1
|
Hasegawa J. New insights into the regulation and roles of phosphatidylinositol 3,4-bisphosphate. J Biochem 2024; 176:339-345. [PMID: 39271134 DOI: 10.1093/jb/mvae063] [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: 07/19/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024] Open
Abstract
Phosphoinositides (PIPs) are phospholipids and components of the cellular membrane. In mammals, seven phosphorylated derivatives of PIPs have been identified. Among them, phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2] is produced by lipid phosphatases (e.g., SHIP2) or by lipid kinases PI3KC2α and PI3KC2β. Although PI(3,4)P2 is undetectable in normal mouse or human tissues and common cell lines, it appears in a mouse prostate cancer model and in cells exposed to oxidative stress, indicating that PI(3,4)P2 is involved in the pathogenesis of some diseases. Here, I summarize recent findings on the cellular roles and pathophysiological significance of PI(3,4)P2.
Collapse
Affiliation(s)
- Junya Hasegawa
- Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|
2
|
Chan V, Camardi C, Zhang K, Orofiamma LA, Anderson KE, Hoque J, Bone LN, Awadeh Y, Lee DKC, Fu NJ, Chow JTS, Salmena L, Stephens LR, Hawkins PT, Antonescu CN, Botelho RJ. The LCLAT1/LYCAT acyltransferase is required for EGF-mediated phosphatidylinositol-3,4,5-trisphosphate generation and Akt signaling. Mol Biol Cell 2024; 35:ar118. [PMID: 39024272 PMCID: PMC11449395 DOI: 10.1091/mbc.e23-09-0361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
Receptor tyrosine kinases such as EGF receptor (EGFR) stimulate phosphoinositide 3 kinases to convert phosphatidylinositol-4,5-bisphosophate [PtdIns(4,5)P2] into phosphatidylinositol-3,4,5-trisphosphate [PtdIns(3,4,5)P3]. PtdIns(3,4,5)P3 then remodels actin and gene expression, and boosts cell survival and proliferation. PtdIns(3,4,5)P3 partly achieves these functions by triggering activation of the kinase Akt, which phosphorylates targets like Tsc2 and GSK3β. Consequently, unchecked upregulation of PtdIns(3,4,5)P3-Akt signaling promotes tumor progression. Interestingly, 50-70% of PtdIns and PtdInsPs have stearate and arachidonate at sn-1 and sn-2 positions of glycerol, respectively, forming a species known as 38:4-PtdIns/PtdInsPs. LCLAT1 and MBOAT7 acyltransferases partly enrich PtdIns in this acyl format. We previously showed that disruption of LCLAT1 lowered PtdIns(4,5)P2 levels and perturbed endocytosis and endocytic trafficking. However, the role of LCLAT1 in receptor tyrosine kinase and PtdIns(3,4,5)P3 signaling was not explored. Here, we show that LCLAT1 silencing in MDA-MB-231 and ARPE-19 cells abated the levels of PtdIns(3,4,5)P3 in response to EGF signaling. Importantly, LCLAT1-silenced cells were also impaired for EGF-driven and insulin-driven Akt activation and downstream signaling. Thus, our work provides first evidence that the LCLAT1 acyltransferase is required for receptor tyrosine kinase signaling.
Collapse
Affiliation(s)
- Victoria Chan
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Cristina Camardi
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Kai Zhang
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Laura A. Orofiamma
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Karen E. Anderson
- Signalling Programme, Babraham Institute, Cambridge CB22 4AT, United Kingdom
| | - Jafarul Hoque
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Leslie N. Bone
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Yasmin Awadeh
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Daniel K. C. Lee
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Norman J. Fu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Jonathan T. S. Chow
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Leonardo Salmena
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Len R. Stephens
- Signalling Programme, Babraham Institute, Cambridge CB22 4AT, United Kingdom
| | - Phillip T. Hawkins
- Signalling Programme, Babraham Institute, Cambridge CB22 4AT, United Kingdom
| | - Costin N. Antonescu
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| | - Roberto J. Botelho
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario M5B2K3, Canada
| |
Collapse
|
3
|
Zhang K, Chan V, Botelho RJ, Antonescu CN. A tail of their own: regulation of cardiolipin and phosphatidylinositol fatty acyl profile by the acyltransferase LCLAT1. Biochem Soc Trans 2023; 51:1765-1776. [PMID: 37737061 DOI: 10.1042/bst20220603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/17/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023]
Abstract
Cardiolipin and phosphatidylinositol along with the latter's phosphorylated derivative phosphoinositides, control a wide range of cellular functions from signal transduction, membrane traffic, mitochondrial function, cytoskeletal dynamics, and cell metabolism. An emerging dimension to these lipids is the specificity of their fatty acyl chains that is remarkably distinct from that of other glycerophospholipids. Cardiolipin and phosphatidylinositol undergo acyl remodeling involving the sequential actions of phospholipase A to hydrolyze acyl chains and key acyltransferases that re-acylate with specific acyl groups. LCLAT1 (also known as LYCAT, AGPAT8, LPLAT6, or ALCAT1) is an acyltransferase that contributes to specific acyl profiles for phosphatidylinositol, phosphoinositides, and cardiolipin. As such, perturbations of LCLAT1 lead to alterations in cardiolipin-dependent phenomena such as mitochondrial respiration and dynamics and phosphoinositide-dependent processes such as endocytic membrane traffic and receptor signaling. Here we examine the biochemical and cellular actions of LCLAT1, as well as the contribution of this acyltransferase to the development and specific diseases.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
| | - Victoria Chan
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
- Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
| | - Roberto J Botelho
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
- Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
| | - Costin N Antonescu
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
- Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada M5B 2K3
| |
Collapse
|
4
|
Loncke J, Luyten T, Ramos AR, Erneux C, Bultynck G. Loss of INPP5K attenuates IP 3-induced Ca 2+ responses in the glioblastoma cell line U-251 MG cells. BBA ADVANCES 2023; 4:100105. [PMID: 37842182 PMCID: PMC10568277 DOI: 10.1016/j.bbadva.2023.100105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/17/2023] Open
Abstract
INPP5K (inositol polyphosphate 5-phosphatase K) is an endoplasmic reticulum (ER)-resident enzyme that acts as a phosphoinositide (PI) 5-phosphatase, capable of dephosphorylating various PIs including PI 4,5-bisphosphate (PI(4,5)P2), a key phosphoinositide found in the plasma membrane. Given its ER localization and substrate specificity, INPP5K may play a role in ER-plasma membrane contact sites. Furthermore, PI(4,5)P2 serves as a substrate for phospholipase C, an enzyme activated downstream of extracellular agonists acting on Gq-coupled receptors or tyrosine-kinase receptors, leading to IP3 production and subsequent release of Ca2+ from the ER, the primary intracellular Ca2+ storage organelle. In this study, we investigated the impact of INPP5K on ER Ca2+ dynamics using a previously established INPP5K-knockdown U-251 MG glioblastoma cell model. We here describe that loss of INPP5K impairs agonist-induced, IP3 receptor (IP3R)-mediated Ca2+ mobilization in intact cells, while the ER Ca2+ content and store-operated Ca2+ influx remain unaffected. To further elucidate the underlying mechanisms, we examined Ca2+ release in permeabilized cells stimulated with exogenous IP3. Interestingly, the absence of INPP5K also disrupted IP3-induced Ca2+ release events. These results suggest that INPP5K may directly influence IP3R activity through mechanisms yet to be resolved. The findings from this study point towards role of INPP5K in modulating ER calcium dynamics, particularly in relation to IP3-mediated signaling pathways. However, further work is needed to establish the general nature of our findings and to unravel the exact molecular mechanisms underlying the interplay between INNP5K function and Ca2+ signaling.
Collapse
Affiliation(s)
- Jens Loncke
- Laboratory of Molecular and Cellular Signaling, KU Leuven, Leuven, Belgium
| | - Tomas Luyten
- Laboratory of Molecular and Cellular Signaling, KU Leuven, Leuven, Belgium
| | - Ana Raquel Ramos
- ULB, IRIBHM, Campus Erasme, Bâtiment C, 808 Route de Lennik, Bruxelles 1070, Belgium
| | - Christophe Erneux
- ULB, IRIBHM, Campus Erasme, Bâtiment C, 808 Route de Lennik, Bruxelles 1070, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Morleo M, Venditti R, Theodorou E, Briere LC, Rosello M, Tirozzi A, Tammaro R, Al-Badri N, High FA, Shi J, Putti E, Ferrante L, Cetrangolo V, Torella A, Walker MA, Tenconi R, Iascone M, Mei D, Guerrini R, van der Smagt J, Kroes HY, van Gassen KLI, Bilal M, Umair M, Pingault V, Attie-Bitach T, Amiel J, Ejaz R, Rodan L, Zollino M, Agrawal PB, Del Bene F, Nigro V, Sweetser DA, Franco B. De novo missense variants in phosphatidylinositol kinase PIP5KIγ underlie a neurodevelopmental syndrome associated with altered phosphoinositide signaling. Am J Hum Genet 2023; 110:1377-1393. [PMID: 37451268 PMCID: PMC10432144 DOI: 10.1016/j.ajhg.2023.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/22/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023] Open
Abstract
Phosphoinositides (PIs) are membrane phospholipids produced through the local activity of PI kinases and phosphatases that selectively add or remove phosphate groups from the inositol head group. PIs control membrane composition and play key roles in many cellular processes including actin dynamics, endosomal trafficking, autophagy, and nuclear functions. Mutations in phosphatidylinositol 4,5 bisphosphate [PI(4,5)P2] phosphatases cause a broad spectrum of neurodevelopmental disorders such as Lowe and Joubert syndromes and congenital muscular dystrophy with cataracts and intellectual disability, which are thus associated with increased levels of PI(4,5)P2. Here, we describe a neurodevelopmental disorder associated with an increase in the production of PI(4,5)P2 and with PI-signaling dysfunction. We identified three de novo heterozygous missense variants in PIP5K1C, which encodes an isoform of the phosphatidylinositol 4-phosphate 5-kinase (PIP5KIγ), in nine unrelated children exhibiting intellectual disability, developmental delay, acquired microcephaly, seizures, visual abnormalities, and dysmorphic features. We provide evidence that the PIP5K1C variants result in an increase of the endosomal PI(4,5)P2 pool, giving rise to ectopic recruitment of filamentous actin at early endosomes (EEs) that in turn causes dysfunction in EE trafficking. In addition, we generated an in vivo zebrafish model that recapitulates the disorder we describe with developmental defects affecting the forebrain, including the eyes, as well as craniofacial abnormalities, further demonstrating the pathogenic effect of the PIP5K1C variants.
Collapse
Affiliation(s)
- Manuela Morleo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy; Department of Precision Medicine, University of Campania "Luigi Vanvitelli," Naples, Italy.
| | - Rossella Venditti
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II," Medical School, Naples, Italy
| | - Evangelos Theodorou
- Center for Genomic Medicine, Divisions of Pediatric Hematology/Oncology and Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lauren C Briere
- Center for Genomic Medicine, Divisions of Pediatric Hematology/Oncology and Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marion Rosello
- Sorbonne Université, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Alfonsina Tirozzi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli," Naples, Italy; Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy
| | - Roberta Tammaro
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
| | - Nour Al-Badri
- Sorbonne Université, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Frances A High
- Division of Medical Genetics & Metabolism, Massachusetts General Hospital for Children, Boston, MA 02114, USA
| | - Jiahai Shi
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elena Putti
- Sorbonne Université, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Luigi Ferrante
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
| | - Viviana Cetrangolo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
| | - Annalaura Torella
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy; Department of Precision Medicine, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Melissa A Walker
- Department of Neurology, Division of Neurogenetics, Child Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Romano Tenconi
- Clinical Genetics Unit, Department of Women and Children's Health, University of Padova, Padova, Italy
| | - Maria Iascone
- Medical Genetics, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Davide Mei
- Meyer Children's Hospital IRCCS, Neuroscience Department, Florence, Italy
| | - Renzo Guerrini
- Meyer Children's Hospital IRCCS, Neuroscience Department, Florence, Italy
| | - Jasper van der Smagt
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hester Y Kroes
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Koen L I van Gassen
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Muhammad Bilal
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center & King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Veronica Pingault
- Service de Médecine Génomique des Maladies Rares, et Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
| | - Tania Attie-Bitach
- Service de Médecine Génomique des Maladies Rares, et Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
| | - Jeannine Amiel
- Service de Médecine Génomique des Maladies Rares, et Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
| | - Resham Ejaz
- Division of Genetics, Department of Pediatrics, McMaster Children's Hospital, Hamilton, ON, Canada
| | - Lance Rodan
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Marcella Zollino
- Institute of Medical Genetics, A. Gemelli School of Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Pankaj B Agrawal
- Divisions of Newborn Medicine, Boston Children's Hospital, Boston, MA, USA; Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Filippo Del Bene
- Sorbonne Université, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Vincenzo Nigro
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy; Department of Precision Medicine, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - David A Sweetser
- Center for Genomic Medicine, Divisions of Pediatric Hematology/Oncology and Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Brunella Franco
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy; Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, Naples, Italy; Medical Genetics, Department of Translational Medicine, University of Naples "Federico II," Via Sergio Pansini, 80131 Naples, Italy
| |
Collapse
|
6
|
Davies EM, Gurung R, Le KQ, Roan KT, Harvey RP, Mitchell GM, Schwarz Q, Mitchell CA. PI(4,5)P 2-dependent regulation of endothelial tip cell specification contributes to angiogenesis. SCIENCE ADVANCES 2023; 9:eadd6911. [PMID: 37000875 PMCID: PMC10065449 DOI: 10.1126/sciadv.add6911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Dynamic positioning of endothelial tip and stalk cells, via the interplay between VEGFR2 and NOTCH signaling, is essential for angiogenesis. VEGFR2 activates PI3K, which phosphorylates PI(4,5)P2 to PI(3,4,5)P3, activating AKT; however, PI3K/AKT does not direct tip cell specification. We report that PI(4,5)P2 hydrolysis by the phosphoinositide-5-phosphatase, INPP5K, contributes to angiogenesis. INPP5K ablation disrupted tip cell specification and impaired embryonic angiogenesis associated with enhanced DLL4/NOTCH signaling. INPP5K degraded a pool of PI(4,5)P2 generated by PIP5K1C phosphorylation of PI(4)P in endothelial cells. INPP5K ablation increased PI(4,5)P2, thereby releasing β-catenin from the plasma membrane, and concurrently increased PI(3,4,5)P3-dependent AKT activation, conditions that licensed DLL4/NOTCH transcription. Suppression of PI(4,5)P2 in INPP5K-siRNA cells by PIP5K1C-siRNA, restored β-catenin membrane localization and normalized AKT signaling. Pharmacological NOTCH or AKT inhibition in vivo or genetic β-catenin attenuation rescued angiogenesis defects in INPP5K-null mice. Therefore, PI(4,5)P2 is critical for β-catenin/DLL4/NOTCH signaling, which governs tip cell specification during angiogenesis.
Collapse
Affiliation(s)
- Elizabeth M. Davies
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Rajendra Gurung
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Kai Qin Le
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Katherine T. T. Roan
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia
- School of Clinical Medicine and School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Geraldine M. Mitchell
- O’Brien Institute Department of St Vincent’s Institute and University of Melbourne, Department of Surgery, St. Vincent’s Hospital, Fitzroy, Victoria 3065, Australia
- Health Sciences Faculty, Australian Catholic University, Fitzroy, Victoria 3065, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia 5001, Australia
| | - Christina A. Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| |
Collapse
|
7
|
Jacquemyn J, Kuenen S, Swerts J, Pavie B, Vijayan V, Kilic A, Chabot D, Wang YC, Schoovaerts N, Corthout N, Verstreken P. Parkinsonism mutations in DNAJC6 cause lipid defects and neurodegeneration that are rescued by Synj1. NPJ Parkinsons Dis 2023; 9:19. [PMID: 36739293 PMCID: PMC9899244 DOI: 10.1038/s41531-023-00459-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 01/16/2023] [Indexed: 02/06/2023] Open
Abstract
Recent evidence links dysfunctional lipid metabolism to the pathogenesis of Parkinson's disease, but the mechanisms are not resolved. Here, we generated a new Drosophila knock-in model of DNAJC6/Auxilin and find that the pathogenic mutation causes synaptic dysfunction, neurological defects and neurodegeneration, as well as specific lipid metabolism alterations. In these mutants, membrane lipids containing long-chain polyunsaturated fatty acids, including phosphatidylinositol lipid species that are key for synaptic vesicle recycling and organelle function, are reduced. Overexpression of another protein mutated in Parkinson's disease, Synaptojanin-1, known to bind and metabolize specific phosphoinositides, rescues the DNAJC6/Auxilin lipid alterations, the neuronal function defects and neurodegeneration. Our work reveals a functional relation between two proteins mutated in Parkinsonism and implicates deregulated phosphoinositide metabolism in the maintenance of neuronal integrity and neuronal survival.
Collapse
Affiliation(s)
- Julie Jacquemyn
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium
- Neuroscience and Mental Health Institute, University of Alberta, Department of Physiology, Department of Cell Biology, Group on Molecular and Cell Biology of Lipids, Edmonton, Alberta, Canada
| | - Sabine Kuenen
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium
| | - Jef Swerts
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium
| | - Benjamin Pavie
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium
- VIB-Bioimaging Core, 3000, Leuven, Belgium
| | - Vinoy Vijayan
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium
| | - Ayse Kilic
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium
| | - Dries Chabot
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium
| | - Yu-Chun Wang
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium
- VIB Technology Watch, Technology Innovation Laboratory, VIB, Gent, Belgium
| | - Nils Schoovaerts
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium
| | - Nikky Corthout
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium
- VIB-Bioimaging Core, 3000, Leuven, Belgium
| | - Patrik Verstreken
- VIB-KU Leuven Center for Brain & Disease Research, 3000, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, 3000, Leuven, Belgium.
| |
Collapse
|
8
|
Droubi A, Wallis C, Anderson KE, Rahman S, de Sa A, Rahman T, Stephens LR, Hawkins PT, Lowe M. The inositol 5-phosphatase INPP5B regulates B cell receptor clustering and signaling. J Cell Biol 2022; 221:e202112018. [PMID: 35878408 PMCID: PMC9351708 DOI: 10.1083/jcb.202112018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 05/27/2022] [Accepted: 07/05/2022] [Indexed: 11/22/2022] Open
Abstract
Upon antigen binding, the B cell receptor (BCR) undergoes clustering to form a signalosome that propagates downstream signaling required for normal B cell development and physiology. BCR clustering is dependent on remodeling of the cortical actin network, but the mechanisms that regulate actin remodeling in this context remain poorly defined. In this study, we identify the inositol 5-phosphatase INPP5B as a key regulator of actin remodeling, BCR clustering, and downstream signaling in antigen-stimulated B cells. INPP5B acts via dephosphorylation of the inositol lipid PI(4,5)P2 that in turn is necessary for actin disassembly, BCR mobilization, and cell spreading on immobilized surface antigen. These effects can be explained by increased actin severing by cofilin and loss of actin linking to the plasma membrane by ezrin, both of which are sensitive to INPP5B-dependent PI(4,5)P2 hydrolysis. INPP5B is therefore a new player in BCR signaling and may represent an attractive target for treatment of B cell malignancies caused by aberrant BCR signaling.
Collapse
Affiliation(s)
- Alaa Droubi
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Connor Wallis
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Saifur Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Aloka de Sa
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | | | - Martin Lowe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
9
|
Sun J, Harion R, Naito T, Saheki Y. INPP5K and Atlastin-1 maintain the nonuniform distribution of ER-plasma membrane contacts in neurons. Life Sci Alliance 2021; 4:4/11/e202101092. [PMID: 34556534 PMCID: PMC8507493 DOI: 10.26508/lsa.202101092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/03/2021] [Accepted: 09/11/2021] [Indexed: 02/04/2023] Open
Abstract
In neurons, the ER extends throughout all cellular processes, forming multiple contacts with the plasma membrane (PM) to fine-tune neuronal physiology. However, the mechanisms that regulate the distribution of neuronal ER-PM contacts are not known. Here, we used the Caenorhabditis elegans DA9 motor neuron as our model system and found that neuronal ER-PM contacts are enriched in soma and dendrite and mostly absent in axons. Using forward genetic screen, we identified that the inositol 5-phosphatase, CIL-1 (human INPP5K), and the dynamin-like GTPase, ATLN-1 (human Atlastin-1), help to maintain the non-uniform, somatodendritic enrichment of neuronal ER-PM contacts. Mechanistically, CIL-1 acts upstream of ATLN-1 to maintain the balance between ER tubules and sheets. In mutants of CIL-1 or ATLN-1, ER sheets expand and invade into the axon. This is accompanied by the ectopic formation of axonal ER-PM contacts and defects in axon regeneration following laser-induced axotomy. As INPP5K and Atlastin-1 have been linked to neurological disorders, the unique distribution of neuronal ER-PM contacts maintained by these proteins may support neuronal resilience during the onset and progression of these diseases.
Collapse
Affiliation(s)
- Jingbo Sun
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Raihanah Harion
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Tomoki Naito
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Yasunori Saheki
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore .,Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
10
|
Hathazi D, Cox D, D'Amico A, Tasca G, Charlton R, Carlier RY, Baumann J, Kollipara L, Zahedi RP, Feldmann I, Deleuze JF, Torella A, Cohn R, Robinson E, Ricci F, Jungbluth H, Fattori F, Boland A, O’Connor E, Horvath R, Barresi R, Lochmüller H, Urtizberea A, Jacquemont ML, Nelson I, Swan L, Bonne G, Roos A. INPP5K and SIL1 associated pathologies with overlapping clinical phenotypes converge through dysregulation of PHGDH. Brain 2021; 144:2427-2442. [PMID: 33792664 PMCID: PMC8418339 DOI: 10.1093/brain/awab133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/12/2021] [Accepted: 01/30/2021] [Indexed: 12/22/2022] Open
Abstract
Marinesco-Sjögren syndrome is a rare human disorder caused by biallelic mutations in SIL1 characterized by cataracts in infancy, myopathy and ataxia, symptoms which are also associated with a novel disorder caused by mutations in INPP5K. While these phenotypic similarities may suggest commonalties at a molecular level, an overlapping pathomechanism has not been established yet. In this study, we present six new INPP5K patients and expand the current mutational and phenotypical spectrum of the disease showing the clinical overlap between Marinesco-Sjögren syndrome and the INPP5K phenotype. We applied unbiased proteomic profiling on cells derived from Marinesco-Sjögren syndrome and INPP5K patients and identified alterations in d-3-PHGDH as a common molecular feature. d-3-PHGDH modulates the production of l-serine and mutations in this enzyme were previously associated with a neurological phenotype, which clinically overlaps with Marinesco-Sjögren syndrome and INPP5K disease. As l-serine administration represents a promising therapeutic strategy for d-3-PHGDH patients, we tested the effect of l-serine in generated sil1, phgdh and inpp5k a+b zebrafish models, which showed an improvement in their neuronal phenotype. Thus, our study defines a core phenotypical feature underpinning a key common molecular mechanism in three rare diseases and reveals a common and novel therapeutic target for these patients.
Collapse
Affiliation(s)
- Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Dan Cox
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Newcastle upon Tyne, NE1 3BZ, UK
| | - Adele D'Amico
- Laboratory of Molecular Medicine for Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children’s Hospital, 00146 Rome, Italy
| | - Giorgio Tasca
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Richard Charlton
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Newcastle upon Tyne, NE1 3BZ, UK
| | - Robert-Yves Carlier
- AP-HP, Service d’Imagerie Médicale, Raymond Poincaré Hospital, 92380 Garches, France
- Inserm U 1179, University of Versailles Saint-Quentin-en-Yvelines (UVSQ), 78180 Versailles, France
| | - Jennifer Baumann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
| | | | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Ingo Feldmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
| | - Jean-Francois Deleuze
- Centre National de Recherche en Génomique Humaine (CNRGH) (A.B., J.F.D.), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, 91000 Evry, France
| | - Annalaura Torella
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Ronald Cohn
- SickKids Research Institute, Department of Paediatrics and Molecular Genetics, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Emily Robinson
- Department of molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Francesco Ricci
- Department of molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Heinz Jungbluth
- Guy’s and St Thomas’ NHS Trust, King’s College London, London, SE1 7EH, UK
| | - Fabiana Fattori
- Laboratory of Molecular Medicine for Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children’s Hospital, 00146 Rome, Italy
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine (CNRGH) (A.B., J.F.D.), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, 91000 Evry, France
| | - Emily O’Connor
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 5B2, Canada
| | - Rita Horvath
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Rita Barresi
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Newcastle upon Tyne, NE1 3BZ, UK
| | - Hanns Lochmüller
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 5B2, Canada
- Department of Neuropediatrics and Muscle Disorders, Medical Center—University of Freiburg, Faculty of Medicine, 79095 Freiburg, Germany
| | | | - Marie-Line Jacquemont
- Unité de Génétique Médicale, Pôle Femme-Mère-Enfant, Groupe Hospitalier Sud Réunion, CHU de La Réunion, 97410 La Réunion, France
| | - Isabelle Nelson
- Sorbonne Université, Inserm UMRS974, Centre de Recherche en Myologie, Institut de Myologie, 75013 Paris, France
| | - Laura Swan
- Department of molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Gisèle Bonne
- Sorbonne Université, Inserm UMRS974, Centre de Recherche en Myologie, Institut de Myologie, 75013 Paris, France
| | - Andreas Roos
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 5B2, Canada
- Department of Pediatric Neurology, University Hospital Essen, University of Duisburg-Essen, Faculty of Medicine, 45147 Essen, Germany
| |
Collapse
|
11
|
A 3D Renal Proximal Tubule on Chip Model Phenocopies Lowe Syndrome and Dent II Disease Tubulopathy. Int J Mol Sci 2021; 22:ijms22105361. [PMID: 34069732 PMCID: PMC8161077 DOI: 10.3390/ijms22105361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 12/28/2022] Open
Abstract
Lowe syndrome and Dent II disease are X-linked monogenetic diseases characterised by a renal reabsorption defect in the proximal tubules and caused by mutations in the OCRL gene, which codes for an inositol-5-phosphatase. The life expectancy of patients suffering from Lowe syndrome is largely reduced because of the development of chronic kidney disease and related complications. There is a need for physiological human in vitro models for Lowe syndrome/Dent II disease to study the underpinning disease mechanisms and to identify and characterise potential drugs and drug targets. Here, we describe a proximal tubule organ on chip model combining a 3D tubule architecture with fluid flow shear stress that phenocopies hallmarks of Lowe syndrome/Dent II disease. We demonstrate the high suitability of our in vitro model for drug target validation. Furthermore, using this model, we demonstrate that proximal tubule cells lacking OCRL expression upregulate markers typical for epithelial–mesenchymal transition (EMT), including the transcription factor SNAI2/Slug, and show increased collagen expression and deposition, which potentially contributes to interstitial fibrosis and disease progression as observed in Lowe syndrome and Dent II disease.
Collapse
|
12
|
Kowal TJ, Prosseda PP, Ning K, Wang B, Alvarado J, Sendayen BE, Jabbehdari S, Stamer WD, Hu Y, Sun Y. Optogenetic Modulation of Intraocular Pressure in a Glucocorticoid-Induced Ocular Hypertension Mouse Model. Transl Vis Sci Technol 2021; 10:10. [PMID: 34111256 PMCID: PMC8107493 DOI: 10.1167/tvst.10.6.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/22/2021] [Indexed: 01/03/2023] Open
Abstract
Purpose Steroid-induced glaucoma is a common form of secondary open angle glaucoma characterized by ocular hypertension (elevated intraocular pressure [IOP]) in response to prolonged glucocorticoid exposure. Elevated IOP occurs with increased outflow resistance and altered trabecular meshwork (TM) function. Recently, we used an optogenetic approach in TM to regulate the 5-phosphatase, OCRL, which contributes to regulating PI(4,5)P2 levels. Here, we applied this system with the aim of reversing compromised outflow function in a steroid-induced ocular hypertension mouse model. Methods Elevated IOP was induced by chronic subconjunctival dexamethasone injections in wild-type C57Bl/6j mice. AAV2 viruses containing optogenetic modules of cryptochrome 2 (Cry2)-OCRL-5ptase and CIBN-GFP were injected into the anterior chamber. Four weeks after viral expression and dexamethasone exposure, IOP was measured by tonometer and outflow facility was measured by perfusion apparatus. Human TM cells were treated with dexamethasone, stimulated by light and treated with rhodamine-phalloidin to analyze actin structure. Results Dexamethasone treatment elevated IOP and decreased outflow facility in wild-type mice. Optogenetic constructs were expressed in the TM of mouse eyes. Light stimulation caused CRY2-OCRL-5ptase to translocate to plasma membrane (CIBN-CAAX-GFP) and cilia (CIBN-SSTR3-GFP) in TM cells, which rescued the IOP and outflow facility. In addition, aberrant actin structures formed by dexamethasone treatment were reduced by optogenetic stimulation in human TM cells in culture. Conclusions Subcellular targeting of inositol phosphatases to remove PIP2 represents a promising strategy to reverse defective TM function in steroid-induced ocular hypertension. Translational Relevance Targeted modulation of OCRL may be used to decrease steroid-induced elevated IOP.
Collapse
Affiliation(s)
- Tia J. Kowal
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Philipp P. Prosseda
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Ke Ning
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Biao Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jorge Alvarado
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Brent E. Sendayen
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sayena Jabbehdari
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - W. Daniel Stamer
- Duke Eye Center, Department of Ophthalmology, Duke University, Durham, NC, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
- Palo Alto Veterans Administration, Palo Alto, CA, USA
| |
Collapse
|
13
|
Role of protons in calcium signaling. Biochem J 2021; 478:895-910. [PMID: 33635336 DOI: 10.1042/bcj20200971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 02/03/2023]
Abstract
Thirty-six years after the publication of the important article by Busa and Nuccitelli on the variability of intracellular pH (pHi) and the interdependence of pHi and intracellular Ca2+ concentration ([Ca2+]i), little research has been carried out on pHi and calcium signaling. Moreover, the results appear to be contradictory. Some authors claim that the increase in [Ca2+]i is due to a reduction in pHi, others that it is caused by an increase in pHi. The reasons for these conflicting results have not yet been discussed and clarified in an exhaustive manner. The idea that variations in pHi are insignificant, because cellular buffers quickly stabilize the pHi, may be a limiting and fundamentally wrong concept. In fact, it has been shown that protons can move and react in the cell before they are neutralized. Variations in pHi have a remarkable impact on [Ca2+]i and hence on some of the basic biochemical mechanisms of calcium signaling. This paper focuses on the possible triggering role of protons during their short cellular cycle and it suggests a new hypothesis for an IP3 proton dependent mechanism of action.
Collapse
|
14
|
The emerging roles of OSBP-related proteins in cancer: Impacts through phosphoinositide metabolism and protein-protein interactions. Biochem Pharmacol 2021; 196:114455. [PMID: 33556339 DOI: 10.1016/j.bcp.2021.114455] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 01/04/2023]
Abstract
Oxysterol-binding protein -related proteins (ORPs) form a large family of intracellular lipid binding/transfer proteins. A number of ORPs are implicated in inter-organelle lipid transfer over membrane contacts sites, their mode of action involving in several cases the transfer of two lipids in opposite directions, termed countercurrent lipid transfer. A unifying feature appears to be the capacity to bind phosphatidylinositol polyphosphates (PIPs). These lipids are in some cases transported by ORPs from one organelle to another to drive the transfer of another lipid against its concentration gradient, while they in other cases may act as allosteric regulators of ORPs, or an ORP may introduce a PIP to an enzyme for catalysis. Dysregulation of several ORP family members is implicated in cancers, ORP3, -4, -5 and -8 being thus far the most studied examples. The most likely mechanisms underlying their associations with malignant growth are (i) impacts on PIP-mediated signaling events resulting in altered Ca2+ homeostasis, bioenergetics, cell survival, proliferation, and migration, (ii) protein-protein interactions affecting the activity of signaling factors, and (iii) modification of cellular lipid transport in a way that facilitates the proliferation of malignant cells. In this review I discuss the existing functional evidence for the involvement of ORPs in cancerous growth, discuss the findings in the light of the putative mechanisms outlined above and the possibility of employing ORPs as targets of anti-cancer therapy.
Collapse
|
15
|
Schurmans S, Vande Catsyne CA, Desmet C, Moës B. The phosphoinositide 5-phosphatase INPP5K: From gene structure to in vivo functions. Adv Biol Regul 2021; 79:100760. [PMID: 33060052 DOI: 10.1016/j.jbior.2020.100760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/05/2020] [Accepted: 10/09/2020] [Indexed: 06/11/2023]
Abstract
INPP5K (Inositol Polyphosphate 5-Phosphatase K, or SKIP (for Skeletal muscle and Kidney enriched Inositol Phosphatase) is a member of the phosphoinositide 5-phosphatases family. Its protein structure is comprised of a N-terminal catalytic domain which hydrolyses both PtdIns(4,5)P2 and PtdIns(3,4,5)P3, followed by a SKICH domain at the C-terminus which is responsible for protein-protein interactions and subcellular localization of INPP5K. Strikingly, INPP5K is mostly concentrated in the endoplasmic reticulum, although it is also detected at the plasma membrane, in the cytosol and the nucleus. Recently, mutations in INPP5K have been detected in patients with a rare form of autosomal recessive congenital muscular dystrophy with cataract, short stature and intellectual disability. INPP5K functions extend from control of insulin signaling, endoplasmic reticulum stress response and structural integrity, myoblast differentiation, cytoskeleton organization, cell adhesion and migration, renal osmoregulation, to cancer. The goal of this review is thus to summarize and comment recent and less recent data in the literature on INPP5K, in particular on the structure, expression, intracellular localization, interactions and functions of this specific member of the 5-phosphatases family.
Collapse
Affiliation(s)
- Stéphane Schurmans
- Laboratoire de Génétique Fonctionnelle, GIGA-Research Centre, Building B34, CHU Sart-Tilman, Université de Liège, Avenue de l'Hôpital 11, 4000-Liège, Belgium; Secteur de Biochimie Métabolique Vétérinaire, Département des Sciences Fonctionnelles, Faculté de Médecine Vétérinaire, Building B42, Université de Liège, Quartier Vallée 2, Avenue de Cureghem 7A-7D, 4000-Liège, Belgium.
| | - Charles-Andrew Vande Catsyne
- Laboratoire de Génétique Fonctionnelle, GIGA-Research Centre, Building B34, CHU Sart-Tilman, Université de Liège, Avenue de l'Hôpital 11, 4000-Liège, Belgium
| | - Christophe Desmet
- Laboratory of Cellular and Molecular Immunology, GIGA-Research Centre, Building B34, CHU Sart-Tilman, Université de Liège, Avenue de l'Hôpital 11, 4000-Liège, Belgium
| | - Bastien Moës
- Laboratoire de Génétique Fonctionnelle, GIGA-Research Centre, Building B34, CHU Sart-Tilman, Université de Liège, Avenue de l'Hôpital 11, 4000-Liège, Belgium
| |
Collapse
|
16
|
Paesmans J, Martin E, Deckers B, Berghmans M, Sethi R, Loeys Y, Pardon E, Steyaert J, Verstreken P, Galicia C, Versées W. A structure of substrate-bound Synaptojanin1 provides new insights in its mechanism and the effect of disease mutations. eLife 2020; 9:64922. [PMID: 33349335 PMCID: PMC7781601 DOI: 10.7554/elife.64922] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Synaptojanin1 (Synj1) is a phosphoinositide phosphatase, important in clathrin uncoating during endocytosis of presynaptic vesicles. It was identified as a potential drug target for Alzheimer's disease, Down syndrome, and TBC1D24-associated epilepsy, while also loss-of-function mutations in Synj1 are associated with epilepsy and Parkinson's disease. Despite its involvement in a range of disorders, structural, and detailed mechanistic information regarding the enzyme is lacking. Here, we report the crystal structure of the 5-phosphatase domain of Synj1. Moreover, we also present a structure of this domain bound to the substrate diC8-PI(3,4,5)P3, providing the first image of a 5-phosphatase with a trapped substrate in its active site. Together with an analysis of the contribution of the different inositide phosphate groups to catalysis, these structures provide new insights in the Synj1 mechanism. Finally, we analysed the effect of three clinical missense mutations (Y793C, R800C, Y849C) on catalysis, unveiling the molecular mechanisms underlying Synj1-associated disease.
Collapse
Affiliation(s)
- Jone Paesmans
- VIB-VUB Center for Structural Biology, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ella Martin
- VIB-VUB Center for Structural Biology, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Babette Deckers
- VIB-VUB Center for Structural Biology, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marjolijn Berghmans
- VIB-VUB Center for Structural Biology, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ritika Sethi
- VIB-VUB Center for Structural Biology, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick Loeys
- VIB-VUB Center for Structural Biology, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Els Pardon
- VIB-VUB Center for Structural Biology, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jan Steyaert
- VIB-VUB Center for Structural Biology, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Patrik Verstreken
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Christian Galicia
- VIB-VUB Center for Structural Biology, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wim Versées
- VIB-VUB Center for Structural Biology, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
17
|
Csolle MP, Ooms LM, Papa A, Mitchell CA. PTEN and Other PtdIns(3,4,5)P 3 Lipid Phosphatases in Breast Cancer. Int J Mol Sci 2020; 21:ijms21239189. [PMID: 33276499 PMCID: PMC7730566 DOI: 10.3390/ijms21239189] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
The phosphoinositide 3-kinase (PI3K)/AKT signalling pathway is hyperactivated in ~70% of breast cancers. Class I PI3K generates PtdIns(3,4,5)P3 at the plasma membrane in response to growth factor stimulation, leading to AKT activation to drive cell proliferation, survival and migration. PTEN negatively regulates PI3K/AKT signalling by dephosphorylating PtdIns(3,4,5)P3 to form PtdIns(4,5)P2. PtdIns(3,4,5)P3 can also be hydrolysed by the inositol polyphosphate 5-phosphatases (5-phosphatases) to produce PtdIns(3,4)P2. Interestingly, while PTEN is a bona fide tumour suppressor and is frequently mutated/lost in breast cancer, 5-phosphatases such as PIPP, SHIP2 and SYNJ2, have demonstrated more diverse roles in regulating mammary tumourigenesis. Reduced PIPP expression is associated with triple negative breast cancers and reduced relapse-free and overall survival. Although PIPP depletion enhances AKT phosphorylation and supports tumour growth, this also inhibits cell migration and metastasis in vivo, in a breast cancer oncogene-driven murine model. Paradoxically, SHIP2 and SYNJ2 are increased in primary breast tumours, which correlates with invasive disease and reduced survival. SHIP2 or SYNJ2 overexpression promotes breast tumourigenesis via AKT-dependent and independent mechanisms. This review will discuss how PTEN, PIPP, SHIP2 and SYNJ2 distinctly regulate multiple functional targets, and the mechanisms by which dysregulation of these distinct phosphoinositide phosphatases differentially affect breast cancer progression.
Collapse
|
18
|
Conduit SE, Vanhaesebroeck B. Phosphoinositide lipids in primary cilia biology. Biochem J 2020; 477:3541-3565. [PMID: 32970140 PMCID: PMC7518857 DOI: 10.1042/bcj20200277] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/30/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022]
Abstract
Primary cilia are solitary signalling organelles projecting from the surface of most cell types. Although the ciliary membrane is continuous with the plasma membrane it exhibits a unique phospholipid composition, a feature essential for normal cilia formation and function. Recent studies have illustrated that distinct phosphoinositide lipid species localise to specific cilia subdomains, and have begun to build a 'phosphoinositide map' of the cilium. The abundance and localisation of phosphoinositides are tightly regulated by the opposing actions of lipid kinases and lipid phosphatases that have also been recently discovered at cilia. The critical role of phosphoinositides in cilia biology is highlighted by the devastating consequences of genetic defects in cilia-associated phosphoinositide regulatory enzymes leading to ciliopathy phenotypes in humans and experimental mouse and zebrafish models. Here we provide a general introduction to primary cilia and the roles phosphoinositides play in cilia biology. In addition to increasing our understanding of fundamental cilia biology, this rapidly expanding field may inform novel approaches to treat ciliopathy syndromes caused by deregulated phosphoinositide metabolism.
Collapse
Affiliation(s)
- Sarah E. Conduit
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, U.K
| | - Bart Vanhaesebroeck
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6BT, U.K
| |
Collapse
|
19
|
Bozelli JC, Epand RM. Specificity of Acyl Chain Composition of Phosphatidylinositols. Proteomics 2020; 19:e1900138. [PMID: 31381272 DOI: 10.1002/pmic.201900138] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/30/2019] [Indexed: 01/15/2023]
Abstract
Phosphatidylinositol (PI) lipids have a predominance of a single molecular species present through the organism. In healthy mammals this molecular species is 1-stearoyl-2-arachidonoyl (18:0/20:4) PI. Although the importance of PI lipids for cell physiology has long been appreciated, less is known about the biological role of enriching PI lipids with 18:0/20:4 acyl chains. In conditions with dysfunctional lipid metabolism, the predominance of 18:0/20:4 acyl chains is lost. Recently, molecular mechanisms underpinning the enrichment or alteration of these acyl chains in PI lipids have begun to emerge. In the majority of the cases a common feature is the presence of enzymes bearing substrate acyl chain specificity. However, in cancer cells, it has been shown that one (not the only) of the mechanisms responsible for the loss in this acyl chain enrichment is mutation on the transcription factor p53 gene, which is one of the most highly mutated genes in cancers. There is a compelling need for a global picture of the specificity of the acyl chain composition of PIs. This can be possible once high-resolution spatio-temporal information is gathered in a cellular context; which can ultimately lead to potential novel targets to combat conditions with altered PI acyl chain profiles.
Collapse
Affiliation(s)
- José Carlos Bozelli
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences Centre, Hamilton, Ontario, L8S 4K1, Canada
| | - Richard M Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences Centre, Hamilton, Ontario, L8S 4K1, Canada
| |
Collapse
|
20
|
The Role of Membrane Surface Charge in Phagocytosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1246:43-54. [DOI: 10.1007/978-3-030-40406-2_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
21
|
Nakada-Tsukui K, Watanabe N, Maehama T, Nozaki T. Phosphatidylinositol Kinases and Phosphatases in Entamoeba histolytica. Front Cell Infect Microbiol 2019; 9:150. [PMID: 31245297 PMCID: PMC6563779 DOI: 10.3389/fcimb.2019.00150] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Phosphatidylinositol (PtdIns) metabolism is indispensable in eukaryotes. Phosphoinositides (PIs) are phosphorylated derivatives of PtdIns and consist of seven species generated by reversible phosphorylation of the inositol moieties at the positions 3, 4, and 5. Each of the seven PIs has a unique subcellular and membrane domain distribution. In the enteric protozoan parasite Entamoeba histolytica, it has been previously shown that the PIs phosphatidylinositol 3-phosphate (PtdIns3P), PtdIns(4,5)P2, and PtdIns(3,4,5)P3 are localized to phagosomes/phagocytic cups, plasma membrane, and phagocytic cups, respectively. The localization of these PIs in E. histolytica is similar to that in mammalian cells, suggesting that PIs have orthologous functions in E. histolytica. In contrast, the conservation of the enzymes that metabolize PIs in this organism has not been well-documented. In this review, we summarized the full repertoire of the PI kinases and PI phosphatases found in E. histolytica via a genome-wide survey of the current genomic information. E. histolytica appears to have 10 PI kinases and 23 PI phosphatases. It has a panel of evolutionarily conserved enzymes that generate all the seven PI species. However, class II PI 3-kinases, type II PI 4-kinases, type III PI 5-phosphatases, and PI 4P-specific phosphatases are not present. Additionally, regulatory subunits of class I PI 3-kinases and type III PI 4-kinases have not been identified. Instead, homologs of class I PI 3-kinases and PTEN, a PI 3-phosphatase, exist as multiple isoforms, which likely reflects that elaborate signaling cascades mediated by PtdIns(3,4,5)P3 are present in this organism. There are several enzymes that have the nuclear localization signal: one phosphatidylinositol phosphate (PIP) kinase, two PI 3-phosphatases, and one PI 5-phosphatase; this suggests that PI metabolism also has conserved roles related to nuclear functions in E. histolytica, as it does in model organisms.
Collapse
Affiliation(s)
- Kumiko Nakada-Tsukui
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Natsuki Watanabe
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan.,Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Tomohiko Maehama
- Division of Molecular and Cellular Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Tomoyoshi Nozaki
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
22
|
Ijuin T. Phosphoinositide phosphatases in cancer cell dynamics-Beyond PI3K and PTEN. Semin Cancer Biol 2019; 59:50-65. [PMID: 30922959 DOI: 10.1016/j.semcancer.2019.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 03/18/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022]
Abstract
Phosphoinositides are a group of lipids that regulate intracellular signaling and subcellular biological events. The signaling by phosphatidylinositol-3,4,5-trisphosphate and Akt mediates the action of growth factors that are essential for cell proliferation, gene transcription, cell migration, and polarity. The hyperactivation of this signaling has been identified in different cancer cells; and, it has been implicated in oncogenic transformation and cancer cell malignancy. Recent studies have argued the role of phosphoinositides in cancer cell dynamics, including actin cytoskeletal rearrangement at the plasma membrane and the organization of intracellular compartments. The focus of this review is to summarize the impact of the activities of phosphoinositide phosphatases on intracellular signaling related to cancer cell dynamics and to discuss how the abnormalities in the activities of the enzymes alter the levels of phosphoinositides in cancer cells.
Collapse
Affiliation(s)
- Takeshi Ijuin
- Division of Biochemistry, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chu-o, Kobe 650-0017, Japan.
| |
Collapse
|
23
|
IPIP27 Coordinates PtdIns(4,5)P 2 Homeostasis for Successful Cytokinesis. Curr Biol 2019; 29:775-789.e7. [PMID: 30799246 PMCID: PMC6408333 DOI: 10.1016/j.cub.2019.01.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 12/03/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
During cytokinesis, an actomyosin contractile ring drives the separation of the two daughter cells. A key molecule in this process is the inositol lipid PtdIns(4,5)P2, which recruits numerous factors to the equatorial region for contractile ring assembly. Despite the importance of PtdIns(4,5)P2 in cytokinesis, the regulation of this lipid in cell division remains poorly understood. Here, we identify a role for IPIP27 in mediating cellular PtdIns(4,5)P2 homeostasis. IPIP27 scaffolds the inositol phosphatase oculocerebrorenal syndrome of Lowe (OCRL) by coupling it to endocytic BAR domain proteins. Loss of IPIP27 causes accumulation of PtdIns(4,5)P2 on aberrant endomembrane vacuoles, mislocalization of the cytokinetic machinery, and extensive cortical membrane blebbing. This phenotype is observed in Drosophila and human cells and can result in cytokinesis failure. We have therefore identified IPIP27 as a key modulator of cellular PtdIns(4,5)P2 homeostasis required for normal cytokinesis. The results indicate that scaffolding of inositol phosphatase activity is critical for maintaining PtdIns(4,5)P2 homeostasis and highlight a critical role for this process in cell division. IPIP27 scaffolds the inositol phosphatase OCRL via coupling to BAR domain proteins IPIP27 scaffolding of OCRL is critical for cellular PtdIns(4,5)P2 homeostasis IPIP27 is required for cortical actin and membrane stability during cytokinesis IPIP27 function is conserved from flies to humans
Collapse
|
24
|
Ramos AR, Ghosh S, Dedobbeleer M, Robe PA, Rogister B, Erneux C. Lipid phosphatases SKIP and SHIP2 regulate fibronectin-dependent cell migration in glioblastoma. FEBS J 2019; 286:1120-1135. [PMID: 30695232 DOI: 10.1111/febs.14769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 11/08/2018] [Accepted: 01/25/2019] [Indexed: 12/19/2022]
Abstract
Cell migration is an important process that occurs during development and has also been linked to the motility of cancer cells. Cytoskeleton reorganization takes place in the migration process leading to lamellipodia formation. Understanding the molecular underpinnings of cell migration is particularly important in studies of glioblastoma, a highly invasive and aggressive cancer type. Two members of the phosphoinositide 5-phosphatase family, SKIP and SHIP2, have been associated with cell migration in glioblastoma; however, the precise role these enzymes play in the process-and whether they work in concert-remains unclear. Here, we compared phosphoinositide 5-phosphatases expression in glioblastoma primary cells and cell lines and showed that SHIP2 and SKIP expression greatly varies between different cell types, while OCRL, another phosphoinositide 5-phosphatase, is constitutively expressed. Upon adhesion of U-251 MG cells to fibronectin, SHIP2, SKIP, and PI(4,5)P2 colocalized in membrane ruffles. Upregulation of PI(4,5)P2 was observed in SKIP-depleted U-251 MG cells compared to control cells, but only when cells were adhered to fibronectin. Both PTEN-deficient (U-251) and PTEN-containing (LN229) glioblastoma cells showed a decrease in cell migration velocity in response to SKIP downregulation. Moreover, a SHIP2 catalytic inhibitor lowered cell migration velocity in the U-251 MG cell line. We conclude that integrin activation in U-251 cells leads to colocalization of both SKIP and SHIP2 in ruffles, where they act as potential drivers of cell migration. Depending on their expression levels in glioblastoma, phosphoinositide 5-phosphatases could cooperate and synergize in the regulation of cell migration and adhesion.
Collapse
Affiliation(s)
| | | | | | - Pierre A Robe
- Department of Neurology and Neurosurgery, Utrecht University Medical Center, The Netherlands
| | - Bernard Rogister
- GIGA-Neurosciences Research Center, Université de Liège, Belgium
| | | |
Collapse
|
25
|
Abstract
Phosphoinositides (PIs) play pivotal roles in the regulation of many biological processes. The quality and quantity of PIs is regulated in time and space by the activity of PI kinases and PI phosphatases. The number of PI-metabolizing enzymes exceeds the number of PIs with, in many cases, more than one enzyme controlling the same biochemical step. This would suggest that the PI system has an intrinsic ability to buffer and compensate for the absence of a specific enzymatic activity. However, there are several examples of severe inherited human diseases caused by mutations in one of the PI enzymes, although other enzymes with the same activity are fully functional. The kidney depends strictly on PIs for physiological processes, such as cell polarization, filtration, solute reabsorption, and signal transduction. Indeed, alteration of the PI system in the kidney very often results in pathological conditions, both inherited and acquired. Most of the knowledge of the roles that PIs play in the kidney comes from the study of KO animal models for genes encoding PI enzymes and from the study of human genetic diseases, such as Lowe syndrome/Dent disease 2 and Joubert syndrome, caused by mutations in the genes encoding the PI phosphatases, OCRL and INPP5E, respectively.
Collapse
Affiliation(s)
- Leopoldo Staiano
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy
| | - Maria Antonietta De Matteis
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy .,University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
26
|
Dong R, Zhu T, Benedetti L, Gowrishankar S, Deng H, Cai Y, Wang X, Shen K, De Camilli P. The inositol 5-phosphatase INPP5K participates in the fine control of ER organization. J Cell Biol 2018; 217:3577-3592. [PMID: 30087126 PMCID: PMC6168264 DOI: 10.1083/jcb.201802125] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/26/2018] [Accepted: 07/11/2018] [Indexed: 12/19/2022] Open
Abstract
INPP5K (SKIP) is an inositol 5-phosphatase that localizes in part to the endoplasmic reticulum (ER). We show that recruitment of INPP5K to the ER is mediated by ARL6IP1, which shares features of ER-shaping proteins. Like ARL6IP1, INPP5K is preferentially localized in ER tubules and enriched, relative to other ER resident proteins (Sec61β, VAPB, and Sac1), in newly formed tubules that grow along microtubule tracks. Depletion of either INPP5K or ARL6IP1 results in the increase of ER sheets. In a convergent but independent study, a screen for mutations affecting the distribution of the ER network in dendrites of the PVD neurons of Caenorhabditis elegans led to the isolation of mutants in CIL-1, which encodes the INPP5K worm orthologue. The mutant phenotype was rescued by expression of wild type, but not of catalytically inactive CIL-1. Our results reveal an unexpected role of an ER localized polyphosphoinositide phosphatase in the fine control of ER network organization.
Collapse
Affiliation(s)
- Rui Dong
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, New Haven, CT
| | - Ting Zhu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lorena Benedetti
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, New Haven, CT
| | - Swetha Gowrishankar
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, New Haven, CT
| | - Huichao Deng
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiying Cai
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, New Haven, CT
| | - Xiangming Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Kang Shen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Department of Biology, Stanford University School of Medicine, Stanford, CA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, New Haven, CT
- Kavli Institute for Neurosciences, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
27
|
Ramos AR, Ghosh S, Erneux C. The impact of phosphoinositide 5-phosphatases on phosphoinositides in cell function and human disease. J Lipid Res 2018; 60:276-286. [PMID: 30194087 DOI: 10.1194/jlr.r087908] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/01/2018] [Indexed: 02/06/2023] Open
Abstract
Phosphoinositides (PIs) are recognized as major signaling molecules in many different functions of eukaryotic cells. PIs can be dephosphorylated by multiple phosphatase activities at the 5-, 4-, and 3- positions. Human PI 5-phosphatases belong to a family of 10 members. Except for inositol polyphosphate 5-phosphatase A, they all catalyze the dephosphorylation of PI(4,5)P2 and/or PI(3,4,5)P3 at the 5- position. PI 5-phosphatases thus directly control the levels of PI(3,4,5)P3 and participate in the fine-tuning regulatory mechanisms of PI(3,4)P2 and PI(4,5)P2 Second messenger functions have been demonstrated for PI(3,4)P2 in invadopodium maturation and lamellipodia formation. PI 5-phosphatases can use several substrates on isolated enzymes, and it has been challenging to establish their real substrate in vivo. PI(4,5)P2 has multiple functions in signaling, including interacting with scaffold proteins, ion channels, and cytoskeleton proteins. PI 5-phosphatase isoenzymes have been individually implicated in human diseases, such as the oculocerebrorenal syndrome of Lowe, through mechanisms that include lipid control. Oncogenic and tumor-suppressive functions of PI 5-phosphatases have also been reported in different cell contexts. The mechanisms responsible for genetic diseases and for oncogenic or tumor-suppressive functions are not fully understood. The regulation of PI 5-phosphatases is thus crucial in understanding cell functions.
Collapse
Affiliation(s)
- Ana Raquel Ramos
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Somadri Ghosh
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Christophe Erneux
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, 1070 Brussels, Belgium
| |
Collapse
|
28
|
Ramos AR, Elong Edimo W, Erneux C. Phosphoinositide 5-phosphatase activities control cell motility in glioblastoma: Two phosphoinositides PI(4,5)P2 and PI(3,4)P2 are involved. Adv Biol Regul 2018; 67:40-48. [PMID: 28916189 DOI: 10.1016/j.jbior.2017.09.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/01/2017] [Accepted: 09/01/2017] [Indexed: 05/15/2023]
Abstract
Inositol polyphosphate 5-phosphatases or phosphoinositide 5-phosphatases (PI 5-phosphatases) are enzymes that can act on soluble inositol phosphates and/or phosphoinositides (PIs). Several PI 5-phosphatases have been linked to human genetic diseases, in particular the Lowe protein or OCRL which is mutated in the Lowe syndrome. There are 10 different members of this family and 9 of them can use PIs as substrate. One of these substrates, PI(3,4,5)P3 binds to specific PH domains and recruits as effectors specific proteins to signaling complexes. Protein kinase B is one target protein and activation of the kinase will have a major impact on cell proliferation, survival and cell metabolism. Two other PIs, PI(4,5)P2 and PI(3,4)P2, are produced or used as substrates of PI 5-phosphatases (OCRL, INPP5B, SHIP1/2, SYNJ1/2, INPP5K, INPP5J, INPP5E). The inositol lipids may influence many aspects of cytoskeletal organization, lamellipodia formation and F-actin polymerization. PI 5-phosphatases have been reported to control cell migration, adhesion, polarity and cell invasion particularly in cancer cells. In glioblastoma, reducing SHIP2 expression can positively or negatively affect the speed of cell migration depending on the glioblastoma cell type. The two PI 5-phosphatases SHIP2 or SKIP could be localized at the plasma membrane and can reduce either PI(3,4,5)P3 or PI(4,5)P2 abundance. In the glioblastoma 1321 N1 cells, SHIP2 controls plasma membrane PI(4,5)P2 thereby participating in the control of cell migration.
Collapse
Affiliation(s)
- Ana Raquel Ramos
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, Campus Erasme, Bldg C, 808 Route de Lennik, 1070 Brussels, Belgium
| | - William's Elong Edimo
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, Campus Erasme, Bldg C, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Christophe Erneux
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, Campus Erasme, Bldg C, 808 Route de Lennik, 1070 Brussels, Belgium.
| |
Collapse
|
29
|
Choy CH, Han BK, Botelho RJ. Phosphoinositide Diversity, Distribution, and Effector Function: Stepping Out of the Box. Bioessays 2017; 39. [PMID: 28977683 DOI: 10.1002/bies.201700121] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/31/2017] [Indexed: 12/26/2022]
Abstract
Phosphoinositides (PtdInsPs) modulate a plethora of functions including signal transduction and membrane trafficking. PtdInsPs are thought to consist of seven interconvertible species that localize to a specific organelle, to which they recruit a set of cognate effector proteins. Here, in reviewing the literature, we argue that this model needs revision. First, PtdInsPs can carry a variety of acyl chains, greatly boosting their molecular diversity. Second, PtdInsPs are more promiscuous in their localization than is usually acknowledged. Third, PtdInsP interconversion is likely achieved through kinase-phosphatase enzyme complexes that coordinate their activities and channel substrates without affecting bulk substrate population. Additionally, we contend that despite hundreds of PtdInsP effectors, our attention is biased toward few proteins. Lastly, we recognize that PtdInsPs can act to nucleate coincidence detection at the effector level, as in PDK1 and Akt. Overall, better integrated models of PtdInsP regulation and function are not only possible but needed.
Collapse
Affiliation(s)
- Christopher H Choy
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada M5B2K3.,Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada M5B2K3
| | - Bong-Kwan Han
- The Intelligent Synthetic Biology Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Roberto J Botelho
- Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada M5B2K3.,Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada M5B2K3
| |
Collapse
|
30
|
Flaig TW, Salzmann-Sullivan M, Su LJ, Zhang Z, Joshi M, Gijón MA, Kim J, Arcaroli JJ, Van Bokhoven A, Lucia MS, La Rosa FG, Schlaepfer IR. Lipid catabolism inhibition sensitizes prostate cancer cells to antiandrogen blockade. Oncotarget 2017; 8:56051-56065. [PMID: 28915573 PMCID: PMC5593544 DOI: 10.18632/oncotarget.17359] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/10/2017] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer (PCa) is the most common malignancy among Western men and the second leading-cause of cancer related deaths. For men who develop metastatic castration resistant PCa (mCRPC), survival is limited, making the identification of novel therapies for mCRPC critical. We have found that deficient lipid oxidation via carnitine palmitoyltransferase (CPT1) results in decreased growth and invasion, underscoring the role of lipid oxidation to fuel PCa growth. Using immunohistochemistry we have found that the CPT1A isoform is abundant in PCa compared to benign tissue (n=39, p<0.001) especially in those with high-grade tumors. Since lipid oxidation is stimulated by androgens, we have evaluated the synergistic effects of combining CPT1A inhibition and anti-androgen therapy. Mechanistically, we have found that decreased CPT1A expression is associated with decreased AKT content and activation, likely driven by a breakdown of membrane phospholipids and activation of the INPP5K phosphatase. This results in increased androgen receptor (AR) action and increased sensitivity to the anti-androgen enzalutamide. To better understand the clinical implications of these findings, we have evaluated fat oxidation inhibitors (etomoxir, ranolazine and perhexiline) in combination with enzalutamide in PCa cell models. We have observed a robust growth inhibitory effect of the combinations, including in enzalutamide-resistant cells and mouse TRAMPC1 cells, a more neuroendocrine PCa model. Lastly, using a xenograft mouse model, we have observed decreased tumor growth with a systemic combination treatment of enzalutamide and ranolazine. In conclusion, our results show that improved anti-cancer efficacy can be achieved by co-targeting the AR axis and fat oxidation via CPT1A, which may have clinical implications, especially in the mCRPC setting.
Collapse
Affiliation(s)
- Thomas W. Flaig
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Maren Salzmann-Sullivan
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lih-Jen Su
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Zhiyong Zhang
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Molishree Joshi
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Miguel A. Gijón
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jihye Kim
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - John J. Arcaroli
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Adrie Van Bokhoven
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - M. Scott Lucia
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Francisco G. La Rosa
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Isabel R. Schlaepfer
- Division of Medical Oncology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
31
|
Abstract
Lowe syndrome is an X-linked disease that is characterized by congenital cataracts, central hypotonia, intellectual disability and renal Fanconi syndrome. The disease is caused by mutations in OCRL, which encodes an inositol polyphosphate 5-phosphatase (OCRL) that acts on phosphoinositides - quantitatively minor constituents of cell membranes that are nonetheless pivotal regulators of intracellular trafficking. In this Review we summarize the considerable progress made over the past decade in understanding the cellular roles of OCRL in regulating phosphoinositide balance along the endolysosomal pathway, a fundamental system for the reabsorption of proteins and solutes by proximal tubular cells. We discuss how studies of OCRL have led to important discoveries about the basic mechanisms of membrane trafficking and describe the key features and limitations of the currently available animal models of Lowe syndrome. Mutations in OCRL can also give rise to a milder pathology, Dent disease 2, which is characterized by renal Fanconi syndrome in the absence of extrarenal pathologies. Understanding how mutations in OCRL give rise to two clinical entities with differing extrarenal manifestations represents an opportunity to identify molecular pathways that could be targeted to develop treatments for these conditions.
Collapse
|
32
|
Segawa T, Hazeki K, Nigorikawa K, Nukuda A, Tanizawa T, Miyamoto K, Morioka S, Hazeki O. Inhibitory receptor FcγRIIb mediates the effects of IgG on a phagosome acidification and a sequential dephosphorylation system comprising SHIPs and Inpp4a. Innate Immun 2017; 23:401-409. [DOI: 10.1177/1753425917701553] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The relative abundance of phosphoinositide (PI) species on the phagosome membrane fluctuates over the course of phagocytosis. PtdIns(3,4,5)P3 and PtdIns(3,4)P2 rapidly increase in the forming of the phagocytic cup, following which they disappear after sealing of the cup. In the present study, we monitored the clearance of these PI species using the enhanced green fluorescent protein-fused pleckstrin homology domain of Akt, a fluorescence probe that binds both PtdIns(3,4,5)P3 and PtdIns(3,4)P2 in Raw 264.7 macrophages. The clearance of PIs was much faster when the phagocytosed particles were coated with IgG. The effect of IgG was not observed in the macrophages deficient in FcγRIIb, an inhibitory IgG receptor. To identify the lipid phosphatases responsible for the FcγRIIb-accelerated PI clearance, we prepared a panel of lipid phosphatase-deficient cells. The lack of a PI 5-phosphatase Src homology 2 domain-containing inositol-5-phosphatase (SHIP)1 or SHIP2 impaired the FcγRIIb-accelerated clearance of PIs. The lack of a PI 4-phosphatase Inpp4a also impaired the accelerated PIs clearance. In the FcγRIIb- and Inpp4a-deficient cells, acidification of the formed phagosome was slowed. These results suggested that FcγRIIb drives the sequential dephosphorylation system comprising SHIPs and Inpp4a, and accelerates phagosome acidification.
Collapse
Affiliation(s)
- Tomohiro Segawa
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kaoru Hazeki
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kiyomi Nigorikawa
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Atsuko Nukuda
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoki Tanizawa
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kenshiro Miyamoto
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shin Morioka
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Osamu Hazeki
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
33
|
Wiessner M, Roos A, Munn CJ, Viswanathan R, Whyte T, Cox D, Schoser B, Sewry C, Roper H, Phadke R, Marini Bettolo C, Barresi R, Charlton R, Bönnemann CG, Abath Neto O, Reed UC, Zanoteli E, Araújo Martins Moreno C, Ertl-Wagner B, Stucka R, De Goede C, Borges da Silva T, Hathazi D, Dell’Aica M, Zahedi RP, Thiele S, Müller J, Kingston H, Müller S, Curtis E, Walter MC, Strom TM, Straub V, Bushby K, Muntoni F, Swan LE, Lochmüller H, Senderek J. Mutations in INPP5K, Encoding a Phosphoinositide 5-Phosphatase, Cause Congenital Muscular Dystrophy with Cataracts and Mild Cognitive Impairment. Am J Hum Genet 2017; 100:523-536. [PMID: 28190456 PMCID: PMC5339217 DOI: 10.1016/j.ajhg.2017.01.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/12/2017] [Indexed: 12/26/2022] Open
Abstract
Phosphoinositides are small phospholipids that control diverse cellular downstream signaling events. Their spatial and temporal availability is tightly regulated by a set of specific lipid kinases and phosphatases. Congenital muscular dystrophies are hereditary disorders characterized by hypotonia and weakness from birth with variable eye and central nervous system involvement. In individuals exhibiting congenital muscular dystrophy, early-onset cataracts, and mild intellectual disability but normal cranial magnetic resonance imaging, we identified bi-allelic mutations in INPP5K, encoding inositol polyphosphate-5-phosphatase K. Mutations impaired phosphatase activity toward the phosphoinositide phosphatidylinositol (4,5)-bisphosphate or altered the subcellular localization of INPP5K. Downregulation of INPP5K orthologs in zebrafish embryos disrupted muscle fiber morphology and resulted in abnormal eye development. These data link congenital muscular dystrophies to defective phosphoinositide 5-phosphatase activity that is becoming increasingly recognized for its role in mediating pivotal cellular mechanisms contributing to disease.
Collapse
|
34
|
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: 4.3] [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
|
35
|
Epand RM. Features of the Phosphatidylinositol Cycle and its Role in Signal Transduction. J Membr Biol 2016; 250:353-366. [PMID: 27278236 DOI: 10.1007/s00232-016-9909-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 05/26/2016] [Indexed: 01/03/2023]
Abstract
The phosphatidylinositol cycle (PI-cycle) has a central role in cell signaling. It is the major pathway for the synthesis of phosphatidylinositol and its phosphorylated forms. In addition, some lipid intermediates of the PI-cycle, including diacylglycerol and phosphatidic acid, are also important lipid signaling agents. The PI-cycle has some features that are important for the understanding of its role in the cell. As a cycle, the intermediates will be regenerated. The PI-cycle requires a large amount of metabolic energy. There are different steps of the cycle that occur in two different membranes, the plasma membrane and the endoplasmic reticulum. In order to complete the PI-cycle lipid must be transferred between the two membranes. The role of the Nir proteins in the process has recently been elucidated. The lipid intermediates of the PI-cycle are normally highly enriched with 1-stearoyl-2-arachidonoyl molecular species in mammals. This enrichment will be retained as long as the intermediates are segregated from other lipids of the cell. However, there is a significant fraction (>15 %) of lipids in the PI-cycle of normal cells that have other acyl chains. Phosphatidylinositol largely devoid of arachidonoyl chains are found in cancer cells. Phosphatidylinositol species with less unsaturation will not be as readily converted to phosphatidylinositol-3,4,5-trisphosphate, the lipid required for the activation of Akt with resulting effects on cell proliferation. Thus, the cyclical nature of the PI-cycle, its dependence on acyl chain composition and its requirement for lipid transfer between two membranes, explain many of the biological properties of this cycle.
Collapse
Affiliation(s)
- Richard M Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
| |
Collapse
|
36
|
Naguib A. Following the trail of lipids: Signals initiated by PI3K function at multiple cellular membranes. Sci Signal 2016; 9:re4. [DOI: 10.1126/scisignal.aad7885] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
37
|
Mills SJ, Silvander C, Cozier G, Trésaugues L, Nordlund P, Potter BVL. Crystal Structures of Type-II Inositol Polyphosphate 5-Phosphatase INPP5B with Synthetic Inositol Polyphosphate Surrogates Reveal New Mechanistic Insights for the Inositol 5-Phosphatase Family. Biochemistry 2016; 55:1384-97. [PMID: 26854536 PMCID: PMC4785718 DOI: 10.1021/acs.biochem.5b00838] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
The inositol polyphosphate 5-phosphatase
INPP5B hydrolyzes the
5-phosphate group from water- and lipid-soluble signaling messengers.
Two synthetic benzene and biphenyl polyphosphates (BzP/BiPhPs), simplified
surrogates of inositol phosphates and phospholipid headgroups, were
identified by thermodynamic studies as potent INPP5B ligands. The
X-ray structure of the complex between INPP5B and biphenyl 3,3′,4,4′,5,5′-hexakisphosphate
[BiPh(3,3′,4,4′,5,5′)P6, IC50 5.5 μM] was determined at 2.89 Å resolution. One inhibitor
pole locates in the phospholipid headgroup binding site and the second
solvent-exposed ring binds to the His-Tag of another INPP5B molecule,
while a molecule of inorganic phosphate is also present in the active
site. Benzene 1,2,3-trisphosphate [Bz(1,2,3)P3] [one ring
of BiPh(3,3′,4,4′,5,5′)P6] inhibits
INPP5B ca. 6-fold less potently. Co-crystallization with benzene 1,2,4,5-tetrakisphosphate
[Bz(1,2,4,5)P4, IC50 = 6.3 μM] yielded
a structure refined at 2.9 Å resolution. Conserved residues among
the 5-phosphatase family mediate interactions with Bz(1,2,4,5)P4 and BiPh(3,3′,4,4′,5,5′)P6 similar to those with the polar groups present in positions 1, 4,
5, and 6 on the inositol ring of the substrate. 5-Phosphatase specificity
most likely resides in the variable zone located close to the 2- and
3-positions of the inositol ring, offering insights to inhibitor design.
We propose that the inorganic phosphate present in the INPP5B–BiPh(3,3′,4,4′,5,5′)P6 complex mimics the postcleavage substrate 5-phosphate released
by INPP5B in the catalytic site, allowing elucidation of two new key
features in the catalytic mechanism proposed for the family of phosphoinositide
5-phosphatases: first, the involvement of the conserved Arg-451 in
the interaction with the 5-phosphate and second, identification of
the water molecule that initiates 5-phosphate hydrolysis. Our model
also has implications for the proposed “moving metal”
mechanism.
Collapse
Affiliation(s)
- Stephen J Mills
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath , Bath BA2 7AY, U.K
| | - Camilla Silvander
- Structural Genomics Consortium, Karolinska Institutet , 17177 Stockholm, Sweden
| | - Gyles Cozier
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath , Bath BA2 7AY, U.K
| | - Lionel Trésaugues
- Structural Genomics Consortium, Karolinska Institutet , 17177 Stockholm, Sweden.,Division of Biophysics, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , 17177 Stockholm, Sweden
| | - Pär Nordlund
- Structural Genomics Consortium, Karolinska Institutet , 17177 Stockholm, Sweden.,Division of Biophysics, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , 17177 Stockholm, Sweden.,School of Biological Sciences, Nanyang Technological University , 637551 Singapore
| | - Barry V L Potter
- Department of Pharmacology, University of Oxford , Mansfield Road, Oxford OX1 3QT, U.K.,Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath , Bath BA2 7AY, U.K
| |
Collapse
|
38
|
Regulation of PtdIns(3,4,5)P3/Akt signalling by inositol polyphosphate 5-phosphatases. Biochem Soc Trans 2016; 44:240-52. [DOI: 10.1042/bst20150214] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The phosphoinositide 3-kinase (PI3K) generated lipid signals, PtdIns(3,4,5)P3 and PtdIns(3,4)P2, are both required for the maximal activation of the serine/threonine kinase proto-oncogene Akt. The inositol polyphosphate 5-phosphatases (5-phosphatases) hydrolyse the 5-position phosphate from the inositol head group of PtdIns(3,4,5)P3 to yield PtdIns(3,4)P2. Extensive work has revealed several 5-phosphatases inhibit PI3K-driven Akt signalling, by decreasing PtdIns(3,4,5)P3 despite increasing cellular levels of PtdIns(3,4)P2. The roles that 5-phosphatases play in suppressing cell proliferation and transformation are slow to emerge; however, the 5-phosphatase PIPP [proline-rich inositol polyphosphate 5-phosphatase; inositol polyphosphate 5-phosphatase (INPP5J)] has recently been identified as a putative tumour suppressor in melanoma and breast cancer and SHIP1 [SH2 (Src homology 2)-containing inositol phosphatase 1] inhibits haematopoietic cell proliferation. INPP5E regulates cilia stability and INPP5E mutations have been implicated ciliopathy syndromes. This review will examine 5-phosphatase regulation of PI3K/Akt signalling, focussing on the role PtdIns(3,4,5)P3 5-phosphatases play in developmental diseases and cancer.
Collapse
|
39
|
Campa CC, Martini M, De Santis MC, Hirsch E. How PI3K-derived lipids control cell division. Front Cell Dev Biol 2015; 3:61. [PMID: 26484344 PMCID: PMC4588110 DOI: 10.3389/fcell.2015.00061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/14/2015] [Indexed: 01/18/2023] Open
Abstract
To succeed in cell division, intense cytoskeletal and membrane remodeling are required to allow accurate chromosome segregation and cytoplasm partitioning. Spatial restriction of the actin dynamics and vesicle trafficking define the cell symmetry and equivalent membrane scission events, respectively. Protein complexes coordinating mitosis are recruited to membrane microdomains characterized by the presence of the phosphatidylinositol lipid members (PtdIns), like PtdIns(3,4,5)P3,PtdIns(4,5)P2, and PtdIns(3)P. These PtdIns represent a minor component of cell membranes, defining membrane domain identity, ultimately controlling cytoskeleton and membrane dynamics during mitosis. The coordinated presence of PtdIns(3,4,5)P3 at the cell poles and PtdIns(4,5)P2 at the cleavage furrow controls the polarity of the actin cytoskeleton leading to symmetrical cell division. In the endosomal compartment, the trafficking of PtdIns(3)P positive vesicles allows the recruitment of the protein machinery required for the abscission.
Collapse
Affiliation(s)
- Carlo C Campa
- Department of Molecular Biotechnology and Health Sciences, University of Turin Torino, Italy
| | - Miriam Martini
- Department of Molecular Biotechnology and Health Sciences, University of Turin Torino, Italy
| | - Maria C De Santis
- Department of Molecular Biotechnology and Health Sciences, University of Turin Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Turin Torino, Italy
| |
Collapse
|
40
|
Rudge SA, Wakelam MJO. Phosphatidylinositolphosphate phosphatase activities and cancer. J Lipid Res 2015; 57:176-92. [PMID: 26302980 DOI: 10.1194/jlr.r059154] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Indexed: 12/13/2022] Open
Abstract
Signaling through the phosphoinositide 3-kinase pathways mediates the actions of a plethora of hormones, growth factors, cytokines, and neurotransmitters upon their target cells following receptor occupation. Overactivation of these pathways has been implicated in a number of pathologies, in particular a range of malignancies. The tight regulation of signaling pathways necessitates the involvement of both stimulatory and terminating enzymes; inappropriate activation of a pathway can thus result from activation or inhibition of the two signaling arms. The focus of this review is to discuss, in detail, the activities of the identified families of phosphoinositide phosphatase expressed in humans, and how they regulate the levels of phosphoinositides implicated in promoting malignancy.
Collapse
Affiliation(s)
- Simon A Rudge
- Signalling Programme, Babraham Institute, Cambridge CB22 3AT, United Kingdom
| | - Michael J O Wakelam
- Signalling Programme, Babraham Institute, Cambridge CB22 3AT, United Kingdom
| |
Collapse
|
41
|
Hsu F, Mao Y. The structure of phosphoinositide phosphatases: Insights into substrate specificity and catalysis. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:698-710. [PMID: 25264170 DOI: 10.1016/j.bbalip.2014.09.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/10/2014] [Accepted: 09/17/2014] [Indexed: 12/17/2022]
Abstract
Phosphoinositides (PIs) are a group of key signaling and structural lipid molecules involved in a myriad of cellular processes. PI phosphatases, together with PI kinases, are responsible for the conversion of PIs between distinctive phosphorylation states. PI phosphatases are a large collection of enzymes that are evolved from at least two disparate ancestors. One group is distantly related to endonucleases, which apply divalent metal ions for phosphoryl transfer. The other group is related to protein tyrosine phosphatases, which contain a highly conserved active site motif Cys-X5-Arg (CX5R). In this review, we focus on structural insights to illustrate current understandings of the molecular mechanisms of each PI phosphatase family, with emphasis on their structural basis for substrate specificity determinants and catalytic mechanisms. This article is part of a Special Issue entitled Phosphoinositides.
Collapse
Affiliation(s)
- FoSheng Hsu
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Yuxin Mao
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
42
|
Differential SKIP expression in PTEN-deficient glioblastoma regulates cellular proliferation and migration. Oncogene 2014; 34:3711-27. [DOI: 10.1038/onc.2014.303] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 07/18/2014] [Accepted: 08/13/2014] [Indexed: 12/25/2022]
|
43
|
Abstract
The specific interaction of phosphoinositides with proteins is critical for a plethora of cellular processes, including cytoskeleton remodelling, mitogenic signalling, ion channel regulation and membrane traffic. The spatiotemporal restriction of different phosphoinositide species helps to define compartments within the cell, and this is particularly important for membrane trafficking within both the secretory and endocytic pathways. Phosphoinositide homoeostasis is tightly regulated by a large number of inositol kinases and phosphatases, which respectively phosphorylate and dephosphorylate distinct phosphoinositide species. Many of these enzymes have been implicated in regulating membrane trafficking and, accordingly, their dysregulation has been linked to a number of human diseases. In the present review, we focus on the inositol phosphatases, concentrating on their roles in membrane trafficking and the human diseases with which they have been associated.
Collapse
|
44
|
Pirruccello M, Nandez R, Idevall-Hagren O, Alcazar-Roman A, Abriola L, Berwick SA, Lucast L, Morel D, De Camilli P. Identification of inhibitors of inositol 5-phosphatases through multiple screening strategies. ACS Chem Biol 2014; 9:1359-68. [PMID: 24742366 PMCID: PMC4076014 DOI: 10.1021/cb500161z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
![]()
Phosphoinositides are low abundance
membrane phospholipids that
have key roles in signaling, membrane trafficking, and cytoskeletal
dynamics in all cells. Until recently, strategies for robust and quantitative
development of pharmacological tools for manipulating phosphoinositide
levels have focused selectively on PI(3,4,5)P3 due to the
importance of this lipid in growth factor signaling and cell proliferation.
However, drugs that affect levels of other phosphoinositides have
potential therapeutic applications and will be powerful research tools.
Here, we describe methodology for the high-throughput screening of
small molecule modulators of the inositol 5-phosphatases, which dephosphorylate
PI(4,5)P2 (the precursor for PI(3,4,5)P3) and
PI(3,4,5)P3). We developed three complementary in vitro activity assays, tested hit compounds on a panel
of 5-phosphatases, and monitored efficacy toward various substrates.
Two prominent chemical scaffolds were identified with high nanomolar/low
micromolar activity, with one class showing inhibitory activity toward
all 5-phosphatases tested and the other selective activity toward
OCRL and INPP5B, which are closely related to each other. One highly
soluble OCRL/INPP5B-specific inhibitor shows a direct interaction
with the catalytic domain of INPP5B. The efficacy of this compound
in living cells was validated through its property to enhance actin
nucleation at the cell cortex, a PI(4,5)P2 dependent process,
and to inhibit PI(4,5)P2 dephosphorylation by OCRL (both
overexpressed and endogenous enzyme). The assays and screening strategies
described here are applicable to other phosphoinositide-metabolizing
enzymes, at least several of which have major clinical relevance.
Most importantly, this study identifies the first OCRL/INPP5B specific
inhibitor and provides a platform for the design of more potent inhibitors
of this family of enzymes.
Collapse
Affiliation(s)
- Michelle Pirruccello
- Department
of Cell Biology, Howard Hughes Medical Institute and Program in Cellular
Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven Connecticut 06510, United States
| | - Ramiro Nandez
- Department
of Cell Biology, Howard Hughes Medical Institute and Program in Cellular
Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven Connecticut 06510, United States
| | - Olof Idevall-Hagren
- Department
of Cell Biology, Howard Hughes Medical Institute and Program in Cellular
Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven Connecticut 06510, United States
| | - Abel Alcazar-Roman
- Department
of Cell Biology, Howard Hughes Medical Institute and Program in Cellular
Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven Connecticut 06510, United States
| | - Laura Abriola
- Yale
Center for Molecular Discovery, Yale University, West Haven, Connecticut 06516, United States
| | - Shana Alexandra Berwick
- Department
of Cell Biology, Howard Hughes Medical Institute and Program in Cellular
Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven Connecticut 06510, United States
| | - Louise Lucast
- Department
of Cell Biology, Howard Hughes Medical Institute and Program in Cellular
Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven Connecticut 06510, United States
| | - Dayna Morel
- Department
of Cell Biology, Howard Hughes Medical Institute and Program in Cellular
Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven Connecticut 06510, United States
| | - Pietro De Camilli
- Department
of Cell Biology, Howard Hughes Medical Institute and Program in Cellular
Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven Connecticut 06510, United States
| |
Collapse
|
45
|
Trésaugues L, Silvander C, Flodin S, Welin M, Nyman T, Gräslund S, Hammarström M, Berglund H, Nordlund P. Structural basis for phosphoinositide substrate recognition, catalysis, and membrane interactions in human inositol polyphosphate 5-phosphatases. Structure 2014; 22:744-55. [PMID: 24704254 DOI: 10.1016/j.str.2014.01.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 01/24/2014] [Accepted: 01/24/2014] [Indexed: 11/15/2022]
Abstract
SHIP2, OCRL, and INPP5B belong to inositol polyphosphate 5-phophatase subfamilies involved in insulin regulation and Lowes syndrome. The structural basis for membrane recognition, substrate specificity, and regulation of inositol polyphosphate 5-phophatases is still poorly understood. We determined the crystal structures of human SHIP2, OCRL, and INPP5B, the latter in complex with phosphoinositide substrate analogs, which revealed a membrane interaction patch likely to assist in sequestering substrates from the lipid bilayer. Residues recognizing the 1-phosphate of the substrates are highly conserved among human family members, suggesting similar substrate binding modes. However, 3- and 4-phosphate recognition varies and determines individual substrate specificity profiles. The high conservation of the environment of the scissile 5-phosphate suggests a common reaction geometry for all members of the human 5-phosphatase family.
Collapse
Affiliation(s)
- Lionel Trésaugues
- Structural Genomics Consortium, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Camilla Silvander
- Structural Genomics Consortium, Karolinska Institutet, 17177 Stockholm, Sweden.
| | - Susanne Flodin
- Structural Genomics Consortium, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Martin Welin
- Structural Genomics Consortium, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Tomas Nyman
- Structural Genomics Consortium, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Susanne Gräslund
- Structural Genomics Consortium, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Martin Hammarström
- Structural Genomics Consortium, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Helena Berglund
- Structural Genomics Consortium, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Pär Nordlund
- Structural Genomics Consortium, Karolinska Institutet, 17177 Stockholm, Sweden; Division of Biophysics, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Centre for Biomedical Structural Biology, School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| |
Collapse
|
46
|
Mehta ZB, Pietka G, Lowe M. The cellular and physiological functions of the Lowe syndrome protein OCRL1. Traffic 2014; 15:471-87. [PMID: 24499450 PMCID: PMC4278560 DOI: 10.1111/tra.12160] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/03/2014] [Accepted: 02/05/2014] [Indexed: 12/17/2022]
Abstract
Phosphoinositide lipids play a key role in cellular physiology, participating in a wide array of cellular processes. Consequently, mutation of phosphoinositide-metabolizing enzymes is responsible for a growing number of diseases in humans. Two related disorders, oculocerebrorenal syndrome of Lowe (OCRL) and Dent-2 disease, are caused by mutation of the inositol 5-phosphatase OCRL1. Here, we review recent advances in our understanding of OCRL1 function. OCRL1 appears to regulate many processes within the cell, most of which depend upon coordination of membrane dynamics with remodeling of the actin cytoskeleton. Recently developed animal models have managed to recapitulate features of Lowe syndrome and Dent-2 disease, and revealed new insights into the underlying mechanisms of these disorders. The continued use of both cell-based approaches and animal models will be key to fully unraveling OCRL1 function, how its loss leads to disease and, importantly, the development of therapeutics to treat patients.
Collapse
Affiliation(s)
- Zenobia B Mehta
- Faculty of Life Sciences, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK; Current address: Faculty of Medicine, Imperial College, London, UK
| | | | | |
Collapse
|
47
|
Goto T, Terada N, Inoue T, Nakayama K, Okada Y, Yoshikawa T, Miyazaki Y, Uegaki M, Sumiyoshi S, Kobayashi T, Kamba T, Yoshimura K, Ogawa O. The expression profile of phosphatidylinositol in high spatial resolution imaging mass spectrometry as a potential biomarker for prostate cancer. PLoS One 2014; 9:e90242. [PMID: 24587297 PMCID: PMC3938652 DOI: 10.1371/journal.pone.0090242] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 01/27/2014] [Indexed: 01/23/2023] Open
Abstract
High-resolution matrix-assisted laser desorption/ionization imaging mass spectrometry (HR-MALDI-IMS) is an emerging application for the comprehensive and detailed analysis of the spatial distribution of ionized molecules in situ on tissue slides. HR-MALDI-IMS in negative mode in a mass range of m/z 500–1000 was performed on optimal cutting temperature (OCT) compound-embedded human prostate tissue samples obtained from patients with prostate cancer at the time of radical prostatectomy. HR-MALDI-IMS analysis of the 14 samples in the discovery set identified 26 molecules as highly expressed in the prostate. Tandem mass spectrometry (MS/MS) showed that these molecules included 14 phosphatidylinositols (PIs), 3 phosphatidylethanolamines (PEs) and 3 phosphatidic acids (PAs). Among the PIs, the expression of PI(18:0/18:1), PI(18:0/20:3) and PI(18:0/20:2) were significantly higher in cancer tissue than in benign epithelium. A biomarker algorithm for prostate cancer was formulated by analyzing the expression profiles of PIs in cancer tissue and benign epithelium of the discovery set using orthogonal partial least squares discriminant analysis (OPLS-DA). The sensitivity and specificity of this algorithm for prostate cancer diagnosis in the 24 validation set samples were 87.5 and 91.7%, respectively. In conclusion, HR-MALDI-IMS identified several PIs as being more highly expressed in prostate cancer than benign prostate epithelium. These differences in PI expression profiles may serve as a novel diagnostic tool for prostate cancer.
Collapse
Affiliation(s)
- Takayuki Goto
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naoki Terada
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Inoue
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Nakayama
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiyuki Okada
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Yoshikawa
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yu Miyazaki
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masayuki Uegaki
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Sumiyoshi
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Takashi Kobayashi
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomomi Kamba
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Yoshimura
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
48
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: enzymes. Br J Pharmacol 2013; 170:1797-867. [PMID: 24528243 PMCID: PMC3892293 DOI: 10.1111/bph.12451] [Citation(s) in RCA: 415] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Enzymes are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, nuclear hormone receptors, catalytic receptors and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
49
|
Dallaire-Dufresne S, Barbeau X, Sarty D, Tanaka KH, Denoncourt AM, Lagüe P, Reith ME, Charette SJ. Aeromonas salmonicida Ati2 is an effector protein of the type three secretion system. Microbiology (Reading) 2013; 159:1937-1945. [DOI: 10.1099/mic.0.067959-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Stéphanie Dallaire-Dufresne
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, Quebec G1V 4G5, Canada
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Institut de biologie intégrative et des systèmes, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
| | - Xavier Barbeau
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Institut de biologie intégrative et des systèmes, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
| | - Darren Sarty
- Aquatic and Crop Resource Development, National Research Council Canada, Halifax, Nova Scotia B3H 3Z1, Canada
| | - Katherine H. Tanaka
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, Quebec G1V 4G5, Canada
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Institut de biologie intégrative et des systèmes, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
| | - Alix M. Denoncourt
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, Quebec G1V 4G5, Canada
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Institut de biologie intégrative et des systèmes, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
| | - Patrick Lagüe
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Institut de biologie intégrative et des systèmes, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
| | - Michael E. Reith
- Aquatic and Crop Resource Development, National Research Council Canada, Halifax, Nova Scotia B3H 3Z1, Canada
| | - Steve J. Charette
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Quebec City, Quebec G1V 4G5, Canada
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
- Institut de biologie intégrative et des systèmes, Faculté des sciences et de génie, Université Laval, Quebec City, Quebec G1V 0A6, Canada
| |
Collapse
|
50
|
Abstract
Phosphoinositides (PIs) make up only a small fraction of cellular phospholipids, yet they control almost all aspects of a cell's life and death. These lipids gained tremendous research interest as plasma membrane signaling molecules when discovered in the 1970s and 1980s. Research in the last 15 years has added a wide range of biological processes regulated by PIs, turning these lipids into one of the most universal signaling entities in eukaryotic cells. PIs control organelle biology by regulating vesicular trafficking, but they also modulate lipid distribution and metabolism via their close relationship with lipid transfer proteins. PIs regulate ion channels, pumps, and transporters and control both endocytic and exocytic processes. The nuclear phosphoinositides have grown from being an epiphenomenon to a research area of its own. As expected from such pleiotropic regulators, derangements of phosphoinositide metabolism are responsible for a number of human diseases ranging from rare genetic disorders to the most common ones such as cancer, obesity, and diabetes. Moreover, it is increasingly evident that a number of infectious agents hijack the PI regulatory systems of host cells for their intracellular movements, replication, and assembly. As a result, PI converting enzymes began to be noticed by pharmaceutical companies as potential therapeutic targets. This review is an attempt to give an overview of this enormous research field focusing on major developments in diverse areas of basic science linked to cellular physiology and disease.
Collapse
Affiliation(s)
- Tamas Balla
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|