1
|
Bradshaw WJ, Kennedy EC, Moreira T, Smith LA, Chalk R, Katis VL, Benesch JLP, Brennan PE, Murphy EJ, Gileadi O. Regulation of inositol 5-phosphatase activity by the C2 domain of SHIP1 and SHIP2. Structure 2024; 32:453-466.e6. [PMID: 38309262 PMCID: PMC10997489 DOI: 10.1016/j.str.2024.01.005] [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: 08/01/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 02/05/2024]
Abstract
SHIP1, an inositol 5-phosphatase, plays a central role in cellular signaling. As such, it has been implicated in many conditions. Exploiting SHIP1 as a drug target will require structural knowledge and the design of selective small molecules. We have determined apo, and magnesium and phosphate-bound structures of the phosphatase and C2 domains of SHIP1. The C2 domains of SHIP1 and the related SHIP2 modulate the activity of the phosphatase domain. To understand the mechanism, we performed activity assays, hydrogen-deuterium exchange mass spectrometry, and molecular dynamics on SHIP1 and SHIP2. Our findings demonstrate that the influence of the C2 domain is more pronounced for SHIP2 than SHIP1. We determined 91 structures of SHIP1 with fragments bound, with some near the interface between the two domains. We performed a mass spectrometry screen and determined four structures with covalent fragments. These structures could act as starting points for the development of potent, selective probes.
Collapse
Affiliation(s)
- William J Bradshaw
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK.
| | - Emma C Kennedy
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Tiago Moreira
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Luke A Smith
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Rod Chalk
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Vittorio L Katis
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Justin L P Benesch
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Paul E Brennan
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Emma J Murphy
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK
| | - Opher Gileadi
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Oxford OX3 7FZ, UK.
| |
Collapse
|
2
|
Elbatsh AMO, Amin-Mansour A, Haberkorn A, Textor C, Ebel N, Renard E, Koch LM, Groenveld FC, Piquet M, Naumann U, Ruddy DA, Romanet V, Martínez Gómez JM, Shirley MD, Wipfli P, Schnell C, Wartmann M, Rausch M, Jager MJ, Levesque MP, Maira SM, Manchado E. INPP5A phosphatase is a synthetic lethal target in GNAQ and GNA11-mutant melanomas. NATURE CANCER 2024; 5:481-499. [PMID: 38233483 DOI: 10.1038/s43018-023-00710-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/14/2023] [Indexed: 01/19/2024]
Abstract
Activating mutations in GNAQ/GNA11 occur in over 90% of uveal melanomas (UMs), the most lethal melanoma subtype; however, targeting these oncogenes has proven challenging and inhibiting their downstream effectors show limited clinical efficacy. Here, we performed genome-scale CRISPR screens along with computational analyses of cancer dependency and gene expression datasets to identify the inositol-metabolizing phosphatase INPP5A as a selective dependency in GNAQ/11-mutant UM cells in vitro and in vivo. Mutant cells intrinsically produce high levels of the second messenger inositol 1,4,5 trisphosphate (IP3) that accumulate upon suppression of INPP5A, resulting in hyperactivation of IP3-receptor signaling, increased cytosolic calcium and p53-dependent apoptosis. Finally, we show that GNAQ/11-mutant UM cells and patients' tumors exhibit elevated levels of IP4, a biomarker of enhanced IP3 production; these high levels are abolished by GNAQ/11 inhibition and correlate with sensitivity to INPP5A depletion. Our findings uncover INPP5A as a synthetic lethal vulnerability and a potential therapeutic target for GNAQ/11-mutant-driven cancers.
Collapse
Affiliation(s)
- Ahmed M O Elbatsh
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Ali Amin-Mansour
- Oncology, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Anne Haberkorn
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Claudia Textor
- PK Sciences, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Nicolas Ebel
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Emilie Renard
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Lisa M Koch
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Femke C Groenveld
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Michelle Piquet
- Oncology, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Ulrike Naumann
- Chemical Biology and Therapeutics, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - David A Ruddy
- Oncology, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Vincent Romanet
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Julia M Martínez Gómez
- Dermatology Department, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthew D Shirley
- Oncology, Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Peter Wipfli
- PK Sciences, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Christian Schnell
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Markus Wartmann
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Martin Rausch
- Chemical Biology and Therapeutics, Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mitchell P Levesque
- Dermatology Department, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Eusebio Manchado
- Oncology, Novartis Institute for Biomedical Research, Basel, Switzerland.
| |
Collapse
|
3
|
Shukla D, Gural BM, Cauley ES, Battula N, Mowla S, Karas BF, Roberts LE, Cavallo L, Turkalj L, Moody SA, Swan LE, Manzini MC. Duplicated zebrafish (Danio rerio) inositol phosphatases inpp5ka and inpp5kb diverged in expression pattern and function. Dev Genes Evol 2023; 233:25-34. [PMID: 37184573 PMCID: PMC10239392 DOI: 10.1007/s00427-023-00703-z] [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: 08/31/2022] [Accepted: 04/27/2023] [Indexed: 05/16/2023]
Abstract
One hurdle in the development of zebrafish models of human disease is the presence of multiple zebrafish orthologs resulting from whole genome duplication in teleosts. Mutations in inositol polyphosphate 5-phosphatase K (INPP5K) lead to a syndrome characterized by variable presentation of intellectual disability, brain abnormalities, cataracts, muscle disease, and short stature. INPP5K is a phosphatase acting at position 5 of phosphoinositides to control their homeostasis and is involved in insulin signaling, cytoskeletal regulation, and protein trafficking. Previously, our group and others have replicated the human phenotypes in zebrafish knockdown models by targeting both INPP5K orthologs inpp5ka and inpp5kb. Here, we show that inpp5ka is the more closely related orthologue to human INPP5K. While both inpp5ka and inpp5kb mRNA expression levels follow a similar trend in the developing head, eyes, and tail, inpp5ka is much more abundantly expressed in these tissues than inpp5kb. In situ hybridization revealed a similar trend, also showing unique localization of inpp5kb in the pineal gland and retina indicating different transcriptional regulation. We also found that inpp5kb has lost its catalytic activity against its preferred substrate, PtdIns(4,5)P2. Since most human mutations are missense changes disrupting phosphatase activity, we propose that loss of inpp5ka alone can be targeted to recapitulate the human presentation. In addition, we show that the function of inpp5kb has diverged from inpp5ka and may play a novel role in the zebrafish.
Collapse
Affiliation(s)
- Dhyanam Shukla
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Brian M Gural
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Edmund S Cauley
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Namarata Battula
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Shorbon Mowla
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Brittany F Karas
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Llion E Roberts
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Luca Cavallo
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Luka Turkalj
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Sally A Moody
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Laura E Swan
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - M Chiara Manzini
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
4
|
Lee JJ, Ramadesikan S, Black AF, Christoffer C, Pacheco AFP, Subramanian S, Hanna CB, Barth G, Stauffacher CV, Kihara D, Aguilar RC. Heterogeneity in Lowe Syndrome: Mutations Affecting the Phosphatase Domain of OCRL1 Differ in Impact on Enzymatic Activity and Severity of Cellular Phenotypes. Biomolecules 2023; 13:615. [PMID: 37189363 PMCID: PMC10135975 DOI: 10.3390/biom13040615] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 04/01/2023] Open
Abstract
Lowe Syndrome (LS) is a condition due to mutations in the OCRL1 gene, characterized by congenital cataracts, intellectual disability, and kidney malfunction. Unfortunately, patients succumb to renal failure after adolescence. This study is centered in investigating the biochemical and phenotypic impact of patient's OCRL1 variants (OCRL1VAR). Specifically, we tested the hypothesis that some OCRL1VAR are stabilized in a non-functional conformation by focusing on missense mutations affecting the phosphatase domain, but not changing residues involved in binding/catalysis. The pathogenic and conformational characteristics of the selected variants were evaluated in silico and our results revealed some OCRL1VAR to be benign, while others are pathogenic. Then we proceeded to monitor the enzymatic activity and function in kidney cells of the different OCRL1VAR. Based on their enzymatic activity and presence/absence of phenotypes, the variants segregated into two categories that also correlated with the severity of the condition they induce. Overall, these two groups mapped to opposite sides of the phosphatase domain. In summary, our findings highlight that not every mutation affecting the catalytic domain impairs OCRL1's enzymatic activity. Importantly, data support the inactive-conformation hypothesis. Finally, our results contribute to establishing the molecular and structural basis for the observed heterogeneity in severity/symptomatology displayed by patients.
Collapse
Affiliation(s)
- Jennifer J. Lee
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.J.L.); (A.F.B.); (A.F.P.P.); (S.S.); (C.B.H.); (G.B.); (C.V.S.); (D.K.)
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Swetha Ramadesikan
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.J.L.); (A.F.B.); (A.F.P.P.); (S.S.); (C.B.H.); (G.B.); (C.V.S.); (D.K.)
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Adrianna F. Black
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.J.L.); (A.F.B.); (A.F.P.P.); (S.S.); (C.B.H.); (G.B.); (C.V.S.); (D.K.)
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Charles Christoffer
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA;
| | - Andres F. Pacheco Pacheco
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.J.L.); (A.F.B.); (A.F.P.P.); (S.S.); (C.B.H.); (G.B.); (C.V.S.); (D.K.)
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Sneha Subramanian
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.J.L.); (A.F.B.); (A.F.P.P.); (S.S.); (C.B.H.); (G.B.); (C.V.S.); (D.K.)
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Claudia B. Hanna
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.J.L.); (A.F.B.); (A.F.P.P.); (S.S.); (C.B.H.); (G.B.); (C.V.S.); (D.K.)
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Gillian Barth
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.J.L.); (A.F.B.); (A.F.P.P.); (S.S.); (C.B.H.); (G.B.); (C.V.S.); (D.K.)
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Cynthia V. Stauffacher
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.J.L.); (A.F.B.); (A.F.P.P.); (S.S.); (C.B.H.); (G.B.); (C.V.S.); (D.K.)
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Daisuke Kihara
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.J.L.); (A.F.B.); (A.F.P.P.); (S.S.); (C.B.H.); (G.B.); (C.V.S.); (D.K.)
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA;
| | - Ruben Claudio Aguilar
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.J.L.); (A.F.B.); (A.F.P.P.); (S.S.); (C.B.H.); (G.B.); (C.V.S.); (D.K.)
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
5
|
Mizutani N, Kawanabe A, Jinno Y, Narita H, Yonezawa T, Nakagawa A, Okamura Y. Interaction between S4 and the phosphatase domain mediates electrochemical coupling in voltage-sensing phosphatase (VSP). Proc Natl Acad Sci U S A 2022; 119:e2200364119. [PMID: 35733115 PMCID: PMC9245683 DOI: 10.1073/pnas.2200364119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/13/2022] [Indexed: 11/18/2022] Open
Abstract
Voltage-sensing phosphatase (VSP) consists of a voltage sensor domain (VSD) and a cytoplasmic catalytic region (CCR), which is similar to phosphatase and tensin homolog (PTEN). How the VSD regulates the innate enzyme component of VSP remains unclear. Here, we took a combined approach that entailed the use of electrophysiology, fluorometry, and structural modeling to study the electrochemical coupling in Ciona intestinalis VSP. We found that two hydrophobic residues at the lowest part of S4 play an essential role in the later transition of VSD-CCR coupling. Voltage clamp fluorometry and disulfide bond locking indicated that S4 and its neighboring linker move as one helix (S4-linker helix) and approach the hydrophobic spine in the CCR, a structure located near the cell membrane and also conserved in PTEN. We propose that the hydrophobic spine operates as a hub for translating an electrical signal into a chemical one in VSP.
Collapse
Affiliation(s)
- Natsuki Mizutani
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Akira Kawanabe
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuka Jinno
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hirotaka Narita
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tomoko Yonezawa
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Atsushi Nakagawa
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yasushi Okamura
- Laboratory of Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
6
|
Deng J, Lin X, Li Q, Cai XY, Wu LW, Wang W, Zhang B, Li YL, Hu J, Lin NM. Decreased INPP5B expression predicts poor prognosis in lung adenocarcinoma. Cancer Cell Int 2022; 22:189. [PMID: 35568951 PMCID: PMC9107680 DOI: 10.1186/s12935-022-02609-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/06/2022] [Indexed: 12/03/2022] Open
Abstract
Background Inositol Polyphosphate-5-Phosphatase B (INPP5B), a inositol 5-phosphatase, plays an important role in many biological processes through phosphorylating PI(4,5)P2 and/or PI(3,4,5)P3 at the 5-position. Nevertheless, little is known about its function and cellular pathways in tumors. This study aims to investigate the potential role of INPP5B as a diagnostic and prognostic biomarker for lung adenocarcinoma (LUAD), as well as its biological functions and molecular mechanisms in LUAD. Methods TCGA, GEO, CTPAC, and HPA datasets were used for differential expression analysis and pathological stratification comparison. The prognostic and diagnostic role of INPP5B was determined by Kaplan–Meier curves, univariate and multivariate Cox regression analysis, and receiver operating characteristics (ROC) curve analyses. The potential mechanism of INPP5B was explored through GO, KEGG, and GSEA enrichment analysis, as well as GeneMANIA and STRING protein–protein interaction (PPI) network. PicTar, PITA, and miRmap databases were used for exploring miRNA targeting INPP5B. In molecular biology experiments, immunohistochemical analyses and Western blot analyses were used to determine protein expression. Co-immunoprecipitation assay was used to detect protein–protein interactions. CCK8 assays and colony formation assays were used for the measurement of cell proliferation. Cell cycle was assessed by PI staining with flow cytometry. Cell migration was performed by Transwell assays and wound healing assays. Result INPP5B was decreased in LUAD tissues compared with normal adjacent tissues. And the low expression of INPP5B was associated with late-stage pathological features. In addition, INPP5B was found to be a significant independent prognostic and diagnostic factor for LUAD patients. Hsa-miR-582-5p was predicted as a negative regulator of INPP5B mRNA expression. INPP5B was significantly correlated with the expression of PTEN and the activity of PI3K/AKT signaling pathways, as determined by enrichment analysis and PPI network. In vitro experiments partially confirmed the aforementioned findings. INPP5B could interact directly with PTEN. INPP5B overexpression inhibited LUAD cell proliferation and migration while downregulating the AKT pathway. Conclusion Our results demonstrated that INPP5B could inhibit the proliferation and metastasis of LUAD cells. It could serve as a novel diagnostic and prognostic biomarker for LUAD patients. Trial registration LUAD tissues and corresponding para-cancerous tissues were collected from 10 different LUAD patients at Hangzhou First People’s Hospital. The Ethics Committee of Hangzhou First People’s Hospital has approved this study. (registration number: IIT-20210907-0031-01; registration date: 2021.09.13) Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02609-8.
Collapse
Affiliation(s)
- Jun Deng
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xu Lin
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qi Li
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Xiao-Yu Cai
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Department of Clinical Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lin-Wen Wu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Department of Clinical Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Wei Wang
- Department of Pathology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Department of Clinical Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yang-Ling Li
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.,Department of Clinical Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Neng-Ming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China. .,Department of Clinical Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China. .,Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, 310024, China. .,Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
7
|
Abd El-Aziz TM, Kaur A, Shapiro MS, Stockand JD, Archer CR. Optogenetic Control of PIP2 Interactions Shaping ENaC Activity. Int J Mol Sci 2022; 23:ijms23073884. [PMID: 35409240 PMCID: PMC8998630 DOI: 10.3390/ijms23073884] [Citation(s) in RCA: 4] [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: 03/14/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
The activity of the epithelial Na+ Channel (ENaC) is strongly dependent on the membrane phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2). PIP2 binds two distinct cationic clusters within the N termini of β- and γ-ENaC subunits (βN1 and γN2). The affinities of these sites were previously determined using short synthetic peptides, yet their role in sensitizing ENaC to changes in PIP2 levels in the cellular system is not well established. We addressed this question by comparing the effects of PIP2 depletion and recovery on ENaC channel activity and intracellular Na+ levels [Na+]i. We tested effects on ENaC activity with mutations to the PIP2 binding sites using the optogenetic system CIBN/CRY2-OCRL to selectively deplete PIP2. We monitored changes of [Na+]i by measuring the fluorescent Na+ indicator, CoroNa Green AM, and changes in channel activity by performing patch clamp electrophysiology. Whole cell patch clamp measurements showed a complete lack of response to PIP2 depletion and recovery in ENaC with mutations to βN1 or γN2 or both sites, compared to wild type ENaC. Whereas mutant βN1 also had no change in CoroNa Green fluorescence in response to PIP2 depletion, γN2 did have reduced [Na+]i, which was explained by having shorter CoroNa Green uptake and half-life. These results suggest that CoroNa Green measurements should be interpreted with caution. Importantly, the electrophysiology results show that the βN1 and γN2 sites on ENaC are each necessary to permit maximal ENaC activity in the presence of PIP2.
Collapse
Affiliation(s)
- Tarek Mohamed Abd El-Aziz
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78228, USA; (T.M.A.E.-A.); (M.S.S.); (J.D.S.)
- Faculty of Science, Zoology Department, Minia University, El-Minia 61519, Egypt
| | - Amanpreet Kaur
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA;
| | - Mark S. Shapiro
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78228, USA; (T.M.A.E.-A.); (M.S.S.); (J.D.S.)
| | - James D. Stockand
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78228, USA; (T.M.A.E.-A.); (M.S.S.); (J.D.S.)
| | - Crystal R. Archer
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78228, USA; (T.M.A.E.-A.); (M.S.S.); (J.D.S.)
- Correspondence:
| |
Collapse
|
8
|
Whitfield H, Hemmings AM, Mills SJ, Baker K, White G, Rushworth S, Riley AM, Potter BVL, Brearley CA. Allosteric Site on SHIP2 Identified Through Fluorescent Ligand Screening and Crystallography: A Potential New Target for Intervention. J Med Chem 2021; 64:3813-3826. [PMID: 33724834 PMCID: PMC7610569 DOI: 10.1021/acs.jmedchem.0c01944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Src homology 2 domain-containing inositol phosphate phosphatase 2 (SHIP2) is one of the 10 human inositol phosphate 5-phosphatases. One of its physiological functions is dephosphorylation of phosphatidylinositol 3,4,5-trisphosphate, PtdIns(3,4,5)P3. It is therefore a therapeutic target for pathophysiologies dependent on PtdIns(3,4,5)P3 and PtdIns(3,4)P2. Therapeutic interventions are limited by the dearth of crystallographic data describing ligand/inhibitor binding. An active site-directed fluorescent probe facilitated screening of compound libraries for SHIP2 ligands. With two additional orthogonal assays, several ligands including galloflavin were identified as low micromolar Ki inhibitors. One ligand, an oxo-linked ethylene-bridged dimer of benzene 1,2,4-trisphosphate, was shown to be an uncompetitive inhibitor that binds to a regulatory site on the catalytic domain. We posit that binding of ligands to this site restrains L4 loop motions that are key to interdomain communications that accompany high catalytic activity with phosphoinositide substrate. This site may, therefore, be a future druggable target for medicinal chemistry.
Collapse
Affiliation(s)
- Hayley Whitfield
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Andrew M Hemmings
- School of Chemistry, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Stephen J Mills
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Kendall Baker
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Gaye White
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| | - Stuart Rushworth
- Department of Molecular Haematology; Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, U.K
| | - Andrew M Riley
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Barry V L Potter
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| | - Charles A Brearley
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, U.K
| |
Collapse
|
9
|
Targeting SHIP1 and SHIP2 in Cancer. Cancers (Basel) 2021; 13:cancers13040890. [PMID: 33672717 PMCID: PMC7924360 DOI: 10.3390/cancers13040890] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Phosphoinositol signaling pathways and their dysregulation have been shown to have a fundamental role in health and disease, respectively. The SH2-containing 5′ inositol phosphatases, SHIP1 and SHIP2, are regulators of the PI3K/AKT pathway that have crucial roles in cancer progression. This review aims to summarize the role of SHIP1 and SHIP2 in cancer signaling and the immune response to cancer, the discovery and use of SHIP inhibitors and agonists as possible cancer therapeutics. Abstract Membrane-anchored and soluble inositol phospholipid species are critical mediators of intracellular cell signaling cascades. Alterations in their normal production or degradation are implicated in the pathology of a number of disorders including cancer and pro-inflammatory conditions. The SH2-containing 5′ inositol phosphatases, SHIP1 and SHIP2, play a fundamental role in these processes by depleting PI(3,4,5)P3, but also by producing PI(3,4)P2 at the inner leaflet of the plasma membrane. With the intent of targeting SHIP1 or SHIP2 selectively, or both paralogs simultaneously, small molecule inhibitors and agonists have been developed and tested in vitro and in vivo over the last decade in various disease models. These studies have shown promising results in various pre-clinical models of disease including cancer and tumor immunotherapy. In this review the potential use of SHIP inhibitors in cancer is discussed with particular attention to the molecular structure, binding site and efficacy of these SHIP inhibitors.
Collapse
|
10
|
Ramadesikan S, Skiba L, Lee J, Madhivanan K, Sarkar D, De La Fuente A, Hanna CB, Terashi G, Hazbun T, Kihara D, Aguilar RC. Genotype & phenotype in Lowe Syndrome: specific OCRL1 patient mutations differentially impact cellular phenotypes. Hum Mol Genet 2021; 30:198-212. [PMID: 33517444 DOI: 10.1093/hmg/ddab025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/15/2020] [Accepted: 01/08/2021] [Indexed: 12/26/2022] Open
Abstract
Lowe Syndrome (LS) is a lethal genetic disorder caused by mutations in the OCRL1 gene which encodes the lipid 5' phosphatase Ocrl1. Patients exhibit a characteristic triad of symptoms including eye, brain and kidney abnormalities with renal failure as the most common cause of premature death. Over 200 OCRL1 mutations have been identified in LS, but their specific impact on cellular processes is unknown. Despite observations of heterogeneity in patient symptom severity, there is little understanding of the correlation between genotype and its impact on phenotype. Here, we show that different mutations had diverse effects on protein localization and on triggering LS cellular phenotypes. In addition, some mutations affecting specific domains imparted unique characteristics to the resulting mutated protein. We also propose that certain mutations conformationally affect the 5'-phosphatase domain of the protein, resulting in loss of enzymatic activity and causing common and specific phenotypes (a conformational disease scenario). This study is the first to show the differential effect of patient 5'-phosphatase mutations on cellular phenotypes and introduces a conformational disease component in LS. This work provides a framework that explains symptom heterogeneity and can help stratify patients as well as to produce a more accurate prognosis depending on the nature and location of the mutation within the OCRL1 gene.
Collapse
Affiliation(s)
- Swetha Ramadesikan
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Lisette Skiba
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jennifer Lee
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | | - Daipayan Sarkar
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | | - Claudia B Hanna
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Genki Terashi
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Tony Hazbun
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Daisuke Kihara
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA.,Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA
| | - R Claudio Aguilar
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
11
|
Zhou H, Yue X, Wang Z, Li S, Zhu J, Yang Y, Liu M. Expression, purification and characterization of the RhoA-binding domain of human SHIP2 in E.coli. Protein Expr Purif 2021; 180:105821. [PMID: 33421554 DOI: 10.1016/j.pep.2021.105821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/02/2020] [Accepted: 01/03/2021] [Indexed: 11/26/2022]
Abstract
Human SH2-containing inositol 5-phosphatase 2 (SHIP2) is a multi-domain protein playing essential roles in various physiological and pathological processes. In cell polarization and migration, SHIP2 serves as a RhoA effector for manipulating the level of phosphatidylinositol 3,4,5-trisphosphate. The domain between SH2 and a potential PH-R domain of SHIP2 was suggested to bind with GTP-bound form of RhoA. However, the structure of this RhoA-binding domain (RBD) of SHIP2 and the mechanism for its binding with RhoA remain unknown. In this study, SHIP2118-298 and SHIP2176-298, two truncated proteins harboring the RBD were designed, expressed, and purified successfully in E. coli. Unexpectedly, both SHIP2118-298 and SHIP2176-298 were determined to exist as homo-dimers in solution by multi-angle light scattering. Circular dichroism spectra indicated that both proteins predominantly consisted of α-helix structure. Moreover, in pull-down experiments, both proteins could bind with GTP-bound RhoA and RhoAQ63L, a mutant mimicing the state of GTP-bound RhoA. Importantly, in silico analysis showed that the shorter truncation, SHIP2176-298, contained all ordered residues between the SH2 and the PH-R domain, and matched the RhoA effector motif 1 of PKN1 well in sequence alignment, suggesting that SHIP2176-298 is sufficient for further studies on the structure and RhoA binding of SHIP2. This work shortens and confirms the main region of SHIP2 interacting with RhoA, provides the method for sample preparation, and presents preliminary information for SHIP2-RBD structure, which will facilitate the comprehensive understanding of the structure and function of SHIP2.
Collapse
Affiliation(s)
- Heng Zhou
- Department of Chemistry, College of Science, Huazhong Agricultural University, Wuhan, 430070, China; State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China
| | - Xiali Yue
- Department of Chemistry, College of Science, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Zi Wang
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuangli Li
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China
| | - Jiang Zhu
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China.
| | - Yunhuang Yang
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
12
|
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
|
13
|
Paley EL. Discovery of Gut Bacteria Specific to Alzheimer's Associated Diseases is a Clue to Understanding Disease Etiology: Meta-Analysis of Population-Based Data on Human Gut Metagenomics and Metabolomics. J Alzheimers Dis 2020; 72:319-355. [PMID: 31561379 DOI: 10.3233/jad-190873] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD)-associated sequence (ADAS) of cultured fecal bacteria was discovered in human gut targeted screening. This study provides important information to expand our current understanding of the structure/activity relationship of ADAS and putative inhibitors/activators that are potentially involved in ADAS appearance/disappearance. The NCBI database analysis revealed that ADAS presents at a large proportion in American Indian Oklahoman (C&A) with a high prevalence of obesity/diabetes and in colorectal cancer (CRC) patients from the US and China. An Oklahoman non-native group (NNI) showed no ADAS. Comparison of two large US populations reveals that ADAS is more frequent in individuals aged ≥66 and in females. Prevalence and levels of fecal metabolites are altered in the C&A and CRC groups versus controls. Biogenic amines (histamine, tryptamine, tyramine, phenylethylamine, cadaverine, putrescine, agmatine, spermidine) that present in food and are produced by gut microbiota are significantly higher in C&A (e.g., histamine/histidine 95-fold) versus NNI (histamine/histidine 16-fold). The majority of these bio-amines are cytotoxic at concentrations found in food. Inositol phosphate signaling implicated in AD is altered in C&A and CRC. Tryptamine stimulated accumulation of inositol phosphate. The seizure-eliciting tryptamine induced cytoplasmic vacuolization and vesiculation with cell fragmentation. Present additions of ADAS-carriers at different ages including infants led to an ADAS-comprising human sample size of 2,830 from 27 studies from four continents (North America, Australia, Asia, Europe). Levels of food-derived monoamine oxidase inhibitors and anti-bacterial compounds, the potential modulators of ADAS-bacteria growth and biogenic amine production, were altered in C&A versus NNI. ADAS is attributable to potentially modifiable risk factors of AD associated diseases.
Collapse
Affiliation(s)
- Elena L Paley
- Expert Biomed, Inc., Miami, FL, USA.,Stop Alzheimers Corp, Miami, FL, USA
| |
Collapse
|
14
|
Pinto PH, Kroupova A, Schleiffer A, Mechtler K, Jinek M, Weitzer S, Martinez J. ANGEL2 is a member of the CCR4 family of deadenylases with 2',3'-cyclic phosphatase activity. Science 2020; 369:524-530. [PMID: 32732418 DOI: 10.1126/science.aba9763] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022]
Abstract
RNA molecules are frequently modified with a terminal 2',3'-cyclic phosphate group as a result of endonuclease cleavage, exonuclease trimming, or de novo synthesis. During pre-transfer RNA (tRNA) and unconventional messenger RNA (mRNA) splicing, 2',3'-cyclic phosphates are substrates of the tRNA ligase complex, and their removal is critical for recycling of tRNAs upon ribosome stalling. We identified the predicted deadenylase angel homolog 2 (ANGEL2) as a human phosphatase that converts 2',3'-cyclic phosphates into 2',3'-OH nucleotides. We analyzed ANGEL2's substrate preference, structure, and reaction mechanism. Perturbing ANGEL2 expression affected the efficiency of pre-tRNA processing, X-box-binding protein 1 (XBP1) mRNA splicing during the unfolded protein response, and tRNA nucleotidyltransferase 1 (TRNT1)-mediated CCA addition onto tRNAs. Our results indicate that ANGEL2 is involved in RNA pathways that rely on the ligation or hydrolysis of 2',3'-cyclic phosphates.
Collapse
Affiliation(s)
- Paola H Pinto
- Max Perutz Labs, Medical University of Vienna, Vienna BioCenter, 1030 Vienna, Austria
| | - Alena Kroupova
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Alexander Schleiffer
- IMP/IMBA Bioinformatics Core Facility, Research Institute of Molecular Pathology, Vienna BioCenter, 1030 Vienna, Austria
| | - Karl Mechtler
- Research Institute of Molecular Pathology, Vienna BioCenter, 1030 Vienna, Austria.,Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna BioCenter, 1030 Vienna, Austria
| | - Martin Jinek
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Stefan Weitzer
- Max Perutz Labs, Medical University of Vienna, Vienna BioCenter, 1030 Vienna, Austria.
| | - Javier Martinez
- Max Perutz Labs, Medical University of Vienna, Vienna BioCenter, 1030 Vienna, Austria.
| |
Collapse
|
15
|
Pons BJ, Loiseau N, Hashim S, Tadrist S, Mirey G, Vignard J. Functional Study of Haemophilus ducreyi Cytolethal Distending Toxin Subunit B. Toxins (Basel) 2020; 12:toxins12090530. [PMID: 32825080 PMCID: PMC7551728 DOI: 10.3390/toxins12090530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
The Cytolethal Distending Toxin (CDT) is produced by many Gram-negative pathogenic bacteria responsible for major foodborne diseases worldwide. CDT induces DNA damage and cell cycle arrest in host-cells, eventually leading to senescence or apoptosis. According to structural and sequence comparison, the catalytic subunit CdtB is suggested to possess both nuclease and phosphatase activities, carried by a single catalytic site. However, the impact of each activity on cell-host toxicity is yet to be characterized. Here, we analyze the consequences of cell exposure to different CDT mutated on key CdtB residues, focusing on cell viability, cell cycle defects, and DNA damage induction. A first class of mutant, devoid of any activity, targets putative catalytic (H160A), metal binding (D273R), and DNA binding residues (R117A-R144A-N201A). The second class of mutants (A163R, F156-T158, and the newly identified G114T), which gathers mutations on residues potentially involved in lipid substrate binding, has only partially lost its toxic effects. However, their defects are alleviated when CdtB is artificially introduced inside cells, except for the F156-T158 double mutant that is defective in nuclear addressing. Therefore, our data reveal that CDT toxicity is mainly correlated to CdtB nuclease activity, whereas phosphatase activity may probably be involved in CdtB intracellular trafficking.
Collapse
Affiliation(s)
| | | | | | | | - Gladys Mirey
- Correspondence: (G.M.); (J.V.); Tel.: +33-582-066-338 (G.M.)
| | - Julien Vignard
- Correspondence: (G.M.); (J.V.); Tel.: +33-582-066-338 (G.M.)
| |
Collapse
|
16
|
Jenkins K, Mateeva T, Szabó I, Melnik A, Picotti P, Csikász-Nagy A, Rosta E. Combining data integration and molecular dynamics for target identification in α-Synuclein-aggregating neurodegenerative diseases: Structural insights on Synaptojanin-1 (Synj1). Comput Struct Biotechnol J 2020; 18:1032-1042. [PMID: 32419904 PMCID: PMC7215115 DOI: 10.1016/j.csbj.2020.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 12/19/2022] Open
Abstract
Parkinson’s disease (PD), Alzheimer’s disease (AD) and Amyotrophic lateral sclerosis (ALS) are neurodegenerative diseases hallmarked by the formation of toxic protein aggregates. However, targeting these aggregates therapeutically have thus far shown no success. The treatment of AD has remained particularly problematic since no new drugs have been approved in the last 15 years. Therefore, novel therapeutic targets need to be identified and explored. Here, through the integration of genomic and proteomic data, a set of proteins with strong links to α-synuclein-aggregating neurodegenerative diseases was identified. We propose 17 protein targets that are likely implicated in neurodegeneration and could serve as potential targets. The human phosphatidylinositol 5-phosphatase synaptojanin-1, which has already been independently confirmed to be implicated in Parkinson’s and Alzheimer’s disease, was among those identified. Despite its involvement in PD and AD, structural aspects are currently missing at the molecular level. We present the first atomistic model of the 5-phosphatase domain of synaptojanin-1 and its binding to its substrate phosphatidylinositol 4,5-bisphosphate (PIP2). We determine structural information on the active site including membrane-embedded molecular dynamics simulations. Deficiency of charge within the active site of the protein is observed, which suggests that a second divalent cation is required to complete dephosphorylation of the substrate. The findings in this work shed light on the protein’s binding to phosphatidylinositol 4,5-bisphosphate (PIP2) and give additional insight for future targeting of the protein active site, which might be of interest in neurodegenerative diseases where synaptojanin-1 is overexpressed.
Collapse
Affiliation(s)
- Kirsten Jenkins
- Randall Division of Cell and Molecular Biophysics, Institute for Mathematical and Molecular Biomedicine, King's College London, London SE1 1UL, UK
| | - Teodora Mateeva
- Department of Chemistry, King's College London, London SE1 1DB, UK
| | - István Szabó
- Department of Chemistry, King's College London, London SE1 1DB, UK
| | - Andre Melnik
- Institute of Biochemistry, Department of Biology, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Paola Picotti
- Institute of Biochemistry, Department of Biology, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Attila Csikász-Nagy
- Randall Division of Cell and Molecular Biophysics, Institute for Mathematical and Molecular Biomedicine, King's College London, London SE1 1UL, UK.,Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, 1083 Budapest, Hungary
| | - Edina Rosta
- Department of Chemistry, King's College London, London SE1 1DB, UK
| |
Collapse
|
17
|
Nelson N, Wundenberg T, Lin H, Rehbach C, Horn S, Windhorst S, Jücker M. Characterization of the substrate specificity of the inositol 5-phosphatase SHIP1. Biochem Biophys Res Commun 2020; 524:366-370. [PMID: 32005521 DOI: 10.1016/j.bbrc.2020.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 01/02/2020] [Indexed: 10/25/2022]
Abstract
SHIP1 is an inositol 5-phosphatase which is well established for its tumour suppressor potential in leukaemia. Enzymatically, two SHIP1 substrates, PtdIns(3,4,5)P3 and Ins(1,3,4,5)P4 have been identified to date. Additional substrates were found for the homologue SHIP2. In this study, we identified new inositol phosphate (InsP) substrates of SHIP1 by metal dye detection high-performance liquid chromatography and compared the substrate profiles of SHIP1 and SHIP2. We were able to verify Ins(1,3,4,5)P4 as a substrate of SHIP1 and interestingly found Ins(1,2,3,4,5)P5 and Ins(2,3,4,5)P4 to be preferably used as substrates and Ins(1,4,5,6)P4 and Ins(2,4,5,6)P4 to be weak substrates. All of those except Ins(2,3,4,5)P4 are also known substrates of SHIP2 indicating a possible exclusive role of Ins(2,3,4,5)P4 hydrolysis for SHIP1 but not SHIP2 function.
Collapse
Affiliation(s)
- Nina Nelson
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Torsten Wundenberg
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | | | - Christoph Rehbach
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Stefan Horn
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany; Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Sabine Windhorst
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| |
Collapse
|
18
|
Small molecule targeting of SHIP1 and SHIP2. Biochem Soc Trans 2020; 48:291-300. [DOI: 10.1042/bst20190775] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
Modulating the activity of the Src Homology 2 (SH2) — containing Inositol 5′-Phosphatase (SHIP) enzyme family with small molecule inhibitors provides a useful and unconventional method of influencing cell signaling in the PI3K pathway. The development of small molecules that selectively target one of the SHIP paralogs (SHIP1 or SHIP2) as well as inhibitors that simultaneously target both enzymes have provided promising data linking the phosphatase activity of the SHIP enzymes to disorders and disease states that are in dire need of new therapeutic targets. These include cancer, immunotherapy, diabetes, obesity, and Alzheimer's disease. In this mini-review, we will provide a brief overview of research in these areas that support targeting SHIP1, SHIP2 or both enzymes for therapeutic purposes.
Collapse
|
19
|
Nuclear Phosphatidylinositol 5-Phosphatase Is Essential for Allelic Exclusion of Variant Surface Glycoprotein Genes in Trypanosomes. Mol Cell Biol 2019; 39:MCB.00395-18. [PMID: 30420356 DOI: 10.1128/mcb.00395-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/23/2018] [Indexed: 11/20/2022] Open
Abstract
Allelic exclusion of variant surface glycoprotein (VSG) genes is essential for African trypanosomes to evade the host antibody response by antigenic variation. The mechanisms by which this parasite expresses only one of its ∼2,000 VSG genes at a time are unknown. We show that nuclear phosphatidylinositol 5-phosphatase (PIP5Pase) interacts with repressor activator protein 1 (RAP1) in a multiprotein complex and functions in the control of VSG allelic exclusion. RAP1 binds PIP5Pase substrate phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3], and catalytic mutation of PIP5Pase that inhibits PI(3,4,5)P3 dephosphorylation results in simultaneous transcription of VSGs from all telomeric expression sites (ESs) and from silent subtelomeric VSG arrays. PIP5Pase and RAP1 bind to telomeric ESs, especially at 70-bp repeats and telomeres, and their binding is altered by PIP5Pase inactivation or knockdown, implying changes in ES chromatin organization. Our data suggest a model whereby PIP5Pase controls PI(3,4,5)P3 binding by RAP1 and, thus, RAP1 silencing of telomeric and subtelomeric VSG genes. Hence, allelic exclusion of VSG genes may entail control of nuclear phosphoinositides.
Collapse
|
20
|
Goulden BD, Pacheco J, Dull A, Zewe JP, Deiters A, Hammond GRV. A high-avidity biosensor reveals plasma membrane PI(3,4)P 2 is predominantly a class I PI3K signaling product. J Cell Biol 2018; 218:1066-1079. [PMID: 30591513 PMCID: PMC6400549 DOI: 10.1083/jcb.201809026] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/20/2018] [Accepted: 12/07/2018] [Indexed: 12/21/2022] Open
Abstract
Class I phosphoinositide 3-OH kinase (PI3K) signaling is central to animal growth and metabolism, and pathological disruption of this pathway affects cancer and diabetes. However, the specific spatial/temporal dynamics and signaling roles of its minor lipid messenger, phosphatidylinositol (3,4)-bisphosphate (PI(3,4)P2), are not well understood. This owes principally to a lack of tools to study this scarce lipid. Here we developed a high-sensitivity genetically encoded biosensor for PI(3,4)P2, demonstrating high selectivity and specificity of the sensor for the lipid. We show that despite clear evidence for class II PI3K in PI(3,4)P2-driven function, the overwhelming majority of the lipid accumulates through degradation of class I PI3K-produced PIP3 However, we show that PI(3,4)P2 is also subject to hydrolysis by the tumor suppressor lipid phosphatase PTEN. Collectively, our results show that PI(3,4)P2 is potentially an important driver of class I PI3K-driven signaling and provides powerful new tools to begin to resolve the biological functions of this lipid downstream of class I and II PI3K.
Collapse
Affiliation(s)
- Brady D Goulden
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jonathan Pacheco
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Allyson Dull
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - James P Zewe
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
21
|
Voth KA, Chung IYW, van Straaten K, Li L, Boniecki MT, Cygler M. The structure of Legionella effector protein LpnE provides insights into its interaction with Oculocerebrorenal syndrome of Lowe (OCRL) protein. FEBS J 2018; 286:710-725. [PMID: 30479037 DOI: 10.1111/febs.14710] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/18/2018] [Accepted: 11/23/2018] [Indexed: 12/17/2022]
Abstract
Legionella pneumophila is a freshwater bacterium that replicates in predatory amoeba and alveolar macrophage. The ability of L. pneumophila to thrive in eukaryotic host cells is conferred by the Legionella containing vacuole (LCV). Formation and intracellular trafficking of the LCV are governed by an arsenal of effector proteins, many of which are secreted by the Icm/Dot Type 4 Secretion System. One such effector, known as LpnE (L. pneumophila Entry), has been implicated in facilitating bacterial entry into host cells, LCV trafficking, and substrate translocation. LpnE belongs to a subfamily of tetratricopeptide repeat proteins known as Sel1-like repeats (SLRs). All eight of the predicted SLRs in LpnE are required to promote host cell invasion. Herein, we report that LpnE(1-375) localizes to cis-Golgi in HEK293 cells via its signal peptide (aa 1-22). We further verify the interaction of LpnE(73-375) and LpnE(22-375) with Oculocerebrorenal syndrome of Lowe protein (OCRL) residues 10-208, restricting the known interacting residues for both proteins. To further characterize the SLR region of LpnE, we solved the crystal structure of LpnE(73-375) to 1.75Å resolution. This construct comprises all SLRs, which are arranged in a superhelical fold. The α-helices forming the inner concave surface of the LpnE superhelix suggest a potential protein-protein interaction interface. DATABASE: Coordinates and structure factors were deposited in the Protein Data Bank with the accession number 6DEH.
Collapse
Affiliation(s)
- Kevin A Voth
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Canada
| | - Ivy Yeuk Wah Chung
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Canada
| | - Karin van Straaten
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Canada
| | - Lei Li
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Canada
| | - Michal T Boniecki
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Canada
| | - Miroslaw Cygler
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
22
|
Kawanabe A, Hashimoto M, Nishizawa M, Nishizawa K, Narita H, Yonezawa T, Jinno Y, Sakata S, Nakagawa A, Okamura Y. The hydrophobic nature of a novel membrane interface regulates the enzyme activity of a voltage-sensing phosphatase. eLife 2018; 7:41653. [PMID: 30484774 PMCID: PMC6298786 DOI: 10.7554/elife.41653] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 11/28/2018] [Indexed: 01/24/2023] Open
Abstract
Voltage-sensing phosphatases (VSP) contain a voltage sensor domain (VSD) similar to that of voltage-gated ion channels but lack a pore-gate domain. A VSD in a VSP regulates the cytoplasmic catalytic region (CCR). However, the mechanisms by which the VSD couples to the CCR remain elusive. Here we report a membrane interface (named ‘the hydrophobic spine’), which is essential for the coupling of the VSD and CCR. Our molecular dynamics simulations suggest that the hydrophobic spine of Ciona intestinalis VSP (Ci-VSP) provides a hinge-like motion for the CCR through the loose membrane association of the phosphatase domain. Electrophysiological experiments indicate that the voltage-dependent phosphatase activity of Ci-VSP depends on the hydrophobicity and presence of an aromatic ring in the hydrophobic spine. Analysis of conformational changes in the VSD and CCR suggests that the VSP has two states with distinct enzyme activities and that the second transition depends on the hydrophobic spine.
Collapse
Affiliation(s)
- Akira Kawanabe
- Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaki Hashimoto
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | | | | | - Hirotaka Narita
- Institute for Protein Research, Osaka University, Osaka, Japan
| | - Tomoko Yonezawa
- Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuka Jinno
- Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Souhei Sakata
- Department of Physiology, Division of Life Sciences, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | | | - Yasushi Okamura
- Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| |
Collapse
|
23
|
Synaptojanin regulates Hedgehog signalling by modulating phosphatidylinositol 4-phosphate levels. J Biosci 2018. [DOI: 10.1007/s12038-018-9799-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
24
|
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
|
25
|
Wang Z, Nie Y, Zhang K, Xu H, Ramelot TA, Kennedy MA, Liu M, Zhu J, Yang Y. Solution structure of SHIP2 SH2 domain and its interaction with a phosphotyrosine peptide from c-MET. Arch Biochem Biophys 2018; 656:31-37. [PMID: 30165040 DOI: 10.1016/j.abb.2018.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/10/2018] [Accepted: 08/26/2018] [Indexed: 12/12/2022]
Abstract
SH2 domain-containing inositol 5-phosphatase 2 (SHIP2) binds with the Y1356-phosphorylated hepatocyte growth factor (HGF) receptor, c-MET, through its SH2 domain, which is essential for the role of SHIP2 in HGF-induced cell scattering and cell spreading. Previously, the experimental structure of the SH2 domain from SHIP2 (SHIP2-SH2) had not been reported, and its interaction with the Y1356-phosphorylated c-MET had not been investigated from a structural point of view. In this study, the solution structure of SHIP2-SH2 was determined by NMR spectroscopy, where it was found to adopt a typical SH2-domain fold that contains a positively-charged pocket for binding to phosphotyrosine (pY). The interaction between SHIP2-SH2 and a pY-containing peptide from c-MET (Y1356 phosphorylated) was investigated through NMR titrations. The results showed that the binding affinity of SHIP2-SH2 with the phosphopeptide is at low micromolar level, and the binding interface consists of the positively-charged pocket and its surrounding regions. Furthermore, R28, S49 and R70 were identified as key residues for the binding and may directly interact with the pY. Taken together, these findings provide structural insights into the binding of SHIP2-SH2 with the Y1356-phosphorylated c-MET, and lay a foundation for further studies of the interactions between SHIP2-SH2 and its various binding partners.
Collapse
Affiliation(s)
- Zi Wang
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yao Nie
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kunxiao Zhang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Huaihai Institute of Technology, Lianyungang, 222005, China
| | - Henghao Xu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Huaihai Institute of Technology, Lianyungang, 222005, China
| | - Theresa A Ramelot
- Department of Chemistry and Biochemistry, The Northeast Structural Genomics Consortium, Miami University, Oxford, OH, 45056, United States
| | - Michael A Kennedy
- Department of Chemistry and Biochemistry, The Northeast Structural Genomics Consortium, Miami University, Oxford, OH, 45056, United States
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jiang Zhu
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Yunhuang Yang
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
26
|
Neuwald AF, Aravind L, Altschul SF. Inferring joint sequence-structural determinants of protein functional specificity. eLife 2018; 7. [PMID: 29336305 PMCID: PMC5770160 DOI: 10.7554/elife.29880] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 12/22/2017] [Indexed: 01/05/2023] Open
Abstract
Residues responsible for allostery, cooperativity, and other subtle but functionally important interactions remain difficult to detect. To aid such detection, we employ statistical inference based on the assumption that residues distinguishing a protein subgroup from evolutionarily divergent subgroups often constitute an interacting functional network. We identify such networks with the aid of two measures of statistical significance. One measure aids identification of divergent subgroups based on distinguishing residue patterns. For each subgroup, a second measure identifies structural interactions involving pattern residues. Such interactions are derived either from atomic coordinates or from Direct Coupling Analysis scores, used as surrogates for structural distances. Applying this approach to N-acetyltransferases, P-loop GTPases, RNA helicases, synaptojanin-superfamily phosphatases and nucleases, and thymine/uracil DNA glycosylases yielded results congruent with biochemical understanding of these proteins, and also revealed striking sequence-structural features overlooked by other methods. These and similar analyses can aid the design of drugs targeting allosteric sites.
Collapse
Affiliation(s)
- Andrew F Neuwald
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, United States.,Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, United States
| | - L Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - Stephen F Altschul
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| |
Collapse
|
27
|
Willett R, Martina JA, Zewe JP, Wills R, Hammond GRV, Puertollano R. TFEB regulates lysosomal positioning by modulating TMEM55B expression and JIP4 recruitment to lysosomes. Nat Commun 2017; 8:1580. [PMID: 29146937 PMCID: PMC5691037 DOI: 10.1038/s41467-017-01871-z] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 10/20/2017] [Indexed: 12/27/2022] Open
Abstract
Lysosomal distribution is linked to the role of lysosomes in many cellular functions, including autophagosome degradation, cholesterol homeostasis, antigen presentation, and cell invasion. Alterations in lysosomal positioning contribute to different human pathologies, such as cancer, neurodegeneration, and lysosomal storage diseases. Here we report the identification of a novel mechanism of lysosomal trafficking regulation. We found that the lysosomal transmembrane protein TMEM55B recruits JIP4 to the lysosomal surface, inducing dynein-dependent transport of lysosomes toward the microtubules minus-end. TMEM55B overexpression causes lysosomes to collapse into the cell center, whereas depletion of either TMEM55B or JIP4 results in dispersion toward the cell periphery. TMEM55B levels are transcriptionally upregulated following TFEB and TFE3 activation by starvation or cholesterol-induced lysosomal stress. TMEM55B or JIP4 depletion abolishes starvation-induced retrograde lysosomal transport and prevents autophagosome-lysosome fusion. Overall our data suggest that the TFEB/TMEM55B/JIP4 pathway coordinates lysosome movement in response to a variety of stress conditions.
Collapse
Affiliation(s)
- Rose Willett
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Room 3537, Bethesda, MD, 20892, USA
| | - José A Martina
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Room 3537, Bethesda, MD, 20892, USA
| | - James P Zewe
- Department of Cell Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Room S332 Biomedical Sciences Tower, Pittsburgh, PA, 15213, USA
| | - Rachel Wills
- Department of Cell Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Room S332 Biomedical Sciences Tower, Pittsburgh, PA, 15213, USA
| | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Room S332 Biomedical Sciences Tower, Pittsburgh, PA, 15213, USA
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Room 3537, Bethesda, MD, 20892, USA.
| |
Collapse
|
28
|
Le Coq J, Camacho-Artacho M, Velázquez JV, Santiveri CM, Gallego LH, Campos-Olivas R, Dölker N, Lietha D. Structural basis for interdomain communication in SHIP2 providing high phosphatase activity. eLife 2017; 6. [PMID: 28792888 PMCID: PMC5550278 DOI: 10.7554/elife.26640] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/24/2017] [Indexed: 12/17/2022] Open
Abstract
SH2-containing-inositol-5-phosphatases (SHIPs) dephosphorylate the 5-phosphate of phosphatidylinositol-3,4,5-trisphosphate (PI(3,4,5)P3) and play important roles in regulating the PI3K/Akt pathway in physiology and disease. Aiming to uncover interdomain regulatory mechanisms in SHIP2, we determined crystal structures containing the 5-phosphatase and a proximal region adopting a C2 fold. This reveals an extensive interface between the two domains, which results in significant structural changes in the phosphatase domain. Both the phosphatase and C2 domains bind phosphatidylserine lipids, which likely helps to position the active site towards its substrate. Although located distant to the active site, the C2 domain greatly enhances catalytic turnover. Employing molecular dynamics, mutagenesis and cell biology, we identify two distinct allosteric signaling pathways, emanating from hydrophobic or polar interdomain interactions, differentially affecting lipid chain or headgroup moieties of PI(3,4,5)P3. Together, this study reveals details of multilayered C2-mediated effects important for SHIP2 activity and points towards interesting new possibilities for therapeutic interventions. DOI:http://dx.doi.org/10.7554/eLife.26640.001
Collapse
Affiliation(s)
- Johanne Le Coq
- Cell Signalling and Adhesion Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Marta Camacho-Artacho
- Cell Signalling and Adhesion Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - José Vicente Velázquez
- Cell Signalling and Adhesion Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Clara M Santiveri
- Spectroscopy and Nuclear Magnetic Resonance Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Luis Heredia Gallego
- Cell Signalling and Adhesion Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Ramón Campos-Olivas
- Spectroscopy and Nuclear Magnetic Resonance Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Nicole Dölker
- Structural Computational Biology Group, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Daniel Lietha
- Cell Signalling and Adhesion Group, Spanish National Cancer Research Centre, Madrid, Spain
| |
Collapse
|
29
|
Ocular Pathology of Oculocerebrorenal Syndrome of Lowe: Novel Mutations and Genotype-Phenotype Analysis. Sci Rep 2017; 7:1442. [PMID: 28473699 PMCID: PMC5431454 DOI: 10.1038/s41598-017-01447-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/28/2017] [Indexed: 12/02/2022] Open
Abstract
Mutations in the OCRL1 gene result in the oculocerebrorenal syndrome of Lowe, with symptoms including congenital bilateral cataracts, glaucoma, renal failure, and neurological impairments. OCRL1 encodes an inositol polyphosphate 5-phosphatase which preferentially dephosphorylates phosphatidylinositide 4,5 bisphosphate (PI(4,5)P2). We have identified two novel mutations in two unrelated Lowe syndrome patients with congenital glaucoma. Novel deletion mutations are detected at c.739-742delAAAG in Lowe patient 1 and c.1595-1631del in Lowe patient 2. End stage glaucoma in patient 2 resulted in the enucleation of the eye, which on histology demonstrated corneal keloid, fibrous infiltration of the angle, ectropion uvea, retinal gliosis, and retinal ganglion cell loss. We measured OCRL protein levels in patient keratinocytes and found that Lowe 1 patient cells had significantly reduced OCRL protein as compared to the control keratinocytes. Genotype-phenotype correlation of OCRL1 mutations associated with congenital glaucoma revealed clustering of missense and deletion mutations in the 5-phosphatase domain and the RhoGAP-like domain. In conclusion, we report novel OCRL1 mutations in Lowe syndrome patients and the corresponding histopathologic analysis of one patient’s ocular pathology.
Collapse
|
30
|
Thomas MP, Erneux C, Potter BVL. SHIP2: Structure, Function and Inhibition. Chembiochem 2017; 18:233-247. [DOI: 10.1002/cbic.201600541] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Mark P. Thomas
- Department of Pharmacy and Pharmacology; University of Bath; Claverton Down Bath BA2 7AY UK
| | - Christophe Erneux
- I.R.I.B.H.M.; Université Libre de Bruxelles; Campus Erasme 808 Route de Lennik 1070 Brussels Belgium
| | - Barry V. L. Potter
- Drug Discovery and Medicinal Chemistry; Department of Pharmacology; University of Oxford; Mansfield Road Oxford OX1 3QT UK
| |
Collapse
|
31
|
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
|
32
|
Alonso A, Pulido R. The extended human PTPome: a growing tyrosine phosphatase family. FEBS J 2015; 283:1404-29. [PMID: 26573778 DOI: 10.1111/febs.13600] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 10/02/2015] [Accepted: 11/13/2015] [Indexed: 12/13/2022]
Abstract
Tyr phosphatases are, by definition, enzymes that dephosphorylate phospho-Tyr (pTyr) from proteins. This activity is found in several structurally diverse protein families, including the protein Tyr phosphatase (PTP), arsenate reductase, rhodanese, haloacid dehalogenase (HAD) and His phosphatase (HP) families. Most of these families include members with substrate specificity for non-pTyr substrates, such as phospho-Ser/phospho-Thr, phosphoinositides, phosphorylated carbohydrates, mRNAs, or inorganic moieties. A Cys is essential for catalysis in PTPs, rhodanese and arsenate reductase enzymes, whereas this work is performed by an Asp in HAD phosphatases and by a His in HPs, via a catalytic mechanism shared by all of the different families. The category that contains most Tyr phosphatases is the PTP family, which, although it received its name from this activity, includes Ser, Thr, inositide, carbohydrate and RNA phosphatases, as well as some inactive pseudophosphatase proteins. Here, we propose an extended collection of human Tyr phosphatases, which we call the extended human PTPome. The addition of new members (SACs, paladin, INPP4s, TMEM55s, SSU72, and acid phosphatases) to the currently categorized PTP group of enzymes means that the extended human PTPome contains up to 125 proteins, of which ~ 40 are selective for pTyr. We set criteria to ascribe proteins to the extended PTPome, and summarize the more important features of the new PTPome members in the context of their phosphatase activity and their relationship with human disease.
Collapse
Affiliation(s)
- Andrés Alonso
- Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Rafael Pulido
- Biocruces Health Research Institute, Barakaldo, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
33
|
Braun W, Schein CH. Membrane interaction and functional plasticity of inositol polyphosphate 5-phosphatases. Structure 2015; 22:664-6. [PMID: 24807076 DOI: 10.1016/j.str.2014.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In this issue of Structure, Trésaugues and colleagues determined the interaction of membrane-bound phosphoinositides with three clinically significant human inositol polyphosphate 5-phosphatases (I5Ps). A comparison to the structures determined with soluble substrates revealed differences in the binding mode and suggested how the I5Ps and apurinic endonuclease (APE1) activities evolved from the same metal-binding active center.
Collapse
Affiliation(s)
- Werner Braun
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Catherine H Schein
- Foundation for Applied Molecular Evolution, 720 SW 2nd Avenue Suite 201, Gainesville, FL 32601, USA
| |
Collapse
|
34
|
Inositol phosphate pathway controls transcription of telomeric expression sites in trypanosomes. Proc Natl Acad Sci U S A 2015; 112:E2803-12. [PMID: 25964327 DOI: 10.1073/pnas.1501206112] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
African trypanosomes evade clearance by host antibodies by periodically changing their variant surface glycoprotein (VSG) coat. They transcribe only one VSG gene at a time from 1 of about 20 telomeric expression sites (ESs). They undergo antigenic variation by switching transcription between telomeric ESs or by recombination of the VSG gene expressed. We show that the inositol phosphate (IP) pathway controls transcription of telomeric ESs and VSG antigenic switching in Trypanosoma brucei. Conditional knockdown of phosphatidylinositol 5-kinase (TbPIP5K) or phosphatidylinositol 5-phosphatase (TbPIP5Pase) or overexpression of phospholipase C (TbPLC) derepresses numerous silent ESs in T. brucei bloodstream forms. The derepression is specific to telomeric ESs, and it coincides with an increase in the number of colocalizing telomeric and RNA polymerase I foci in the nucleus. Monoallelic VSG transcription resumes after reexpression of TbPIP5K; however, most of the resultant cells switched the VSG gene expressed. TbPIP5K, TbPLC, their substrates, and products localize to the plasma membrane, whereas TbPIP5Pase localizes to the nucleus proximal to telomeres. TbPIP5Pase associates with repressor/activator protein 1 (TbRAP1), and their telomeric silencing function is altered by TbPIP5K knockdown. These results show that specific steps in the IP pathway control ES transcription and antigenic switching in T. brucei by epigenetic regulation of telomere silencing.
Collapse
|
35
|
Waugh MG. PIPs in neurological diseases. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1066-82. [PMID: 25680866 DOI: 10.1016/j.bbalip.2015.02.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/29/2015] [Accepted: 02/01/2015] [Indexed: 12/19/2022]
Abstract
Phosphoinositide (PIP) lipids regulate many aspects of cell function in the nervous system including receptor signalling, secretion, endocytosis, migration and survival. Levels of PIPs such as PI4P, PI(4,5)P2 and PI(3,4,5)P3 are normally tightly regulated by phosphoinositide kinases and phosphatases. Deregulation of these biochemical pathways leads to lipid imbalances, usually on intracellular endosomal membranes, and these changes have been linked to a number of major neurological diseases including Alzheimer's, Parkinson's, epilepsy, stroke, cancer and a range of rarer inherited disorders including brain overgrowth syndromes, Charcot-Marie-Tooth neuropathies and neurodevelopmental conditions such as Lowe's syndrome. This article analyses recent progress in this area and explains how PIP lipids are involved, to varying degrees, in almost every class of neurological disease. This article is part of a Special Issue entitled Brain Lipids.
Collapse
Affiliation(s)
- Mark G Waugh
- Lipid and Membrane Biology Group, Institute for Liver and Digestive Health, UCL, Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom.
| |
Collapse
|
36
|
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
|
37
|
Wang F, Cheung S, Mui A. SHIP2 phosphatase domain structure suggests sites of regulation (retrospective on DOI 10.1002/bies.201100195). Bioessays 2014; 36:1019. [PMID: 25251772 DOI: 10.1002/bies.201400158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Frank Wang
- University of British Columbia, Surgery, Biochemistry and Molecular Biology, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
38
|
Emptage RP, Tonthat NK, York JD, Schumacher MA, Zhou P. Structural basis of lipid binding for the membrane-embedded tetraacyldisaccharide-1-phosphate 4'-kinase LpxK. J Biol Chem 2014; 289:24059-68. [PMID: 25023290 DOI: 10.1074/jbc.m114.589986] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The membrane-bound tetraacyldisaccharide-1-phosphate 4'-kinase, LpxK, catalyzes the sixth step of the lipid A (Raetz) biosynthetic pathway and is a viable antibiotic target against emerging Gram-negative pathogens. We report the crystal structure of lipid IVA, the LpxK product, bound to the enzyme, providing a rare glimpse into interfacial catalysis and the surface scanning strategy by which many poorly understood lipid modification enzymes operate. Unlike the few previously structurally characterized proteins that bind lipid A or its precursors, LpxK binds almost exclusively to the glucosamine/phosphate moieties of the lipid molecule. Steady-state kinetic analysis of multiple point mutants of the lipid-binding pocket pinpoints critical residues involved in substrate binding, and characterization of N-terminal helix truncation mutants uncovers the role of this substructure as a hydrophobic membrane anchor. These studies make critical contributions to the limited knowledge surrounding membrane-bound enzymes that act upon lipid substrates and provide a structural template for designing small molecule inhibitors targeting this essential kinase.
Collapse
Affiliation(s)
- Ryan P Emptage
- From the Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710 and
| | - Nam K Tonthat
- From the Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710 and
| | - John D York
- the Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37205
| | - Maria A Schumacher
- From the Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710 and
| | - Pei Zhou
- From the Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710 and
| |
Collapse
|
39
|
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
|