit PRKAG2. We identify six ULK-dependent phosphorylation sites on VPS15, mutation of which reduces autophagosome formation in cells and VPS34 activity in vitro. Mutation of serine 861, the major VPS15 phosphosite, decreases both autophagy initiation and autophagic flux. Analysis of VPS15 knockout cells reveals two novel ULK-dependent phenotypes downstream of VPS15 removal that can be partially recapitulated by chronic VPS34 inhibition, starvation-independent accumulation of ULK substrates and kinase activity-regulated recruitment of autophagy proteins to ubiquitin-positive structures.
Collapse
Affiliation(s)
| | | | - Stefan Boeing
- Bioinformatics and BiostatisticsThe Francis Crick InstituteLondonUK
| | | | - Stefano De Tito
- Molecular Cell Biology of AutophagyThe Francis Crick InstituteLondonUK
- Institute of Experimental Endocrinology and Oncology (IEOS)National Research CouncilNaplesItaly
| | - Helen Flynn
- Institute of Experimental Endocrinology and Oncology (IEOS)National Research CouncilNaplesItaly
| | | | - Wenxin Zhang
- Molecular Cell Biology of AutophagyThe Francis Crick InstituteLondonUK
| | - Martina Wirth
- Molecular Cell Biology of AutophagyThe Francis Crick InstituteLondonUK
| | - David Frith
- ProteomicsThe Francis Crick InstituteLondonUK
| | | | | | - Sharon A Tooze
- Molecular Cell Biology of AutophagyThe Francis Crick InstituteLondonUK
| |
Collapse
|
8
|
Achzet LM, Davison CJ, Shea M, Sturgeon I, Jackson DA. Oxidative Stress Underlies the Ischemia/Reperfusion-Induced Internalization and Degradation of AMPA Receptors. Int J Mol Sci 2021; 22:E717. [PMID: 33450848 PMCID: PMC7828337 DOI: 10.3390/ijms22020717] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/27/2022] Open
Abstract
Stroke is the fifth leading cause of death annually in the United States. Ischemic stroke occurs when a blood vessel supplying the brain is occluded. The hippocampus is particularly susceptible to AMPA receptor-mediated delayed neuronal death as a result of ischemic/reperfusion injury. AMPA receptors composed of a GluA2 subunit are impermeable to calcium due to a post-transcriptional modification in the channel pore of the GluA2 subunit. GluA2 undergoes internalization and is subsequently degraded following ischemia/reperfusion. The subsequent increase in the expression of GluA2-lacking, Ca2+-permeable AMPARs results in excitotoxicity and eventually delayed neuronal death. Following ischemia/reperfusion, there is increased production of superoxide radicals. This study describes how the internalization and degradation of GluA1 and GluA2 AMPAR subunits following ischemia/reperfusion is mediated through an oxidative stress signaling cascade. U251-MG cells were transiently transfected with fluorescently tagged GluA1 and GluA2, and different Rab proteins to observe AMPAR endocytic trafficking following oxygen glucose-deprivation/reperfusion (OGD/R), an in vitro model for ischemia/reperfusion. Pretreatment with Mn(III)tetrakis(1-methyl-4-pyridyl)porphyrin (MnTMPyP), a superoxide dismutase mimetic, ameliorated the OGD/R-induced, but not agonist-induced, internalization and degradation of GluA1 and GluA2 AMPAR subunits. Specifically, MnTMPyP prevented the increased colocalization of GluA1 and GluA2 with Rab5, an early endosomal marker, and with Rab7, a late endosomal marker, but did not affect the colocalization of GluA1 with Rab11, a marker for recycling endosomes. These data indicate that oxidative stress may play a vital role in AMPAR-mediated cell death following ischemic/reperfusion injury.
Collapse
Affiliation(s)
- Lindsay M. Achzet
- Department of Pharmaceutical Sciences and Molecular Medicine, Washington State University-Health Sciences, Spokane, WA 99201, USA;
| | - Clara J. Davison
- Department of Biomedical Sciences, University of Montana, Missoula, MT 59802, USA; (C.J.D.); (M.S.); (I.S.)
| | - Moira Shea
- Department of Biomedical Sciences, University of Montana, Missoula, MT 59802, USA; (C.J.D.); (M.S.); (I.S.)
| | - Isabella Sturgeon
- Department of Biomedical Sciences, University of Montana, Missoula, MT 59802, USA; (C.J.D.); (M.S.); (I.S.)
| | - Darrell A. Jackson
- Department of Pharmaceutical Sciences and Molecular Medicine, Washington State University-Health Sciences, Spokane, WA 99201, USA;
| |
Collapse
|
9
|
Bertović I, Kurelić R, Milošević I, Bender M, Krauss M, Haucke V, Jurak Begonja A. Vps34 derived phosphatidylinositol 3-monophosphate modulates megakaryocyte maturation and proplatelet production through late endosomes/lysosomes. J Thromb Haemost 2020; 18:1756-1772. [PMID: 32056354 DOI: 10.1111/jth.14764] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/28/2020] [Accepted: 02/07/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Development of platelet precursor cells, megakaryocytes (MKs), implies an increase in their size; formation of the elaborate demarcation membrane system (DMS); and extension of branched cytoplasmic structures, proplatelets, that will release platelets. The membrane source(s) for MK expansion and proplatelet formation have remained elusive. OBJECTIVE We hypothesized that traffic of membranes regulated by phosphatidylinositol 3-monophosphate (PI3P) contributes to MK maturation and proplatelet formation. RESULTS In immature MKs, PI3P produced by the lipid kinase Vps34 is confined to perinuclear early endosomes (EE), while in mature MKs PI3P shifts to late endosomes and lysosomes (LE/Lys). PI3P partially colocalized with the plasma membrane marker phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2 ) and with LE/Lys in mature MKs, suggests that PI3P-containing LE/Lys membranes contribute to MK expansion and proplatelet formation. Consistently, we found that sequestration of PI3P, specific pharmacological inhibition of Vps34-mediated PI3P production, or depletion of PI3P by PI3-phosphatase (MTM1)-mediated hydrolysis potently blocked proplatelet formation. Moreover, Vps34 inhibition led to the intracellular accumulation of enlarged LE/Lys, and decreased expression of surface LE/Lys markers. Inhibiting Vps34 at earlier MK stages caused aberrant DMS development. Finally, inhibition of LE/Lys membrane fusion by a dominant negative mutant of the small GTPase Rab7 or pharmacological inhibition of PI3P conversion into PI(3,5)P2 led to enlarged LE/Lys, reduced surface levels of LE/Lys markers, and decreased proplatelet formation. CONCLUSION Our results suggest that PI3P-positive LE/Lys contribute to the membrane growth and proplatelet formation in MKs by their translocation to the cell periphery and fusion with the plasma membrane.
Collapse
Affiliation(s)
- Ivana Bertović
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Roberta Kurelić
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Ira Milošević
- European Neuroscience Institute (ENI), University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Markus Bender
- Institute of Experimental Biomedicine, University Hospital, and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Michael Krauss
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | | |
Collapse
|
10
|
He F, Nichols RM, Kailasam L, Wensel TG, Agosto MA. Critical Role for Phosphatidylinositol-3 Kinase Vps34/PIK3C3 in ON-Bipolar Cells. Invest Ophthalmol Vis Sci 2019; 60:2861-2874. [PMID: 31260037 PMCID: PMC6607926 DOI: 10.1167/iovs.19-26586] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Purpose Phosphatidylinositol-3-phosphate (PI(3)P), and Vps34, the type III phosphatidylinositol 3-kinase primarily responsible for its production, are important for function and survival of sensory neurons, where they have key roles in membrane processing events, such as autophagy, endosome processing, and fusion of membranes bearing ubiquitinated cargos with lysosomes. We examined their roles in the most abundant class of secondary neurons in the vertebrate retina, the ON-bipolar cells (ON-BCs). Methods A conditional Vps34 knockout mouse line was generated by crossing Vps34 floxed mice with transgenic mice expressing Cre recombinase in ON-BCs. Structural changes in the retina were determined by immunofluorescence and electron microscopy, and bipolar cell function was determined by electroretinography. Results Vps34 deletion led to selective death of ON-BCs, a thinning of the inner nuclear layer, and a progressive decline of electroretinogram b-wave amplitudes. There was no evidence for loss of other retinal neurons, or disruption of rod-horizontal cell contacts in the outer plexiform layer. Loss of Vps34 led to aberrant accumulation of membranes positive for autophagy markers LC3, p62, and ubiquitin, accumulation of endosomal membranes positive for Rab7, and accumulation of lysosomes. Similar effects were observed in Purkinje cells of the cerebellum, leading to severe and progressive ataxia. Conclusions These results support an essential role for PI(3)P in fusion of autophagosomes with lysosomes and in late endosome maturation. The cell death resulting from Vps34 knockout suggests that these processes are essential for the health of ON-BCs.
Collapse
Affiliation(s)
- Feng He
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States
| | - Ralph M Nichols
- Department of Ophthalmology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States
| | - Lavanya Kailasam
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States
| | - Theodore G Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States.,Department of Ophthalmology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, United States
| | - Melina A Agosto
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
11
|
Wei F, Duan Y. Crosstalk between Autophagy and Nanomaterials: Internalization, Activation, Termination. ACTA ACUST UNITED AC 2018; 3:e1800259. [PMID: 32627344 DOI: 10.1002/adbi.201800259] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Nanomaterials (NMs) are comprehensively applied in biomedicine due to their unique physical and chemical properties. Autophagy, as an evolutionarily conserved cellular quality control process, is closely associated with the effect of NMs on cells. In this review, the recent advances in NM-induced/inhibited autophagy (NM-phagy) are summarized, with an aim to present a comprehensive description of the mechanisms of NM-phagy from the perspective of internalization, activation, and termination, thereby bridging autophagy and nanomaterials. Several possible mechanisms are extensively reviewed including the endocytosis pathway of NMs and the related cross components (clathrin and adaptor protein 2 (AP-2), adenosine diphosphate (ADP)-ribosylation factor 6 (Arf6), Rab, UV radiation resistance associated gene (UVRAG)), three main stress mechanisms (oxidative stress, damaged organelles stress, and toxicity stress), and several signal pathway-related molecules. The mechanistic insight is beneficial to understand the autophagic response to NMs or NMs' regulation of autophagy. The challenges currently encountered and research trend in the field of NM-phagy are also highlighted. It is hoped that the NM-phagy discussion in this review with the focus on the mechanistic aspects may serve as a guideline for future research in this field.
Collapse
Affiliation(s)
- Fujing Wei
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-enviroment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P. R. China
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-enviroment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P. R. China
| |
Collapse
|
12
|
Mejlvang J, Olsvik H, Svenning S, Bruun JA, Abudu YP, Larsen KB, Brech A, Hansen TE, Brenne H, Hansen T, Stenmark H, Johansen T. Starvation induces rapid degradation of selective autophagy receptors by endosomal microautophagy. J Cell Biol 2018; 217:3640-3655. [PMID: 30018090 PMCID: PMC6168274 DOI: 10.1083/jcb.201711002] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/20/2018] [Accepted: 06/27/2018] [Indexed: 01/02/2023] Open
Abstract
It is not clear to what extent starvation-induced autophagy affects the proteome on a global scale and whether it is selective. In this study, we report based on quantitative proteomics that cells during the first 4 h of acute starvation elicit lysosomal degradation of up to 2-3% of the proteome. The most significant changes are caused by an immediate autophagic response elicited by shortage of amino acids but executed independently of mechanistic target of rapamycin and macroautophagy. Intriguingly, the autophagy receptors p62/SQSTM1, NBR1, TAX1BP1, NDP52, and NCOA4 are among the most efficiently degraded substrates. Already 1 h after induction of starvation, they are rapidly degraded by a process that selectively delivers autophagy receptors to vesicles inside late endosomes/multivesicular bodies depending on the endosomal sorting complex required for transport III (ESCRT-III). Our data support a model in which amino acid deprivation elicits endocytosis of specific membrane receptors, induction of macroautophagy, and rapid degradation of autophagy receptors by endosomal microautophagy.
Collapse
Affiliation(s)
- Jakob Mejlvang
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Hallvard Olsvik
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Steingrim Svenning
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Jack-Ansgar Bruun
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Yakubu Princely Abudu
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Kenneth Bowitz Larsen
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Andreas Brech
- Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Tom E Hansen
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Hanne Brenne
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Terkel Hansen
- Department of Pharmacy, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Harald Stenmark
- Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
13
|
Filippakis H, Alesi N, Ogorek B, Nijmeh J, Khabibullin D, Gutierrez C, Valvezan AJ, Cunningham J, Priolo C, Henske EP. Lysosomal regulation of cholesterol homeostasis in tuberous sclerosis complex is mediated via NPC1 and LDL-R. Oncotarget 2018; 8:38099-38112. [PMID: 28498820 PMCID: PMC5503518 DOI: 10.18632/oncotarget.17485] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/15/2017] [Indexed: 12/13/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a multisystem disease associated with hyperactive mTORC1. The impact of TSC1/2 deficiency on lysosome-mediated processes is not fully understood. We report here that inhibition of lysosomal function using chloroquine (CQ) upregulates cholesterol homeostasis genes in TSC2-deficient cells. This TSC2-dependent transcriptional signature is associated with increased accumulation and intracellular levels of both total cholesterol and cholesterol esters. Unexpectedly, engaging this CQ-induced cholesterol uptake pathway together with inhibition of de novo cholesterol synthesis allows survival of TSC2-deficient, but not TSC2-expressing cells. The underlying mechanism of TSC2-deficient cell survival is dependent on exogenous cholesterol uptake via LDL-R, and endosomal trafficking mediated by Vps34. Simultaneous inhibition of lysosomal and endosomal trafficking inhibits uptake of esterified cholesterol and cell growth in TSC2-deficient, but not TSC2-expressing cells, highlighting the TSC-dependent lysosome-mediated regulation of cholesterol homeostasis and pointing toward the translational potential of these pathways for the therapy of TSC.
Collapse
Affiliation(s)
- Harilaos Filippakis
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicola Alesi
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Barbara Ogorek
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julie Nijmeh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Damir Khabibullin
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Catherine Gutierrez
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexander J Valvezan
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA, USA
| | - James Cunningham
- Department of Medicine, Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carmen Priolo
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth P Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Law F, Rocheleau CE. Vps34 and the Armus/TBC-2 Rab GAPs: Putting the brakes on the endosomal Rab5 and Rab7 GTPases. CELLULAR LOGISTICS 2017; 7:e1403530. [PMID: 29296513 PMCID: PMC5739090 DOI: 10.1080/21592799.2017.1403530] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/07/2017] [Indexed: 11/30/2022]
Abstract
Rab5 and Rab7 GTPases are key regulators of endosome maturation and lysosome fusion. They activate the class III phosphoinositide 3-kinase (PI3K) Vps34 to generate pools of phosphatidylinositol-3 phosphate [PI(3)P] on endosomes. Together PI(3)P and the GTP-bound Rabs coordinate the recruitment of endosomal regulators to drive early to late endosome maturation and ultimately lysosome fusion. Counterintuitively, loss of Vps34 results in enlarged endosomes, like those seen from expressing activated Rab GTPases. Two recent papers in the Journal of Cell Science, Jaber et al., 2016 and Law, Seo et al., 2017, demonstrate that a function of Vps34 is to inactive the Rab5 and Rab7 GTPases via recruitment of the TBC1D2 family of Rab GTPase Activating Proteins (GAPs).
Collapse
Affiliation(s)
- Fiona Law
- Division of Endocrinology and Metabolism, Department of Medicine and the Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada.,Program in Metabolic Disorders and Complications, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Christian E Rocheleau
- Division of Endocrinology and Metabolism, Department of Medicine and the Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada.,Program in Metabolic Disorders and Complications, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Chen CH, Changou CA, Hsieh TH, Lee YC, Chu CY, Hsu KC, Wang HC, Lin YC, Lo YN, Liu YR, Liou JP, Yen Y. Dual Inhibition of PIK3C3 and FGFR as a New Therapeutic Approach to Treat Bladder Cancer. Clin Cancer Res 2017; 24:1176-1189. [PMID: 29222162 DOI: 10.1158/1078-0432.ccr-17-2066] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/05/2017] [Accepted: 12/04/2017] [Indexed: 11/16/2022]
Abstract
Purpose: MPT0L145 has been developed as a FGFR inhibitor exhibiting significant anti-bladder cancer activity in vitro and in vivo via promoting autophagy-dependent cell death. Here, we aim to elucidate the underlying mechanisms.Experimental Design: Autophagy flux, morphology, and intracellular organelles were evaluated by Western blotting, transmission electron microscope, and fluorescence microscope. Molecular docking and surface plasmon resonance assay were performed to identify drug-protein interaction. Lentiviral delivery of cDNA or shRNA and CRISPR/Cas9-mediated genome editing was used to modulate gene expression. Mitochondrial oxygen consumption rate was measured by a Seahorse XFe24 extracellular flux analyzer, and ROS level was measured by flow cytometry.Results: MPT0L145 persistently increased incomplete autophagy and phase-lucent vacuoles at the perinuclear region, which were identified as enlarged and alkalinized late-endosomes. Screening of a panel of lipid kinases revealed that MPT0L145 strongly inhibits PIK3C3 with a Kd value of 0.53 nmol/L. Ectopic expression of PIK3C3 reversed MPT0L145-increased cell death and incomplete autophagy. Four residues (Y670, F684, I760, D761) at the ATP-binding site of PIK3C3 are important for the binding of MPT0L145. In addition, MPT0L145 promotes mitochondrial dysfunction, ROS production, and DNA damage, which may in part, contribute to cell death. ATG5-knockout rescued MPT0L145-induced cell death, suggesting simultaneous induction of autophagy is crucial to its anticancer activity. Finally, our data demonstrated that MPT0L145 is able to overcome cisplatin resistance in bladder cancer cells.Conclusions: MPT0L145 is a first-in-class PIK3C3/FGFR inhibitor, providing an innovative strategy to design new compounds that increase autophagy, but simultaneously perturb its process to promote bladder cancer cell death. Clin Cancer Res; 24(5); 1176-89. ©2017 AACR.
Collapse
Affiliation(s)
- Chun-Han Chen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun A Changou
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Translational Medicine, Taipei Medical University, Taipei, Taiwan.,Integrated Laboratory, Center of Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsung-Han Hsieh
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ching Lee
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Ying Chu
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kai-Cheng Hsu
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hao-Ching Wang
- Graduate Institute of Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen Lin
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yan-Ni Lo
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yun Yen
- The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
16
|
Ravanan P, Srikumar IF, Talwar P. Autophagy: The spotlight for cellular stress responses. Life Sci 2017; 188:53-67. [PMID: 28866100 DOI: 10.1016/j.lfs.2017.08.029] [Citation(s) in RCA: 459] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/05/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Abstract
Autophagy is an essential cellular mechanism which plays "housekeeping" role in normal physiological processes including removing of long lived, aggregated and misfolded proteins, clearing damaged organelles, growth regulation and aging. Autophagy is also involved in a variety of biological functions like development, cellular differentiation, defense against pathogens and nutritional starvation. The integration of autophagy into these biological functions and other stress responses is determined by the transcriptional factors that undertake the regulatory mechanism. This review discusses the machinery of autophagy, the molecular web that connects autophagy to various stress responses like inflammation, hypoxia, ER stress, and various other pathologic conditions. Defects in autophagy regulation play a central role in number of diseases, including neurodegenerative diseases, cancer, pathogen infection and metabolic diseases. Similarly, inhibiting autophagy would contribute in the treatment of cancer. However, understanding the biology of autophagy regulation requires pharmacologically active compounds which modulate the autophagy process. Inducers of autophagy are currently receiving considerable attention as autophagy upregulation may be a therapeutic benefit for certain neurodegenerative diseases (via removal of protein aggregates) while the inhibitors are being investigated for the treatment of cancers. Both induction and inhibition of autophagy have been proven to be beneficial in the treatment of cancer. This dual role of autophagy in cancers is now getting uncovered by the advancement in the research findings and development of effective autophagy modulators.
Collapse
Affiliation(s)
- Palaniyandi Ravanan
- Apoptosis and Cell Survival Research Laboratory, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu-632014, India.
| | - Ida Florance Srikumar
- Apoptosis and Cell Survival Research Laboratory, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu-632014, India
| | - Priti Talwar
- Apoptosis and Cell Survival Research Laboratory, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu-632014, India
| |
Collapse
|
17
|
Vacuole-inducing compounds that disrupt endolysosomal trafficking stimulate production of exosomes by glioblastoma cells. Mol Cell Biochem 2017; 439:1-9. [PMID: 28770472 DOI: 10.1007/s11010-017-3130-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 07/26/2017] [Indexed: 12/13/2022]
Abstract
Exosomes are produced from mammalian cells when multivesicular endosomes fuse with the plasma membrane, releasing their intralumenal vesicles. In this study we assessed the effects of MOPIPP, a novel indole-based chalcone, and vacuolin-1, a distinct triazine-based compound, on exosome production in cultured glioblastoma and 293T cells. Both compounds promote vacuolization of late endosome compartments and interfere with trafficking of late endosomes to lysosomes, without significant cytotoxicity. The results show that vacuolated cells treated with these compounds release exosomes with morphologies similar to untreated controls. However, both compounds trigger multi-fold increases in release of exosome marker proteins (e.g., CD63, Alix) in exosome fractions collected from equivalent numbers of cells. Despite the marked increase in exosome production, the profiles of selected miRNA cargoes carried by the exosomes were generally similar in cells treated with the compounds. Insofar as MOPIPP and vacuolin-1 seem able to increase the overall yield of exosomes from cultured cells, they might be useful for efforts to develop exosome-based therapeutics.
Collapse
|
18
|
Logan AM, Mammel AE, Robinson DC, Chin AL, Condon AF, Robinson FL. Schwann cell-specific deletion of the endosomal PI 3-kinase Vps34 leads to delayed radial sorting of axons, arrested myelination, and abnormal ErbB2-ErbB3 tyrosine kinase signaling. Glia 2017; 65:1452-1470. [PMID: 28617998 DOI: 10.1002/glia.23173] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 05/04/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022]
Abstract
The PI 3-kinase Vps34 (Pik3c3) synthesizes phosphatidylinositol 3-phosphate (PI3P), a lipid critical for both endosomal membrane traffic and macroautophagy. Human genetics have implicated PI3P dysregulation, and endosomal trafficking in general, as a recurring cause of demyelinating Charcot-Marie-Tooth (CMT) peripheral neuropathy. Here, we investigated the role of Vps34, and PI3P, in mouse Schwann cells by selectively deleting Vps34 in this cell type. Vps34-Schwann cell knockout (Vps34SCKO ) mice show severe hypomyelination in peripheral nerves. Vps34-/- Schwann cells interact abnormally with axons, and there is a delay in radial sorting, a process by which large axons are selected for myelination. Upon reaching the promyelinating stage, Vps34-/- Schwann cells are significantly impaired in the elaboration of myelin. Nerves from Vps34SCKO mice contain elevated levels of the LC3 and p62 proteins, indicating impaired autophagy. However, in the light of recent demonstrations that autophagy is dispensable for myelination, it is unlikely that hypomyelination in Vps34SCKO mice is caused by impaired autophagy. Endosomal trafficking is also disturbed in Vps34-/- Schwann cells. We investigated the activation of the ErbB2/3 receptor tyrosine kinases in Vps34SCKO nerves, as these proteins, which play essential roles in Schwann cell myelination, are known to traffic through endosomes. In Vps34SCKO nerves, ErbB3 was hyperphosphorylated on a tyrosine known to be phosphorylated in response to neuregulin 1 exposure. ErbB2 protein levels were also decreased during myelination. Our findings suggest that the loss of Vps34 alters the trafficking of ErbB2/3 through endosomes. Abnormal ErbB2/3 signaling to downstream targets may contribute to the hypomyelination observed in Vps34SCKO mice.
Collapse
Affiliation(s)
- Anne M Logan
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239.,Neuroscience Graduate Program, Oregon Health and Science University, Portland, Oregon, 97239
| | - Anna E Mammel
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239.,Cell, Developmental and Cancer Biology Graduate Program, Oregon Health and Science University, Portland, Oregon, 97239
| | - Danielle C Robinson
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239.,Neuroscience Graduate Program, Oregon Health and Science University, Portland, Oregon, 97239
| | - Andrea L Chin
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239
| | - Alec F Condon
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239.,Neuroscience Graduate Program, Oregon Health and Science University, Portland, Oregon, 97239
| | - Fred L Robinson
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Mail Code L623, Portland, Oregon, 97239.,Vollum Institute, Oregon Health and Science University, Portland, Oregon, 97239
| |
Collapse
|
19
|
Shi B, Huang QQ, Birkett R, Doyle R, Dorfleutner A, Stehlik C, He C, Pope RM. SNAPIN is critical for lysosomal acidification and autophagosome maturation in macrophages. Autophagy 2016; 13:285-301. [PMID: 27929705 DOI: 10.1080/15548627.2016.1261238] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
We previously observed that SNAPIN, which is an adaptor protein in the SNARE core complex, was highly expressed in rheumatoid arthritis synovial tissue macrophages, but its role in macrophages and autoimmunity is unknown. To identify SNAPIN's role in these cells, we employed siRNA to silence the expression of SNAPIN in primary human macrophages. Silencing SNAPIN resulted in swollen lysosomes with impaired CTSD (cathepsin D) activation, although total CTSD was not reduced. Neither endosome cargo delivery nor lysosomal fusion with endosomes or autophagosomes was inhibited following the forced silencing of SNAPIN. The acidification of lysosomes and accumulation of autolysosomes in SNAPIN-silenced cells was inhibited, resulting in incomplete lysosomal hydrolysis and impaired macroautophagy/autophagy flux. Mechanistic studies employing ratiometric color fluorescence on living cells demonstrated that the reduction of SNAPIN resulted in a modest reduction of H+ pump activity; however, the more critical mechanism was a lysosomal proton leak. Overall, our results demonstrate that SNAPIN is critical in the maintenance of healthy lysosomes and autophagy through its role in lysosome acidification and autophagosome maturation in macrophages largely through preventing proton leak. These observations suggest an important role for SNAPIN and autophagy in the homeostasis of macrophages, particularly long-lived tissue resident macrophages.
Collapse
Affiliation(s)
- Bo Shi
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Qi-Quan Huang
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Robert Birkett
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Renee Doyle
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Andrea Dorfleutner
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Christian Stehlik
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Congcong He
- b Department of Cell and Molecular Biology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Richard M Pope
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| |
Collapse
|
20
|
Fassy F, Dureuil C, Lamberton A, Mathieu M, Michot N, Ronan B, Pasquier B. In Vitro Characterization of VPS34 Lipid Kinase Inhibition by Small Molecules. Methods Enzymol 2016; 587:447-464. [PMID: 28253972 DOI: 10.1016/bs.mie.2016.09.070] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
VPS34 is a class III phosphoinositide 3-kinase that acts on vesicle trafficking. This kinase has recently attracted significant attention because of the function it plays in the machinery involved in the early steps of autophagy. Moreover, because significant progress had been made in the optimization of specific kinase inhibitors, its potential to be targeted by catalytic inhibitors has been investigated by different groups. The aim of this review is to present the key in vitro assays necessary for characterizing inhibitors of the catalytic activity of VPS34. The review covers catalytic (IC50 on purified recombinant protein) and binding assays (KD, ka, kd on purified recombinant protein), and a cell-based assay (IC50 in GFP-FYVE expressing cell line). The methodology for crystallization of VPS34 protein is also presented as it can provide guidance for the design by medicinal chemistry of small molecular mass kinase inhibitor.
Collapse
Affiliation(s)
- F Fassy
- Sanofi, Quai Jules Guesde, Vitry Sur Seine, Paris, France
| | - C Dureuil
- Sanofi, Quai Jules Guesde, Vitry Sur Seine, Paris, France
| | - A Lamberton
- Sanofi, Quai Jules Guesde, Vitry Sur Seine, Paris, France
| | - M Mathieu
- Sanofi, Quai Jules Guesde, Vitry Sur Seine, Paris, France
| | - N Michot
- Sanofi, Quai Jules Guesde, Vitry Sur Seine, Paris, France
| | - B Ronan
- Sanofi, Quai Jules Guesde, Vitry Sur Seine, Paris, France
| | - B Pasquier
- Sanofi, Quai Jules Guesde, Vitry Sur Seine, Paris, France.
| |
Collapse
|
21
|
Bone LN, Dayam RM, Lee M, Kono N, Fairn GD, Arai H, Botelho RJ, Antonescu CN. The acyltransferase LYCAT controls specific phosphoinositides and related membrane traffic. Mol Biol Cell 2016; 28:161-172. [PMID: 28035047 PMCID: PMC5221620 DOI: 10.1091/mbc.e16-09-0668] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 11/18/2022] Open
Abstract
Phosphoinositides (PIPs) control membrane traffic. PIPs have an acyl profile unique among phospholipids. The acyltransferase LYCAT localizes to phosphatidylinositol synthase vesicles, selectively regulates levels and locale of PIPs, and controls related membrane traffic, indicating that dynamic acyl remodeling selectively controls certain PIPs. Phosphoinositides (PIPs) are key regulators of membrane traffic and signaling. The interconversion of PIPs by lipid kinases and phosphatases regulates their functionality. Phosphatidylinositol (PI) and PIPs have a unique enrichment of 1-stearoyl-2-arachidonyl acyl species; however, the regulation and function of this specific acyl profile remains poorly understood. We examined the role of the PI acyltransferase LYCAT in control of PIPs and PIP-dependent membrane traffic. LYCAT silencing selectively perturbed the levels and localization of phosphatidylinositol-4,5-bisphosphate [PI(4,5)P2] and phosphatidylinositol-3-phosphate and the membrane traffic dependent on these specific PIPs but was without effect on phosphatidylinositol-4-phosphate or biosynthetic membrane traffic. The acyl profile of PI(4,5)P2 was selectively altered in LYCAT-deficient cells, whereas LYCAT localized with phosphatidylinositol synthase. We propose that LYCAT remodels the acyl chains of PI, which is then channeled into PI(4,5)P2. Our observations suggest that the PIP acyl chain profile may exert broad control of cell physiology.
Collapse
Affiliation(s)
- Leslie N Bone
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Roya M Dayam
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada.,Graduate Program in Molecular Science, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Minhyoung Lee
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Nozomu Kono
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Gregory D Fairn
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.,Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| | - Hiroyuki Arai
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutionary Science and Technology, Tokyo 113-0033, Japan
| | - Roberto J Botelho
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada .,Graduate Program in Molecular Science, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada .,Graduate Program in Molecular Science, Ryerson University, Toronto, ON M5B 2K3, Canada.,Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| |
Collapse
|
22
|
Mbah NE, Overmeyer JH, Maltese WA. Disruption of endolysosomal trafficking pathways in glioma cells by methuosis-inducing indole-based chalcones. Cell Biol Toxicol 2016; 33:263-282. [PMID: 27822587 DOI: 10.1007/s10565-016-9369-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/26/2016] [Indexed: 12/21/2022]
Abstract
Methuosis is a form of non-apoptotic cell death involving massive vacuolization of macropinosome-derived endocytic compartments, followed by a decline in metabolic activity and loss of membrane integrity. To explore the induction of methuosis as a potential therapeutic strategy for killing cancer cells, we have developed small molecules (indole-based chalcones) that trigger this form of cell death in glioblastoma and other cancer cell lines. Here, we report that in addition to causing fusion and expansion of macropinosome compartments, the lead compound, 3-(5-methoxy-2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propen-1-one (MOMIPP), disrupts vesicular trafficking at the lysosomal nexus, manifested by impaired degradation of EGF and LDL receptors, defective processing of procathepsins, and accumulation of autophagosomes. In contrast, secretion of the ectodomain derived from a prototypical type-I membrane glycoprotein, β-amyloid precursor protein, is increased rather than diminished. A closely related MOMIPP analog, which causes substantial vacuolization without reducing cell viability, also impedes cathepsin processing and autophagic flux, but has more modest effects on receptor degradation. A third analog, which causes neither vacuolization nor loss of viability, has no effect on endolysosomal trafficking. The results suggest that differential cytotoxicity of structurally similar indole-based chalcones is related, at least in part, to the severity of their effects on endolysosomal trafficking pathways.
Collapse
Affiliation(s)
- Nneka E Mbah
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA
| | - Jean H Overmeyer
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA
| | - William A Maltese
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA.
| |
Collapse
|
23
|
Jaber N, Mohd-Naim N, Wang Z, DeLeon JL, Kim S, Zhong H, Sheshadri N, Dou Z, Edinger AL, Du G, Braga VMM, Zong WX. Vps34 regulates Rab7 and late endocytic trafficking through recruitment of the GTPase-activating protein Armus. J Cell Sci 2016; 129:4424-4435. [PMID: 27793976 DOI: 10.1242/jcs.192260] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 10/21/2016] [Indexed: 01/03/2023] Open
Abstract
The class III phosphoinositide 3-kinase (PI3K) Vps34 (also known as PIK3C3 in mammals) produces phosphatidylinositol 3-phosphate [PI(3)P] on both early and late endosome membranes to control membrane dynamics. We used Vps34-deficient cells to delineate whether Vps34 has additional roles in endocytic trafficking. In Vps34-/- mouse embryonic fibroblasts (MEFs), transferrin recycling and EEA1 membrane localization were unaffected despite elevated Rab5-GTP levels. Strikingly, a large increase in Rab7-GTP levels, an accumulation of enlarged late endosomes, and decreased EGFR degradation were observed in Vps34-deficient cells. The hyperactivation of Rab7 in Vps34-deficient cells stemmed from the failure to recruit the Rab7 GTPase-activating protein (GAP) Armus (also known as TBC1D2), which binds to PI(3)P, to late endosomes. Protein-lipid overlay and liposome-binding assays reveal that the putative pleckstrin homology (PH) domain in Armus can directly bind to PI(3)P. Elevated Rab7-GTP led to the failure of intraluminal vesicle (ILV) formation and lysosomal maturation. Rab7 silencing and Armus overexpression alleviated the vacuolization seen in Vps34-deficient cells. Taken together, these results demonstrate that Vps34 has a previously unknown role in regulating Rab7 activity and late endosomal trafficking.
Collapse
Affiliation(s)
- Nadia Jaber
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY11794, USA
| | - Noor Mohd-Naim
- Molecular Medicine, NHLI, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Ziqing Wang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jennifer L DeLeon
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY11794, USA
| | - Seong Kim
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Hua Zhong
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, 164 Frelinghuysen Road, Piscataway NJ08854, USA
| | - Namratha Sheshadri
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY11794, USA
| | - Zhixun Dou
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY11794, USA
| | - Aimee L Edinger
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Guangwei Du
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Vania M M Braga
- Molecular Medicine, NHLI, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Wei-Xing Zong
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY11794, USA .,Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, 164 Frelinghuysen Road, Piscataway NJ08854, USA.,Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick NJ08903, USA
| |
Collapse
|
24
|
Compton LM, Ikonomov OC, Sbrissa D, Garg P, Shisheva A. Active vacuolar H+ ATPase and functional cycle of Rab5 are required for the vacuolation defect triggered by PtdIns(3,5)P2 loss under PIKfyve or Vps34 deficiency. Am J Physiol Cell Physiol 2016; 311:C366-77. [PMID: 27335171 DOI: 10.1152/ajpcell.00104.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/17/2016] [Indexed: 01/12/2023]
Abstract
The two evolutionarily conserved mammalian lipid kinases Vps34 and PIKfyve are involved in an important physiological relationship, whereby the former produces phosphatidylinositol (PtdIns) 3P that is used as a substrate for PtdIns(3,5)P2 synthesis by the latter. Reduced production of PtdIns(3,5)P2 in proliferating mammalian cells is phenotypically manifested by the formation of multiple translucent cytoplasmic vacuoles, readily rescued upon exogenous delivery of PtdIns(3,5)P2 or overproduction of PIKfyve. Although the aberrant vacuolation phenomenon has been frequently used as a sensitive functional measure of localized PtdIns(3,5)P2 reduction, cellular factors governing the appearance of cytoplasmic vacuoles under PtdIns3P-PtdIns(3,5)P2 loss remain elusive. To gain further mechanistic insight about the vacuolation process following PtdIns(3,5)P2 reduction, in this study we sought for cellular mechanisms required for manifestation of the aberrant endomembrane vacuoles triggered by PIKfyve or Vps34 dysfunction. The latter was achieved by various means such as pharmacological inhibition, gene disruption, or dominant-interference in several proliferating mammalian cell types. We report here that inhibition of V-ATPase with bafilomycin A1 as well as inactivation of the GTP-GDP cycle of Rab5a GTPase phenotypically rescued or completely precluded the cytoplasmic vacuolization despite the continued presence of inactivated PIKfyve or Vps34. Bafilomycin A1 also restored the aberrant EEA1-positive endosomes, enlarged upon short PIKfyve inhibition with YM201636. Together, our work identifies for the first time that factors such as active V-ATPase or functional Rab5a cycle are acting coincidentally with the PtdIns(3,5)P2 reduction in triggering formation of aberrant cytoplasmic vacuoles under PIKfyve or Vps34 dysfunction.
Collapse
Affiliation(s)
- Lauren M Compton
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan; and
| | - Ognian C Ikonomov
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan; and
| | - Diego Sbrissa
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan; and
| | - Puneet Garg
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Assia Shisheva
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan; and
| |
Collapse
|
25
|
Abstract
Autophagy is a lysosome-dependent mechanism of intracellular degradation. The cellular and molecular mechanisms underlying this process are highly complex and involve multiple proteins, including the kinases ULK1 and Vps34. The main function of autophagy is the maintenance of cell survival when modifications occur in the cellular environment. During the past decade, extensive studies have greatly improved our knowledge and autophagy has exploded as a research field. This process is now widely implicated in pathophysiological processes such as cancer, metabolic, and neurodegenerative disorders, making it an attractive target for drug discovery. In this review, we will summarize the different types of inhibitors that affect the autophagy machinery and provide some potential therapeutic perspectives.
Collapse
Affiliation(s)
- Benoit Pasquier
- Sanofi, 13, Quai Jules Guesde, 94403, Vitry Sur Seine, France.
| |
Collapse
|
26
|
Posor Y, Eichhorn-Grünig M, Haucke V. Phosphoinositides in endocytosis. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:794-804. [DOI: 10.1016/j.bbalip.2014.09.014] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 08/21/2014] [Accepted: 09/17/2014] [Indexed: 02/04/2023]
|
27
|
Naguib A, Bencze G, Cho H, Zheng W, Tocilj A, Elkayam E, Faehnle CR, Jaber N, Pratt CP, Chen M, Zong WX, Marks MS, Joshua-Tor L, Pappin DJ, Trotman LC. PTEN functions by recruitment to cytoplasmic vesicles. Mol Cell 2015; 58:255-68. [PMID: 25866245 DOI: 10.1016/j.molcel.2015.03.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/18/2015] [Accepted: 03/05/2015] [Indexed: 12/21/2022]
Abstract
PTEN is proposed to function at the plasma membrane, where receptor tyrosine kinases are activated. However, the majority of PTEN is located throughout the cytoplasm. Here, we show that cytoplasmic PTEN is distributed along microtubules, tethered to vesicles via phosphatidylinositol 3-phosphate (PI(3)P), the signature lipid of endosomes. We demonstrate that the non-catalytic C2 domain of PTEN specifically binds PI(3)P through the CBR3 loop. Mutations render this loop incapable of PI(3)P binding and abrogate PTEN-mediated inhibition of PI 3-kinase/AKT signaling. This loss of function is rescued by fusion of the loop mutant PTEN to FYVE, the canonical PI(3)P binding domain, demonstrating the functional importance of targeting PTEN to endosomal membranes. Beyond revealing an upstream activation mechanism of PTEN, our data introduce the concept of PI 3-kinase signal activation on the vast plasma membrane that is contrasted by PTEN-mediated signal termination on the small, discrete surfaces of internalized vesicles.
Collapse
Affiliation(s)
- Adam Naguib
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Gyula Bencze
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Hyejin Cho
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Wu Zheng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Ante Tocilj
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; W. M. Keck Structural Biology Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Elad Elkayam
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; W. M. Keck Structural Biology Laboratory, Cold Spring Harbor, NY 11724, USA; Howard Hughes Medical Institute, Cold Spring Harbor, NY 11724, USA
| | - Christopher R Faehnle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; W. M. Keck Structural Biology Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Nadia Jaber
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY 11794, USA
| | | | - Muhan Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Wei-Xing Zong
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Michael S Marks
- Department of Pathology & Laboratory of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology & Laboratory of Medicine and Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leemor Joshua-Tor
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; W. M. Keck Structural Biology Laboratory, Cold Spring Harbor, NY 11724, USA; Howard Hughes Medical Institute, Cold Spring Harbor, NY 11724, USA
| | - Darryl J Pappin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Lloyd C Trotman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
28
|
Phosphatidylinositol 4-phosphate and phosphatidylinositol 3-phosphate regulate phagolysosome biogenesis. Proc Natl Acad Sci U S A 2015; 112:4636-41. [PMID: 25825728 DOI: 10.1073/pnas.1423456112] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Professional phagocytic cells ingest microbial intruders by engulfing them into phagosomes, which subsequently mature into microbicidal phagolysosomes. Phagosome maturation requires sequential fusion of the phagosome with early endosomes, late endosomes, and lysosomes. Although various phosphoinositides (PIPs) have been detected on phagosomes, it remained unclear which PIPs actually govern phagosome maturation. Here, we analyzed the involvement of PIPs in fusion of phagosomes with various endocytic compartments and identified phosphatidylinositol 4-phosphate [PI(4)P], phosphatidylinositol 3-phosphate [PI(3)P], and the lipid kinases that generate these PIPs, as mediators of phagosome-lysosome fusion. Phagosome-early endosome fusion required PI(3)P, yet did not depend on PI(4)P. Thus, PI(3)P regulates phagosome maturation at early and late stages, whereas PI(4)P is selectively required late in the pathway.
Collapse
|
29
|
Shubin AV, Demidyuk IV, Lunina NA, Komissarov AA, Roschina MP, Leonova OG, Kostrov SV. Protease 3C of hepatitis A virus induces vacuolization of lysosomal/endosomal organelles and caspase-independent cell death. BMC Cell Biol 2015; 16:4. [PMID: 25886889 PMCID: PMC4355371 DOI: 10.1186/s12860-015-0050-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 01/26/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND 3C proteases, the main proteases of picornaviruses, play the key role in viral life cycle by processing polyproteins. In addition, 3C proteases digest certain host cell proteins to suppress antiviral defense, transcription, and translation. The activity of 3C proteases per se induces host cell death, which makes them critical factors of viral cytotoxicity. To date, cytotoxic effects have been studied for several 3C proteases, all of which induce apoptosis. This study for the first time describes the cytotoxic effect of 3C protease of human hepatitis A virus (3Cpro), the only proteolytic enzyme of the virus. RESULTS Individual expression of 3Cpro induced catalytic activity-dependent cell death, which was not abrogated by the pan-caspase inhibitor (z-VAD-fmk) and was not accompanied by phosphatidylserine externalization in contrast to other picornaviral 3C proteases. The cell survival was also not affected by the inhibitors of cysteine proteases (z-FA-fmk) and RIP1 kinase (necrostatin-1), critical enzymes involved in non-apoptotic cell death. A substantial fraction of dying cells demonstrated numerous non-acidic cytoplasmic vacuoles with not previously described features and originating from several types of endosomal/lysosomal organelles. The lysosomal protein Lamp1 and GTPases Rab5, Rab7, Rab9, and Rab11 were associated with the vacuolar membranes. The vacuolization was completely blocked by the vacuolar ATPase inhibitor (bafilomycin A1) and did not depend on the activity of the principal factors of endosomal transport, GTPases Rab5 and Rab7, as well as on autophagy and macropinocytosis. CONCLUSIONS 3Cpro, apart from other picornaviral 3C proteases, induces caspase-independent cell death, accompanying by cytoplasmic vacuolization. 3Cpro-induced vacuoles have unique properties and are formed from several organelle types of the endosomal/lysosomal compartment. The data obtained demonstrate previously undocumented morphological characters of the 3Cpro-induced cell death, which can reflect unknown aspects of the human hepatitis A virus-host cell interaction.
Collapse
Affiliation(s)
- Andrey V Shubin
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Ilya V Demidyuk
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Nataliya A Lunina
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Alexey A Komissarov
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Marina P Roschina
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Olga G Leonova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119992, Russia.
| | - Sergey V Kostrov
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
- National Research Center "Kurchatov Institute", Moscow, 123182, Russia.
| |
Collapse
|
30
|
Ikonomov OC, Sbrissa D, Venkatareddy M, Tisdale E, Garg P, Shisheva A. Class III PI 3-kinase is the main source of PtdIns3P substrate and membrane recruitment signal for PIKfyve constitutive function in podocyte endomembrane homeostasis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1240-50. [PMID: 25619930 DOI: 10.1016/j.bbamcr.2015.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/22/2014] [Accepted: 01/16/2015] [Indexed: 11/19/2022]
Abstract
The evolutionarily conserved PIKfyve, which synthesizes PtdIns5P from PtdIns, and PtdIns(3,5)P2 from PtdIns3P, requires PtdIns3P as both an enzyme substrate and a membrane recruitment signal. Whereas the PtdIns3P source is undetermined, class III PI3K (Vps34), the only evolutionarily conserved of the eight mammalian PI3Ks, is presumed as a main candidate. A hallmark of PIKfyve deficiency is formation of multiple translucent cytoplasmic vacuoles seen by light microscopy in cells cultured in complete media. Such an aberrant phenotype is often observed in cells from conditional Vps34 knockout (KO) mice. To clarify the mechanism of Vps34 KO-triggered vacuolation and the PtdIns3P source for PIKfyve functionality, here we have characterized a podocyte cell type derived from Vps34fl/fl mice, which, upon Cre-mediated gene KO, robustly formed cytoplasmic vacuoles resembling those in PikfyveKO MEFs. Vps34wt, expressed in Vps34KO podocytes restored the normal morphology, but only if the endogenous PIKfyve activity was intact. Conversely, expressed PIKfyvewt rescued completely the vacuolation only in PikfyveKO MEFs but not in Vps34KO podocytes. Analyses of phosphoinositide profiles by HPLC and localization patterns by a PtdIns3P biosensor revealed that Vps34 is the main supplier of localized PtdIns3P not only for PIKfyve activity but also for membrane recruitment. Concordantly, Vps34KO podocytes had severely reduced steady-state levels of both PtdIns(3,5)P2 and PtdIns5P, along with PtdIns3P. We further revealed a plausible physiologically-relevant Vps34-independent PtdIns3P supply for PIKfyve, operating through activated class I PI3Ks. Our data provide the first evidence that the vacuolation phenotype in Vps34KO podocytes is due to PIKfyve dysfunction and that Vps34 is a main PtdIns3P source for constitutive PIKfyve functionality.
Collapse
Affiliation(s)
- Ognian C Ikonomov
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Diego Sbrissa
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | - Ellen Tisdale
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Puneet Garg
- Division of Nephrology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Assia Shisheva
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
31
|
Pasquier B, El-Ahmad Y, Filoche-Rommé B, Dureuil C, Fassy F, Abecassis PY, Mathieu M, Bertrand T, Benard T, Barrière C, El Batti S, Letallec JP, Sonnefraud V, Brollo M, Delbarre L, Loyau V, Pilorge F, Bertin L, Richepin P, Arigon J, Labrosse JR, Clément J, Durand F, Combet R, Perraut P, Leroy V, Gay F, Lefrançois D, Bretin F, Marquette JP, Michot N, Caron A, Castell C, Schio L, McCort G, Goulaouic H, Garcia-Echeverria C, Ronan B. Discovery of (2S)-8-[(3R)-3-Methylmorpholin-4-yl]-1-(3-methyl-2-oxobutyl)-2-(trifluoromethyl)-3,4-dihydro-2H-pyrimido[1,2-a]pyrimidin-6-one: A Novel Potent and Selective Inhibitor of Vps34 for the Treatment of Solid Tumors. J Med Chem 2014; 58:376-400. [DOI: 10.1021/jm5013352] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jérôme Arigon
- Lead Generation Candidate Realization (LGCR C&BD), Sanofi, 371 Rue du Professeur Joseph Blayac, 34184 Montpellier, France
| | - Jean-Robert Labrosse
- Lead Generation Candidate Realization (LGCR C&BD), Sanofi, 371 Rue du Professeur Joseph Blayac, 34184 Montpellier, France
| | - Jacques Clément
- Lead Generation Candidate Realization (LGCR C&BD), Sanofi, 371 Rue du Professeur Joseph Blayac, 34184 Montpellier, France
| | - Florence Durand
- Lead Generation Candidate Realization (LGCR C&BD), Sanofi, 371 Rue du Professeur Joseph Blayac, 34184 Montpellier, France
| | - Romain Combet
- Lead Generation Candidate Realization (LGCR C&BD), Sanofi, 371 Rue du Professeur Joseph Blayac, 34184 Montpellier, France
| | - Pierre Perraut
- Lead Generation Candidate Realization (LGCR C&BD), Sanofi, 371 Rue du Professeur Joseph Blayac, 34184 Montpellier, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
A highly potent and selective Vps34 inhibitor alters vesicle trafficking and autophagy. Nat Chem Biol 2014; 10:1013-9. [PMID: 25326666 DOI: 10.1038/nchembio.1681] [Citation(s) in RCA: 372] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 09/23/2014] [Indexed: 12/24/2022]
Abstract
Vps34 is a phosphoinositide 3-kinase (PI3K) class III isoform that has attracted major attention over the recent years because of its role in autophagy. Herein we describe the biological characterization of SAR405, which is a low-molecular-mass kinase inhibitor of Vps34 (KD 1.5 nM). This compound has an exquisite protein and lipid kinase selectivity profile that is explained by its unique binding mode and molecular interactions within the ATP binding cleft of human Vps34. To the best of our knowledge, this is the first potent and specific Vps34 inhibitor described so far. Our results demonstrate that inhibition of Vps34 kinase activity by SAR405 affects both late endosome-lysosome compartments and prevents autophagy. Moreover, we show that the concomitant inhibition of Vps34 and mTOR, with SAR405 and the US Food and Drug Administration-approved mTOR inhibitor everolimus, results in synergistic antiproliferative activity in renal tumor cell lines, indicating a potential clinical application in cancer.
Collapse
|
33
|
Beclin-1 deficiency in the murine ovary results in the reduction of progesterone production to promote preterm labor. Proc Natl Acad Sci U S A 2014; 111:E4194-203. [PMID: 25246579 DOI: 10.1073/pnas.1409323111] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Autophagy is an important cellular process that serves as a companion pathway to the ubiquitin-proteasome system to degrade long-lived proteins and organelles to maintain cell homeostasis. Although initially characterized in yeast, autophagy is being realized as an important regulator of development and disease in mammals. Beclin1 (Becn1) is a putative tumor suppressor gene that has been shown to undergo a loss of heterozygosity in 40-75% of human breast, ovarian, and prostate cancers. Because Becn1 is a key regulator of autophagy, we sought to investigate its role in female reproduction by using a conditional knockout approach in mice. We find that pregnant females lacking Becn1 in the ovarian granulosa cell population have a defect in progesterone production and a subsequent preterm labor phenotype. Luteal cells in this model exhibit defective autophagy and a failure to accumulate lipid droplets needed for steroidogenesis. Collectively, we show that Becn1 provides essential functions in the ovary that are essential for mammalian reproduction.
Collapse
|
34
|
Reifler A, Li X, Archambeau AJ, McDade JR, Sabha N, Michele DE, Dowling JJ. Conditional knockout of pik3c3 causes a murine muscular dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1819-30. [PMID: 24726497 DOI: 10.1016/j.ajpath.2014.02.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 02/07/2014] [Accepted: 02/18/2014] [Indexed: 12/28/2022]
Abstract
Abnormalities in phosphoinositide metabolism are an emerging theme in human neurodegenerative disease. Myotubular myopathy is a prototypical disorder of phosphoinositide dysregulation that is characterized by profound muscle pathology and weakness and that is caused by mutations in MTM1, which encodes a phosphatase that targets 3-position phosphoinositides, including phosphatidylinositol 3-phosphate. Although the association between MTM1 and muscle disease has become increasingly clarified, the normal role(s) of phosphatidylinositol 3-phosphate metabolism in muscle development and homeostasis remain poorly understood. To begin to address the function of phosphatidylinositol 3-phosphate in skeletal muscle, we focused on the primary kinase responsible for its production, and created a muscle-specific conditional knockout of the class III phosphatidylinositol 3-kinase, Pik3c3. Muscle-specific deletion of Pik3c3 did not disturb embryogenesis or early postnatal development, but resulted in progressive disease characterized by reduced activity and death by 2 months of age. Histopathological analysis demonstrated changes consistent with a murine muscular dystrophy. Examination for cellular mechanism(s) responsible for the dystrophic phenotype revealed significant alterations in the autophagolysosomal pathway with mislocation of known dystrophy proteins to the lysosomal compartment. In all, we present the first analysis of Pik3c3 in skeletal muscle, and report a novel association between deletion of Pik3c3 and muscular dystrophy.
Collapse
Affiliation(s)
- Aaron Reifler
- Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, Michigan; Neuroscience Graduate Program, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Xingli Li
- Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Ashley J Archambeau
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Joel R McDade
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Nesrin Sabha
- Department of Neurology and Program of Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Daniel E Michele
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - James J Dowling
- Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, Michigan; Department of Neurology and Program of Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada; Departments of Paediatrics and Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
35
|
Vinod V, Padmakrishnan CJ, Vijayan B, Gopala S. 'How can I halt thee?' The puzzles involved in autophagic inhibition. Pharmacol Res 2014; 82:1-8. [PMID: 24657238 DOI: 10.1016/j.phrs.2014.03.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 03/07/2014] [Accepted: 03/12/2014] [Indexed: 12/20/2022]
Abstract
The strategy for interpreting the role of autophagy on the basis of evidence obtained through autophagic inhibition sounds logical, but is beset with practical constraints. The knock down of autophagy-related (ATG) gene(s) or blockage of class III PI3-Kinase are the most common approaches for inhibiting autophagy. However, during stressful conditions, autophagy may operate in synchrony with other processes such as apoptosis; autophagy-related genes, unlike what their name implies, exert their regulation on apoptosis as well. Knocking down such genes not only blocks autophagy but also renders apoptosis defective, making the interpretation of autophagic roles unreliable. Similarly, class III PI3-Kinase aids in initiating autophagy but it is not a quintessential autophagic regulator. Class III PI3-Kinase also has a role in regulating almost all membrane transport in cells. Blocking it not only inhibits autophagy, but also hampers all the membrane trades, including endosomal transport. The pharmacological inhibitors used to block autophagy by blocking class III PI3-Kinase further compound these limitations with their off-target effects. Knowing the limitations involved in blocking a target or using an autophagy-blocking tool is a prerequisite for designing the experiments meant for analyzing autophagic functions. This review attempts to provide a detailed overview about the practical constraints involved in using autophagic inhibition as a strategy to understand autophagy.
Collapse
Affiliation(s)
- V Vinod
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695011, India
| | - C J Padmakrishnan
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695011, India
| | - Bejoy Vijayan
- Comprehensive Care Centre for Movement Disorders, Department of Neurology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695011, India
| | - Srinivas Gopala
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695011, India.
| |
Collapse
|
36
|
Human cytomegalovirus pUL37x1-induced calcium flux activates PKCα, inducing altered cell shape and accumulation of cytoplasmic vesicles. Proc Natl Acad Sci U S A 2014; 111:E1140-8. [PMID: 24616524 DOI: 10.1073/pnas.1402515111] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The human cytomegalovirus immediate-early protein pUL37x1 induces the release of Ca(2+) stores from the endoplasmic reticulum into the cytosol. This release causes reorganization of the cellular actin cytoskeleton with concomitant cell rounding. Here we demonstrate that pUL37x1 activates Ca(2+)-dependent protein kinase Cα (PKCα). Both PKCα and Rho-associated protein kinases are required for actin reorganization and cell rounding; however, only PKCα is required for the efficient production of virus progeny, arguing that HCMV depends on the kinase for a second function. PKCα activation is also needed for the production of large (1-5 μm) cytoplasmic vesicles late after infection. The production of these vesicles is blocked by inhibition of fatty acid or phosphatidylinositol-3-phosphate biosynthesis, and the failure to produce vesicles is correlated with substantially reduced production of enveloped virus capsids. These results connect earlier work identifying a requirement for lipid synthesis with specific morphological changes, and support the argument that the PKCα-induced large vesicles are either required for the efficient production of mature virus particles or serve as a marker for the process.
Collapse
|
37
|
Peppard JV, Rugg C, Smicker M, Dureuil C, Ronan B, Flamand O, Durand L, Pasquier B. Identifying Small Molecules which Inhibit Autophagy: a Phenotypic Screen Using Image-Based High-Content Cell Analysis. Curr Chem Genom Transl Med 2014; 8:3-15. [PMID: 24596680 PMCID: PMC3941084 DOI: 10.2174/2213988501408010003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 11/10/2013] [Accepted: 11/25/2013] [Indexed: 12/02/2022] Open
Abstract
Autophagy plays an important role in cancer and it has been suggested that it functions not only as a tumor
suppressor pathway to prevent tumor initiation, but also as a pro-survival pathway that helps tumor cells endure metabolic
stress and resist death triggered by chemotherapeutic agents, including acquired resistance. We aimed to identify small-molecule
autophagy inhibitors using a HTS/HCA approach through a phenotypic, cell image-based assay, in order to
screen multiple biological targets simultaneously and to screen compounds in a physiologically relevant environment.
LC3 is a component of the autophagosome, which undergoes a cytoplasmic redistribution from diffuse to punctate dots
during autophagy. We employed HeLa cells stably expressing EGFP-LC3 in a primary phenotypic screen. As a first step,
a “Validation Library” of about 8,000 pre-selected compounds, about 25% of which had known biological activity and the
others representing a range of chemical structures, was run in duplicate both to assess screening suitability and likely hit
rate, and to give a valuable preview of possible active structures or biological targets. The primary screen of about 0.25
million compounds yielded around 10,500 positive compounds. These were tested in a suite of further cellular assays designed
to eliminate unwanted positives, together with the application of chemi- and bioinformatics to pick out compounds
with known biological activity. These processes enabled the selection of compounds that were the most promisingly active
and specific. The screening “tree” identified, amongst others with as yet unidentified targets, chemical series active
against autophagy-relevant biological targets ULK or Vsp34, validating the phenotypic screening methods selected. Finally,
about 400 compounds were fully qualified after following this triage. The development of the assays, compound
screening process and the compound triage is described.
Collapse
Affiliation(s)
- J V Peppard
- Lead Generation and Candidate Realization, R&D, Bridgewater, NJ 07059, U.S.A
| | - C Rugg
- Lead Generation and Candidate Realization, R&D, Bridgewater, NJ 07059, U.S.A
| | - M Smicker
- Lead Generation and Candidate Realization, R&D, Bridgewater, NJ 07059, U.S.A
| | - C Dureuil
- Oncology R&D, Sanofi, 94403, Vitry-Sur-Seine, France
| | - B Ronan
- Oncology R&D, Sanofi, 94403, Vitry-Sur-Seine, France
| | - O Flamand
- Oncology R&D, Sanofi, 94403, Vitry-Sur-Seine, France
| | - L Durand
- Oncology R&D, Sanofi, 94403, Vitry-Sur-Seine, France
| | - B Pasquier
- Oncology R&D, Sanofi, 94403, Vitry-Sur-Seine, France
| |
Collapse
|
38
|
Abstract
Phosphoinositides (PIs) are minor lipid components of cellular membranes that play critical roles in membrane dynamics, trafficking, and cellular signaling. Among the seven naturally occurring PIs, the monophosphate phosphatidylinositol 3-phosphate (PtdIns3P) and the bisphosphate phosphatidylinositol 3,5-bisphosphate [PtdIns(3,5)P2] have been mainly associated with endosomes and endosomal functions. Metabolic labeling and HPLC analysis revealed that a bulk of PtdIns3P is constitutively present in cells, making it the only detectable product of the enzymes phosphoinositide 3-kinases in unstimulated, normal cells. The use of specific tagged-PtdIns3P-binding domains later demonstrated that this constitutive PtdIns3P accumulates in endosomes where it critically regulates trafficking and membrane dynamics.
Collapse
Affiliation(s)
- Tania Maffucci
- Inositide Signalling Group, Centre for Diabetes, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Marco Falasca
- Inositide Signalling Group, Centre for Diabetes, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
39
|
Devereaux K, Dall’Armi C, Alcazar-Roman A, Ogasawara Y, Zhou X, Wang F, Yamamoto A, De Camilli P, Di Paolo G. Regulation of mammalian autophagy by class II and III PI 3-kinases through PI3P synthesis. PLoS One 2013; 8:e76405. [PMID: 24098492 PMCID: PMC3789715 DOI: 10.1371/journal.pone.0076405] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 08/30/2013] [Indexed: 11/19/2022] Open
Abstract
Synthesis of phosphatidylinositol-3-phosphate (PI3P) by Vps34, a class III phosphatidylinositol 3-kinase (PI3K), is critical for the initial steps of autophagosome (AP) biogenesis. Although Vps34 is the sole source of PI3P in budding yeast, mammalian cells can produce PI3P through alternate pathways, including direct synthesis by the class II PI3Ks; however, the physiological relevance of these alternate pathways in the context of autophagy is unknown. Here we generated Vps34 knockout mouse embryonic fibroblasts (MEFs) and using a higher affinity 4x-FYVE finger PI3P-binding probe found a Vps34-independent pool of PI3P accounting for (~)35% of the total amount of this lipid species by biochemical analysis. Importantly, WIPI-1, an autophagy-relevant PI3P probe, still formed some puncta upon starvation-induced autophagy in Vps34 knockout MEFs. Additional characterization of autophagy by electron microscopy as well as protein degradation assays showed that while Vps34 is important for starvation-induced autophagy there is a significant component of functional autophagy occurring in the absence of Vps34. Given these findings, class II PI3Ks (α and β isoforms) were examined as potential positive regulators of autophagy. Depletion of class II PI3Ks reduced recruitment of WIPI-1 and LC3 to AP nucleation sites and caused an accumulation of the autophagy substrate, p62, which was exacerbated upon the concomitant ablation of Vps34. Our studies indicate that while Vps34 is the main PI3P source during autophagy, class II PI3Ks also significantly contribute to PI3P generation and regulate AP biogenesis.
Collapse
Affiliation(s)
- Kelly Devereaux
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, United States of America
| | - Claudia Dall’Armi
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, United States of America
| | - Abel Alcazar-Roman
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Yuta Ogasawara
- Department of Animal Bio-Science, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Xiang Zhou
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Fan Wang
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Akitsugu Yamamoto
- Department of Animal Bio-Science, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Pietro De Camilli
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Gilbert Di Paolo
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
40
|
Giardina BJ, Chiang HL. The key gluconeogenic enzyme fructose-1,6-bisphosphatase is secreted during prolonged glucose starvation and is internalized following glucose re-feeding via the non-classical secretory and internalizing pathways in Saccharomyces cerevisiae. PLANT SIGNALING & BEHAVIOR 2013; 8:24936. [PMID: 23673352 PMCID: PMC3999075 DOI: 10.4161/psb.24936] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/03/2013] [Accepted: 05/04/2013] [Indexed: 06/02/2023]
Abstract
In Saccharomyces cerevisia, the key gluconeogenic enzyme fructose-1,6-bisphosphatase is secreted into the periplasm during prolonged glucose starvation and is internalized into Vid/endosomes following glucose re-feeding. Fructose-1,6-bisphosphatase does not contain signal sequences required for the classical secretory and endocytic pathways. Hence, the secretion and internalization are mediated via the non-classical pathways.
Collapse
|
41
|
Kim J, Kim YC, Fang C, Russell RC, Kim JH, Fan W, Liu R, Zhong Q, Guan KL. Differential regulation of distinct Vps34 complexes by AMPK in nutrient stress and autophagy. Cell 2013; 152:290-303. [PMID: 23332761 DOI: 10.1016/j.cell.2012.12.016] [Citation(s) in RCA: 641] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 11/13/2012] [Accepted: 12/12/2012] [Indexed: 12/14/2022]
Abstract
Autophagy is a stress response protecting cells from unfavorable conditions, such as nutrient starvation. The class III phosphatidylinositol-3 kinase, Vps34, forms multiple complexes and regulates both intracellular vesicle trafficking and autophagy induction. Here, we show that AMPK plays a key role in regulating different Vps34 complexes. AMPK inhibits the nonautophagy Vps34 complex by phosphorylating T163/S165 in Vps34 and therefore suppresses overall PI(3)P production and protects cells from starvation. In parallel, AMPK activates the proautophagy Vps34 complex by phosphorylating S91/S94 in Beclin1 to induce autophagy. Atg14L, an autophagy-essential gene present only in the proautophagy Vps34 complex, inhibits Vps34 phosphorylation but increases Beclin1 phosphorylation by AMPK. As such, Atg14L dictates the differential regulation (either inhibition or activation) of different Vps34 complexes in response to glucose starvation. Our study reveals an intricate molecular regulation of Vps34 complexes by AMPK in nutrient stress response and autophagy.
Collapse
Affiliation(s)
- Joungmok Kim
- Department of Oral Biochemistry and Molecular Biology, Research Center for Tooth and Periodontal Tissue Regeneration, School of Dentistry, Kyung Hee University, Seoul 130-701, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Morishita H, Eguchi S, Kimura H, Sasaki J, Sakamaki Y, Robinson ML, Sasaki T, Mizushima N. Deletion of autophagy-related 5 (Atg5) and Pik3c3 genes in the lens causes cataract independent of programmed organelle degradation. J Biol Chem 2013; 288:11436-47. [PMID: 23479732 DOI: 10.1074/jbc.m112.437103] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The lens of the eye is composed of fiber cells, which differentiate from epithelial cells and undergo programmed organelle degradation during terminal differentiation. Although autophagy, a major intracellular degradation system, is constitutively active in these cells, its physiological role has remained unclear. We have previously shown that Atg5-dependent macroautophagy is not necessary for lens organelle degradation, at least during the embryonic period. Here, we generated lens-specific Atg5 knock-out mice and showed that Atg5 is not required for lens organelle degradation at any period of life. However, deletion of Atg5 in the lens results in age-related cataract, which is accompanied by accumulation of polyubiquitinated and oxidized proteins, p62, and insoluble crystallins, suggesting a defect in intracellular quality control. We also produced lens-specific Pik3c3 knock-out mice to elucidate the possible involvement of Atg5-independent alternative autophagy, which is proposed to be dependent on Pik3c3 (also known as Vps34), in lens organelle degradation. Deletion of Pik3c3 in the lens does not affect lens organelle degradation, but it leads to congenital cataract and a defect in lens development after birth likely due to an impairment of the endocytic pathway. Taken together, these results suggest that clearance of lens organelles is independent of macroautophagy. These findings also clarify the physiological role of Atg5 and Pik3c3 in quality control and development of the lens, respectively.
Collapse
Affiliation(s)
- Hideaki Morishita
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Raiborg C, Schink KO, Stenmark H. Class III phosphatidylinositol 3-kinase and its catalytic product PtdIns3P in regulation of endocytic membrane traffic. FEBS J 2013; 280:2730-42. [PMID: 23289851 DOI: 10.1111/febs.12116] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 12/20/2012] [Accepted: 12/24/2012] [Indexed: 01/01/2023]
Abstract
Endocytosis and subsequent membrane traffic through endosomes are cellular processes that are integral to eukaryotic evolution, and numerous human diseases are associated with their dysfunction. Consequently, it is important to untangle the molecular machineries that regulate membrane dynamics and protein flow in the endocytic pathway. Central in this context is class III phosphatidylinositol 3-kinase, an evolutionarily conserved enzyme complex that phosphorylates phosphatidylinositol into phosphatidylinositol 3-phosphate. Phosphatidylinositol 3-phosphate recruits specific effector proteins, most of which contain FYVE or PX domains, to promote endocytosis, endosome fusion, endosome motility and endosome maturation, as well as cargo sorting to lysosomes, the biosynthetic pathway or the plasma membrane. Here we review the functions of key phosphatidylinositol 3-phosphate effectors in regulation of endocytic membrane dynamics and protein sorting.
Collapse
Affiliation(s)
- Camilla Raiborg
- Centre for Cancer Biomedicine, Faculty of Medicine, Oslo University Hospital, Montebello, Norway
| | | | | |
Collapse
|
44
|
Ng AA, Logan AM, Schmidt EJ, Robinson FL. The CMT4B disease-causing phosphatases Mtmr2 and Mtmr13 localize to the Schwann cell cytoplasm and endomembrane compartments, where they depend upon each other to achieve wild-type levels of protein expression. Hum Mol Genet 2013; 22:1493-506. [PMID: 23297362 DOI: 10.1093/hmg/dds562] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The demyelinating peripheral neuropathy Charcot-Marie-Tooth type 4B (CMT4B) is characterized by axonal degeneration and myelin outfoldings. CMT4B results from mutations in either myotubularin-related protein 2 (MTMR2; CMT4B1) or MTMR13 (CMT4B2), phosphoinositide (PI) 3-phosphatases that dephosphorylate phosphatidylinositol 3-phosphate (PtdIns3P) and PtdIns(3,5)P2, lipids which regulate endo-lysosomal membrane traffic. The catalytically active MTMR2 and catalytically inactive MTMR13 physically associate, although the significance of this association is not well understood. Here we show that Mtmr13 loss leads to axonal degeneration in sciatic nerves of older mice. In addition, CMT4B2-like myelin outfoldings are present in Mtmr13(-/-) nerves at postnatal day 3. Thus, Mtmr13(-/-) mice show both the initial dysmyelination and later degenerative pathology of CMT4B2. Given the key role of PI 3-kinase-Akt signaling in myelination, we investigated the state of the pathway in nerves of CMT4B models. We found that Akt activation is unaltered in Mtmr13(-/-) and Mtmr2(-/-) mice. Mtmr2 and Mtmr13 are found within the Schwann cell cytoplasm, where the proteins are partially localized to punctate compartments, suggesting that Mtmr2-Mtmr13 may dephosphorylate their substrates on specific intracellular compartments. Mtmr2-Mtmr13 substrates play essential roles in endo-lysosomal membrane traffic. However, endosomes and lysosomes of Mtmr13(-/-) and Mtmr2(-/-) Schwann cells are morphologically indistinguishable from those of controls, indicating that loss of these proteins does not cause wholesale dysregulation of the endo-lysosomal system. Notably, Mtmr2 and Mtmr13 depend upon each other to achieve wild-type levels of protein expression. Mtmr2 stabilizes Mtmr13 on membranes, indicating that the Mtmr13 pseudophosphatase is regulated by its catalytically active binding partner.
Collapse
Affiliation(s)
- Aubree A Ng
- The Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
45
|
Endosomal maturation, Rab7 GTPase and phosphoinositides in African swine fever virus entry. PLoS One 2012; 7:e48853. [PMID: 23133661 PMCID: PMC3486801 DOI: 10.1371/journal.pone.0048853] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Accepted: 10/02/2012] [Indexed: 11/22/2022] Open
Abstract
Here we analyzed the dependence of African swine fever virus (ASFV) infection on the integrity of the endosomal pathway. Using confocal immunofluorescence with antibodies against viral capsid proteins, we found colocalization of incoming viral particles with early endosomes (EE) during the first minutes of infection. Conversely, viral capsid protein was not detected in acidic late endosomal compartments, multivesicular bodies (MVBs), late endosomes (LEs) or lysosomes (LY). Using an antibody against a viral inner core protein, we found colocalization of viral cores with late compartments from 30 to 60 minutes postinfection. The absence of capsid protein staining in LEs and LYs suggested that virus desencapsidation would take place at the acid pH of these organelles. In fact, inhibitors of intraluminal acidification of endosomes caused retention of viral capsid staining virions in Rab7 expressing endosomes and more importantly, severely impaired subsequent viral protein production. Endosomal acidification in the first hour after virus entry was essential for successful infection but not thereafter. In addition, altering the balance of phosphoinositides (PIs) which are responsible of the maintenance of the endocytic pathway impaired ASFV infection. Early infection steps were dependent on the production of phosphatidylinositol 3-phosphate (PtdIns3P) which is involved in EE maturation and multivesicular body (MVB) biogenesis and on the interconversion of PtdIns3P to phosphatidylinositol 3, 5-biphosphate (PtdIns(3,5)P2). Likewise, GTPase Rab7 activity should remain intact, as well as processes related to LE compartment physiology, which are crucial during early infection. Our data demonstrate that the EE and LE compartments and the integrity of the endosomal maturation pathway orchestrated by Rab proteins and PIs play a central role during early stages of ASFV infection.
Collapse
|
46
|
Endothelial PI3K-C2α, a class II PI3K, has an essential role in angiogenesis and vascular barrier function. Nat Med 2012; 18:1560-9. [PMID: 22983395 DOI: 10.1038/nm.2928] [Citation(s) in RCA: 175] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 08/10/2012] [Indexed: 12/11/2022]
Abstract
The class II α-isoform of phosphatidylinositol 3-kinase (PI3K-C2α) is localized in endosomes, the trans-Golgi network and clathrin-coated vesicles; however, its functional role is not well understood. Global or endothelial-cell-specific deficiency of PI3K-C2α resulted in embryonic lethality caused by defects in sprouting angiogenesis and vascular maturation. PI3K-C2α knockdown in endothelial cells resulted in a decrease in the number of PI3-phosphate-enriched endosomes, impaired endosomal trafficking, defective delivery of VE-cadherin to endothelial cell junctions and defective junction assembly. PI3K-C2α knockdown also impaired endothelial cell signaling, including vascular endothelial growth factor receptor internalization and endosomal RhoA activation. Together, the effects of PI3K-C2α knockdown led to defective endothelial cell migration, proliferation, tube formation and barrier integrity. Endothelial PI3K-C2α deficiency in vivo suppressed postischemic and tumor angiogenesis and diminished vascular barrier function with a greatly augmented susceptibility to anaphylaxis and a higher incidence of dissecting aortic aneurysm formation in response to angiotensin II infusion. Thus, PI3K-C2α has a crucial role in vascular formation and barrier integrity and represents a new therapeutic target for vascular disease.
Collapse
|
47
|
Thi EP, Lambertz U, Reiner NE. Class IA phosphatidylinositol 3-kinase p110α regulates phagosome maturation. PLoS One 2012; 7:e43668. [PMID: 22928013 PMCID: PMC3425514 DOI: 10.1371/journal.pone.0043668] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 07/24/2012] [Indexed: 12/31/2022] Open
Abstract
Of the various phosphatidylinositol 3- kinases (PI3Ks), only the class III enzyme Vps34 has been shown to regulate phagosome maturation. During studies of phagosome maturation in THP-1 cells deficient in class IA PI3K p110α, we discovered that this PI3K isoform is required for vacuole maturation to progress beyond acquisition of Rab7 leading to delivery of lysosomal markers. Bead phagosomes from THP-1 cells acquired p110α and contained PI3P and PI(3,4,5)P3; however, p110α and PI(3,4,5)P3 levels in phagosomes from p110α knockdown cells were decreased. Phagosomes from p110α knock down cells showed normal acquisition of both Rab5 and EEA-1, but were markedly deficient in the lysosomal markers LAMP-1 and LAMP-2, and the lysosomal hydrolase, β-galactosidase. Phagosomes from p110α deficient cells also displayed impaired fusion with Texas Red dextran-loaded lysosomes. Despite lacking lysosomal components, phagosomes from p110α deficient cells recruited normal levels of Rab7, Rab-interacting lysosomal protein (RILP) and homotypic vacuole fusion and protein sorting (HOPs) components Vps41 and Vps16. The latter observations demonstrated that phagosomal Rab7 was active and capable of recruiting effectors involved in membrane fusion. Nevertheless, active Rab7 was not sufficient to bring about the delivery of lysosomal proteins to the maturing vacuole, which is shown for the first time to be dependent on a class I PI3K.
Collapse
Affiliation(s)
- Emily P. Thi
- Departments of Medicine, Experimental Medicine Program, Division of Infectious Diseases, University of British Columbia and the Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, B.C., Canada
| | - Ulrike Lambertz
- Departments of Medicine, Experimental Medicine Program, Division of Infectious Diseases, University of British Columbia and the Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, B.C., Canada
| | - Neil E. Reiner
- Departments of Medicine, Experimental Medicine Program, Division of Infectious Diseases, University of British Columbia and the Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, B.C., Canada
- Microbiology and Immunology, University of British Columbia and the Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, B.C., Canada
- * E-mail:
| |
Collapse
|
48
|
Wiczer BM, Thomas G. Phospholipase D and mTORC1: Nutrients Are What Bring Them Together. Sci Signal 2012; 5:pe13. [DOI: 10.1126/scisignal.2003019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
49
|
Cytoplasmic vacuolization during exposure to drugs and other substances. Cell Biol Toxicol 2012; 28:125-31. [DOI: 10.1007/s10565-012-9212-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 01/19/2012] [Indexed: 01/11/2023]
|
50
|
Class III PI3K Vps34 plays an essential role in autophagy and in heart and liver function. Proc Natl Acad Sci U S A 2012; 109:2003-8. [PMID: 22308354 DOI: 10.1073/pnas.1112848109] [Citation(s) in RCA: 305] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A critical regulator of autophagy is the Class III PI3K Vps34 (also called PIK3C3). Although Vps34 is known to play an essential role in autophagy in yeast, its role in mammals remains elusive. To elucidate the physiological function of Vps34 and to determine its precise role in autophagy, we have generated Vps34(f/f) mice, in which expression of Cre recombinase results in a deletion of exon 4 of Vps34 and a frame shift causing a deletion of 755 of the 887 amino acids of Vps34. Acute ablation of Vps34 in MEFs upon adenoviral Cre infection results in a diminishment of localized generation of phosphatidylinositol 3-phosphate and blockade of both endocytic and autophagic degradation. Starvation-induced autophagosome formation is blocked in both Vps34-null MEFs and liver. Liver-specific Albumin-Cre;Vps34(f/f) mice developed hepatomegaly and hepatic steatosis, and impaired protein turnover. Ablation of Vps34 in the heart of muscle creatine kinase-Cre;Vps34(f/f) mice led to cardiomegaly and decreased contractility. In addition, while amino acid-stimulated mTOR activation was suppressed in the absence of Vps34, the steady-state level of mTOR signaling was not affected in Vps34-null MEFs, liver, or cardiomyocytes. Taken together, our results indicate that Vps34 plays an essential role in regulating functional autophagy and is indispensable for normal liver and heart function.
Collapse
|