Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
JCI Insight ; 9(9)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38716728

ABSTRACT

The importance of the proper localization of most receptors at the cell surface is often underestimated, although this feature is essential for optimal receptor response. Endospanin 1 (Endo1) (also known as OBRGRP or LEPROT) is a protein generated from the same gene as the human leptin receptor and regulates the trafficking of proteins to the surface, including the leptin receptor. The systemic role of Endo1 on whole-body metabolism has not been studied so far. Here, we report that general Endo1-KO mice fed a high-fat diet develop metabolically healthy obesity with lipid repartitioning in organs and preferential accumulation of fat in adipose tissue, limited systematic inflammation, and better controlled glucose homeostasis. Mechanistically, Endo1 interacts with the lipid translocase CD36, thus regulating its surface abundance and lipid uptake in adipocytes. In humans, the level of Endo1 transcripts is increased in the adipose tissue of patients with obesity, but low levels rather correlate with a profile of metabolically healthy obesity. We suggest here that Endo1, most likely by controlling CD36 cell surface abundance and lipid uptake in adipocytes, dissociates obesity from diabetes and that its absence participates in metabolically healthy obesity.


Subject(s)
Adipose Tissue , CD36 Antigens , Diet, High-Fat , Mice, Knockout , Obesity , Animals , Female , Humans , Male , Mice , Adipocytes/metabolism , Adipose Tissue/metabolism , CD36 Antigens/metabolism , CD36 Antigens/genetics , Diet, High-Fat/adverse effects , Glucose/metabolism , Lipid Metabolism/genetics , Mice, Inbred C57BL , Obesity/metabolism , Obesity/genetics
2.
Nat Commun ; 15(1): 1718, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38409097

ABSTRACT

Foxo family transcription factors are critically involved in multiple processes, such as metabolism, quiescence, cell survival and cell differentiation. Although continuous, high activity of Foxo transcription factors extends the life span of some species, the involvement of Foxo proteins in mammalian aging remains to be determined. Here, we show that Foxo1 is down-regulated with age in mouse T cells. This down-regulation of Foxo1 in T cells may contribute to the disruption of naive T-cell homeostasis with age, leading to an increase in the number of memory T cells. Foxo1 down-regulation is also associated with the up-regulation of co-inhibitory receptors by memory T cells and exhaustion in aged mice. Using adoptive transfer experiments, we show that the age-dependent down-regulation of Foxo1 in T cells is mediated by T-cell-extrinsic cues, including type 1 interferons. Taken together, our data suggest that type 1 interferon-induced Foxo1 down-regulation is likely to contribute significantly to T-cell dysfunction in aged mice.


Subject(s)
Forkhead Transcription Factors , T-Cell Exhaustion , Mice , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Down-Regulation , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Cell Differentiation , Proteins/metabolism , Interferons/metabolism , Mammals/metabolism
3.
Circulation ; 147(8): 650-666, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36515093

ABSTRACT

BACKGROUND: Senescent cells (SCs) are involved in proliferative disorders, but their role in pulmonary hypertension remains undefined. We investigated SCs in patients with pulmonary arterial hypertension and the role of SCs in animal pulmonary hypertension models. METHODS: We investigated senescence (p16, p21) and DNA damage (γ-H2AX, 53BP1) markers in patients with pulmonary arterial hypertension and murine models. We monitored p16 activation by luminescence imaging in p16-luciferase (p16LUC/+) knock-in mice. SC clearance was obtained by a suicide gene (p16 promoter-driven killer gene construct in p16-ATTAC mice), senolytic drugs (ABT263 and cell-permeable FOXO4-p53 interfering peptide [FOXO4-DRI]), and p16 inactivation in p16LUC/LUC mice. We investigated pulmonary hypertension in mice exposed to normoxia, chronic hypoxia, or hypoxia+Sugen, mice overexpressing the serotonin transporter (SM22-5-HTT+), and rats given monocrotaline. RESULTS: Patients with pulmonary arterial hypertension compared with controls exhibited high lung p16, p21, and γ-H2AX protein levels, with abundant vascular cells costained for p16, γ-H2AX, and 53BP1. Hypoxia increased thoracic bioluminescence in p16LUC/+ mice. In wild-type mice, hypoxia increased lung levels of senescence and DNA-damage markers, senescence-associated secretory phenotype components, and p16 staining of pulmonary endothelial cells (P-ECs, 30% of lung SCs in normoxia), and pulmonary artery smooth muscle cells. SC elimination by suicide gene or ABT263 increased the right ventricular systolic pressure and hypertrophy index, increased vessel remodeling (higher dividing proliferating cell nuclear antigen-stained vascular cell counts during both normoxia and hypoxia), and markedly decreased lung P-ECs. Pulmonary hemodynamic alterations and lung P-EC loss occurred in older p16LUC/LUC mice, wild-type mice exposed to Sugen or hypoxia+Sugen, and SM22-5-HTT+ mice given either ABT263 or FOXO4-DRI, compared with relevant controls. The severity of monocrotaline-induced pulmonary hypertension in rats was decreased slightly by ABT263 for 1 week but was aggravated at 3 weeks, with loss of P-ECs. CONCLUSIONS: Elimination of senescent P-ECs by senolytic interventions may worsen pulmonary hemodynamics. These results invite consideration of the potential impact on pulmonary vessels of strategies aimed at controlling cell senescence in various contexts.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Mice , Rats , Animals , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Pulmonary Arterial Hypertension/metabolism , Endothelial Cells/metabolism , Monocrotaline/metabolism , Senotherapeutics , Pulmonary Artery , Familial Primary Pulmonary Hypertension/metabolism , Hypoxia/metabolism , Cellular Senescence , Forkhead Transcription Factors/metabolism
4.
F S Sci ; 3(1): 49-63, 2022 02.
Article in English | MEDLINE | ID: mdl-35559995

ABSTRACT

OBJECTIVE: To study the cyclic fertilin peptide effects on preimplantation human embryogenesis. Cyclic fertilin peptide reproduces the structure of the binding site of the sperm Fertilin ß (also named A Disintegrin and Metalloprotease 2: ADAM2) disintegrin domain. It binds to the oocyte membrane and increases sperm-oocyte fusion index in human and fertilization rate in mouse, providing healthy pups. It also improves human oocyte maturation and chromosome segregation in meiosis I and binds to human embryo blastomeres, suggesting that it has a membrane receptor. DESIGN: Thawed human embryos at the 3 to 4 cells stage were randomly included in a dose-response study with cyclic fertilin peptide. Inner cell mass (ICM), trophectoderm (TE), and total cell numbers were evaluated in top- and good-quality blastocysts. SETTING: The study was performed in an academic hospital and research laboratory. PATIENT(S): Human embryos donated for research. This project was approved by the French "Agence de la Biomédecine." INTERVENTION(S): Immunofluorescence and tissue-specific gene expression analysis, using Clariom D microarrays, were performed to study its mechanism of action. MAIN OUTCOME MEASURE(S): Cyclic fertilin peptide improves blastocyst formation by almost 20%, the concentration of 1 µM being the lowest most efficient concentration. It significantly increases twice the TE cell number, without modifying the ICM. It increases the in vitro hatching rate from 14% to 45%. RESULT(S): Cyclic fertilin peptide stimulates TE growth. In the ICM, it induces transcriptional activation of intracellular protein and vesicle-mediated transport. CONCLUSION(S): Cyclic fertilin peptide dramatically improves human embryo development potential. It could be used to supplement culture medium and improve the in vitro human embryo development. Starting supplementation immediately after fertilization, instead of day 2, could significantly upgrade assisted reproductive technology outcome.


Subject(s)
Disintegrins , Peptides, Cyclic , ADAM Proteins , Embryonic Development , Fertilins , Humans , Membrane Glycoproteins/chemistry , Peptides, Cyclic/pharmacology
5.
Arthritis Rheumatol ; 74(8): 1387-1398, 2022 08.
Article in English | MEDLINE | ID: mdl-35255201

ABSTRACT

OBJECTIVE: Systemic sclerosis (SSc) is a debilitating autoimmune disease characterized by severe lung outcomes resulting in reduced life expectancy. Fra-2-transgenic mice offer the opportunity to decipher the relationships between the immune system and lung fibrosis. This study was undertaken to investigate whether the Fra-2-transgenic mouse lung phenotype may result from an imbalance between the effector and regulatory arms in the CD4+ T cell compartment. METHODS: We first used multicolor flow cytometry to extensively characterize homeostasis and the phenotype of peripheral CD4+ T cells from Fra-2-transgenic mice and control mice. We then tested different treatments for their effectiveness in restoring CD4+ Treg cell homeostasis, including adoptive transfer of Treg cells and treatment with low-dose interleukin-2 (IL-2). RESULTS: Fra-2-transgenic mice demonstrated a marked decrease in the proportion and absolute number of peripheral Treg cells that preceded accumulation of activated, T helper cell type 2-polarized, CD4+ T cells. This defect in Treg cell homeostasis was derived from a combination of mechanisms including impaired generation of these cells in both the thymus and the periphery. The impaired ability of peripheral conventional CD4+ T cells to produce IL-2 may greatly contribute to Treg cell deficiency in Fra-2-transgenic mice. Notably, adoptive transfer of Treg cells, low-dose IL-2 therapy, or combination therapy changed the phenotype of Fra-2-transgenic mice, resulting in a significant reduction in pulmonary parenchymal fibrosis and vascular remodeling in the lungs. CONCLUSION: Immunotherapies for restoring Treg cell homeostasis could be relevant in SSc. An intervention based on low-dose IL-2 injections, as is already proposed in other autoimmune diseases, could be the most suitable treatment modality for restoring Treg cell homeostasis for future research.


Subject(s)
Pulmonary Fibrosis , Scleroderma, Systemic , Animals , CD4-Positive T-Lymphocytes , Disease Models, Animal , Interleukin-2 , Mice , Mice, Transgenic , Pulmonary Fibrosis/metabolism , T-Lymphocytes, Regulatory , Vascular Remodeling
6.
Nat Commun ; 13(1): 1039, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35210422

ABSTRACT

The contractile properties of adult myofibers are shaped by their Myosin heavy chain isoform content. Here, we identify by snATAC-seq a 42 kb super-enhancer at the locus regrouping the fast Myosin genes. By 4C-seq we show that active fast Myosin promoters interact with this super-enhancer by DNA looping, leading to the activation of a single promoter per nucleus. A rainbow mouse transgenic model of the locus including the super-enhancer recapitulates the endogenous spatio-temporal expression of adult fast Myosin genes. In situ deletion of the super-enhancer by CRISPR/Cas9 editing demonstrates its major role in the control of associated fast Myosin genes, and deletion of two fast Myosin genes at the locus reveals an active competition of the promoters for the shared super-enhancer. Last, by disrupting the organization of fast Myosin, we uncover positional heterogeneity within limb skeletal muscles that may underlie selective muscle susceptibility to damage in certain myopathies.


Subject(s)
Muscle Fibers, Skeletal , Myosins , Animals , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosins/genetics , Myosins/metabolism , Phenotype
7.
J Clin Invest ; 130(12): 6379-6394, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32790646

ABSTRACT

Gain-of-function mutations in with no lysine (K) 1 (WNK1) and WNK4 genes are responsible for familial hyperkalemic hypertension (FHHt), a rare, inherited disorder characterized by arterial hypertension and hyperkalemia with metabolic acidosis. More recently, FHHt-causing mutations in the Kelch-like 3-Cullin 3 (KLHL3-CUL3) E3 ubiquitin ligase complex have shed light on the importance of WNK's cellular degradation on renal ion transport. Using full exome sequencing for a 4-generation family and then targeted sequencing in other suspected cases, we have identified new missense variants in the WNK1 gene clustering in the short conserved acidic motif known to interact with the KLHL3-CUL3 ubiquitin complex. Affected subjects had an early onset of a hyperkalemic hyperchloremic phenotype, but normal blood pressure values"Functional experiments in Xenopus laevis oocytes and HEK293T cells demonstrated that these mutations strongly decrease the ubiquitination of the kidney-specific isoform KS-WNK1 by the KLHL3-CUL3 complex rather than the long ubiquitous catalytically active L-WNK1 isoform. A corresponding CRISPR/Cas9 engineered mouse model recapitulated both the clinical and biological phenotypes. Renal investigations showed increased activation of the Ste20 proline alanine-rich kinase-Na+-Cl- cotransporter (SPAK-NCC) phosphorylation cascade, associated with impaired ROMK apical expression in the distal part of the renal tubule. Together, these new WNK1 genetic variants highlight the importance of the KS-WNK1 isoform abundance on potassium homeostasis.


Subject(s)
Acidosis/metabolism , Kidney Tubules, Distal/metabolism , Mutation , Pseudohypoaldosteronism/metabolism , WNK Lysine-Deficient Protein Kinase 1/metabolism , Acidosis/genetics , Acidosis/pathology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Animals , Cullin Proteins/genetics , Cullin Proteins/metabolism , HEK293 Cells , Humans , Kidney Tubules, Distal/pathology , Mice , Mice, Mutant Strains , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pseudohypoaldosteronism/genetics , Pseudohypoaldosteronism/pathology , WNK Lysine-Deficient Protein Kinase 1/genetics , Xenopus laevis
8.
Am J Physiol Lung Cell Mol Physiol ; 319(1): L71-L81, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32374670

ABSTRACT

SPARC/osteonectin, cwcv and kazal-like domains proteoglycan 2 (SPOCK2) was previously associated with genetic susceptibility to bronchopulmonary dysplasia in a French population of very preterm neonates. Its expression increases during lung development and is increased after exposure of rat pups to hyperoxia compared with controls bred in room air. To further investigate the role of SPOCK2 during lung development, we designed two mouse models, one that uses a specific anti-Spock2 antibody and one that reproduces the hyperoxia-induced Spock2 expression with a transgenic mouse model resulting in a conditional and lung-targeted overexpression of Spock2. When mice were bred under hyperoxic conditions, treatment with anti-Spock2 antibodies significantly improved alveolarization. Lung overexpression of Spock2 altered alveolar development in pups bred in room air and worsened hyperoxia-induced lesions. Neither treatment with anti-Spock2 antibody nor overexpression of Spock2 was associated with abnormal activation of matrix metalloproteinase-2. These two models did not alter the expression of known players in alveolar development. This study brings strong arguments for the deleterious role of SPOCK2 on lung alveolar development especially after lung injury, suggesting its role in bronchopulmonary dysplasia susceptibility. These effects are not mediated by a deregulation in metalloproteases activity and in expression of factors essential to normal alveolarization. The balance between types 1 and 2 epithelial alveolar cells may be involved.


Subject(s)
Hyperoxia/pathology , Proteoglycans/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Animals , Antibodies/metabolism , Enzyme Activation , Hyperoxia/metabolism , Matrix Metalloproteinase 2/metabolism , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
9.
Sci Rep ; 10(1): 5335, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210282

ABSTRACT

Three genes are known to be essential for gamete adhesion/fusion (Cd9, Izumo1 and Juno). Here, we confirmed that Spaca6 null males are infertile and showed that their sperm accumulate in the perivitelline space but are unable to fuse with oocyte. Like IZUMO1, SPACA6 which is expressed by human sperm, is remained on the equatorial segment after acrosomal reaction and is involved in human fertilization since an anti-SPACA6 antibody inhibited it. Despite the similarity of the phenotypes caused by Spaca6 and Izumo1 knockouts, these are not redundant and the essential relocation of IZUMO1 is not affected by the lack of SPACA6. We propose a model in which IZUMO1 and SPACA6 would be part of a molecular complex necessary for gamete fusion and that their concomitant presence would be required for the recruitment of another essential molecular actor, such as a fusogen, for the fusion to take place.


Subject(s)
Seminal Plasma Proteins/metabolism , Sperm-Ovum Interactions/physiology , Spermatozoa/physiology , Acrosome Reaction , Animals , COS Cells , Chlorocebus aethiops , Female , Fertilization in Vitro , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Infertility, Male/genetics , Infertility, Male/therapy , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Seminal Plasma Proteins/genetics , Sperm Head/physiology , Sperm Injections, Intracytoplasmic
10.
Am J Hum Genet ; 105(6): 1148-1167, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31735292

ABSTRACT

In humans, structural or functional defects of the sperm flagellum induce asthenozoospermia, which accounts for the main sperm defect encountered in infertile men. Herein we focused on morphological abnormalities of the sperm flagellum (MMAF), a phenotype also termed "short tails," which constitutes one of the most severe sperm morphological defects resulting in asthenozoospermia. In previous work based on whole-exome sequencing of a cohort of 167 MMAF-affected individuals, we identified bi-allelic loss-of-function mutations in more than 30% of the tested subjects. In this study, we further analyzed this cohort and identified five individuals with homozygous truncating variants in TTC29, a gene preferentially and highly expressed in the testis, and encoding a tetratricopeptide repeat-containing protein related to the intraflagellar transport (IFT). One individual carried a frameshift variant, another one carried a homozygous stop-gain variant, and three carried the same splicing variant affecting a consensus donor site. The deleterious effect of this last variant was confirmed on the corresponding transcript and protein product. In addition, we produced and analyzed TTC29 loss-of-function models in the flagellated protist T. brucei and in M. musculus. Both models confirmed the importance of TTC29 for flagellar beating. We showed that in T. brucei the TPR structural motifs, highly conserved between the studied orthologs, are critical for TTC29 axonemal localization and flagellar beating. Overall our work demonstrates that TTC29 is a conserved axonemal protein required for flagellar structure and beating and that TTC29 mutations are a cause of male sterility due to MMAF.


Subject(s)
Asthenozoospermia/etiology , Axoneme/pathology , Flagella/pathology , Infertility, Male/etiology , Microtubule-Associated Proteins/genetics , Mutation , Animals , Asthenozoospermia/metabolism , Asthenozoospermia/pathology , Axoneme/genetics , Axoneme/metabolism , Evolution, Molecular , Female , Fertilization in Vitro , Flagella/genetics , Flagella/metabolism , Humans , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Mice, Inbred C57BL , Trypanosoma brucei brucei/physiology , Trypanosomiasis
11.
Clin Cancer Res ; 25(14): 4504-4515, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31004003

ABSTRACT

PURPOSE: The genesis of all cancers results from an accumulation of mutations, constitutional and/or acquired when induced by external mutagenic factors. High-speed technologies for genome sequencing have completely changed the study of disease genetics, but with limited knowledge of the functional value of most genetic changes. EXPERIMENTAL DESIGN: Here, we proposed an innovative individual approach by studying tissue samples from a young woman with an unusual association of breast cancer, polycythemia vera, and rheumatoid arthritis. We performed genomic analyses for copy number variations and point mutations on laser-microdissected tumor cells from the breast cancer, and on CD34+ cells sorted from bone marrow aspiration, to identify gene abnormalities common to these two types of cell populations. RESULTS: Using ONCOSCAN technology, we identified a constitutional pR988C, c2962C>T mutation of MET. Using CRISPR-Cas9 technology, we established pR988C MET-mutated transgenic mice, which reproduced the autoimmune diseases and myeloproliferation found in our index-case; one of the transgenic mice spontaneously developed a skin squamous cell carcinoma. We also showed that additional mutagenic factors were required to induce cancers, including skin squamous cell carcinoma and thyroid cancer. Using an anti-MET drug, cabozantinib, we demonstrated for the first time the functional role of this mutation in the maintenance of myeloproliferation and rheumatoid arthritis, and in cancer genesis. CONCLUSIONS: Our study opens a considerable field of application in the domain of constitutional genetics, to establish genetic links between cancers and other very different severe diseases.


Subject(s)
Anilides/pharmacology , Arthritis, Rheumatoid/pathology , Autoimmune Diseases/pathology , Breast Neoplasms/pathology , Mutation , Myeloproliferative Disorders/pathology , Proto-Oncogene Proteins c-met/genetics , Pyridines/pharmacology , Adult , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Autoimmune Diseases/drug therapy , Autoimmune Diseases/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Chronic Disease , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Polycythemia Vera/drug therapy , Polycythemia Vera/genetics , Polycythemia Vera/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology
12.
Nat Commun ; 9(1): 1216, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572483

ABSTRACT

Transforming growth factor-ß (TGFß) signaling is initiated by the type I, II TGFß receptor (TßRI/TßRII) complex. Here we report the formation of an alternative complex between TßRI and the orphan GPR50, belonging to the G protein-coupled receptor super-family. The interaction of GPR50 with TßRI induces spontaneous TßRI-dependent Smad and non-Smad signaling by stabilizing the active TßRI conformation and competing for the binding of the negative regulator FKBP12 to TßRI. GPR50 overexpression in MDA-MB-231 cells mimics the anti-proliferative effect of TßRI and decreases tumor growth in a xenograft mouse model. Inversely, targeted deletion of GPR50 in the MMTV/Neu spontaneous mammary cancer model shows decreased survival after tumor onset and increased tumor growth. Low GPR50 expression is associated with poor survival prognosis in human breast cancer irrespective of the breast cancer subtype. This describes a previously unappreciated spontaneous TGFß-independent activation mode of TßRI and identifies GPR50 as a TßRI co-receptor with potential impact on cancer development.


Subject(s)
Mammary Neoplasms, Animal/prevention & control , Nerve Tissue Proteins/physiology , Receptor, Transforming Growth Factor-beta Type I/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Endosomes/metabolism , Female , Gene Expression Profiling , HEK293 Cells , HeLa Cells , Humans , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smad Proteins/metabolism , Tacrolimus Binding Protein 1A/metabolism
13.
Gut ; 67(12): 2192-2203, 2018 12.
Article in English | MEDLINE | ID: mdl-29074727

ABSTRACT

OBJECTIVE: The AAA+ ATPase Reptin is overexpressed in hepatocellular carcinoma and preclinical studies indicate that it could be a relevant therapeutic target. However, its physiological and pathophysiological roles in vivo remain unknown. This study aimed to determine the role of Reptin in mammalian adult liver. DESIGN AND RESULTS: We generated an inducible liver-specific Reptin knockout (RepinLKO ) mouse model. Following Reptin invalidation, mice displayed decreased body and fat mass, hypoglycaemia and hypolipidaemia. This was associated with decreased hepatic mTOR protein abundance. Further experiments in primary hepatocytes demonstrated that Reptin maintains mTOR protein level through its ATPase activity. Unexpectedly, loss or inhibition of Reptin induced an opposite effect on mTORC1 and mTORC2 signalling, with: (1) strong inhibition of hepatic mTORC1 activity, likely responsible for the reduction of hepatocytes cell size, for decreased de novo lipogenesis and cholesterol transcriptional programmes and (2) enhancement of mTORC2 activity associated with inhibition of the gluconeogenesis transcriptional programme and hepatic glucose production. Consequently, the role of hepatic Reptin in the pathogenesis of insulin resistance (IR) and non-alcoholic fatty liver disease consecutive to a high-fat diet was investigated. We found that Reptin deletion completely rescued pathological phenotypes associated with IR, including glucose intolerance, hyperglycaemia, hyperlipidaemia and hepatic steatosis. CONCLUSION: We show here that the AAA +ATPase Reptin is a regulator of mTOR signalling in the liver and global glucido-lipidic homeostasis. Inhibition of hepatic Reptin expression or activity represents a new therapeutic perspective for metabolic syndrome.


Subject(s)
ATPases Associated with Diverse Cellular Activities/physiology , DNA Helicases/physiology , Glucose/metabolism , Lipid Metabolism/physiology , Adenosine Triphosphatases/physiology , Animals , Body Weight/physiology , DNA Helicases/deficiency , DNA Helicases/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Energy Metabolism/physiology , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/prevention & control , Glucose Intolerance/physiopathology , Glucose Intolerance/prevention & control , Hepatocytes/metabolism , Insulin Resistance/physiology , Lipogenesis/physiology , Liver/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Knockout , Signal Transduction/physiology
14.
Cell Rep ; 21(2): 403-416, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29020627

ABSTRACT

While the physiological benefits of the fibroblast growth factor 21 (FGF21) hepatokine are documented in response to fasting, little information is available on Fgf21 regulation in a glucose-overload context. We report that peroxisome-proliferator-activated receptor α (PPARα), a nuclear receptor of the fasting response, is required with the carbohydrate-sensitive transcription factor carbohydrate-responsive element-binding protein (ChREBP) to balance FGF21 glucose response. Microarray analysis indicated that only a few hepatic genes respond to fasting and glucose similarly to Fgf21. Glucose-challenged Chrebp-/- mice exhibit a marked reduction in FGF21 production, a decrease that was rescued by re-expression of an active ChREBP isoform in the liver of Chrebp-/- mice. Unexpectedly, carbohydrate challenge of hepatic Pparα knockout mice also demonstrated a PPARα-dependent glucose response for Fgf21 that was associated with an increased sucrose preference. This blunted response was due to decreased Fgf21 promoter accessibility and diminished ChREBP binding onto Fgf21 carbohydrate-responsive element (ChoRE) in hepatocytes lacking PPARα. Our study reports that PPARα is required for the ChREBP-induced glucose response of FGF21.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucose/metabolism , Nuclear Proteins/metabolism , PPAR alpha/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cells, Cultured , Female , Fibroblast Growth Factors/genetics , Hepatocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , PPAR alpha/genetics , Response Elements , Transcription Factors/genetics
15.
J Clin Endocrinol Metab ; 101(11): 4449-4460, 2016 11.
Article in English | MEDLINE | ID: mdl-27575941

ABSTRACT

CONTEXT: In a cohort of 95 women with multiple breast fibroadenomas (MFAs), we recently identified patients harboring germline heterozygous variants of the prolactin receptor (PRLR) exhibiting constitutive activity (PRLRI146L and PRLRI176V). OBJECTIVE: This study sought to better delineate the potential role of PRLR gain-of-function variants in benign and malignant mammary tumorigenesis. DESIGN: This was an observational study and transgenic mouse model analysis. SETTING: The study took place at the Department of Endocrinology, Reproductive Disorders and Rare Gynecologic Diseases, Pitié Salpêtrière, Paris, and Inserm Unit 1151, Paris. PATIENTS OR OTHER PARTICIPANTS: We generated a second MFA cohort (n = 71) as well as a group of control subjects (n = 496) and a cohort of women with breast cancer (n = 119). We also generated two transgenic mouse models carrying the coding sequences of human PRLRI146L or PRLRWT. INTERVENTION: We aimed to determine the prevalence of PRLR variants in these three populations and to uncover any association of the latter with specific tumor pattern, especially in patients with breast cancer. RESULTS: This study did not highlight a higher prevalence of PRLR variants in the MFA group and in the breast cancer group compared with control subjects. Transgenic mice expressing PRLRI146L exhibited very mild histological mammary phenotype but tumors were never observed. CONCLUSION: PRLRI146L and PRLRI176V variants are not associated with breast cancer or MFA risk. However, one cannot exclude that low but sustained PRLR signaling may facilitate or contribute to pathological development driven by oncogenic pathways. Long-term patient follow-up should help to address this issue.


Subject(s)
Breast Neoplasms/genetics , Fibroadenoma/genetics , Receptors, Prolactin/genetics , Adolescent , Adult , Animals , Cohort Studies , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Young Adult
16.
Cancer Cell ; 20(1): 25-38, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21723201

ABSTRACT

Loss-of-function mutations affecting one or both copies of the Ten-Eleven-translocation (TET)2 gene have been described in various human myeloid malignancies. We report that inactivation of Tet2 in mouse perturbs both early and late steps of hematopoiesis including myeloid and lymphoid differentiation in a cell-autonomous manner, endows the cells with competitive advantage, and eventually leads to the development of malignancies. We subsequently observed TET2 mutations in human lymphoid disorders. TET2 mutations could be detected in immature progenitors endowed with myeloid colony-forming potential. Our results show that the mutations present in lymphoid tumor cells may occur at both early and later steps of lymphoid development and indicate that impairment of TET2 function or/and expression predisposes to the development of hematological malignancies.


Subject(s)
DNA-Binding Proteins/genetics , Gene Silencing , Hematopoiesis , Lymphoma/pathology , Precancerous Conditions/pathology , Proto-Oncogene Proteins/genetics , Animals , Antigens, CD34/metabolism , Cell Lineage , Dioxygenases , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Homeostasis , Humans , Lymphoma/metabolism , Mice , Models, Animal , Mutation/genetics , Myeloid Cells/metabolism , Myeloid Cells/pathology , Precancerous Conditions/metabolism
17.
Proc Natl Acad Sci U S A ; 108(14): 5825-30, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21436032

ABSTRACT

The mechanotransducer channels of auditory hair cells are gated by tip-links, oblique filaments that interconnect the stereocilia of the hair bundle. Tip-links stretch from the tips of stereocilia in the short and middle rows to the sides of neighboring, taller stereocilia. They are made of cadherin-23 and protocadherin-15, products of the Usher syndrome type 1 genes USH1D and USH1F, respectively. In this study we address the role of sans, a putative scaffold protein and product of the USH1G gene. In Ush1g(-/-) mice, the cohesion of stereocilia is disrupted, and both the amplitude and the sensitivity of the transduction currents are reduced. In Ush1g(fl/fl)Myo15-cre(+/-) mice, the loss of sans occurs postnatally and the stereocilia remain cohesive. In these mice, there is a decrease in the amplitude of the total transducer current with no loss in sensitivity, and the tips of the stereocilia in the short and middle rows lose their prolate shape, features that can be attributed to the loss of tip-links. Furthermore, stereocilia from these rows undergo a dramatic reduction in length, suggesting that the mechanotransduction machinery has a positive effect on F-actin polymerization. Sans interacts with the cytoplasmic domains of cadherin-23 and protocadherin-15 in vitro and is absent from the hair bundle in mice defective for either of the two cadherins. Because sans localizes mainly to the tips of short- and middle-row stereocilia in vivo, we conclude that it belongs to a molecular complex at the lower end of the tip-link and plays a critical role in the maintenance of this link.


Subject(s)
Actins/metabolism , Hair Cells, Auditory/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Cadherin Related Proteins , Cadherins/metabolism , Cilia/metabolism , Electrophysiology , Fluorescent Antibody Technique , Genetic Vectors/genetics , Hair Cells, Auditory/ultrastructure , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Nerve Tissue Proteins/genetics , Polymerization , Protein Precursors/metabolism , Signal Transduction/genetics
18.
FASEB J ; 25(1): 337-47, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20881209

ABSTRACT

AMP-activated protein kinase (AMPK) is an αßγ heterotrimer conserved throughout evolution and important for energy sensing in all eukaryote cells. AMPK controls metabolism and various cellular events in response to both hormones and changes in cellular energy status. The γ subunit senses intracellular energy status through the competitive binding of AMP and ATP. We show here that targeted disruption of the mouse AMPKγ1 gene (Prkag1) causes regenerative hemolytic anemia by increasing the sequestration of abnormal erythrocytes. Prkag1(-/-) mice displayed splenomegaly and iron accumulation due to compensatory splenic erythropoiesis and erythrophagocytosis. Moreover, AMPKγ1-deficient erythrocytes were highly resistant to osmotic hemolysis and poorly deformable in response to increasing shear stress, consistent with greater membrane rigidity. No change in cytoskeletal protein composition was observed; however, the phosphorylation level of adducin, a protein promoting the binding of spectrin to actin, was higher in AMPKγ1-deficient erythrocytes. Together, these results demonstrate that AMPKγ1 subunit is required for the maintenance of erythrocyte membrane elasticity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anemia/enzymology , Erythrocyte Membrane/metabolism , Splenomegaly/enzymology , AMP-Activated Protein Kinases/genetics , Anemia/blood , Anemia/genetics , Anemia, Hemolytic/enzymology , Anemia, Hemolytic/genetics , Animals , Blotting, Western , Elasticity , Erythroblasts/metabolism , Erythroblasts/pathology , Erythrocyte Count , Erythrocyte Deformability , Female , Hyperplasia , Iron/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Spleen/metabolism , Spleen/pathology , Splenomegaly/blood , Splenomegaly/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...