Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Nat Commun ; 14(1): 4675, 2023 08 04.
Article de Anglais | MEDLINE | ID: mdl-37542026

RÉSUMÉ

To maintain and restore skeletal muscle mass and function is essential for healthy aging. We have found that myonectin acts as a cardioprotective myokine. Here, we investigate the effect of myonectin on skeletal muscle atrophy in various male mouse models of muscle dysfunction. Disruption of myonectin exacerbates skeletal muscle atrophy in age-associated, sciatic denervation-induced or dexamethasone (DEX)-induced muscle atrophy models. Myonectin deficiency also contributes to exacerbated mitochondrial dysfunction and reduces expression of mitochondrial biogenesis-associated genes including PGC1α in denervated muscle. Myonectin supplementation attenuates denervation-induced muscle atrophy via activation of AMPK. Myonectin also reverses DEX-induced atrophy of cultured myotubes through the AMPK/PGC1α signaling. Furthermore, myonectin treatment suppresses muscle atrophy in senescence-accelerated mouse prone (SAMP) 8 mouse model of accelerated aging or mdx mouse model of Duchenne muscular dystrophy. These data indicate that myonectin can ameliorate skeletal muscle dysfunction through AMPK/PGC1α-dependent mechanisms, suggesting that myonectin could represent a therapeutic target of muscle atrophy.


Sujet(s)
AMP-Activated Protein Kinases , Muscles squelettiques , Animaux , Mâle , Souris , AMP-Activated Protein Kinases/métabolisme , Souris de lignée mdx , Muscles squelettiques/métabolisme , Amyotrophie/prévention et contrôle , Amyotrophie/induit chimiquement , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme
2.
iScience ; 26(5): 106591, 2023 May 19.
Article de Anglais | MEDLINE | ID: mdl-37250342

RÉSUMÉ

Although chronic kidney disease (CKD) is a major health problem worldwide, its underlining mechanism is incompletely understood. We previously identified adipolin as an adipokine which provides benefits for cardiometabolic diseases. Here, we investigated the role of adipolin in the development of CKD. Adipolin-deficiency exacerbated urinary albumin excretion, tubulointerstitial fibrosis and oxidative stress of remnant kidneys in mice after subtotal nephrectomy through inflammasome activation. Adipolin positively regulated the production of ketone body, ß-hydroxybutyrate (BHB) and expression of a catalytic enzyme producing BHB, HMGCS2 in the remnant kidney. Treatment of proximal tubular cells with adipolin attenuated inflammasome activation through the PPARα/HMGCS2-dependent pathway. Furthermore, systemic administration of adipolin to wild-type mice with subtotal nephrectomy ameliorated renal injury, and these protective effects of adipolin were diminished in PPARα-deficient mice. Thus, adipolin protects against renal injury by reducing renal inflammasome activation through its ability to induce HMGCS2-dependent ketone body production via PPARα activation.

3.
Physiol Rep ; 10(5): e15218, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35262272

RÉSUMÉ

Chronic kidney disease (CKD) is an increasing and life-threatening disease worldwide. Recent evidence indicates that blood coagulation factors promote renal dysfunction in CKD patients. Activated factor X (FXa) inhibitors are safe and first-line drugs for the prevention of thrombosis in patients with atrial fibrillation. Here, we investigated the therapeutic effects of edoxaban on CKD using the mouse 5/6 nephrectomy model. Eight-week-old wild-type mice were subjected to 5/6 nephrectomy surgery and randomly assigned to two groups, edoxaban or vehicle admixture diet. Edoxaban treatment led to reduction of urinary albumin excretion and plasma UN levels compared with vehicle group, which was accompanied with reduced glomerular cross-sectional area and cell number. Edoxaban treatment also attenuated fibrinogen positive area in the remnant kidneys after subtotal nephrectomy. Moreover, edoxaban treatment resulted in attenuated tubulointerstitial fibrosis after 5/6 nephrectomy, which was accompanied by reduced expression levels of epithelial-mesenchymal transition (EMT) markers, inflammatory mediators, and oxidative stress markers in the remnant kidneys. Treatment of cultured proximal tubular cells, HK-2 cells, with FXa protein led to increased expression levels of EMT markers, inflammatory mediators, and oxidative stress markers, which were abolished by pretreatment with edoxaban. Treatment of HK-2 cells with edoxaban attenuated FXa-stimulated phosphorylation levels of extracellular signal-regulated kinase (ERK) and NF-κB. Our findings indicate that edoxaban can improve renal injury after subtotal nephrectomy by reducing EMT and inflammatory response, suggesting that FXa inhibition could be a novel therapeutic target for CKD patients with atrial fibrillation.


Sujet(s)
Fibrillation auriculaire , Insuffisance rénale chronique , Animaux , Souris , Fibrillation auriculaire/anatomopathologie , Modèles animaux de maladie humaine , Transition épithélio-mésenchymateuse , Inhibiteurs du facteur Xa/pharmacologie , Inhibiteurs du facteur Xa/usage thérapeutique , Fibrose , Médiateurs de l'inflammation/pharmacologie , Rein , Néphrectomie/effets indésirables , Pyridines , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/étiologie , Insuffisance rénale chronique/anatomopathologie , Thiazoles
4.
Biochem Biophys Res Commun ; 593: 5-12, 2022 02 19.
Article de Anglais | MEDLINE | ID: mdl-35051783

RÉSUMÉ

Skeletal muscle atrophy caused by various conditions including aging, nerve damage, and steroid administration, is a serious health problem worldwide. We recently reported that neuron-derived neurotrophic factor (NDNF) functions as a muscle-derived secreted factor, also known as myokine, which exerts protective actions on endothelial cell and cardiomyocyte function. Here, we investigated whether NDNF regulates skeletal muscle atrophy induced by steroid administration and sciatic denervation. NDNF-knockout (KO) mice and age-matched wild-type (WT) mice were subjected to continuous dexamethasone (DEX) treatment or sciatic denervation. NDNF-KO mice exhibited decreased gastrocnemius muscle weight and reduced cross sectional area of myocyte fiber after DEX treatment or sciatic denervation compared with WT mice. Administration of an adenoviral vector expressing NDNF (Ad-NDNF) or recombinant NDNF protein to gastrocnemius muscle of WT mice increased gastrocnemius muscle weight after DEX treatment. NDNF-KO mice showed increased expression of ubiquitin E3-ligases, including atrogin-1 and MuRF-1, in gastrocnemius muscle after DEX treatment, whereas Ad-NDNF reduced expression of atrogin-1 and MuRF-1 in gastrocnemius muscle of WT mice after DEX treatment. Pretreatment of cultured C2C12 myocytes with NDNF protein reversed reduced myotube diameter and increased expression of atrogin-1 and MuRF-1 after DEX stimulation. Treatment of C2C12 myocytes increased Akt phosphorylation. Pretreatment of C2C12 myotubes with the PI3-kinase/Akt inhibitor reversed NDNF-induced increase in myotube fiber diameter after DEX treatment. In conclusion, our findings indicated that NDNF prevents skeletal muscle atrophy in vivo and in vitro through reduction of ubiquitin E3-ligases expression, suggesting that NDNF could be a novel therapeutic target of muscle atrophy.


Sujet(s)
Dexaméthasone/toxicité , Muscles squelettiques/effets des médicaments et des substances chimiques , Amyotrophie/prévention et contrôle , Facteurs de croissance nerveuse/pharmacologie , Neurones/effets des médicaments et des substances chimiques , Agents protecteurs/métabolisme , Animaux , Anti-inflammatoires/toxicité , Femelle , Régulation de l'expression des gènes , Souris , Souris de lignée C57BL , Souris knockout , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Amyotrophie/induit chimiquement , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Neurones/métabolisme , Neurones/anatomopathologie , Phosphorylation
5.
J Cardiol ; 78(6): 524-532, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34334268

RÉSUMÉ

BACKGROUND: Follistatin-like 1 (FSTL1) is a myocyte-secreted glycoprotein that could play a role in myocardial maintenance in response to harmful stimuli. We investigated the association between serum FSTL1 levels, especially focused on transcardiac gradient and the hemodynamics, to explore the prognostic impact of FSTL1 levels in patients with dilated cardiomyopathy (DCM). METHODS: Thirty-two ambulatory patients with DCM (23 men; mean age 59 years) were prospectively enrolled. Blood samples were simultaneously collected from the aortic root (Ao), coronary sinus (CS), as well as from the peripheral vein during cardiac catheterization in stable conditions. The transcardiac gradient of FSTL1 was calculated by the difference between serum FSTL1 levels of CS and Ao (FSTL1CS-Ao). Patients were divided into two groups based on the median of FSTL1CS-Ao: Low FSTL1CS-Ao group, <0 ng/mL; High FSTL1CS-Ao group, ≥0 ng/mL. Cardiac events were defined as a composite of cardiac deaths and hospitalizations for worsening heart failure. RESULTS: Mean left ventricular ejection fraction and median plasma B-type natriuretic peptide levels were 30.9% and 92.3 pg/mL, respectively. FSTL1CS-Ao was negatively correlated with pulmonary capillary wedge pressure (r = -0.400, p = 0.023). Kaplan-Meier survival analysis showed that event-free survival rate was significantly lower in the Low FSTL1CS-Ao group than in the High FSTL1CS-Ao group (p = 0.013). Cox regression analyses revealed that the transcardiac gradient of FSTL1 was an independent predictor for cardiac events. Receiver operating characteristic curve analysis showed that the cut-off value of FSTL1CS-Ao for the prediction of cardiac events was -4.09 ng/mL with sensitivity of 82% and specificity of 86% (area under the curve, 0.87). CONCLUSIONS: Fifty percent of patients had negative transcardiac gradient of FSTL1. Reduced transcardiac gradient of FSTL1 might be a novel prognostic predictor in DCM patients with impaired hemodynamics.


Sujet(s)
Cardiomyopathie dilatée , Protéines apparentées à la follistatine , Défaillance cardiaque , Follistatine , Hémodynamique , Humains , Mâle , Adulte d'âge moyen , Pronostic , Débit systolique , Fonction ventriculaire gauche
6.
PLoS One ; 15(12): e0243483, 2020.
Article de Anglais | MEDLINE | ID: mdl-33275602

RÉSUMÉ

BACKGROUND: Myocardial infarction (MI) is a leading cause of death worldwide. We previously identified adipolin, also known as C1q/Tnf-related protein 12, as an anti-inflammatory adipokine with protective features against metabolic and vascular disorders. Here, we investigated the effect of adipolin on myocardial remodeling in a mouse model of MI. METHODS: Male adipolin-knockout (APL-KO) and wild-type (WT) mice were subjected to the permanent ligation of the left anterior descending coronary artery to create MI. RESULTS: APL-KO mice exhibited increased ratios of heart weight/body weight and lung weight/body weight after MI compared with WT mice. APL-KO mice showed increased left ventricular diastolic diameter and decreased fractional shortening after MI compared with WT mice. APL-KO mice exhibited increased expression of pro-inflammatory mediators and enhanced cardiomyocyte apoptosis in the post-MI hearts compared with WT mice. Systemic administration of adenoviral vectors expressing adipolin to WT mice after MI surgery improved left ventricular contractile dysfunction and reduced cardiac expression of pro-inflammatory genes. Treatment of cultured cardiomyocytes with adipolin protein reduced lipopolysaccharide-induced expression of pro-inflammatory mediators and hypoxia-induced apoptosis. Treatment with adipolin protein increased Akt phosphorylation in cardiomyocytes. Inhibition of PI3 kinase/Akt signaling reversed the anti-inflammatory and anti-apoptotic effects of adipolin in cardiomyocytes. CONCLUSION: Our data indicate that adipolin ameliorates pathological remodeling of myocardium after MI, at least in part, by its ability to reduce myocardial inflammatory response and apoptosis.


Sujet(s)
Adipokines/métabolisme , Infarctus du myocarde/anatomopathologie , Remodelage ventriculaire , Adipokines/déficit , Adipokines/génétique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Médiateurs de l'inflammation/métabolisme , Lipopolysaccharides/pharmacologie , Souris , Souris de lignée C57BL , Souris knockout , Infarctus du myocarde/métabolisme , Myocarde/métabolisme , Myocarde/anatomopathologie , Myocytes cardiaques/cytologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Rats , Protéines recombinantes/biosynthèse , Protéines recombinantes/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme
7.
Cell Rep ; 29(1): 162-175.e9, 2019 10 01.
Article de Anglais | MEDLINE | ID: mdl-31577946

RÉSUMÉ

Type I interferons (IFNs) play important roles in antitumor immunity. We generated IFN-α-producing cells by genetically engineered induced pluripotent stem cell (iPSC)-derived proliferating myeloid cells (iPSC-pMCs). Local administration of IFN-α-producing iPSC-pMCs (IFN-α-iPSC-pMCs) alters the tumor microenvironment and propagates the molecular signature associated with type I IFN. The gene-modified cell actively influences host XCR1+ dendritic cells to enhance CD8+ T cell priming, resulting in CXCR3-dependent and STING-IRF3 pathway-independent systemic tumor control. Administration of IFN-α-iPSC-pMCs in combination with immune checkpoint blockade overcomes resistance to single-treatment modalities and generates long-lasting antitumor immunity. These preclinical data suggest that IFN-α-iPSC-pMCs might constitute effective immune-stimulating agents for cancer that are refractory to checkpoint blockade.


Sujet(s)
Cellules dendritiques/immunologie , Immunité/immunologie , Cellules souches pluripotentes induites/immunologie , Interféron de type I/immunologie , Cellules myéloïdes/immunologie , Récepteurs aux chimiokines/immunologie , Animaux , Immunothérapie/méthodes , Facteur-3 de régulation d'interféron/immunologie , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Tumeurs/immunologie , Récepteurs CXCR3/immunologie , Microenvironnement tumoral/immunologie
8.
Stem Cell Reports ; 10(6): 1935-1946, 2018 06 05.
Article de Anglais | MEDLINE | ID: mdl-29805109

RÉSUMÉ

CD4+ T helper (Th) cell activation is essential for inducing cytotoxic T lymphocyte (CTL) responses against malignancy. We reprogrammed a Th clone specific for chronic myelogenous leukemia (CML)-derived b3a2 peptide to pluripotency and re-differentiated the cells into original TCR-expressing T-lineage cells (iPS-T cells) with gene expression patterns resembling those of group 1 innate lymphoid cells. CD4 gene transduction into iPS-T cells enhanced b3a2 peptide-specific responses via b3a2 peptide-specific TCR. iPS-T cells upregulated CD40 ligand (CD40L) expression in response to interleukin-2 and interleukin-15. In the presence of Wilms tumor 1 (WT1) peptide, antigen-specific dendritic cells (DCs) conditioned by CD4-modified CD40Lhigh iPS-T cells stimulated WT1-specific CTL priming, which eliminated WT1 peptide-expressing CML cells in vitro and in vivo. Thus, CD4 modification of CD40Lhigh iPS-T cells generates innate lymphoid helper-like cells inducing bcr-abl-specific TCR signaling that mediates effectiveanti-leukemic CTL responses via DC maturation, showing potential for adjuvant immunotherapy against leukemia.


Sujet(s)
Expression des gènes , Immunité innée , Récepteurs aux antigènes des cellules T/génétique , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/métabolisme , Lymphocytes T auxiliaires/immunologie , Lymphocytes T auxiliaires/métabolisme , Marqueurs biologiques , Ligand de CD40/métabolisme , Différenciation cellulaire , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Humains , Immunophénotypage , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/immunologie , Cellules souches pluripotentes induites/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/immunologie , Leucémie myéloïde chronique BCR-ABL positive/mortalité , Spécificité antigénique des récepteurs des lymphocytes T/immunologie , Protéines WT1/immunologie
9.
Cell Mol Immunol ; 15(1): 15-26, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-27181332

RÉSUMÉ

The advent of tyrosine kinase inhibitor (TKI) therapy markedly improved the outcome of patients with chronic-phase chronic myeloid leukemia (CML). However, the poor prognosis of patients with advanced-phase CML and the lifelong dependency on TKIs are remaining challenges; therefore, an effective therapeutic has been sought. The BCR-ABL p210 fusion protein's junction region represents a leukemia-specific neoantigen and is thus an attractive target for antigen-specific T-cell immunotherapy. BCR-ABL p210 fusion-region-specific CD4+ T-helper (Th) cells possess antileukemic potential, but their function remains unclear. In this study, we established a BCR-ABL p210 b3a2 fusion-region-specific CD4+ Th-cell clone (b3a2-specific Th clone) and examined its dendritic cell (DC)-mediated antileukemic potential. The b3a2-specific Th clone recognized the b3a2 peptide in the context of HLA-DRB1*09:01 and exhibited a Th1 profile. Activation of this clone through T-cell antigen receptor stimulation triggered DC maturation, as indicated by upregulated production of CD86 and IL-12p70 by DCs, which depended on CD40 ligation by CD40L expressed on b3a2-specific Th cells. Moreover, in the presence of HLA-A*24:02-restricted Wilms tumor 1 (WT1)235-243 peptide, DCs conditioned by b3a2-specific Th cells efficiently stimulated the primary expansion of WTI-specific cytotoxic T lymphocytes (CTLs). The expanded CTLs were cytotoxic toward WT1235-243-peptide-loaded HLA-A*24:02-positive cell lines and exerted a potent antileukemic effect in vivo. However, the b3a2-specific Th-clone-mediated antileukemic CTL responses were strongly inhibited by both TKIs and interferon-α. Our findings indicate a crucial role of b3a2-specific Th cells in leukemia antigen-specific CTL-mediated immunity and provide an experimental basis for establishing novel CML immunotherapies.


Sujet(s)
Cross-priming/immunologie , Cellules dendritiques/immunologie , Épitopes/immunologie , Protéines de fusion bcr-abl/métabolisme , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T auxiliaires/immunologie , Séquence d'acides aminés , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Clones cellulaires , Cross-priming/effets des médicaments et des substances chimiques , Cellules dendritiques/effets des médicaments et des substances chimiques , Sous-types sérologiques HLA-DR/métabolisme , Humains , Interféron alpha/pharmacologie , Interleukine-12/biosynthèse , Leucémies/anatomopathologie , Souris , Souris de lignée BALB C , Peptides/pharmacologie , Phénotype , Inhibiteurs de protéines kinases/pharmacologie , Récepteurs aux antigènes des cellules T/composition chimique , Récepteurs aux antigènes des cellules T/métabolisme , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Lymphocytes T auxiliaires/effets des médicaments et des substances chimiques
10.
Stem Cell Reports ; 6(2): 213-27, 2016 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-26862702

RÉSUMÉ

Vα24 invariant natural killer T (iNKT) cells are a subset of T lymphocytes implicated in the regulation of broad immune responses. They recognize lipid antigens presented by CD1d on antigen-presenting cells and induce both innate and adaptive immune responses, which enhance effective immunity against cancer. Conversely, reduced iNKT cell numbers and function have been observed in many patients with cancer. To recover these numbers, we reprogrammed human iNKT cells to pluripotency and then re-differentiated them into regenerated iNKT cells in vitro through an IL-7/IL-15-based optimized cytokine combination. The re-differentiated iNKT cells showed proliferation and IFN-γ production in response to α-galactosylceramide, induced dendritic cell maturation and downstream activation of both cytotoxic T lymphocytes and NK cells, and exhibited NKG2D- and DNAM-1-mediated NK cell-like cytotoxicity against cancer cell lines. The immunological features of re-differentiated iNKT cells and their unlimited availability from induced pluripotent stem cells offer a potentially effective immunotherapy against cancer.


Sujet(s)
Adjuvants immunologiques/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules souches pluripotentes induites/cytologie , Cellules tueuses naturelles/cytologie , Récepteurs aux antigènes des cellules T/métabolisme , Mort cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Humains , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/métabolisme
11.
Cancer Immunol Res ; 3(6): 668-77, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25672396

RÉSUMÉ

The use of dendritic cells (DC) to prime tumor-associated antigen-specific T-cell responses provides a promising approach to cancer immunotherapy. Embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) can differentiate into functional DCs, thus providing an unlimited source of DCs. However, the previously established methods of generating practical volumes of DCs from pluripotent stem cells (PSC) require a large number of PSCs at the start of the differentiation culture. In this study, we generated mouse proliferating myeloid cells (pMC) as a source of antigen-presenting cells (APC) using lentivirus-mediated transduction of the c-Myc gene into mouse PSC-derived myeloid cells. The pMCs could propagate almost indefinitely in a cytokine-dependent manner, while retaining their potential to differentiate into functional APCs. After treatment with IL4 plus GM-CSF, the pMCs showed impaired proliferation and differentiated into immature DC-like cells (pMC-DC) expressing low levels of major histocompatibility complex (MHC)-I, MHC-II, CD40, CD80, and CD86. In addition, exposure to maturation stimuli induced the production of TNFα and IL12p70, and enhanced the expression of MHC-II, CD40, and CD86, which is thus suggestive of typical DC maturation. Similar to bone marrow-derived DCs, they stimulated a primary mixed lymphocyte reaction. Furthermore, the in vivo transfer of pMC-DCs pulsed with H-2K(b)-restricted OVA257-264 peptide primed OVA-specific cytotoxic T cells and elicited protection in mice against challenge with OVA-expressing melanoma. Overall, myeloid cells exhibiting cytokine-dependent proliferation and DC-like differentiation may be used to address issues associated with the preparation of DCs.


Sujet(s)
Cellules présentatrices d'antigène/cytologie , Cellules présentatrices d'antigène/immunologie , Différenciation cellulaire , Cellules myéloïdes/cytologie , Cellules myéloïdes/immunologie , Cellules souches pluripotentes/cytologie , Transfert adoptif , Animaux , Présentation d'antigène , Cellules présentatrices d'antigène/métabolisme , Antigènes néoplasiques/immunologie , Antigènes de surface/métabolisme , Prolifération cellulaire , Cellules cultivées , Cytokines/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Femelle , Immunophénotypage , Mélanome/immunologie , Mélanome/anatomopathologie , Mélanome/thérapie , Souris , Cellules myéloïdes/métabolisme , Tumeurs/immunologie , Peptides/immunologie , Phénotype , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE