Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cell ; 177(5): 1201-1216.e19, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31031005

ABSTRACT

Innate immune responses are intricately linked with intracellular metabolism of myeloid cells. Toll-like receptor (TLR) stimulation shifts intracellular metabolism toward glycolysis, while anti-inflammatory signals depend on enhanced mitochondrial respiration. How exogenous metabolic signals affect the immune response is unknown. We demonstrate that TLR-dependent responses of dendritic cells (DCs) are exacerbated by a high-fatty-acid (FA) metabolic environment. FAs suppress the TLR-induced hexokinase activity and perturb tricarboxylic acid cycle metabolism. These metabolic changes enhance mitochondrial reactive oxygen species (mtROS) production and, in turn, the unfolded protein response (UPR), leading to a distinct transcriptomic signature with IL-23 as hallmark. Interestingly, chemical or genetic suppression of glycolysis was sufficient to induce this specific immune response. Conversely, reducing mtROS production or DC-specific deficiency in XBP1 attenuated IL-23 expression and skin inflammation in an IL-23-dependent model of psoriasis. Thus, fine-tuning of innate immunity depends on optimization of metabolic demands and minimization of mtROS-induced UPR.


Subject(s)
Cellular Microenvironment/immunology , Dendritic Cells/immunology , Immunity, Innate , Mitochondria/immunology , Reactive Oxygen Species/immunology , Unfolded Protein Response/immunology , Animals , Cellular Microenvironment/genetics , Citric Acid Cycle/genetics , Citric Acid Cycle/immunology , Dendritic Cells/pathology , Hexokinase/genetics , Hexokinase/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Unfolded Protein Response/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology
3.
Arterioscler Thromb Vasc Biol ; 36(12): 2324-2333, 2016 12.
Article in English | MEDLINE | ID: mdl-27758768

ABSTRACT

OBJECTIVE: Although initially seemingly paradoxical because of the lack of nucleus, platelets possess many transcription factors that regulate their function through DNA-independent mechanisms. These include the farnesoid X receptor (FXR), a member of the superfamily of ligand-activated transcription factors, that has been identified as a bile acid receptor. In this study, we show that FXR is present in human platelets and FXR ligands, GW4064 and 6α-ethyl-chenodeoxycholic acid, modulate platelet activation nongenomically. APPROACH AND RESULTS: FXR ligands inhibited the activation of platelets in response to stimulation of collagen or thrombin receptors, resulting in diminished intracellular calcium mobilization, secretion, fibrinogen binding, and aggregation. Exposure to FXR ligands also reduced integrin αIIbß3 outside-in signaling and thereby reduced the ability of platelets to spread and to stimulate clot retraction. FXR function in platelets was found to be associated with the modulation of cyclic guanosine monophosphate levels in platelets and associated downstream inhibitory signaling. Platelets from FXR-deficient mice were refractory to the actions of FXR agonists on platelet function and cyclic nucleotide signaling, firmly linking the nongenomic actions of these ligands to the FXR. CONCLUSIONS: This study provides support for the ability of FXR ligands to modulate platelet activation. The atheroprotective effects of GW4064, with its novel antiplatelet effects, indicate FXR as a potential target for the prevention of atherothrombotic disease.


Subject(s)
Blood Platelets/drug effects , Chenodeoxycholic Acid/analogs & derivatives , Hemostasis/drug effects , Isoxazoles/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Thrombosis/prevention & control , Animals , Blood Platelets/metabolism , Calcium Signaling/drug effects , Chenodeoxycholic Acid/pharmacology , Cyclic GMP/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrinogen/metabolism , Genotype , Humans , Ligands , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Cytoplasmic and Nuclear/blood , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Thrombosis/blood , Time Factors
4.
J Allergy Clin Immunol ; 138(5): 1309-1318.e11, 2016 11.
Article in English | MEDLINE | ID: mdl-27177781

ABSTRACT

BACKGROUND: Epidemiologic and clinical observations identify obesity as an important risk factor for asthma exacerbation, but the underlying mechanisms remain poorly understood. Type 2 innate lymphoid cells (ILC2s) and type 3 innate lymphoid cells (ILC3s) have been implicated, respectively, in asthma and adipose tissue homeostasis and in obesity-associated airway hyperresponsiveness (AHR). OBJECTIVE: We sought to determine the potential involvement of innate lymphoid cells (ILCs) in allergic airway disease exacerbation caused by high-fat diet (HFD)-induced obesity. METHODS: Obesity was induced by means of HFD feeding, and allergic airway inflammation was subsequently induced by means of intranasal administration of house dust mite (HDM) extract. AHR, lung and visceral adipose tissue inflammation, humoral response, cytokines, and innate and adaptive lymphoid populations were analyzed in the presence or absence of ILCs. RESULTS: HFD feeding exacerbated allergic airway disease features, including humoral response, airway and tissue eosinophilia, AHR, and TH2 and TH17 pulmonary profiles. Notably, nonsensitized obese mice already exhibited increased lung ILC counts and tissue eosinophil infiltration compared with values in lean mice in the absence of AHR. The numbers of total and cytokine-expressing lung ILC2s and ILC3s further increased in HDM-challenged obese mice compared with those in HDM-challenged lean mice, and this was accompanied by high IL-33 and IL-1ß levels and decreased ILC markers in visceral adipose tissue. Furthermore, depletion of ILCs with an anti-CD90 antibody, followed by T-cell reconstitution, led to a profound decrease in allergic airway inflammatory features in obese mice, including TH2 and TH17 infiltration. CONCLUSION: These results indicate that HFD-induced obesity might exacerbate allergic airway inflammation through mechanisms involving ILC2s and ILC3s.


Subject(s)
Asthma/immunology , Lymphocytes/immunology , Obesity/immunology , Animals , Antigens, Dermatophagoides/immunology , Asthma/blood , Asthma/physiopathology , Cytokines/immunology , Diet, High-Fat , Immunity, Innate , Immunoglobulin E/blood , Lung/immunology , Mice, Inbred C57BL , Mice, Transgenic , Obesity/blood , Obesity/physiopathology , Spleen/cytology
5.
Clin Sci (Lond) ; 127(7): 507-18, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24766485

ABSTRACT

Non-alcoholic fatty liver (steatosis) and steatohepatitis [non-alcoholic steatohepatitis (NASH)] are hepatic complications of the metabolic syndrome. Endoplasmic reticulum (ER) stress is proposed as a crucial disease mechanism in obese and insulin-resistant animals (such as ob/ob mice) with simple steatosis, but its role in NASH remains controversial. We therefore evaluated the role of ER stress as a disease mechanism in foz/foz mice, which develop both the metabolic and histological features that mimic human NASH. We explored ER stress markers in the liver of foz/foz mice in response to a high-fat diet (HFD) at several time points. We then evaluated the effect of treatment with an ER stress inducer tunicamycin, or conversely with the ER protectant tauroursodeoxycholic acid (TUDCA), on the metabolic and hepatic features. foz/foz mice are obese, glucose intolerant and develop NASH characterized by steatosis, inflammation, ballooned hepatocytes and apoptosis from 6 weeks of HFD feeding. This was not associated with activation of the upstream unfolded protein response [phospho-eukaryotic initiation factor 2α (eIF2α), inositol-requiring enzyme 1α (IRE1α) activity and spliced X-box-binding protein 1 (Xbp1)]. Activation of c-Jun N-terminal kinase (JNK) and up-regulation of activating transcription factor-4 (Atf4) and CCAAT/enhancer-binding protein-homologous protein (Chop) transcripts were however compatible with a 'pathological' response to ER stress. We tested this by using intervention experiments. Induction of chronic ER stress failed to worsen obesity, glucose intolerance and NASH pathology in HFD-fed foz/foz mice. In addition, the ER protectant TUDCA, although reducing steatosis, failed to improve glucose intolerance, hepatic inflammation and apoptosis in HFD-fed foz/foz mice. These results show that signals driving hepatic inflammation, apoptosis and insulin resistance are independent of ER stress in obese diabetic mice with steatohepatitis.


Subject(s)
Diet, High-Fat , Endoplasmic Reticulum Stress , Fatty Liver/metabolism , Insulin Resistance , Animals , Blood Glucose , Cell Cycle Proteins , DNA-Binding Proteins/genetics , Fatty Liver/pathology , Female , Male , Mice , Mice, Inbred NOD , Mice, Obese , Non-alcoholic Fatty Liver Disease , Phenotype
6.
Med Sci (Paris) ; 40(3): 275-282, 2024 Mar.
Article in French | MEDLINE | ID: mdl-38520103

ABSTRACT

Cells can be reprogrammed into senescence to adapt to a variety of stresses, most often affecting the genome integrity. Senescent cells accumulate with age or upon various insults in almost all tissues, and contribute to the development of several age-associated pathologies. Studying the molecular pathways involved in senescence induction, maintenance, or escape is challenged by the heterogeneity in the level of commitment to senescence, and by the pollution of senescent cell populations by proliferating pre- or post-senescent cells. We coped with these difficulties by developing a protocol for sorting senescent cells by flow cytometry, based on three major senescence markers : the SA-ß-Galactosidase activity, the size of the cells, and their granularity reflecting the accumulation of aggregates, lysosomes, and altered mitochondria. We address the issues related to sorting senescent cells, the pitfalls to avoid, and propose solutions for sorting viable cells expressing senescent markers at different extents.


Title: Tri des cellules sénescentes par cytométrie en flux - Des spécificitéset des pièges à éviter. Abstract: La sénescence est un état d'adaptation des cellules au stress qui contribue au vieillissement et au développement de nombreuses maladies. Étudier les voies moléculaires modulant l'induction, le maintien ou l'échappement de la sénescence est compliqué par la contamination des populations de cellules sénescentes par des cellules proliférantes pré- ou post-sénescentes. Pour contourner cette difficulté, nous avons développé un protocole de tri par cytométrie en flux, fondé sur trois marqueurs majeurs de sénescence (l'activité SA-ß-galactosidase, la taille et la granularité des cellules), qui permet de trier des cellules sénescentes viables, à des degrés choisis d'engagement dans le phénotype.


Subject(s)
Cellular Senescence , Lysosomes , Humans , Cellular Senescence/genetics , Flow Cytometry
7.
Cell Death Dis ; 15(6): 391, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830870

ABSTRACT

Tissue injury causes activation of mesenchymal lineage cells into wound-repairing myofibroblasts (MFs), whose uncontrolled activity ultimately leads to fibrosis. Although this process is triggered by deep metabolic and transcriptional reprogramming, functional links between these two key events are not yet understood. Here, we report that the metabolic sensor post-translational modification O-linked ß-D-N-acetylglucosaminylation (O-GlcNAcylation) is increased and required for myofibroblastic activation. Inhibition of protein O-GlcNAcylation impairs archetypal myofibloblast cellular activities including extracellular matrix gene expression and collagen secretion/deposition as defined in vitro and using ex vivo and in vivo murine liver injury models. Mechanistically, a multi-omics approach combining proteomic, epigenomic, and transcriptomic data mining revealed that O-GlcNAcylation controls the MF transcriptional program by targeting the transcription factors Basonuclin 2 (BNC2) and TEA domain transcription factor 4 (TEAD4) together with the Yes-associated protein 1 (YAP1) co-activator. Indeed, inhibition of protein O-GlcNAcylation impedes their stability leading to decreased functionality of the BNC2/TEAD4/YAP1 complex towards promoting activation of the MF transcriptional regulatory landscape. We found that this involves O-GlcNAcylation of BNC2 at Thr455 and Ser490 and of TEAD4 at Ser69 and Ser99. Altogether, this study unravels protein O-GlcNAcylation as a key determinant of myofibroblastic activation and identifies its inhibition as an avenue to intervene with fibrogenic processes.


Subject(s)
Myofibroblasts , Signal Transduction , Myofibroblasts/metabolism , Animals , Mice , Humans , Fibrosis/metabolism , Transcription Factors/metabolism , YAP-Signaling Proteins/metabolism , Mice, Inbred C57BL , TEA Domain Transcription Factors/metabolism , Male , Protein Processing, Post-Translational , Acetylglucosamine/metabolism , Transcription, Genetic , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics
8.
Clin Sci (Lond) ; 125(11): 501-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23758436

ABSTRACT

Alcohol consumption is a major cause of liver disease. It also associates with increased cardiovascular risk and Type 2 diabetes. ALD (alcoholic liver disease) and NAFLD (non-alcoholic fatty liver disease) share pathological features, pathogenic mechanisms and pattern of disease progression. In NAFLD, steatosis, lipotoxicity and liver inflammation participate to hepatic insulin resistance. The aim of the present study was to verify the effect of alcohol on hepatic insulin sensitivity and to evaluate the role of alcohol-induced steatosis and inflammation on glucose homoeostasis. C57BL/6J mice were fed for 20 days a modified Lieber-DeCarli diet in which the alcohol concentration was gradually increased up to 35% of daily caloric intake. OH (alcohol liquid diet)-fed mice had liver steatosis and inflammatory infiltration. In addition, these mice developed insulin resistance in the liver, but not in muscles, as demonstrated by euglycaemic-hyperinsulinaemic clamp and analysis of the insulin signalling cascade. Treatment with the PPAR-α (peroxisome-proliferator-activated receptor-α) agonist Wy14,643 protected against OH-induced steatosis and KC (Kupffer cell) activation and almost abolished OH-induced insulin resistance. As KC activation may modulate insulin sensitivity, we repeated the clamp studies in mice depleted in KC to decipher the role of macrophages. Depletion of KC using liposomes-encapsuled clodronate in OH-fed mice failed both to improve hepatic steatosis and to restore insulin sensitivity as assessed by clamp. Our study shows that chronic alcohol consumption induces steatosis, KC activation and hepatic insulin resistance in mice. PPAR-α agonist treatment that prevents steatosis and dampens hepatic inflammation also prevents alcohol-induced hepatic insulin resistance. However, KC depletion has little impact on OH-induced metabolic disturbances.


Subject(s)
Alcohol Drinking/metabolism , Glucose/metabolism , PPAR alpha/physiology , Animals , Blood Glucose , Clodronic Acid/pharmacology , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Homeostasis , Hydroxides/pharmacology , Insulin Resistance , Kupffer Cells/drug effects , Kupffer Cells/physiology , Macrophage Activation/drug effects , Mice , Mice, Inbred C57BL , Oxidative Stress , PPAR alpha/agonists , PPAR alpha/metabolism , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology
9.
Bio Protoc ; 13(7): e4612, 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-37056241

ABSTRACT

Cellular senescence is a reprogrammed cell state triggered as an adaptative response to a variety of stresses, most often those affecting the genome integrity. Senescent cells accumulate in most tissues with age and contribute to the development of several pathologies. Studying molecular pathways involved in senescence induction and maintenance, or in senescence escape, can be hindered by the heterogeneity of senescent cell populations. Here, we describe a flow cytometry strategy for sorting senescent cells according to three senescence canonical markers whose thresholds can be independently adapted to be more or less stringent: (i) the senescence-associated-ß-galactosidase (SA-ß-Gal) activity, detected using 5-dodecanoylaminofluorescein Di-ß-D-galactopyranoside (C12FDG), a fluorigenic substrate of ß-galactosidase; (ii) cell size, proportional to the forward scatter value, since increased size is one of the major changes observed in senescent cells; and (iii) cell granularity, proportional to the side scatter value, which reflects the accumulation of aggregates, lysosomes, and altered mitochondria in senescent cells. We applied this protocol to the sorting of normal human fibroblasts at the replicative senescence plateau. We highlighted the challenge of sorting these senescent cells because of their large sizes, and established that it requires using sorters equipped with a nozzle of an unusually large diameter: at least 200 µm. We present evidence of the sorting efficiency and sorted cell viability, as well as of the senescent nature of the sorted cells, confirmed by the detection of other senescence markers, including the expression of the CKI p21 and the presence of 53BP1 DNA damage foci. Our protocol makes it possible, for the first time, to sort senescent cells from contaminating proliferating cells and, at the same time, to sort subpopulations of senescent cells featuring senescent markers to different extents. Graphical abstract.

10.
J Mol Endocrinol ; 71(1)2023 07 01.
Article in English | MEDLINE | ID: mdl-36988391

ABSTRACT

The functional versatility of the liver is paramount for organismal homeostasis. Adult liver functions are controlled by a tightly regulated transcription factor network including nuclear receptors (NRs), which orchestrate many aspects of hepatic physiology. NRs are transcription factors sensitive to extracellular cues such as hormones, lipids, xenobiotics, etc. and are modulated by intracellular signaling pathways. While liver functional zonation and adaptability to fluctuating conditions rely on a sophisticated cellular architecture, a comprehensive knowledge of NR functions within liver cell populations is still lacking. As a step toward the accurate mapping of NR functions in the liver, we characterized their levels of expression in the whole liver from C57Bl6/J male mice as a function of time and diet. Nr1d1 (Rev-erba), Nr1d2 (Rev-erbb), Nr1c2 (Pparb/d), and Nr1f3 (Rorg) exhibited a robust cyclical expression in ad libitum-fed mice which was, like most cyclically expressed NRs, reinforced upon time-restricted feeding. In a few instances, cyclical expression was lost or gained as a function of the feeding regimen. NR isoform expression was explored in purified hepatocytes, cholangiocytes, Kupffer cells, hepatic stellate cells, and liver sinusoidal cells. The expression of some NR isoforms, such as Nr1h4 (Fxra) and Nr1b1 (Rara) isoforms, was markedly restricted to a few cell types. Leveraging liver single-cell RNAseq studies yielded a zonation pattern of NRs in hepatocytes, liver sinusoidal cells, and stellate cells, establishing a link between NR subtissular localization and liver functional specialization. In summary, we provide here an up-to-date compendium of NR expression in mouse liver in space and time.


Subject(s)
Hepatocytes , Liver , Male , Mice , Animals , Liver/metabolism , Hepatocytes/metabolism , Gene Expression Regulation , Signal Transduction/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism
11.
iScience ; 26(7): 107231, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37496675

ABSTRACT

Histone deacetylases enzymes (HDACs) are chromatin modifiers that regulate gene expression through deacetylation of lysine residues within specific histone and non-histone proteins. A cell-specific gene expression pattern defines the identity of insulin-producing pancreatic ß cells, yet molecular networks driving this transcriptional specificity are not fully understood. Here, we investigated the HDAC-dependent molecular mechanisms controlling pancreatic ß-cell identity and function using the pan-HDAC inhibitor trichostatin A through chromatin immunoprecipitation assays and RNA sequencing experiments. We observed that TSA alters insulin secretion associated with ß-cell specific transcriptome programming in both mouse and human ß-cell lines, as well as on human pancreatic islets. We also demonstrated that this alternative ß-cell transcriptional program in response to HDAC inhibition is related to an epigenome-wide remodeling at both promoters and enhancers. Our data indicate that HDAC activity could be required to protect against loss of ß-cell identity with unsuitable expression of genes associated with alternative cell fates.

12.
J Am Coll Cardiol ; 81(13): 1263-1278, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36990546

ABSTRACT

BACKGROUND: On-pump cardiac surgery triggers sterile inflammation and postoperative complications such as postoperative atrial fibrillation (POAF). Hematopoietic somatic mosaicism (HSM) is a recently identified risk factor for cardiovascular diseases and results in a shift toward a chronic proinflammatory monocyte transcriptome and phenotype. OBJECTIVES: The aim of this study was to assess the prevalence, characteristics, and impact of HSM on preoperative blood and myocardial myeloid cells as well as on outcomes after cardiac surgery. METHODS: Blood DNA from 104 patients referred for surgical aortic valve replacement (AVR) was genotyped using the HemePACT panel (576 genes). Four screening methods were applied to assess HSM, and postoperative outcomes were explored. In-depth blood and myocardial leukocyte phenotyping was performed in selected patients using mass cytometry and preoperative and postoperative RNA sequencing analysis of classical monocytes. RESULTS: The prevalence of HSM in the patient cohort ranged from 29%, when considering the conventional HSM panel (97 genes) with variant allelic frequencies ≥2%, to 60% when considering the full HemePACT panel and variant allelic frequencies ≥1%. Three of 4 explored HSM definitions were significantly associated with higher risk for POAF. On the basis of the most inclusive definition, HSM carriers exhibited a 3.5-fold higher risk for POAF (age-adjusted OR: 3.5; 95% CI: 1.52-8.03; P = 0.003) and an exaggerated inflammatory response following AVR. HSM carriers presented higher levels of activated CD64+CD14+CD16- circulating monocytes and inflammatory monocyte-derived macrophages in presurgery myocardium. CONCLUSIONS: HSM is frequent in candidates for AVR, is associated with an enrichment of proinflammatory cardiac monocyte-derived macrophages, and predisposes to a higher incidence of POAF. HSM assessment may be useful in the personalized management of patients in the perioperative period. (Post-Operative Myocardial Incident & Atrial Fibrillation [POMI-AF]; NCT03376165).


Subject(s)
Atrial Fibrillation , Cardiac Surgical Procedures , Humans , Atrial Fibrillation/etiology , Atrial Fibrillation/genetics , Mosaicism , Aortic Valve/surgery , Cardiac Surgical Procedures/adverse effects , Risk Factors , Postoperative Complications/epidemiology , Postoperative Complications/genetics , Postoperative Complications/diagnosis
13.
FASEB J ; 25(12): 4301-11, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21873555

ABSTRACT

We aimed to evaluate activation of macrophages in insulin-sensitive tissues (liver, adipose tissue, and muscles) under high-fat diet (HFD) and elucidate the role of Kupffer cells (KC) in HFD-induced insulin resistance. Tissue macrophage populations, insulin signaling, and sensitivity were evaluated in mice fed a HFD for 4 or 16 wk. Selective KC depletion was obtained by intravenous injections of liposome-encapsulated clodronate. Mice fed a HFD for 4 to 16 wk have hepatic and peripheral insulin resistance together with macrophage recruitment in the adipose tissue but not in the liver. Depletion of KC for the last 10 d of the 16 wk experiment fails to improve insulin sensitivity compared to PBS-treated animals. In contrast, preventive KC depletion prior to and during the 4 wk HFD attenuates the development of obesity, adiposity, adipose tissue inflammation (P<0.01 vs. PBS group), and insulin resistance (P<0.01). Interestingly, in mice fed a normal diet, prolonged KC depletion ameliorates insulin sensitivity and decreases adiposity without altering physiological body weight gain or food intake. Preventive and prolonged KC depletion ameliorates insulin sensitivity and prevents adipose tissue inflammation, suggesting a communication between the liver and the adipose tissue in the development of HFD-induced metabolic alterations.


Subject(s)
Diet, High-Fat/adverse effects , Insulin Resistance/physiology , Kupffer Cells/physiology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adiposity , Animals , Clodronic Acid/pharmacology , Fatty Liver/prevention & control , Inflammation/prevention & control , Kupffer Cells/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Obesity/prevention & control
14.
Metabolites ; 12(11)2022 Oct 25.
Article in English | MEDLINE | ID: mdl-36355106

ABSTRACT

Hepatokines (liver secreted proteins with possible distant action) are emerging potential players in insulin resistance in type 2 diabetic patients. Here, we explored the effect of a high-fat diet on the expression of fetuin-A, one of those candidate liver proteins, and its relationship with liver macrophage activation. Mice were fed a normal diet or a high-fat diet for 3 days, known to initiate steatosis and liver insulin resistance. A preventive liver macrophage depletion was obtained by intravenous injection of clodronate-loaded liposomes. The mRNA and protein expression of fetuin-A was evaluated by qPCR, Western blot and immunofluorescence on different insulin-sensitive tissues (liver, adipose tissue, and muscle). Short-term high-fat diet-induced steatosis, liver macrophage activation, and hepatic insulin resistance together with a significantly increased expression of liver AHSG (α2-HS glycoprotein/fetuin-A) mRNA and serum fetuin-A concentration. On immunofluorescence, fetuin-A was mostly expressed in centrilobular hepatocytes. This increase in fetuin-A under high-fat diet was not evidenced in other peripheral insulin-sensitive tissues (skeletal muscle and adipose tissue). The mRNA expression of α2-HS glycoprotein was 800 times higher within the liver compared with the adipose tissue or the muscle. Liver macrophage depletion that significantly ameliorated insulin sensitivity was associated with a significant decrease in α2-HS glycoprotein mRNA expression. In conclusion, this study demonstrated liver fetuin-A overexpression at the initiation of high-fat diet feeding, concurrent with hepatic steatosis and insulin resistance. Targeting liver macrophages in this setting reduced liver α2-HS glycoprotein expression suggesting that fetuin-A acts as an hepatokine with proinsulin resistance effects.

15.
Elife ; 112022 03 18.
Article in English | MEDLINE | ID: mdl-35302491

ABSTRACT

A rare but severe complication of curative-intent radiation therapy is the induction of second primary cancers. These cancers preferentially develop not inside the planning target volume (PTV) but around, over several centimeters, after a latency period of 1-40 years. We show here that normal human or mouse dermal fibroblasts submitted to the out-of-field dose scattering at the margin of a PTV receiving a mimicked patient's treatment do not die but enter in a long-lived senescent state resulting from the accumulation of unrepaired DNA single-strand breaks, in the almost absence of double-strand breaks. Importantly, a few of these senescent cells systematically and spontaneously escape from the cell cycle arrest after a while to generate daughter cells harboring mutations and invasive capacities. These findings highlight single-strand break-induced senescence as the mechanism of second primary cancer initiation, with clinically relevant spatiotemporal specificities. Senescence being pharmacologically targetable, they open the avenue for second primary cancer prevention.


Subject(s)
DNA Repair , Neoplasms, Second Primary , Animals , Carcinogenesis , Cell Transformation, Neoplastic , Cellular Senescence , DNA Breaks, Single-Stranded , DNA Damage , Mice
16.
Am J Physiol Gastrointest Liver Physiol ; 298(1): G107-16, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19875703

ABSTRACT

Recruited adipose tissue macrophages contribute to chronic and low-grade inflammation causing insulin resistance in obesity. Similarly, we hypothesized here that Kupffer cells, the hepatic resident macrophages, play a pathogenic role in hepatic insulin resistance induced by a high-fat diet. Mice were fed a normal diet or high-fat diet for 3 days. Kupffer cell activation was evaluated by immunohistochemistry and quantitative RT-PCR. Insulin sensitivity was assessed in vivo by hyperinsulinemic-euglycemic clamp and insulin-activated signaling was investigated by Western blot. Liposome-encapsulated clodronate was injected intravenously to deplete macrophages prior to a short-term exposure to high-fat diet. Here, we characterized a short-term high-fat diet model in mice and demonstrated early hepatic insulin resistance and steatosis concurrent with Kupffer cell activation. We demonstrated that selective Kupffer cell depletion obtained by intravenous clodronate, without affecting adipose tissue macrophages, was sufficient to enhance insulin-dependent insulin signaling and significantly improve hepatic insulin sensitivity in vivo in this short-term high-fat diet model. Our study clearly shows that hepatic macrophage response participates to the onset of high-fat diet-induced hepatic insulin resistance and may therefore represent an attractive target for prevention and treatment of diet- and obesity-induced insulin resistance.


Subject(s)
Fatty Liver/metabolism , Hepatitis/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Kupffer Cells/metabolism , Obesity/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Clodronic Acid/pharmacology , Dietary Fats/pharmacology , Drug Delivery Systems , Fatty Liver/etiology , Fatty Liver/pathology , Glucose Clamp Technique , Hepatitis/etiology , Hepatitis/pathology , Hyperinsulinism/metabolism , Injections, Intravenous , Kupffer Cells/drug effects , Kupffer Cells/pathology , Liposomes/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
17.
Sci Rep ; 10(1): 21095, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33273527

ABSTRACT

Retinoic acid receptor-related orphan receptor-alpha (RORα) is a transcription factor from the nuclear receptor family expressed by immune cells and involved in the development of obesity, insulin resistance (IR) and non-alcoholic steatohepatitis (NASH). It was recently reported that mice deficient for RORα in macrophages develop more severe NASH upon high fat diet (HFD) feeding due to altered Kupffer cell function. To better understand the role of RORα in obesity and IR, we independently generated a macrophage RORα-deficient mouse line. We report that RORα deletion in macrophages does not impact on HFD-induced obesity and IR. Surprisingly, we did not confirm an effect on NASH development upon HFD feeding nor in the more severe and obesity-independent choline-deficient, L-amino acid-defined diet model. Our results therefore show that RORα deletion in macrophages does not alter the development of obesity and IR and question its role in NASH.


Subject(s)
Insulin Resistance , Macrophages/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Obesity/metabolism , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Gene Deletion , Kupffer Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Obesity/etiology
18.
Commun Biol ; 3(1): 237, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32409640

ABSTRACT

Like all obligate intracellular pathogens, influenza A virus (IAV) reprograms host cell's glucose and lipid metabolism to promote its own replication. However, the impact of influenza infection on white adipose tissue (WAT), a key tissue in the control of systemic energy homeostasis, has not been yet characterized. Here, we show that influenza infection induces alterations in whole-body glucose metabolism that persist long after the virus has been cleared. We report depot-specific changes in the WAT of IAV-infected mice, notably characterized by the appearance of thermogenic brown-like adipocytes within the subcutaneous fat depot. Importantly, viral RNA- and viral antigen-harboring cells are detected in the WAT of infected mice. Using in vitro approaches, we find that IAV infection enhances the expression of brown-adipogenesis-related genes in preadipocytes. Overall, our findings shed light on the role that the white adipose tissue, which lies at the crossroads of nutrition, metabolism and immunity, may play in influenza infection.


Subject(s)
Adipose Tissue, White/metabolism , Energy Metabolism , Orthomyxoviridae Infections/metabolism , Thermogenesis , Adipose Tissue, Brown/metabolism , Animals , Disease Models, Animal , Humans , Influenza, Human/metabolism , Male , Mice , Mice, Inbred C57BL
19.
Neuroendocrinology ; 90(1): 54-66, 2009.
Article in English | MEDLINE | ID: mdl-19276635

ABSTRACT

Numerous data suggest that the development of the sympathoadrenal system is highly sensitive to the perinatal environment. We previously reported that maternal perinatal food restriction by 50% (FR50) altered chromaffin cell (CC) organization and activity in offspring at weaning. This study investigated the effects of FR50 on the postnatal time course of CC functional and structural adaptations. FR50 pups exhibited smaller and more abundant scattered clusters of noradrenergic CCs as early as postnatal day 7 (P7), indicating that morphological changes took place earlier during development. At birth, the adrenaline release was defective in FR50 pups, suggesting that maternal FR50 impaired the non-neurogenic control of catecholamine release. At P4, the catecholamine release in response to insulin-induced hypoglycaemia was also absent in FR50 pups. This was associated with the reduction of adrenal catecholamine contents, indicating that the failure to synthesize catecholamine might lead to impaired secretion. We hypothesized that maternal FR50 accelerated the functional connections between CCs and splanchnic nerve endings, leading to the premature loss of the non-neurogenic response. Acetylcholine-containing synaptic endings seemed more precociously functional in FR50 pups, as suggested by increased levels of acetylcholine esterase activity at P14. At P7, insulin-induced hypoglycaemia caused preferential adrenaline release associated with increased catecholamine contents in both groups. However, the response was accentuated in FR50 pups. At P14, the insulin challenge increased plasma levels of adrenaline in control rats, whereas it markedly enhanced the circulating level of both catecholamines in FR50 pups. We demonstrated that maternal FR50 leads to developmentally impaired noradrenergic CC aggregation and advanced splanchnic neurotransmission maturation associated with altered medulla activity in response to metabolic stress. This might contribute to the long-lasting malprogramming of the adrenal medulla and to the development of chronic adult diseases.


Subject(s)
Adrenal Medulla/growth & development , Chromaffin Cells/physiology , Malnutrition , Mothers , Prenatal Exposure Delayed Effects , Acetylcholinesterase/metabolism , Adrenal Medulla/physiology , Adrenal Medulla/physiopathology , Animals , Animals, Newborn , Catecholamines/blood , Catecholamines/metabolism , Epinephrine/blood , Epinephrine/metabolism , Female , Hypoglycemia/chemically induced , Hypoglycemia/physiopathology , Insulin , Male , Norepinephrine/metabolism , Pregnancy , Random Allocation , Rats , Rats, Wistar , Stress, Physiological/physiology , Synapses/physiology
20.
Nat Metab ; 1(6): 604-614, 2019 06.
Article in English | MEDLINE | ID: mdl-31701087

ABSTRACT

Progression of fatty liver to non-alcoholic steatohepatitis (NASH) is a rapidly growing health problem. Presence of inflammatory infiltrates in the liver and hepatocyte damage distinguish NASH from simple steatosis. However, the underlying molecular mechanisms involved in the development of NASH remain to be fully understood. Here we perform transcriptional and immune profiling of NASH patients before and after lifestyle intervention (LSI). Analysis of liver microarray data from a cohort of patients with histologically assessed NAFLD reveals a hepatic gene signature, which is associated with NASH and is sensitive to regression of NASH activity upon LSI independently of body weight loss. Enrichment analysis reveals the presence of immune-associated genes linked to inflammatory responses, antigen presentation and cytotoxic cells in the NASH-linked gene signature. In an independent cohort, NASH is also associated with alterations in blood immune cell populations, including conventional dendritic cells (cDC) type 1 and 2, and cytotoxic CD8 T cells. Lobular inflammation and ballooning are associated with the accumulation of CD8 T cells in the liver. Progression from simple steatosis to NASH in a mouse model of diet-driven NASH results in a comparable immune-related hepatic expression signature and the accumulation of intra-hepatic cDC and CD8 T cells. These results show that NASH, compared to normal liver or simple steatosis, is associated with a distinct hepatic immune-related gene signature, elevated hepatic CD8 T cells, and altered antigen-presenting and cytotoxic cells in blood. These findings expand our understanding of NASH and may identify potential targets for NASH therapy.


Subject(s)
Liver/immunology , Non-alcoholic Fatty Liver Disease/genetics , Transcription, Genetic , Animals , Diet, High-Fat , Gene Regulatory Networks , Humans , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/therapy
SELECTION OF CITATIONS
SEARCH DETAIL