Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 25(10)2024 May 11.
Article in English | MEDLINE | ID: mdl-38791291

ABSTRACT

The src homology 2 domain-containing inositol 5-phosphatases SHIP1 and SHIP2 are two proteins involved in intracellular signaling pathways and have been linked to the pathogenesis of several diseases. Both protein paralogs are well known for their involvement in the formation of various kinds of cancer. SHIP1, which is expressed predominantly in hematopoietic cells, has been implicated as a tumor suppressor in leukemogenesis especially in myeloid leukemia, whereas SHIP2, which is expressed ubiquitously, has been implicated as an oncogene in a wider variety of cancer types and is suggested to be involved in the process of metastasis of carcinoma cells. However, there are numerous other diseases, such as inflammatory diseases as well as allergic responses, Alzheimer's disease, and stroke, in which SHIP1 can play a role. Moreover, SHIP2 overexpression was shown to correlate with opsismodysplasia and Alzheimer's disease, as well as metabolic diseases. The SHIP1-inhibitor 3-α-aminocholestane (3AC), and SHIP1-activators, such as AQX-435 and AQX-1125, and SHIP2-inhibitors, such as K161 and AS1949490, have been developed and partly tested in clinical trials, which indicates the importance of the SHIP-paralogs as possible targets in the therapy of those diseases. The aim of this article is to provide an overview of the current knowledge about the involvement of SHIP proteins in the pathogenesis of cancer and other human diseases and to create awareness that SHIP1 and SHIP2 are more than just tumor suppressors and oncogenes.


Subject(s)
Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Humans , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Neoplasms/metabolism , Neoplasms/pathology , Animals , src Homology Domains , Signal Transduction , Inositol Polyphosphate 5-Phosphatases/metabolism , Inositol Polyphosphate 5-Phosphatases/genetics
2.
Nat Cancer ; 5(3): 481-499, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38233483

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.


Subject(s)
Melanoma , Humans , Melanoma/drug therapy , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/therapeutic use , Mutation , Signal Transduction , Inositol Polyphosphate 5-Phosphatases/genetics
3.
Iran Biomed J ; 26(6): 440-53, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36437782

ABSTRACT

Background: Background: Type I inositol polyphosphate-5-phosphatase A (INPP5A) is involved in different cellular events, including cell proliferation. Since INPP5A, HLAG1, IL-10, and matrix metalloproteinases (MMP)-21 genes play fundamental roles in esophageal squamous cell carcinoma (ESCC) tumorigenesis, we aimed in this study to clarify the possible interplay of these genes and explore the potential of these chemistries as a predictor marker for diagnosis in ESCC disease. Methods: Methods: Gene expression analysis of INPP5A, HLAG-1, IL-10, and MMP-21 was performed using relative comparative real-time PCR in 56 ESCCs compared to their margin normal tissues. Immunohistochemical staining was accomplished for INPP5A in ESCCs. Analysis of ROC curves and the AUC were applied to evaluate the diagnostic capability of the candidate genes. Results: Results: High levels of HLA-G1, MMP-21, and IL-10 were detected in nearly 23.2%, 62.5%, and 53.5% of ESCCs compared to the normal tissues, respectively, whereas INPP5A underexpression was detected in 19.6% of ESCCs, which all tested genes indicated significant correlations with each other. The protein expression level of INPP5A in ESCC tissues was significantly lower than that of the non-tumor esophageal tissues (p = 0.001). Interestingly, the concomitant expression of the INPP5A/HLA-G1, INPP5A/MMP-21, INPP5A/IL-10, HLA-G1/MMP-21, HLA-G1/IL-10, and MMP-21/IL-10 was significantly correlated with several clinicopathological variables. INPP5A, HLA-G1, MMP-21, and IL-10 showed to be the most appropriate candidates to discriminate tumor/non-tumor groups due to the total AUCs of all combinations (>60%). Conclusion: Conclusion: Our results represent a new regulatory axis containing INPP5A/HLAG-1/IL-10/MMP-21 markers in ESCC development and may provide novel insight into the mechanism of immune evasion mediated by the INPP5A/HLAG-1/IL-10/MMP-21 regulatory network in the disease.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Cell Line, Tumor , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic , HLA-G Antigens/genetics , HLA-G Antigens/metabolism , Inositol Polyphosphate 5-Phosphatases/genetics , Inositol Polyphosphate 5-Phosphatases/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Matrix Metalloproteinases, Secreted/genetics , Matrix Metalloproteinases, Secreted/metabolism
4.
J Cell Biol ; 221(9)2022 09 05.
Article in English | MEDLINE | ID: mdl-35878408

ABSTRACT

Upon antigen binding, the B cell receptor (BCR) undergoes clustering to form a signalosome that propagates downstream signaling required for normal B cell development and physiology. BCR clustering is dependent on remodeling of the cortical actin network, but the mechanisms that regulate actin remodeling in this context remain poorly defined. In this study, we identify the inositol 5-phosphatase INPP5B as a key regulator of actin remodeling, BCR clustering, and downstream signaling in antigen-stimulated B cells. INPP5B acts via dephosphorylation of the inositol lipid PI(4,5)P2 that in turn is necessary for actin disassembly, BCR mobilization, and cell spreading on immobilized surface antigen. These effects can be explained by increased actin severing by cofilin and loss of actin linking to the plasma membrane by ezrin, both of which are sensitive to INPP5B-dependent PI(4,5)P2 hydrolysis. INPP5B is therefore a new player in BCR signaling and may represent an attractive target for treatment of B cell malignancies caused by aberrant BCR signaling.


Subject(s)
Actins , Inositol Polyphosphate 5-Phosphatases , Receptors, Antigen, B-Cell , Actins/metabolism , B-Lymphocytes , Humans , Inositol Polyphosphate 5-Phosphatases/genetics , Inositol Polyphosphate 5-Phosphatases/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphoric Monoester Hydrolases , Receptors, Antigen, B-Cell/metabolism
5.
Nat Plants ; 7(5): 587-597, 2021 05.
Article in English | MEDLINE | ID: mdl-34007035

ABSTRACT

Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is a low-abundance membrane lipid essential for plasma membrane function1,2. In plants, mutations in phosphatidylinositol 4-phosphate (PI4P) 5-kinases (PIP5K) suggest that PI(4,5)P2 production is involved in development, immunity and reproduction3-5. However, phospholipid synthesis is highly intricate6. It is thus likely that steady-state depletion of PI(4,5)P2 triggers confounding indirect effects. Furthermore, inducible tools available in plants allow PI(4,5)P2 to increase7-9 but not decrease, and no PIP5K inhibitors are available. Here, we introduce iDePP (inducible depletion of PI(4,5)P2 in plants), a system for the inducible and tunable depletion of PI(4,5)P2 in plants in less than three hours. Using this strategy, we confirm that PI(4,5)P2 is critical for various aspects of plant development, including root growth, root-hair elongation and organ initiation. We show that PI(4,5)P2 is required to recruit various endocytic proteins, including AP2-µ, to the plasma membrane, and thus to regulate clathrin-mediated endocytosis. Finally, we find that inducible PI(4,5)P2 perturbation impacts the dynamics of the actin cytoskeleton as well as microtubule anisotropy. Together, we propose that iDePP is a simple and efficient genetic tool to test the importance of PI(4,5)P2 in given cellular or developmental responses, and also to evaluate the importance of this lipid in protein localization.


Subject(s)
Arabidopsis/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Arabidopsis/genetics , Arabidopsis/growth & development , Cell Membrane/metabolism , Cytoskeleton/metabolism , Drosophila Proteins/genetics , Inositol Polyphosphate 5-Phosphatases/genetics , Phosphatidylinositol 4,5-Diphosphate/physiology , Phospholipids/metabolism , Plant Roots/growth & development , Plant Roots/metabolism , Plants, Genetically Modified
6.
Nat Commun ; 12(1): 2673, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976123

ABSTRACT

Vesicular traffic and membrane contact sites between organelles enable the exchange of proteins, lipids, and metabolites. Recruitment of tethers to contact sites between the endoplasmic reticulum (ER) and the plasma membrane is often triggered by calcium. Here we reveal a function for calcium in the repression of cholesterol export at membrane contact sites between the ER and the Golgi complex. We show that calcium efflux from ER stores induced by inositol-triphosphate [IP3] accumulation upon loss of the inositol 5-phosphatase INPP5A or receptor signaling triggers depletion of cholesterol and associated Gb3 from the cell surface, resulting in a blockade of clathrin-independent endocytosis (CIE) of Shiga toxin. This phenotype is caused by the calcium-induced dissociation of oxysterol binding protein (OSBP) from the Golgi complex and from VAP-containing membrane contact sites. Our findings reveal a crucial function for INPP5A-mediated IP3 hydrolysis in the control of lipid exchange at membrane contact sites.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Inositol Phosphates/metabolism , Membrane Lipids/metabolism , Animals , Biological Transport , COS Cells , Chlorocebus aethiops , Cholesterol/metabolism , Endocytosis , HEK293 Cells , HeLa Cells , Humans , Inositol Polyphosphate 5-Phosphatases/genetics , Inositol Polyphosphate 5-Phosphatases/metabolism , Microscopy, Confocal , Phosphatidylinositol Phosphates/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Trihexosylceramides/metabolism
7.
Brain ; 144(8): 2427-2442, 2021 09 04.
Article in English | MEDLINE | ID: mdl-33792664

ABSTRACT

Marinesco-Sjögren syndrome is a rare human disorder caused by biallelic mutations in SIL1 characterized by cataracts in infancy, myopathy and ataxia, symptoms which are also associated with a novel disorder caused by mutations in INPP5K. While these phenotypic similarities may suggest commonalties at a molecular level, an overlapping pathomechanism has not been established yet. In this study, we present six new INPP5K patients and expand the current mutational and phenotypical spectrum of the disease showing the clinical overlap between Marinesco-Sjögren syndrome and the INPP5K phenotype. We applied unbiased proteomic profiling on cells derived from Marinesco-Sjögren syndrome and INPP5K patients and identified alterations in d-3-PHGDH as a common molecular feature. d-3-PHGDH modulates the production of l-serine and mutations in this enzyme were previously associated with a neurological phenotype, which clinically overlaps with Marinesco-Sjögren syndrome and INPP5K disease. As l-serine administration represents a promising therapeutic strategy for d-3-PHGDH patients, we tested the effect of l-serine in generated sil1, phgdh and inpp5k a+b zebrafish models, which showed an improvement in their neuronal phenotype. Thus, our study defines a core phenotypical feature underpinning a key common molecular mechanism in three rare diseases and reveals a common and novel therapeutic target for these patients.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Inositol Polyphosphate 5-Phosphatases/genetics , Mutation , Phenotype , Phosphoglycerate Dehydrogenase/genetics , Spinocerebellar Degenerations/genetics , Adolescent , Adult , Animals , Child , Female , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Proteomics , Spinocerebellar Degenerations/pathology , Zebrafish
8.
Am J Med Genet A ; 185(6): 1841-1847, 2021 06.
Article in English | MEDLINE | ID: mdl-33720513

ABSTRACT

Childhood-Onset Schizophrenia (COS) is a very rare and severe psychiatric disorder defined by adult schizophrenia symptoms occurring before the age of 13. We report a microduplication in the 10q26.3 region including part of the Inositol Polyphosphate-5-Phosphatase A (INPP5A) gene that segregates with Schizophrenia Spectrum Disorders (SSDs) in the family of a female patient affected by both COS and Autism Spectrum Disorder (ASD). Phenotyping and genotyping (including CGH-array) were performed for mother, healthy sister, and affected child according to the GenAuDiss protocol (NCT02565524). The duplication size is 324 kb and is present in a patient with COS and in her mother with SSD, but not in the patient's healthy sister. INPP5A encodes a membrane-associated 43 kDa type I inositol 1,4,5-trisphosphate (InsP3) 5-phosphatase. This protein is found both in mouse and human brains and we found that its Drosophila homologue 5PtaseI is specifically expressed in the central nervous system. Hydrolyzed products from InsP3 5-phosphatases mobilize intracellular calcium, which is relevant for dendritic spine morphogenesis in neurons and altered in both schizophrenia and ASD. These may constitute arguments in favor of this gene alteration in the pathophysiology of COS.


Subject(s)
Autism Spectrum Disorder/genetics , Child Development Disorders, Pervasive/genetics , Inositol Polyphosphate 5-Phosphatases/genetics , Schizophrenia, Childhood/genetics , Adolescent , Adult , Animals , Autism Spectrum Disorder/complications , Autism Spectrum Disorder/pathology , Brain/pathology , Child , Child Development Disorders, Pervasive/complications , Child Development Disorders, Pervasive/pathology , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , Male , Mice , Pedigree , Phenotype , Schizophrenia, Childhood/complications , Schizophrenia, Childhood/pathology , Siblings , Young Adult
9.
Adv Biol Regul ; 79: 100760, 2021 01.
Article in English | MEDLINE | ID: mdl-33060052

ABSTRACT

INPP5K (Inositol Polyphosphate 5-Phosphatase K, or SKIP (for Skeletal muscle and Kidney enriched Inositol Phosphatase) is a member of the phosphoinositide 5-phosphatases family. Its protein structure is comprised of a N-terminal catalytic domain which hydrolyses both PtdIns(4,5)P2 and PtdIns(3,4,5)P3, followed by a SKICH domain at the C-terminus which is responsible for protein-protein interactions and subcellular localization of INPP5K. Strikingly, INPP5K is mostly concentrated in the endoplasmic reticulum, although it is also detected at the plasma membrane, in the cytosol and the nucleus. Recently, mutations in INPP5K have been detected in patients with a rare form of autosomal recessive congenital muscular dystrophy with cataract, short stature and intellectual disability. INPP5K functions extend from control of insulin signaling, endoplasmic reticulum stress response and structural integrity, myoblast differentiation, cytoskeleton organization, cell adhesion and migration, renal osmoregulation, to cancer. The goal of this review is thus to summarize and comment recent and less recent data in the literature on INPP5K, in particular on the structure, expression, intracellular localization, interactions and functions of this specific member of the 5-phosphatases family.


Subject(s)
Inositol Polyphosphate 5-Phosphatases/chemistry , Inositol Polyphosphate 5-Phosphatases/metabolism , Animals , Humans , Inositol Polyphosphate 5-Phosphatases/genetics , Mutation , Protein Domains , Protein Transport , Signal Transduction
10.
J Cell Mol Med ; 24(17): 9898-9907, 2020 09.
Article in English | MEDLINE | ID: mdl-32693431

ABSTRACT

Congenital heart disease (CHD) with extracardiac malformations (EM) is the most common multiple malformation, resulting from the interaction between genetic abnormalities and environmental factors. Most studies have attributed the causes of CHD with EM to chromosomal abnormalities. However, multi-system dysplasia is usually caused by both genetic mutations and epigenetic dysregulation. The epigenetic mechanisms underlying the pathogenesis of CHD with EM remain unclear. In this study, we investigated the mechanisms of imprinting alterations, including those of the Small nuclear ribonucleoprotein polypeptide N (SNRPN), PLAG1 like zinc finger 1 (ZAC1) and inositol polyphosphate-5-phosphatase F (INPP5F) genes, in the pathogenesis of CHD with EM. The methylation levels of SNRPN, ZAC1, and INPP5F genes were analysed by the MassARRAY platform in 24 children with CHD with EM and 20 healthy controls. The expression levels of these genes were detected by real-time polymerase chain reaction (PCR). The correlation between methylation regulation and gene expression was confirmed using 5-azacytidine (5-Aza) treated cells. The methylation levels of SNRPN and ZAC1 genes were significantly increased in CHD with EM, while that of INPP5F was decreased. The methylation alterations of these genes were negatively correlated with expression. Risk analysis showed that abnormal hypermethylation of SNRPN and ZAC1 resulted in 5.545 and 7.438 times higher risks of CHD with EM, respectively, and the abnormal hypomethylation of INPP5F was 8.38 times higher than that of the control group. We concluded that abnormally high methylation levels of SNRPN and ZAC1 and decreased levels of INPP5F imply an increased risk of CHD with EM by altering their gene functions. This study provides evidence of imprinted regulation in the pathogenesis of multiple malformations.


Subject(s)
Cell Cycle Proteins/genetics , Heart Defects, Congenital/genetics , Inositol Polyphosphate 5-Phosphatases/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , snRNP Core Proteins/genetics , Azacitidine/chemistry , Child , Child, Preschool , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Female , Gene Expression/genetics , Genomic Imprinting/genetics , Heart Defects, Congenital/epidemiology , Heart Defects, Congenital/pathology , Humans , Infant , Male
11.
Biomed Res Int ; 2020: 9519235, 2020.
Article in English | MEDLINE | ID: mdl-32420386

ABSTRACT

Liver cancer is a devastating disease for humans with poor prognosis. Although the survival rate of patients with liver cancer has improved in the past decades, the recurrence and metastasis of liver cancer are still obstacles for us. Inositol polyphosphate-5-phosphatase K (INPP5K) belongs to the family of phosphoinositide 5-phosphatases (PI 5-phosphatases), which have been reported to be associated with cell migration, polarity, adhesion, and cell invasion, especially in cancers. However, there have been few studies on the correlation of INPP5K and liver cancer. In this study, we explored the prognostic significance of INPP5K in liver cancer through bioinformatics analysis of data collected from The Cancer Genome Atlas (TCGA) database. Chi-square and Fisher exact tests were used to evaluate the relationship between INPP5K expression and clinical characteristics. Our results showed that low INPP5K expression was correlated with poor outcomes in liver cancer patients. Univariate and multivariate Cox analyses demonstrated that low INPP5K mRNA expression played a significant role in shortening overall survival (OS) and relapse-free survival (RFS), which might serve as the useful biomarker and prognostic factor for liver cancer. In conclusion, low INPP5K mRNA expression is an independent risk factor for poor prognosis in liver cancer.


Subject(s)
Databases, Nucleic Acid , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Inositol Polyphosphate 5-Phosphatases , Liver Neoplasms , Neoplasm Proteins , Aged , Disease-Free Survival , Female , Humans , Inositol Polyphosphate 5-Phosphatases/biosynthesis , Inositol Polyphosphate 5-Phosphatases/genetics , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Predictive Value of Tests , Survival Rate
12.
Biochem Biophys Res Commun ; 527(1): 207-212, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446368

ABSTRACT

SH2 domain-containing inositol 5'-phosphatase (SHIP) has critical functions in regulating signal transduction. In additional to its lipid phosphatase activity, SHIP engages in multiple protein-protein interactions, which can serve to localize either SHIP or its binding partners to a particular subcellular domain. Knock-out and knock-down studies have elucidated that SHIP negatively regulates the accumulation of F-actin in leukocytes, usually resulting in inhibition of actin dependent cellular activities such as spreading and migration. Here, we demonstrate that overexpression of SHIP inhibits B cell antigen receptor (BCR)-mediated cell spreading in murine and human B cell lines. B cell stimulation via the BCR or pervanadate induces an interaction between SHIP and Nck, an adaptor protein known to promote actin polymerization. Using a fluorescence recovery after photobleaching (FRAP) assay, we demonstrate that overexpression of SHIP slows F-actin dynamics in BCR-stimulated B cells and this can be overcome by co-overexpression of Nck. Our data supports a role for SHIP in limiting actin turnover and suggests it may do so in part by sequestering Nck.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , B-Lymphocytes/metabolism , Inositol Polyphosphate 5-Phosphatases/metabolism , Oncogene Proteins/metabolism , Animals , Humans , Inositol Polyphosphate 5-Phosphatases/genetics , Mice , Receptors, Antigen, B-Cell/metabolism , Tumor Cells, Cultured , src Homology Domains
13.
Proc Natl Acad Sci U S A ; 117(22): 12428-12434, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32424101

ABSTRACT

Numerous genes whose mutations cause, or increase the risk of, Parkinson's disease (PD) have been identified. An inactivating mutation (R258Q) in the Sac inositol phosphatase domain of synaptojanin 1 (SJ1/PARK20), a phosphoinositide phosphatase implicated in synaptic vesicle recycling, results in PD. The gene encoding Sac2/INPP5F, another Sac domain-containing protein, is located within a PD risk locus identified by genome-wide association studies. Knock-In mice carrying the SJ1 patient mutation (SJ1RQKI) exhibit PD features, while Sac2 knockout mice (Sac2KO) do not have obvious neurologic defects. We report a "synthetic" effect of the SJ1 mutation and the KO of Sac2 in mice. Most mice with both mutations died perinatally. The occasional survivors had stunted growth, died within 3 wk, and showed abnormalities of striatal dopaminergic nerve terminals at an earlier stage than SJ1RQKI mice. The abnormal accumulation of endocytic factors observed at synapses of cultured SJ1RQKI neurons was more severe in double-mutant neurons. Our results suggest that SJ1 and Sac2 have partially overlapping functions and are consistent with a potential role of Sac2 as a PD risk gene.


Subject(s)
Inositol Polyphosphate 5-Phosphatases/genetics , Parkinson Disease/enzymology , Animals , Dopamine/metabolism , Genome-Wide Association Study , Humans , Inositol Polyphosphate 5-Phosphatases/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nerve Endings/metabolism , Parkinson Disease/genetics , Phenotype , Synapses/metabolism
14.
Nat Commun ; 11(1): 1101, 2020 02 27.
Article in English | MEDLINE | ID: mdl-32107387

ABSTRACT

Spinocerebellar ataxias 17 (SCA17) is caused by polyglutamine (polyQ) expansion in the TATA box-binding protein (TBP). The selective neurodegeneration in the cerebellum in SCA17 raises the question of why ubiquitously expressed polyQ proteins can cause neurodegeneration in distinct brain regions in different polyQ diseases. By expressing mutant TBP in different brain regions in adult wild-type mice via stereotaxic injection of adeno-associated virus, we found that adult cerebellar neurons are particularly vulnerable to mutant TBP. In SCA17 knock-in mice, mutant TBP inhibits SP1-mediated gene transcription to down-regulate INPP5A, a protein that is highly abundant in the cerebellum. CRISPR/Cas9-mediated deletion of Inpp5a in the cerebellum of wild-type mice leads to Purkinje cell degeneration, and Inpp5a overexpression decreases inositol 1,4,5-trisphosphate (IP3) levels and ameliorates Purkinje cell degeneration in SCA17 knock-in mice. Our findings demonstrate the important contribution of a tissue-specific protein to the polyQ protein-mediated selective neuropathology.


Subject(s)
Inositol Polyphosphate 5-Phosphatases/genetics , Purkinje Cells/pathology , Spinocerebellar Ataxias/pathology , TATA-Box Binding Protein/genetics , Animals , Disease Models, Animal , Down-Regulation , Gene Knock-In Techniques , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol Polyphosphate 5-Phosphatases/metabolism , Mice , Mice, Transgenic , Peptides/genetics , Peptides/metabolism , Purkinje Cells/metabolism , Sp1 Transcription Factor/metabolism , Spinocerebellar Ataxias/genetics , TATA-Box Binding Protein/metabolism , Trinucleotide Repeat Expansion
15.
Int J Mol Sci ; 21(3)2020 Feb 04.
Article in English | MEDLINE | ID: mdl-32033113

ABSTRACT

Inositol polyphosphate 5-phosphatases (5PTases) function in inositol signaling by regulating the catabolism of phosphoinositol derivatives. Previous reports showed that 5PTases play a critical role in plant development and stress responses. In this study, we identified a novel 5PTase gene, Gs5PTase8, from the salt-tolerance locus of chromosome 3 in wild soybean (Glycine soja). Gs5PTase8 is highly up-regulated under salt treatment. It is localized in the nucleus and plasma membrane with a strong signal in the apoplast. Ectopic expression of Gs5PTase8 significantly increased salt tolerance in transgenic BY-2 cells, soybean hairy roots and Arabidopsis, suggesting Gs5PTase8 could increase salt tolerance in plants. The overexpression of Gs5PTase8 significantly enhanced the activities of catalase and ascorbate peroxidase under salt stress. The seeds of Gs5PTase8-transgenic Arabidopsis germinated earlier than the wild type under abscisic acid treatment, indicating Gs5PTase8 would alter ABA sensitivity. Besides, transcriptional analyses showed that the stress-responsive genes, AtRD22, AtRD29A and AtRD29B, were induced with a higher level in the Gs5PTase8-transgenic Arabidopsis plants than in the wild type under salt stress. These results reveal that Gs5PTase8 play a positive role in salt tolerance and might be a candidate gene for improving soybean adaptation to salt stress.


Subject(s)
Ectopic Gene Expression/genetics , Glycine max/genetics , Inositol Polyphosphate 5-Phosphatases/genetics , Plant Proteins/genetics , Salt Tolerance/genetics , Arabidopsis/genetics , Ascorbate Peroxidases/genetics , Catalase/genetics , Cell Membrane/genetics , Gene Expression Regulation, Plant/genetics , Germination/genetics , Plant Roots/genetics , Plants, Genetically Modified/genetics , Seeds/genetics , Stress, Physiological/genetics , Up-Regulation/genetics
16.
J Am Acad Dermatol ; 82(4): 846-853, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31437542

ABSTRACT

BACKGROUND: Inositol polyphosphate-5-phosphatase (INPP5A) has been shown to play a role in the progression of actinic keratosis to cutaneous squamous cell carcinoma (cSCC) and the progression of localized disease to metastatic disease. Currently, no cSCC biomarkers are able to risk stratify recurrent and metastatic disease. OBJECTIVE: To determine the prognostic value of INPP5A expression in cSCC recurrent and metastatic disease. METHODS: We conducted a multicenter, single-institutional, retrospective cohort study within the Mayo Clinic Health System on the use of immunohistochemical staining to examine cSCC INPP5A protein expression in primary tumors and recurrent and metastatic disease. Dermatologists and dermatopathologists were blinded to outcome. RESULTS: Low staining expression of INPP5A in recurrent and metastatic disease tumors was associated with poor overall survival (OS) (31.0 months for low versus 62.0 months for high expression; P = .0272). A composite risk score (calculated as score of primary tumor + score of recurrent or metastatic disease tumor, with tumors with high expression scoring a zero and low expression a 1, score range 0-2) of 0 was predictive of improved OS compared with a composite risk score of ≥1 (hazard ratio 0.42, 95% confidence interval 0.21-0.84; P = .0113). LIMITATIONS: This is a multicenter but single institution study of a white population. CONCLUSION: Loss of INPP5A expression predicts poor OS in recurrent and metastatic disease of cSCC.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Inositol Polyphosphate 5-Phosphatases/genetics , Neoplasm Recurrence, Local/enzymology , Skin Neoplasms/enzymology , Aged , Biomarkers/analysis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Disease Progression , Female , Gene Expression , Humans , Immunohistochemistry , Inositol Polyphosphate 5-Phosphatases/analysis , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prognosis , Retrospective Studies , Skin Neoplasms/genetics , Skin Neoplasms/pathology
17.
Genes (Basel) ; 10(5)2019 05 22.
Article in English | MEDLINE | ID: mdl-31121965

ABSTRACT

Inositol polyphosphate 5-phosphatase (5PTase), a key enzyme that hydrolyzes the 5` position of the inositol ring, has essential functions in growth, development, and stress responses in plants, yeasts, and animals. However, the evolutionary history and patterns of 5PTases have not been examined systematically. Here, we report a comprehensive molecular evolutionary analysis of the 5PTase gene family and define four groups. These four groups are different from former classifications, which were based on in vitro substrate specificity. Most orthologous groups appear to be conserved as single or low-copy genes in all lineages in Groups II-IV, whereas 5PTase genes in Group I underwent several duplication events in angiosperm, resulting in multiple gene copies. Whole-genome duplication (WGD) was the main mechanism for 5PTase duplications in angiosperm. Plant 5PTases have more members than that of animals, and most plant 5PTase genes appear to have evolved under strong purifying selection. The paralogs have diverged in substrate specificity and expression pattern, showing evidence of selection pressure. Meanwhile, the increase in 5PTases and divergences in sequence, expression, and substrate might have contributed to the divergent functions of 5PTase genes, allowing the angiosperms to successfully adapt to a great number of ecological niches.


Subject(s)
Evolution, Molecular , Inositol Polyphosphate 5-Phosphatases/genetics , Magnoliopsida/genetics , Amino Acid Sequence/genetics , Arabidopsis/genetics , Gene Duplication , Inositol Polyphosphate 5-Phosphatases/metabolism , Substrate Specificity
18.
Gene ; 641: 367-375, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-29111207

ABSTRACT

Identification of new polymorphic variants from RNA-seq data is difficult mainly because of the errors arising during bioinformatic analysis. Therefore, new experiments in this area are very profitable for improving new statistical methods. In our study of the porcine brain transcriptome, we have identified 10966 polymorphic variants, among which 7277 were single nucleotide polymorphisms (SNPs). Further, we have calculated allelic ratios for the SNPs identified and estimated that 52% of genes in porcine brain are subjected to allele-specific expression (ASE), a phenomenon in which one allele is preferentially expressed. Our investigation presents the first estimates of ASE in porcine brain. In addition, we have used the results of RNA-seq for the identification of SNPs in putatively imprinted genes. Finally, we have used these SNPs for the verification of the imprinted status of the INPP5f variant 2, LRRTM1 and HM13 genes in pigs by Sanger sequencing. We observed that INPP5f variant 2 is paternally expressed, while HM13 and LRRTM1 are biallelically expressed in porcine brain. We have also confirmed maternal expression of the MEG3 gene in pigs. Our results present how RNA-seq data may be used for imprinting studies without sequencing of parental genomes.


Subject(s)
Brain/physiology , Gene Expression/genetics , Genomic Imprinting/genetics , Polymorphism, Single Nucleotide/genetics , Alleles , Animals , Female , Inositol Polyphosphate 5-Phosphatases/genetics , Male , Membrane Proteins/genetics , Sequence Analysis, RNA/methods , Swine
19.
Int J Immunopathol Pharmacol ; 30(4): 406-412, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29164959

ABSTRACT

This study aimed to exploit the potential therapeutic value of palmatine in treatment of cardiac hypertrophy and the underlying molecular mechanism. Rat hypertrophy model was established by intraperitoneal isoproterenol (ISO) injection. The hypertrophy was evaluated with cardiac hypertrophic parameters, hemodynamic parameters, lipid profile, and non-specific cardiac markers. The animals were intraperitoneally administrated with either palmatine or vehicle. The relative expressions of ANP, BNP, HDAC2, HDAC5, KLF4, and INPP5F transcripts were determined by real-time polymerase chain reaction (PCR). The relative protein levels of HDAC2, HDAC5, KLF4, and INPP5F were analyzed by immunoblotting. Palmatine treatment significantly attenuated ISO-induced hypertrophy in rats and elicited remarkable repressions in ANP, BNP, and HDAC2 transcriptions but not HDAC5. The downstream effector genes KLF4 and INPP5F were greatly restored in a dose-dependent manner in response to palmatine treatment. Our data demonstrated that palmatine possessed promising therapeutic potential against hypertrophy, which was mediated by modulation of HDAC2-KLF4/INPP5F pathway.


Subject(s)
Berberine Alkaloids/pharmacology , Cardiomegaly/genetics , Cardiotonic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Animals , Atrial Natriuretic Factor/genetics , Berberine Alkaloids/therapeutic use , Cardiomegaly/chemically induced , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Cardiotonic Agents/therapeutic use , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/genetics , Histone Deacetylase Inhibitors/therapeutic use , Inositol Polyphosphate 5-Phosphatases/genetics , Isoproterenol , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Natriuretic Peptide, Brain/genetics , RNA, Messenger/metabolism , Rats, Wistar
20.
Nat Rev Nephrol ; 13(8): 455-470, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28669993

ABSTRACT

Lowe syndrome is an X-linked disease that is characterized by congenital cataracts, central hypotonia, intellectual disability and renal Fanconi syndrome. The disease is caused by mutations in OCRL, which encodes an inositol polyphosphate 5-phosphatase (OCRL) that acts on phosphoinositides - quantitatively minor constituents of cell membranes that are nonetheless pivotal regulators of intracellular trafficking. In this Review we summarize the considerable progress made over the past decade in understanding the cellular roles of OCRL in regulating phosphoinositide balance along the endolysosomal pathway, a fundamental system for the reabsorption of proteins and solutes by proximal tubular cells. We discuss how studies of OCRL have led to important discoveries about the basic mechanisms of membrane trafficking and describe the key features and limitations of the currently available animal models of Lowe syndrome. Mutations in OCRL can also give rise to a milder pathology, Dent disease 2, which is characterized by renal Fanconi syndrome in the absence of extrarenal pathologies. Understanding how mutations in OCRL give rise to two clinical entities with differing extrarenal manifestations represents an opportunity to identify molecular pathways that could be targeted to develop treatments for these conditions.


Subject(s)
Genetic Diseases, X-Linked/genetics , Mutation , Nephrolithiasis/genetics , Oculocerebrorenal Syndrome/genetics , Phosphoric Monoester Hydrolases/genetics , Animals , Clathrin-Coated Vesicles , Disease Models, Animal , Endocytosis , Humans , Inositol Polyphosphate 5-Phosphatases/genetics , Kidney Tubules, Proximal/cytology
SELECTION OF CITATIONS
SEARCH DETAIL