Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 15(1): 20, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38195526

ABSTRACT

In recent years, primary familial brain calcification (PFBC), a rare neurological disease characterized by a wide spectrum of cognitive disorders, has been associated to mutations in the sodium (Na)-Phosphate (Pi) co-transporter SLC20A2. However, the functional roles of the Na-Pi co-transporters in the brain remain still largely elusive. Here we show that Slc20a1 (PiT-1) and Slc20a2 (PiT-2) are the most abundant Na-Pi co-transporters expressed in the brain and are involved in the control of hippocampal-dependent learning and memory. We reveal that Slc20a1 and Slc20a2 are differentially distributed in the hippocampus and associated with independent gene clusters, suggesting that they influence cognition by different mechanisms. Accordingly, using a combination of molecular, electrophysiological and behavioral analyses, we show that while PiT-2 favors hippocampal neuronal branching and survival, PiT-1 promotes synaptic plasticity. The latter relies on a likely Otoferlin-dependent regulation of synaptic vesicle trafficking, which impacts the GABAergic system. These results provide the first demonstration that Na-Pi co-transporters play key albeit distinct roles in the hippocampus pertaining to the control of neuronal plasticity and cognition. These findings could provide the foundation for the development of novel effective therapies for PFBC and cognitive disorders.


Subject(s)
Cognition , Symporters , Ion Transport , Neuronal Plasticity/genetics , Phosphates
2.
J Exp Med ; 219(4)2022 04 04.
Article in English | MEDLINE | ID: mdl-35254402

ABSTRACT

Crouzon syndrome with acanthosis nigricans (CAN, a rare type of craniosynostosis characterized by premature suture fusion and neurological impairments) has been linked to a gain-of-function mutation (p.Ala391Glu) in fibroblast growth factor receptor 3 (FGFR3). To characterize the CAN mutation's impact on the skull and on brain functions, we developed the first mouse model (Fgfr3A385E/+) of this syndrome. Surprisingly, Fgfr3A385E/+ mice did not exhibit craniosynostosis but did show severe memory impairments, a structurally abnormal hippocampus, low activity-dependent synaptic plasticity, and overactivation of MAPK/ERK and Akt signaling pathways in the hippocampus. Systemic or brain-specific pharmacological inhibition of FGFR3 overactivation by BGJ398 injections rescued the memory impairments observed in Fgfr3A385E/+ mice. The present study is the first to have demonstrated cognitive impairments associated with brain FGFR3 overactivation, independently of skull abnormalities. Our results provide a better understanding of FGFR3's functional role and the impact of its gain-of-function mutation on brain functions. The modulation of FGFR3 signaling might be of value for treating the neurological disorders associated with craniosynostosis.


Subject(s)
Acanthosis Nigricans , Craniofacial Dysostosis , Craniosynostoses , Acanthosis Nigricans/complications , Acanthosis Nigricans/genetics , Animals , Brain , Craniofacial Dysostosis/complications , Craniofacial Dysostosis/genetics , Craniosynostoses/genetics , Disease Models, Animal , Memory Disorders/genetics , Mice , Receptor, Fibroblast Growth Factor, Type 3/genetics
3.
Sci Rep ; 9(1): 1808, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30755642

ABSTRACT

PiT1/SLC20A1 is an inorganic phosphate transporter with additional functions including the regulation of TNFα-induced apoptosis, erythropoiesis, cell proliferation and insulin signaling. Recent data suggest a relationship between PiT1 and NF-κB-dependent inflammation: (i) Pit1 mRNA is up-regulated in the context of NF-κB pathway activation; (ii) NF-κB target gene transcription is decreased in PiT1-deficient conditions. This led us to investigate the role of PiT1 in lipopolysaccharide (LPS)-induced inflammation. MCP-1 and IL-6 concentrations were impaired in PiT1-deficient bone marrow derived macrophages (BMDMs) upon LPS stimulation. Lower MCP-1 and IL-6 serum levels were observed in Mx1-Cre; Pit1lox/lox mice dosed intraperitoneally with LPS. Lower PiT1 expression correlated with decreased in vitro wound healing and lower reactive oxygen species levels. Reduced IκB degradation and lower p65 nuclear translocation were observed in PiT1-deficient cells stimulated with LPS. Conversely, PiT1 expression was induced in vitro upon LPS stimulation. Addition of an NF-κB inhibitor abolished LPS-induced PiT1 expression. Furthermore, we showed that p65 expression activated Pit1 promoter activity. Finally, ChIP assays demonstrated that p65 directly binds to the mPit1 promoter in response to LPS. These data demonstrate a completely novel function of PiT1 in the response to LPS and provide mechanistic insights into the regulation of PiT1 expression by NF-κB.


Subject(s)
Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Transcription Factor Pit-1/metabolism , Animals , Apoptosis/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , NADPH Oxidase 2/metabolism , NF-kappa B/metabolism , Peritonitis/chemically induced , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Thioglycolates/toxicity , Transcription Factor Pit-1/genetics , Tumor Necrosis Factor-alpha/metabolism , Wound Healing/drug effects
5.
Blood ; 128(26): 3125-3136, 2016 12 29.
Article in English | MEDLINE | ID: mdl-27702798

ABSTRACT

Metabolic changes drive monocyte differentiation and fate. Although abnormal mitochondria metabolism and innate immune responses participate in the pathogenesis of many inflammatory disorders, molecular events regulating mitochondrial activity to control life and death in monocytes remain poorly understood. We show here that, in human monocytes, microRNA-125b (miR-125b) attenuates the mitochondrial respiration through the silencing of the BH3-only proapoptotic protein BIK and promotes the elongation of the mitochondrial network through the targeting of the mitochondrial fission process 1 protein MTP18, leading to apoptosis. Proinflammatory activation of monocyte-derived macrophages is associated with a concomitant increase in miR-125b expression and decrease in BIK and MTP18 expression, which lead to reduced oxidative phosphorylation and enhanced mitochondrial fusion. In a chronic inflammatory systemic disorder, CD14+ blood monocytes display reduced miR-125b expression as compared with healthy controls, inversely correlated with BIK and MTP18 messenger RNA expression. Our findings not only identify BIK and MTP18 as novel targets for miR-125b that control mitochondrial metabolism and dynamics, respectively, but also reveal a novel function for miR-125b in regulating metabolic adaptation of monocytes to inflammation. Together, these data unravel new molecular mechanisms for a proapoptotic role of miR-125b in monocytes and identify potential targets for interfering with excessive inflammatory activation of monocytes in inflammatory disorders.


Subject(s)
Inflammation/genetics , Inflammation/pathology , MicroRNAs/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Monocytes/metabolism , Monocytes/pathology , Aged , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Polarity/genetics , Cell Respiration/genetics , Female , Gene Expression Regulation , Gene Silencing , HEK293 Cells , Humans , Lipopolysaccharide Receptors/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Biological , Toll-Like Receptor 4/metabolism
6.
Cell Rep ; 16(10): 2736-2748, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27568561

ABSTRACT

The liver plays a central role in whole-body lipid and glucose homeostasis. Increasing dietary fat intake results in increased hepatic fat deposition, which is associated with a risk for development of insulin resistance and type 2 diabetes. In this study, we demonstrate a role for the phosphate inorganic transporter 1 (PiT1/SLC20A1) in regulating metabolism. Specific knockout of Pit1 in hepatocytes significantly improved glucose tolerance and insulin sensitivity, enhanced insulin signaling, and decreased hepatic lipogenesis. We identified USP7 as a PiT1 binding partner and demonstrated that Pit1 deletion inhibited USP7/IRS1 dissociation upon insulin stimulation. This prevented IRS1 ubiquitination and its subsequent proteasomal degradation. As a consequence, delayed insulin negative feedback loop and sustained insulin signaling were observed. Moreover, PiT1-deficient mice were protected against high-fat-diet-induced obesity and diabetes. Our findings indicate that PiT1 has potential as a therapeutic target in the context of metabolic syndrome, obesity, and diabetes.


Subject(s)
Glucose/metabolism , Hepatocytes/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin/metabolism , Signal Transduction , Transcription Factor Pit-1/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , Adipose Tissue/pathology , Aging/pathology , Animals , Diet, High-Fat , Fatty Liver/complications , Fatty Liver/pathology , Fibroblasts/metabolism , Gluconeogenesis , Glucose Tolerance Test , Inflammation/complications , Inflammation/pathology , Insulin Resistance , Mice, Knockout , Obesity/pathology , Organ Specificity , Phenotype , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Ubiquitination , Weight Gain
7.
Nat Cell Biol ; 18(6): 657-67, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27214279

ABSTRACT

Autophagy is an adaptation mechanism that is vital for cellular homeostasis in response to various stress conditions. Previous reports indicate that there is a functional interaction between the primary cilium (PC) and autophagy. The PC, a microtubule-based structure present at the surface of numerous cell types, is a mechanical sensor. Here we show that autophagy induced by fluid flow regulates kidney epithelial cell volume in vitro and in vivo. PC ablation blocked autophagy induction and cell-volume regulation. In addition, inhibition of autophagy in ciliated cells impaired the flow-dependent regulation of cell volume. PC-dependent autophagy can be triggered either by mTOR inhibition or a mechanism dependent on the polycystin 2 channel. Only the LKB1-AMPK-mTOR signalling pathway was required for the flow-dependent regulation of cell volume by autophagy. These findings suggest that therapies regulating autophagy should be considered in developing treatments for PC-related diseases.


Subject(s)
Autophagy , Cell Physiological Phenomena , Cilia/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/physiology , AMP-Activated Protein Kinases/physiology , Animals , Cell Size , Dogs , Immunoblotting , Madin Darby Canine Kidney Cells , Mice , Mice, Knockout , Microscopy, Fluorescence , Protein Serine-Threonine Kinases/physiology , Signal Transduction , TOR Serine-Threonine Kinases/physiology
8.
Proc Natl Acad Sci U S A ; 113(7): 1901-6, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26831115

ABSTRACT

Autophagy is an essential component of innate immunity, enabling the detection and elimination of intracellular pathogens. Legionella pneumophila, an intracellular pathogen that can cause a severe pneumonia in humans, is able to modulate autophagy through the action of effector proteins that are translocated into the host cell by the pathogen's Dot/Icm type IV secretion system. Many of these effectors share structural and sequence similarity with eukaryotic proteins. Indeed, phylogenetic analyses have indicated their acquisition by horizontal gene transfer from a eukaryotic host. Here we report that L. pneumophila translocates the effector protein sphingosine-1 phosphate lyase (LpSpl) to target the host sphingosine biosynthesis and to curtail autophagy. Our structural characterization of LpSpl and its comparison with human SPL reveals high structural conservation, thus supporting prior phylogenetic analysis. We show that LpSpl possesses S1P lyase activity that was abrogated by mutation of the catalytic site residues. L. pneumophila triggers the reduction of several sphingolipids critical for macrophage function in an LpSpl-dependent and -independent manner. LpSpl activity alone was sufficient to prevent an increase in sphingosine levels in infected host cells and to inhibit autophagy during macrophage infection. LpSpl was required for efficient infection of A/J mice, highlighting an important virulence role for this effector. Thus, we have uncovered a previously unidentified mechanism used by intracellular pathogens to inhibit autophagy, namely the disruption of host sphingolipid biosynthesis.


Subject(s)
Aldehyde-Lyases/metabolism , Autophagy , Legionella pneumophila/enzymology , Sphingolipids/metabolism , Aldehyde-Lyases/chemistry , Animals , Catalytic Domain , Crystallography, X-Ray , Legionnaires' Disease/immunology , Mice , Protein Conformation
9.
Blood ; 121(4): 666-78, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23190530

ABSTRACT

The PIT1/SLC20A1 protein, a well-described sodium/phosphate cotransporter and retrovirus receptor, has been identified recently as a modular of proliferation and apoptosis in vitro. The targeted deletion of the PIT1 gene in mice revealed a lethal phenotype due to severe anemia attributed to defects in liver development. However, the presence of immature erythroid cells associated with impaired maturation of the globin switch led us to investigate the role of PIT1 in hematopoietic development. In the present study, specific deletion of PIT1 in the hematopoietic system and fetal liver transplantation experiments demonstrated that anemia was associated with an erythroid cell- autonomous defect. Moreover, anemia was not due to RBC destruction but rather to maturation defects. Because Erythroid Krüppel-like Factor (EKLF)-knockout mice showed similar maturation defects, we investigated the functional link between PIT1 and EKLF. We demonstrated that EKLF increases PIT1 expression during RBC maturation by binding to its promoter in vivo and that shRNA-driven depletion of either PIT1 or EKLF impairs erythroid maturation of G1E cells in vitro, whereas reexpression of PIT1 in EKLF-depleted G1E cells partially restores erythroid maturation. This is the first demonstration of a physiologic involvement of PIT1 in erythroid maturation in vivo.


Subject(s)
Erythroid Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Transcription Factor Pit-1/genetics , Animals , Base Sequence , Cell Differentiation , Colony-Forming Units Assay , Erythroid Cells/cytology , Erythropoiesis/genetics , Gene Deletion , Gene Expression , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Liver/embryology , Liver/metabolism , Mice , Molecular Sequence Data , Phenotype , Promoter Regions, Genetic , Sequence Alignment , Transcriptional Activation
10.
J Biol Chem ; 285(45): 34408-18, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20817733

ABSTRACT

PiT1/SLC20A1 is a sodium-dependent P(i) transporter expressed by most mammalian cells. Interestingly, PiT1 transcription has been shown to be up-regulated by the tumor necrosis factor α (TNF), and we have now investigated the possible involvement of PiT1 in TNF-induced apoptosis. We show that PiT1-depleted cells are more sensitive to the proapoptotic activity of TNF (i.e. when the antiapoptotic NFκB pathway is inactivated). These observations were made in the human HeLa cancer cell line either transiently or stably depleted in PiT1 by RNA interference and in immortalized mouse embryonic fibroblasts isolated from PiT1 knock-out embryos. Depletion of the closely related family member PiT2 had no effect on TNF-induced apoptosis, showing that this effect was specific to PiT1. The increased sensitivity of PiT1-depleted cells was evident regardless of the presence or absence of extracellular P(i), suggesting that a defect in P(i) uptake was not involved in the observed phenotype. Importantly, we show that the re-expression of a P(i) uptake mutant of PiT1 in PiT1(-/-) mouse embryonic fibroblasts delays apoptosis as efficiently as the WT protein, showing that this function of PiT1 is unrelated to its transport activity. Caspase-8 is more activated in PiT1-depleted cells, and our data reveal that the sustained activation of the MAPK JNK is up-regulated in response to TNF. JNK activity is actually involved in PiT1-depleted cell death because specific JNK inhibitors delay apoptosis.


Subject(s)
Apoptosis/drug effects , Fibroblasts/metabolism , Sodium-Phosphate Cotransporter Proteins, Type III/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Animals , Apoptosis/physiology , Biological Transport/drug effects , Biological Transport/physiology , Caspase 8/genetics , Caspase 8/metabolism , Cell Line, Transformed , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fibroblasts/cytology , HeLa Cells , Humans , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , RNA Interference , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...