Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Eur Acad Dermatol Venereol ; 37 Suppl 2: 12-19, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36729401

RESUMEN

INTRODUCTION: Acne is a multifactorial inflammatory disease of the pilosebaceous unit in which Cutibacterium acnes is one of the main triggers. A strong predominance of C. acnes phylotype IA1 is present in acne skin with higher biofilm organization and virulence, promoting local immuno-inflammation, especially the Th17 pathway. OBJECTIVES: We evaluated the single and combined pharmacological properties of the plant extracts, Myrtus communis (Myrtacine®) and Celastrol enriched plant cell culture (CEE) extracts on the C. acnes/Th17 pathway. METHODS: The effect of Myrtacine® on the virulence of C. acnes phylotype IA1 was quantified according to the expression of several related genes. The activity of Myrtacine® and CEE on the inflammatory cascade was assessed using monocytes-derived dendritic cells (Mo-DC) stimulated with membranes or biofilms of the C. acnes phylotype IA1. Finally, the effect of CEE on the Th17 pathway was studied using C. acnes stimulated sebocyte 2D cultures and 3D skin tissue models containing preactivated Th17 cells. RESULTS: Myrtacine® had an anti-virulence effect, evident as a significant and strong inhibition of the expression of several virulence factor genes by 60%-95% compared to untreated controls. Myrtacine® and CEE significantly inhibited proinflammatory cytokine (IL-6, IL-8, IL-12p40 and TNF-α) production by Mo-DC in response to C. acnes phylotype IA1. Interestingly, these two ingredients resulted in synergistic inhibition of most cytokines when used in combination. Finally, we demonstrated an inhibitory effect of CEE, in solution or formulated at 0.3%, specifically on IL-17 release by Th17 lymphocytes in a C. acnes-stimulated sebocyte 2D cultures and by Th17-lymphocytes integrated in a 3D skin models. CONCLUSIONS: 2D and 3D models were developed to represent relevant and specific pathways involved in acne. Myrtacine® and CEE were shown to alter one or more of these pathways, indicating their potential beneficial effects on this disease.


Asunto(s)
Acné Vulgar , Myrtus , Humanos , Myrtus/metabolismo , Acné Vulgar/tratamiento farmacológico , Acné Vulgar/microbiología , Citocinas/metabolismo , Técnicas de Cultivo de Célula , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Propionibacterium acnes
2.
JID Innov ; 1(4): 100057, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34909752

RESUMEN

In adipocytes and sebocytes, lipid droplet proteins control the storage of lipids in organized droplets and their release on demand. The contribution of lipid droplet proteins to the pathogenesis of acne is plausible because they control the levels of comedogenic free fatty acids. The expression of two lipid droplet proteins, CIDEA and PLIN2, was analyzed in the skin of patients with acne by immunohistochemistry and western blotting. The design of clinical protocols allowed correlating the expression of CIDEA and PLIN2 with both comedogenesis and the release of free fatty acids. Both proteins were detected by immunohistochemistry in the sebaceous glands of patients with acne, with a disturbed expression pattern of PLIN2 compared with that in the controls. Higher levels of PLIN2 and CIDEA, as detected by western blotting in the infundibulum, significantly correlated with lower ongoing comedogenesis over 48 weeks of Silybum marianum fruit extract application. Accordingly, free fatty acid release from sebum triglycerides was significantly decreased, as shown with two distinct methods. The data are consistent with the expected role of PLIN2 and CIDEA in the prevention of comedogenic free fatty acid release. Modulation of PLIN2 and CIDEA expression appears as a sound target for the maintenance of low comedogenic sebum and acne-prone skin health.

3.
Front Immunol ; 12: 740540, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34650562

RESUMEN

Acne is a multifactorial disease driven by physiological changes occurring during puberty in the pilosebaceous unit (PSU) that leads to sebum overproduction and a dysbiosis involving notably Cutibacterium acnes. These changes in the PSU microenvironment lead to a shift from a homeostatic to an inflammatory state. Indeed, immunohistochemical analyses have revealed that inflammation and lymphocyte infiltration can be detected even in the infraclinical acneic stages, highlighting the importance of the early stages of the disease. In this study, we utilized a robust multi-pronged approach that included flow cytometry, confocal microscopy, and bioinformatics to comprehensively characterize the evolution of the infiltrating and resident immune cell populations in acneic lesions, beginning in the early stages of their development. Using a discovery cohort of 15 patients, we demonstrated that the composition of immune cell infiltrate is highly dynamic in nature, with the relative abundance of different cell types changing significantly as a function of clinical lesion stage. Within the stages examined, we identified a large population of CD69+ CD4+ T cells, several populations of activated antigen presenting cells, and activated mast cells producing IL-17. IL-17+ mast cells were preferentially located in CD4+ T cell rich areas and we showed that activated CD4+ T cells license mast cells to produce IL-17. Our study reveals that mast cells are the main IL-17 producers in the early stage of acne, underlying the importance of targeting the IL-17+ mast cell/T helper cell axis in therapeutic approaches.


Asunto(s)
Acné Vulgar/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Mastocitos/inmunología , Propionibacterium acnes/fisiología , Células Th17/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Línea Celular , Biología Computacional , Progresión de la Enfermedad , Citometría de Flujo , Humanos , Memoria Inmunológica , Interleucina-17/metabolismo , Lectinas Tipo C/metabolismo , Activación de Linfocitos , Piel/patología
4.
Acta Diabetol ; 58(8): 1035-1049, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33754166

RESUMEN

OBJECTIVE: The intestinal microbiota to immune system crosstalk is a major regulator of metabolism and hence metabolic diseases. An impairment of the chemokine receptor CX3CR1, as a key regulator shaping intestinal microbiota under normal chow feeding, could be one of the early events of dysglycemia. METHODS: We studied the gut microbiota ecology by sequencing the gut and tissue microbiota. We studied its role in energy metabolism in CX3CR1-deficent and control mice using various bioassays notably the glycemic regulation during fasting and the respiratory quotient as two highly sensitive physiological features. We used antibiotics and prebiotics treatments, and germ free mouse colonization. RESULTS: We identify that CX3CR1 disruption impairs gut microbiota ecology and identified a specific signature associated to the genotype. The glycemic control during fasting and the respiratory quotient throughout the day are deeply impaired. A selected four-week prebiotic treatment modifies the dysbiotic microbiota and improves the fasting state glycemic control of the CX3CR1-deficent mice and following a glucose tolerance test. A 4 week antibiotic treatment also improves the glycemic control as well. Eventually, germ free mice colonized with the microbiota from CX3CR1-deficent mice developed glucose intolerance. CONCLUSIONS: CX3CR1 is a molecular mechanism in the control of the gut microbiota ecology ensuring the maintenance of a steady glycemia and energy metabolism. Its impairment could be an early mechanism leading to gut microbiota dysbiosis and the onset of metabolic disease.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/fisiología , Diabetes Mellitus Tipo 2/microbiología , Microbioma Gastrointestinal/fisiología , Animales , Antibacterianos/administración & dosificación , Glucemia/fisiología , Receptor 1 de Quimiocinas CX3C/deficiencia , Disbiosis , Metabolismo Energético , Masculino , Ratones , Ratones Endogámicos C57BL , Prebióticos/administración & dosificación , Factores de Riesgo
5.
Clin Cosmet Investig Dermatol ; 11: 421-429, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30233225

RESUMEN

BACKGROUND: Rosacea is a chronic facial skin disorder characterized by inflammation and vascular abnormalities. The pathophysiology of rosacea involves increased activation of the capsaicin receptor, TRPV1, the vascular endothelial growth factor (VEGF) pathway, and cathelicidin LL-37, MMP-9, and KLKs. We evaluated the activity of four compounds (dextran sulfate, 4-t-butylcyclohexanol [BCH; TRP-regulin®], pongamia oil, and hesperidin methyl chalcone [HMC]) on inflammatory and vascular responses implicated in rosacea. MATERIALS AND METHODS: The anti-inflammatory activity of dextran sulfate was evaluated on PGE2 production after PMA stimulation of NCTC-2544 keratinocytes, and on normal human epidermal keratinocytes (NHEKs) after proinflammatory stimulation to mimic a rosacea environment. The anti-angiogenic activity of dextran sulfate was measured by analyzing pseudotube formation in co-cultured human microvascular endothelial cells/normal human dermal fibroblasts. HMC modulation of vascular responses and IL-8 cytokine production after SP stimulation was evaluated in human skin explants. We also assessed the effect of BCH on TRPV1 activation, and the effect of combined BCH and pongamia oil on the inflammatory response of NHEKs. RESULTS: Dextran sulfate strongly and significantly inhibited PMA-induced PGE2 production, inhibited KLK5 and MMP-9 mRNA expression, and IL-8, IL-1α and VEGF production, and displayed a highly significant inhibitory effect on VEGF-induced pseudotube formation. In SP-stimulated human skin explants, HMC significantly decreased the proportion of dilated vessels, total vessel area, and IL-8 production. BCH significantly and dose-dependently inhibited TRPV1 activation, and BCH and pongamia oil inhibited CXCL1 and CXCL6 mRNA expression and IL-8 production in NHEKs. Combined BCH/pongamia oil inhibited IL-8 production synergistically. CONCLUSION: These in vitro results showed that dextran sulfate, BCH, pongamia oil and HMC, possess complementary soothing and anti-redness properties, supporting their combination in Avène redness-relief cosmetic products for sensitive skin prone to redness, and for topical adjunctive rosacea treatment.

6.
Gut ; 66(5): 872-885, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-26838600

RESUMEN

OBJECTIVE: To identify a causal mechanism responsible for the enhancement of insulin resistance and hyperglycaemia following periodontitis in mice fed a fat-enriched diet. DESIGN: We set-up a unique animal model of periodontitis in C57Bl/6 female mice by infecting the periodontal tissue with specific and alive pathogens like Porphyromonas gingivalis (Pg), Fusobacterium nucleatum and Prevotella intermedia. The mice were then fed with a diabetogenic/non-obesogenic fat-enriched diet for up to 3 months. Alveolar bone loss, periodontal microbiota dysbiosis and features of glucose metabolism were quantified. Eventually, adoptive transfer of cervical (regional) and systemic immune cells was performed to demonstrate the causal role of the cervical immune system. RESULTS: Periodontitis induced a periodontal microbiota dysbiosis without mainly affecting gut microbiota. The disease concomitantly impacted on the regional and systemic immune response impairing glucose metabolism. The transfer of cervical lymph-node cells from infected mice to naive recipients guarded against periodontitis-aggravated metabolic disease. A treatment with inactivated Pg prior to the periodontal infection induced specific antibodies against Pg and protected the mouse from periodontitis-induced dysmetabolism. Finally, a 1-month subcutaneous chronic infusion of low rates of lipopolysaccharides from Pg mimicked the impact of periodontitis on immune and metabolic parameters. CONCLUSIONS: We identified that insulin resistance in the high-fat fed mouse is enhanced by pathogen-induced periodontitis. This is caused by an adaptive immune response specifically directed against pathogens and associated with a periodontal dysbiosis.


Asunto(s)
Inmunidad Adaptativa , Infecciones por Bacteroidaceae/complicaciones , Disbiosis/inmunología , Resistencia a la Insulina/inmunología , Periodontitis/inmunología , Periodontitis/prevención & control , Porphyromonas gingivalis , Animales , Trasplante de Células , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Disbiosis/microbiología , Disbiosis/prevención & control , Femenino , Encía/microbiología , Hiperglucemia/inmunología , Hiperglucemia/microbiología , Interferón gamma/sangre , Interleucina-6/sangre , Lipopolisacáridos/inmunología , Ganglios Linfáticos/citología , Linfocitos , Ratones , Ratones Endogámicos C57BL , Microbiota , Periodontitis/microbiología , Periodontitis/patología , Porphyromonas gingivalis/inmunología , Distribución Aleatoria , Bazo/citología , Vacunación
7.
Mol Metab ; 5(6): 392-403, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27257599

RESUMEN

OBJECTIVE: To demonstrate that glycemia and insulin resistance are controlled by a mechanism involving the adaptive immune system and gut microbiota crosstalk. METHODS: We triggered the immune system with microbial extracts specifically from the intestinal ileum contents of HFD-diabetic mice by the process of immunization. 35 days later, immunized mice were fed a HFD for up to two months in order to challenge the development of metabolic features. The immune responses were quantified. Eventually, adoptive transfer of immune cells from the microbiota-immunized mice to naïve mice was performed to demonstrate the causality of the microbiota-stimulated adaptive immune system on the development of metabolic disease. The gut microbiota of the immunized HFD-fed mice was characterized in order to demonstrate whether the manipulation of the microbiota to immune system interaction reverses the causal deleterious effect of gut microbiota dysbiosis on metabolic disease. RESULTS: Subcutaneous injection (immunization procedure) of ileum microbial extracts prevented hyperglycemia and insulin resistance in a dose-dependent manner in response to a HFD. The immunization enhanced the proliferation of CD4 and CD8 T cells in lymphoid organs, also increased cytokine production and antibody secretion. As a mechanism explaining the metabolic improvement, the immunization procedure reversed gut microbiota dysbiosis. Finally, adoptive transfer of immune cells from immunized mice improved metabolic features in response to HFD. CONCLUSIONS: Glycemia and insulin sensitivity can be regulated by triggering the adaptive immunity to microbiota interaction. This reduces the gut microbiota dysbiosis induced by a fat-enriched diet.

8.
Cell Metab ; 22(1): 100-12, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26154056

RESUMEN

A high-fat diet (HFD) induces metabolic disease and low-grade metabolic inflammation in response to changes in the intestinal microbiota through as-yet-unknown mechanisms. Here, we show that a HFD-derived ileum microbiota is responsible for a decrease in Th17 cells of the lamina propria in axenic colonized mice. The HFD also changed the expression profiles of intestinal antigen-presenting cells and their ability to generate Th17 cells in vitro. Consistent with these data, the metabolic phenotype was mimicked in RORγt-deficient mice, which lack IL17 and IL22 function, and in the adoptive transfer experiment of T cells from RORγt-deficient mice into Rag1-deficient mice. We conclude that the microbiota of the ileum regulates Th17 cell homeostasis in the small intestine and determines the outcome of metabolic disease.


Asunto(s)
Antígenos CD4/inmunología , Linfocitos T CD4-Positivos/microbiología , Diabetes Mellitus Tipo 2/microbiología , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Obesidad/microbiología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/inmunología , Eliminación de Gen , Regulación de la Expresión Génica , Íleon/inmunología , Íleon/metabolismo , Íleon/microbiología , Inmunidad , Interleucina-17/genética , Interleucina-17/inmunología , Masculino , Ratones , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Obesidad/etiología , Obesidad/genética , Obesidad/inmunología , Células Th17/inmunología , Células Th17/metabolismo , Células Th17/microbiología
9.
EMBO Mol Med ; 7(3): 259-74, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25666722

RESUMEN

Pattern recognition receptors link metabolite and bacteria-derived inflammation to insulin resistance during obesity. We demonstrate that NOD2 detection of bacterial cell wall peptidoglycan (PGN) regulates metabolic inflammation and insulin sensitivity. An obesity-promoting high-fat diet (HFD) increased NOD2 in hepatocytes and adipocytes, and NOD2(-/-) mice have increased adipose tissue and liver inflammation and exacerbated insulin resistance during a HFD. This effect is independent of altered adiposity or NOD2 in hematopoietic-derived immune cells. Instead, increased metabolic inflammation and insulin resistance in NOD2(-/-) mice is associated with increased commensal bacterial translocation from the gut into adipose tissue and liver. An intact PGN-NOD2 sensing system regulated gut mucosal bacterial colonization and a metabolic tissue dysbiosis that is a potential trigger for increased metabolic inflammation and insulin resistance. Gut dysbiosis in HFD-fed NOD2(-/-) mice is an independent and transmissible factor that contributes to metabolic inflammation and insulin resistance when transferred to WT, germ-free mice. These findings warrant scrutiny of bacterial component detection, dysbiosis, and protective immune responses in the links between inflammatory gut and metabolic diseases, including diabetes.


Asunto(s)
Bacterias/inmunología , Dieta/métodos , Disbiosis , Inflamación/patología , Resistencia a la Insulina , Proteína Adaptadora de Señalización NOD2/metabolismo , Peptidoglicano/metabolismo , Animales , Pared Celular/química , Ratones , Ratones Noqueados , Peptidoglicano/análisis
10.
Mol Metab ; 2(3): 281-91, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24049740

RESUMEN

Metabolic endotoxemia triggers inflammation, targets cells from the stroma-vascular fraction of adipose depots, and metabolic disease. To identify these cells we here infused mice with lipopolysaccharides and showed by FACS analyses and BrdU staining that the number of small subcutaneous adipocytes, preadipocytes and macrophages increased in wild type but not in CD14-knockout (KO) mice. This mechanism was direct since in CD14KO mice grafted subcutaneously and simultaneously with fat pads from CD14KO and wild-type mice the concentration of cytokine mRNA was increased in the wild-type fat pad only. Conversely, the mRNA concentration of genes involved in glucose and lipid metabolism and the number of large adipocytes was reduced. Eventually, a pretreatment with LPS enhanced HFD-induced metabolic diseases. Altogether, these results show that metabolic endotoxemia increases the proliferation of preadipocytes through a CD14-dependent mechanism directly, without recruiting CD14-positive cells from non-adipose depot origin. This mechanism could precede the onset of metabolic diseases.

11.
Diabetes Obes Metab ; 15 Suppl 3: 61-70, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24003922

RESUMEN

Over the last decade, the research community has revealed the role of a new organ: the intestinal microbiota. It is considered as a symbiont that is part of our organism since, at birth, it educates the immune system and contributes to the development of the intestinal vasculature and most probably the nervous system. With the advent of new generation sequencing techniques, a catalogue of genes that belong to this microbiome has been established that lists more than 5 million non-redundant genes called the metagenome. Using germ free mice colonized with the microbiota from different origins, it has been formally demonstrated that the intestinal microbiota causes the onset of metabolic diseases. Further to the role of point mutations in our genome, the microbiota can explain the on-going worldwide pandemic of obesity and diabetes, its dissemination and family inheritance, as well as the diversity of the associated metabolic phenotypes. More recently, the discovery of bacterial DNA within host tissues, such as the liver, the adipose tissue and the blood, which establishes a tissue microbiota, introduces new opportunities to identify targets and predictive biomarkers based on the host to microbiota interaction, as well as to define new strategies for pharmacological, immunomodulatory vaccines and nutritional applications.


Asunto(s)
Metabolismo/fisiología , Metagenoma/fisiología , Microbiota/fisiología , Animales , Comunicación Celular/fisiología , Especificidad del Huésped/inmunología , Humanos , Intestinos/inmunología , Intestinos/microbiología , Enfermedades Metabólicas/microbiología , Ratones
12.
Stem Cells ; 31(7): 1309-20, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23533182

RESUMEN

Adipose tissue (AT) has become accepted as a source of multipotent progenitor cells, the adipose stromal cells (ASCs). In this regard, considerable work has been performed to harvest and characterize this cell population as well as to investigate the mechanisms by which transplanted ASCs mediate tissue regeneration. In contrast the endogenous release of native ASCs by AT has been poorly investigated. In this work, we show that native ASCs egress from murine AT. Indeed, we demonstrated that the release of native ASCs from AT can be evidenced both using an ex vivo perfusion model that we set up and in vivo. Such a mobilization process is controlled by CXCR4 chemokine receptor. In addition, once mobilized from AT, circulating ASCs were found to navigate through lymph fluid and to home into lymph nodes (LN). Therefore, we demonstrated that, during the LN activation, the fat depot encapsulating the activated LN releases native ASCs, which in turn invade the activated LN. Moreover, the ASCs invading the LN were visualized in close physical interaction with podoplanin and ER-TR7 positive structures corresponding to the stromal network composing the LN. This dynamic was impaired with CXCR4 neutralizing antibody. Taken together, these data provide robust evidences that native ASCs can traffic in vivo and that AT might provide stromal cells to activated LNs.


Asunto(s)
Tejido Adiposo/citología , Ganglios Linfáticos/citología , Células Madre Mesenquimatosas/citología , Tejido Adiposo/metabolismo , Animales , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Quimiocina CXCL12/metabolismo , Inmunofenotipificación , Ganglios Linfáticos/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores CXCR4/metabolismo
13.
J Virol ; 86(13): 7060-71, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22553324

RESUMEN

Persistent viral infections often overburden the immune system and are a major cause of disease in humans. During many persistent infections, antiviral T cells are maintained in a state of immune exhaustion characterized by diminished effector and helper functions. In mammalian systems, an extensive immune regulatory network exists to limit unwanted, potentially fatal immunopathology by inducing T cell exhaustion. However, this regulatory network at times overprotects the host and fosters viral persistence by severely dampening adaptive immune responsiveness. Importantly, recent studies have shown that T cell exhaustion is mediated in part by host immunoregulatory pathways (e.g., programmed death 1 [PD-1], interleukin 10 [IL-10]) and that therapeutic blockade of these pathways either before or during persistent infection can promote viral clearance. Transforming growth factor beta (TGF-ß) is another immunosuppressive cytokine known to impede both self- and tumor-specific T cells, but its role in regulating antiviral immunity is not entirely understood. In this study, we inhibited TGF-ß with three potent antagonists to determine whether neutralization of this regulatory molecule is a viable approach to control a persistent viral infection. Our results revealed that these inhibitors modestly elevate the number of antiviral T cells following infection with a persistent variant of lymphocytic choriomeningitis virus (LCMV) but have no impact on viral clearance. These data suggest that therapeutic neutralization of TGF-ß is not an efficacious means to promote clearance of a persistent viral infection.


Asunto(s)
Infecciones por Arenaviridae/tratamiento farmacológico , Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Factores Inmunológicos/administración & dosificación , Virus de la Coriomeningitis Linfocítica/inmunología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Infecciones por Arenaviridae/virología , Linfocitos T CD4-Positivos/inmunología , Enfermedad Crónica , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Resultado del Tratamiento
14.
Semin Immunol ; 24(1): 67-74, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22265028

RESUMEN

Over the last decades the rising occurrence of metabolic diseases throughout the world points to the failure of preventive and therapeutic strategies and of the corresponding molecular and physiological concepts. Therefore, a new paradigm needs to be elucidated. Very recently the intimate cross talk of the intestinal microbiota with the host immune system has opened new avenues. The large diversity of the intestinal microbes' genome, i.e. the metagenome, and the extreme plasticity of the immune system provide a unique balance which, when finely tuned, maintains a steady homeostasis. The discovery that a new microbiota repertoire is one of the causes responsible for the onset of metabolic disease suggests that the relationship with the immune system is impaired. Therefore, we here review the recent arguments that support the view that an alteration in the microbiota to host immune system balance leads to an increased translocation of bacterial antigens towards metabolically active tissues, and could result in a chronic inflammatory state and consequently impaired metabolic functions such as insulin resistance, hepatic fat deposition, insulin unresponsiveness, and excessive adipose tissue development. This imbalance could be at the onset of metabolic disease, and therefore the early treatment of the microbiota dysbiosis or immunomodulatory strategies should prevent and slow down the epidemic of metabolic diseases and hence the corresponding lethal cardiovascular consequences.


Asunto(s)
Síndrome Metabólico/inmunología , Síndrome Metabólico/microbiología , Metagenoma , Inmunidad Adaptativa , Animales , Dieta Alta en Grasa , Humanos , Inmunidad Innata , Intestinos/inmunología , Intestinos/microbiología , Síndrome Metabólico/genética
15.
J Immunol ; 183(6): 3895-905, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19717517

RESUMEN

Persistence of even the stealthiest viruses can perturb immune function either to the benefit or detriment of the host. Lymphocytic choriomeningitis virus (LCMV) establishes lifelong, systemic persistence when introduced in utero or at birth. Despite a highly evolved host-pathogen relationship, LCMV cannot escape detection by the innate immune system, which results in chronic stimulation of the type 1 IFN pathway in adult carrier mice. In this study we demonstrate that IFN-beta is chronically up-regulated in peripheral lymphoid and nonlymphoid tissues (but not the CNS) of mice persistently infected from birth with LCMV and that dendritic cells (DCs) represent at least one source of IFN-beta. Interestingly, chronic stimulation of this innate pathway significantly elevated MHC class I expression in the CNS as well as the periphery. Elevated MHC I expression was dependent on IFN-alphabeta receptor but not MyD88-dependent signaling, as only genetic deletion of the former reduced MHC I to normal levels. An increase in circulating virus was also observed in the IFN-alphabeta receptor deficient carrier mice, signifying that type I IFN continually exerts anti-viral pressure during a LCMV carrier state. Finally, to determine whether heightened CNS MHC I could be therapeutically corrected, we purged LCMV carrier mice of their persistent infection using adoptive immunotherapy. This treatment significantly reduced CNS MHC I expression. Collectively, these data demonstrate that even a well adapted pathogen can chronically stimulate the innate immune system and consequently alter the expression of Ag presenting machinery in an immunologically specialized compartment like the CNS.


Asunto(s)
Sistema Nervioso Central/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Interferón beta/genética , Receptor de Interferón alfa y beta/metabolismo , Virosis/inmunología , Animales , Enfermedad Crónica , Inmunoterapia , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Receptor de Interferón alfa y beta/inmunología , Regulación hacia Arriba , Virosis/terapia
16.
J Virol ; 83(17): 8905-15, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19553326

RESUMEN

Persistent viral infections are a major health concern worldwide. During persistent infection, overwhelming viral replication and the rapid loss of antiviral T-cell function can prevent immune-mediated clearance of the infection, and therapies to reanimate the immune response and purge persistent viruses have been largely unsuccessful. Adoptive immunotherapy using memory T cells is a highly successful therapeutic approach to eradicate a persistent viral infection. Understanding precisely how therapeutically administered memory T cells achieve clearance should improve our ability to terminate states of viral persistence in humans. Mice persistently infected from birth with lymphocytic choriomeningitis virus are tolerant to the pathogen at the T-cell level and thus provide an excellent model to evaluate immunotherapeutic regimens. Previously, we demonstrated that adoptively transferred memory T cells require recipient dendritic cells to effectively purge an established persistent viral infection. However, the mechanisms that reactivate and sustain memory T-cell responses during clearance of such an infection remain unclear. Here we establish that therapeutic memory T cells require CD80 and CD86 costimulatory signals to efficiently clear an established persistent viral infection in vivo. Early blockade of costimulatory pathways with CTLA-4-Fc decreased the secondary expansion of virus-specific CD8(+) and CD4(+) memory T cells as well as their ability to produce antiviral cytokines and purge the persistent infection. Late costimulation blockade also reduced virus-specific T-cell numbers, illustrating that sustained interactions with costimulatory molecules is required for efficient T-cell expansion. These findings indicate that antiviral memory T cells require costimulation to efficiently clear a persistent viral infection and that costimulatory pathways can be targeted to modulate the magnitude of an adoptive immunotherapeutic regimen.


Asunto(s)
Traslado Adoptivo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunoterapia/métodos , Coriomeningitis Linfocítica/terapia , Virus de la Coriomeningitis Linfocítica/inmunología , Animales , Antígeno B7-1/inmunología , Antígeno B7-2/inmunología , Activación de Linfocitos/inmunología , Ratones
17.
Proc Natl Acad Sci U S A ; 106(9): 3473-8, 2009 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-19208813

RESUMEN

Several arenaviruses cause hemorrhagic fever disease in humans for which no licensed vaccines are available and current therapeutic intervention is limited to the off-label use of the wide-spectrum antiviral ribavirin. However, the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) has proven to be a Rosetta stone for the investigation of virus-host interactions. Arenaviruses have a bisegmented negative-strand RNA genome. The S segment encodes for the virus nucleoprotein and glycoprotein, whereas the L segment encodes for the virus polymerase (L) and Z protein. The ability to generate recombinant LCMV (rLCMV) expressing additional foreign genes of interest would open novel avenues for the study of virus-host interactions and the development of novel vaccine strategies and high-throughput screens to identify antiarenaviral molecules. To this end, we have developed a trisegmented (1L + 2S) rLCMV-based approach (r3LCMV). Each of the two S segments in r3LCMV was altered to replace one of the viral genes by a gene of interest. All r3LCMVs examined expressing different reported genes were stable both genetically and phenotypically and exhibited wild-type growth properties in cultured cells. Reporter gene expression in r3LCMV-infected cells provided an accurate surrogate of levels of virus multiplication. Notably, some r3LCMVs displayed highly attenuated virulence in mice but induced protective immunity against a subsequent lethal challenge with wild-type LCMV, supporting the potential development of r3LCMV-based vaccines.


Asunto(s)
Regulación Viral de la Expresión Génica/genética , Genoma Viral/genética , Virus de la Coriomeningitis Linfocítica/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Modelos Animales de Enfermedad , Expresión Génica , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/genética , Ratones , Fenotipo , Virión/genética , Virión/metabolismo , Replicación Viral
18.
Blood ; 108(7): 2257-64, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16449531

RESUMEN

Accumulating evidence indicates that, in absence of CD8+ T-cell activation, CD4+ T-cell-mediated allograft rejection is associated with a dominant Th2-cell response and eosinophil infiltrates. In this study, we analyzed the mechanisms by which CD8+ T cells regulate alloreactive CD4+ T-cell priming and differentiation into interleukin 4 (IL-4)-producing cells. We showed that interferon gamma (IFN-gamma) production by CD8+ T cells was dispensable for the inhibition of Th2-cell development, as well as tissue eosinophilia and type 2 cytokine production in the rejected grafts. Since we noticed that CD8+ T cells not only suppressed Th2 differentiation, but also down-modulated the overall priming of alloreactive CD4+ T cells, we evaluated whether CD8+ T cells act by limiting the accumulation of donor-derived dendritic cells (DCs) in lymph nodes. We found that indeed, alloreactive CD8+ T cells rapidly eliminated allogeneic DCs from T-cell areas of draining lymph nodes, through a perforin-dependent mechanism. Thus, our data demonstrate that cytotoxic T lymphocyte (CTL)-mediated clearance of allogeneic DCs is a negative feedback mechanism that limits the duration of alloantigen presentation in draining lymph nodes, thereby modulating the amplitude and polarization of the primary alloreactive CD4+ T-cell responses.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/citología , Animales , Células de la Médula Ósea/citología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/fisiología , Movimiento Celular , Trasplante de Células , Regulación hacia Abajo , Eosinofilia/metabolismo , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Metástasis Linfática , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Th2
19.
J Immunol ; 175(8): 5050-7, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16210608

RESUMEN

Myasthenia gravis (MG) is an organ-specific autoimmune disease caused in most cases by autoantibodies against the nicotinic acetylcholine receptor (AChR). It is now well documented that many autoimmune diseases, including MG, are more prevalent in women than in men, and that fluctuations in disease severity occur during pregnancy. These observations raise the question of the potential role of sex hormones, such as estrogens, as mediators of sex differences in autoimmunity. In the present study, we have analyzed the effect of 17beta-estradiol (E2) on the pathogenesis of experimental autoimmune myasthenia gravis (EAMG), an animal model of MG. We show that treatment with E2 before Ag priming is necessary and sufficient to promote AChR-specific Th1 cell expansion in vivo. This time-limited exposure to E2 enhances the production of anti-AChR IgG2a(b) (specific for b allotype; e.g., B6) and IgG2b, but not IgG1, and significantly increases the severity of EAMG in mice. Interestingly, the E2-mediated augmentation in AChR-specific Th1 response correlates with an enhanced production of IL-12 by splenic APCs through the recruitment of CD8alpha(+) dendritic cells. These data provide the first evidence that estrogen enhances EAMG, and sheds some light on the role of sex hormones in immune responses and susceptibility to autoimmune disease in women.


Asunto(s)
Estradiol/fisiología , Miastenia Gravis Autoinmune Experimental/inmunología , Células TH1/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Proliferación Celular , Células Cultivadas , Susceptibilidad a Enfermedades/inducido químicamente , Susceptibilidad a Enfermedades/inmunología , Estradiol/administración & dosificación , Femenino , Interleucina-12/biosíntesis , Ratones , Ratones Endogámicos C57BL , Miastenia Gravis Autoinmune Experimental/fisiopatología , Receptores Colinérgicos/inmunología , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Células TH1/metabolismo
20.
Eur J Immunol ; 35(8): 2284-94, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16021601

RESUMEN

We have investigated the density of peptides required to elicit different biological responses in cytotoxic T lymphocytes (CTL), including trogocytosis (i.e., the phenomenon whereby the lymphocytes actively capture fragments of plasma membrane from those cells with which they establish an immune synapse). We have used two separate mouse models of CTL recognising defined peptides presented by MHC class I molecules. In both systems, triggering of cytotoxicity and capture of membrane components reached saturation with low densities of ligand. On the other hand, down-modulation of cell-surface levels of TCR, induction of IFN-gamma production and detection of peptide captured required much higher ligand densities. Interestingly, fratricide (i.e., killing between CTL sharing the same specificity), a mechanism proposed to account for CTL exhaustion, was detected only at antigen concentrations still well above that second threshold leading to full blown activation. Taken together, our results show that the different thresholds that govern the elicitation of different CTL functions correlate with different proportions of antigen among the target cell components being captured via trogocytosis.


Asunto(s)
Antígenos/biosíntesis , Activación de Linfocitos/inmunología , Células Plasmáticas/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Biotina/metabolismo , Línea Celular Tumoral , Células Cultivadas , Citocinas/metabolismo , Antígenos de Histocompatibilidad/inmunología , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/inmunología , Células Plasmáticas/metabolismo , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/biosíntesis , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Citotóxicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...