Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Immunity ; 43(2): 343-53, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26287682

RESUMEN

Activated retina-specific T cells that have acquired the ability to break through the blood-retinal barrier are thought to be causally involved in autoimmune uveitis, a major cause of human blindness. It is unclear where these autoreactive T cells first become activated, given that their cognate antigens are sequestered within the immune-privileged eye. We demonstrate in a novel mouse model of spontaneous uveitis that activation of retina-specific T cells is dependent on gut commensal microbiota. Retina-specific T cell activation involved signaling through the autoreactive T cell receptor (TCR) in response to non-cognate antigen in the intestine and was independent of the endogenous retinal autoantigen. Our findings not only have implications for the etiology of human uveitis, but also raise the possibility that activation of autoreactive TCRs by commensal microbes might be a more common trigger of autoimmune diseases than is currently appreciated.


Asunto(s)
Intestinos/inmunología , Microbiota/inmunología , Retina/inmunología , Linfocitos T/inmunología , Uveítis/inmunología , Animales , Antígenos Bacterianos/administración & dosificación , Autoantígenos/inmunología , Autoinmunidad , Barrera Hematorretinal/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Proteínas del Ojo/inmunología , Proteínas del Ojo/metabolismo , Tolerancia Inmunológica , Intestinos/microbiología , Activación de Linfocitos , Ratones Endogámicos , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas de Unión al Retinol/genética , Proteínas de Unión al Retinol/inmunología , Proteínas de Unión al Retinol/metabolismo , Uveítis/microbiología
2.
J Autoimmun ; 102: 65-76, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31080013

RESUMEN

IL-22 has opposing effects in different tissues, from pro-inflammatory (skin, joints) to protective (liver, intestine) but little is known about its effects on neuroinflammation. We examined the effect of IL-22 on retinal tissue by using the model of experimental autoimmune uveitis (EAU) in IL-22-/- mice, as well as by intraocular injections of recombinant IL-22 or anti-IL-22 antibodies in wild type animals. During EAU, IL-22 was produced in the eye by CD4+ eye-infiltrating T cells. EAU-challenged IL-22-/- mice, as well as WT mice treated systemically or intraocularly with anti-IL-22 antibodies during the expression phase of disease, developed exacerbated retinal damage. Furthermore, IL-22-/- mice were more susceptible than WT controls to glutamate-induced neurotoxicity, whereas local IL-22 supplementation was protective, suggesting direct or indirect neuroprotective effects. Mechanistic studies revealed that retinal glial Müller cells express IL-22rα1 in vivo, and in vitro IL-22 enhanced their ability to suppress proliferation of effector T cells. Finally, IL-22 injected into the eye concurrently with IL-1, inhibited the (IL-1-induced) expression of multiple proinflammatory and proapoptotic genes in retinal tissue. These findings suggest that IL-22 can function locally within the retina to reduce inflammatory damage and provide neuroprotection by affecting multiple molecular and cellular pathways.


Asunto(s)
Autoinmunidad , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Susceptibilidad a Enfermedades , Interleucinas/metabolismo , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Autoinmunidad/genética , Sistema Nervioso Central/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Células Ependimogliales/inmunología , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Interleucinas/genética , Interleucinas/farmacología , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Neuroprotección/genética , Índice de Severidad de la Enfermedad , Linfocitos T/inmunología , Linfocitos T/metabolismo , Uveítis/etiología , Uveítis/metabolismo , Uveítis/patología , Interleucina-22
3.
J Antimicrob Chemother ; 70(6): 1774-83, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25687643

RESUMEN

OBJECTIVES: Although TB immunotherapy improves the results of conventional drug treatment, the effects of combining chemotherapy and immunotherapy have never been systematically evaluated. We used a comprehensive lung transcriptome analysis to directly compare the activity of combined chemotherapy and immunotherapy with that of single treatments in a mouse model of TB. METHODS: Mycobacterium tuberculosis-infected mice in the chronic phase of the disease (day 30) received: (i) isoniazid and rifampicin (drugs) daily for 30 days; (ii) DNA immunotherapy (DNA), consisting of four 100 µg injections at 10 day intervals; (iii) both therapies (DNA + drugs); or (iv) saline. The effects were evaluated 10 days after the end of treatment (day 70 post-infection). RESULTS: In all groups a systemic reduction in the load of bacilli was observed, bacilli became undetectable in the drugs and DNA + drugs groups, but the whole lung transcriptome analysis showed 867 genes exclusively modulated by the DNA + drugs combination. Gene enrichment analysis indicated that DNA + drugs treatment provided synergistic effects, including the down-regulation of proinflammatory cytokines and mediators of fibrosis, as confirmed by real-time PCR, ELISA, histopathology and hydroxyproline assay. CONCLUSIONS: Our results provide a molecular basis for the advantages of TB treatment using combined chemotherapy and DNA immunotherapy and demonstrate the synergistic effects obtained with this strategy.


Asunto(s)
Terapia Combinada/métodos , Quimioterapia/métodos , Inmunoterapia/métodos , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/inmunología , Tuberculosis/terapia , Animales , Antituberculosos/administración & dosificación , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Isoniazida/administración & dosificación , Pulmón/microbiología , Pulmón/patología , Ratones Endogámicos BALB C , Rifampin/administración & dosificación , Resultado del Tratamiento , Vacunas de ADN/administración & dosificación
4.
J Immunol ; 188(4): 1742-50, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22238462

RESUMEN

Immune privilege is used by the eye, brain, reproductive organs, and gut to preserve structural and functional integrity in the face of inflammation. The eye is arguably the most vulnerable and, therefore, also the most "privileged" of tissues; paradoxically, it remains subject to destructive autoimmunity. It has been proposed, although never proven in vivo, that the eye can induce T regulatory cells (Tregs) locally. Using Foxp3-GFP reporter mice expressing a retina-specific TCR, we now show that uncommitted T cells rapidly convert in the living eye to Foxp3(+) Tregs in a process involving retinal Ag recognition, de novo Foxp3 induction, and proliferation. This takes place within the ocular tissue and is supported by retinoic acid, which is normally present in the eye because of its function in the chemistry of vision. Nonconverted T cells showed evidence of priming but appeared restricted from expressing effector function in the eye. Pre-existing ocular inflammation impeded conversion of uncommitted T cells into Tregs. Importantly, retina-specific T cells primed in vivo before introduction into the eye were resistant to Treg conversion in the ocular environment and, instead, caused severe uveitis. Thus, uncommitted T cells can be disarmed, but immune privilege is unable to protect from uveitogenic T cells that have acquired effector function prior to entering the eye. These findings shed new light on the phenomenon of immune privilege and on its role, as well as its limitations, in actively controlling immune responses in the tissue.


Asunto(s)
Autoinmunidad , Ojo/inmunología , Factores de Transcripción Forkhead/análisis , Linfocitos T Reguladores/inmunología , Linfocitos T/inmunología , Tretinoina/metabolismo , Uveítis/inmunología , Animales , Diferenciación Celular , Proliferación Celular , Proteínas del Ojo/inmunología , Factores de Transcripción Forkhead/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas de Unión al Retinol/inmunología
5.
Cells ; 11(15)2022 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-35892575

RESUMEN

Childhood malnutrition affects physiology and development. It increases infection rates, which may not present clinical signs in severe cases. The World Health Organization recommends prophylactic treatment with cotrimoxazole (SXT) and nutritional recovery to overcome this issue. This treatment is controversial, since evidence of a reduction in morbidity and mortality is not a consensus and could induce the development of antibiotic-resistant bacteria. Moreover, the impact of using this wide-spectrum antibiotic on gut microbiota in a critical period of development, and weakness is unknown. To understand how SXT prophylaxis could affect gut microbiota in undernutrition, we induced protein-energy undernutrition (PEU) in weaning C57BL/6 mice for three weeks and treated animals with SXT for two weeks. Using 16S rRNA gene sequencing, we compared the taxonomic composition and metabolic pathways of control mice, animals submitted to undernutrition (UND), treated with SXT, or undernourished and SXT treated (UND + SXT). We identified that UND mice had a significant increase in predicted pathways related to metabolic syndromes later in life. The prophylactic SXT treatment alone resulted in a significant loss in community richness and beta diversity. Furthermore, we identified the reduction of three genera in SXT treated mice, including the butyrate producers Faecalibacterium and Anaerotruncus. Both UND and double challenge (UND + SXT) resulted in a reduction of the amino acid's biosynthesis pathway related to cell growth. Our results show that the SXT prophylaxis of young mice during an undernourishment period did not re-establish the undernourished microbiota community composition similar to healthy controls but induced a distinct dysbiotic profile with functional metabolic consequences.


Asunto(s)
Disbiosis , Desnutrición , Animales , Antibacterianos , Disbiosis/microbiología , Desnutrición/complicaciones , Desnutrición/tratamiento farmacológico , Desnutrición/microbiología , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética , Combinación Trimetoprim y Sulfametoxazol
6.
World J Gastroenterol ; 28(33): 4762-4772, 2022 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-36156924

RESUMEN

Fecal microbiota transplantation (FMT) is a successful method for treating recurrent Clostridioides difficile (C. difficile) infection (rCDI) with around 90% efficacy. Due to the relative simplicity of this approach, it is being widely used and currently, thousands of patients have been treated with FMT worldwide. Nonetheless, the mechanisms underlying its effects are just beginning to be understood. Data indicate that FMT effectiveness is due to a combination of microbiological direct mechanisms against C. difficile, but also through indirect mechanisms including the production of microbiota-derived metabolites as secondary bile acids and short chain fatty acids. Moreover, the modulation of the strong inflammatory response triggered by C. difficile after FMT seems to rely on a pivotal role of regulatory T cells, which would be responsible for the reduction of several cells and soluble inflammatory mediators, ensuing normalization of the intestinal mucosal immune system. In this minireview, we analyze recent advances in these immunological aspects associated with the efficacy of FMT.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium , Ácidos y Sales Biliares , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/terapia , Ácidos Grasos Volátiles , Trasplante de Microbiota Fecal/efectos adversos , Trasplante de Microbiota Fecal/métodos , Heces/microbiología , Humanos , Mediadores de Inflamación , Recurrencia Local de Neoplasia , Recurrencia , Resultado del Tratamiento
7.
Cells ; 10(2)2021 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-33668457

RESUMEN

The normal composition of the intestinal microbiota is a key factor for maintaining healthy homeostasis, and accordingly, dysbiosis is well known to be present in HIV-1 patients. This article investigates the gut microbiota profile of antiretroviral therapy-naive HIV-1 patients and healthy donors living in Latin America in a cohort of 13 HIV positive patients (six elite controllers, EC, and seven non-controllers, NC) and nine healthy donors (HD). Microbiota compositions in stool samples were determined by sequencing the V3-V4 region of the bacterial 16S rRNA, and functional prediction was inferred using PICRUSt. Several taxa were enriched in EC compared to NC or HD groups, including Acidaminococcus, Clostridium methylpentosum, Barnesiella, Eubacterium coprostanoligenes, and Lachnospiraceae UCG-004. In addition, our data indicate that the route of infection is an important factor associated with changes in gut microbiome composition, and we extend these results by identifying several metabolic pathways associated with each route of infection. Importantly, we observed several bacterial taxa that might be associated with different viral subtypes, such as Succinivibrio, which were more abundant in patients infected by HIV subtype B, and Streptococcus enrichment in patients infected by subtype C. In conclusion, our data brings a significant contribution to the understanding of dysbiosis-associated changes in HIV infection and describes, for the first time, differences in microbiota composition according to HIV subtypes. These results warrant further confirmation in a larger cohort of patients.


Asunto(s)
Microbioma Gastrointestinal , Infecciones por VIH/metabolismo , Infecciones por VIH/microbiología , Redes y Vías Metabólicas , Adulto , Bacterias/clasificación , Análisis Discriminante , Heces/microbiología , Femenino , Infecciones por VIH/epidemiología , Infecciones por VIH/transmisión , VIH-1/fisiología , Humanos , Persona de Mediana Edad
8.
Exp Biol Med (Maywood) ; 234(7): 802-12, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19429853

RESUMEN

Since circulating leukocytes, mainly B and T cells, continuously maintain vigilant and comprehensive immune surveillance, these cells could be used as reporters for signs of infection or other pathologies, including cancer. Activated lymphocyte clones trigger a sensitive transcriptional response, which could be identified by gene expression profiling. To assess this hypothesis, we conducted microarray analysis of the gene expression profile of lymphocytes isolated from immunocompetent BALB/c mice subcutaneously injected with different numbers of tumorigenic B61 fibrosarcoma cells. Flow cytometry demonstrated that the number of circulating T (CD3(+)CD4(+) or CD3(+)CD8(+)) or B (CD19(+)) cells did not change. However, the lymphocytes isolated from tumor cell-injected animals expressed a unique transcriptional profile that was identifiable before the development of a palpable tumor mass. This finding demonstrates that the transcriptional response appears before alterations in the main lymphocyte subsets and that the gene expression profile of peripheral lymphocytes can serve as a sensitive and accurate method for the early detection of cancer.


Asunto(s)
Fibrosarcoma/diagnóstico , Fibrosarcoma/patología , Perfilación de la Expresión Génica , Hibridación Genética/fisiología , Linfocitos/metabolismo , Linfocitos/patología , Modelos Biológicos , Transcripción Genética/fisiología , Adenosina Desaminasa/metabolismo , Animales , Antígenos CD19/metabolismo , Linfocitos B/metabolismo , Linfocitos B/patología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Ratones , Ratones Endogámicos BALB C , Poli(ADP-Ribosa) Polimerasas/metabolismo , Sensibilidad y Especificidad
9.
Gut Microbes ; 10(5): 599-614, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30657007

RESUMEN

HIV-exposed but uninfected (HEU) children represent a growing population and show a significantly higher number of infectious diseases, several immune alterations, compromised growth, and increased mortality rates when compared to HIV-unexposed children. Considering the impact that the gut microbiota has on general host homeostasis and immune system development and modulation, we hypothesized that HEU children present altered gut microbiota that is linked to the increased morbidity and the immune system disorders faced by them. Our experiments revealed no differences in beta and alpha diversity of the gut microbiota between HEU and unexposed children or between HIV-infected and uninfected mothers. However, there were differences in the abundance of several taxa from the gut microbiota between HEU and unexposed children and between HIV-infected and uninfected mothers. Functional prediction based on 16S rRNA sequences also indicated differences between HEU and unexposed children and between infected and uninfected mothers. In addition, we detected no differences between HEU and unexposed children in relation to weight, weight-for-age z scores, albumin serum levels, or microbial translocation and inflammation markers. In summary, HIV-infected mothers and their HIV-exposed children present alterations in the abundance of several taxa in the gut microbiome and the predicted functional metagenome when compared to uninfected mothers and unexposed children. Knowledge about the gut microbiome of HEU children in different settings is essential in order to determine better treatments for this susceptible population.


Asunto(s)
Microbioma Gastrointestinal , Infecciones por VIH , Complicaciones Infecciosas del Embarazo , Efectos Tardíos de la Exposición Prenatal/microbiología , Adulto , Bacterias/clasificación , Bacterias/genética , Bacterias/crecimiento & desarrollo , Bacterias/aislamiento & purificación , Niño , Femenino , Microbioma Gastrointestinal/genética , Infecciones por VIH/microbiología , Humanos , Metagenoma , Madres , Embarazo , Complicaciones Infecciosas del Embarazo/microbiología , ARN Ribosómico 16S/genética , Adulto Joven
10.
Behav Brain Res ; 370: 111966, 2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31125622

RESUMEN

The Spontaneously Hypertensive Rat (SHR) has been proposed as a good model to study the pathways related to neurodegenerative diseases and glucose intolerance. Our research group developed the SLA16 (SHR.LEW-Anxrr16) congenic strain, which is genetically identical to the SHR strain, except for a locus on chromosome 4 (DGR). We applied in silico analysis on DGR to evaluate the association of their genes with neurobiological and metabolic pathways. After, we characterized cholesterol, triglycerides, metabolism of glucose and the behavioral performance of young (2 months old) and adult (8 months old) SHR and SLA16 rats in the open field, object location and water maze tasks. Finally, naïve young rats were repeatedly treated with metformin (200 mg/kg; v.o.) and evaluated in the same tests. Bioinformatics analysis showed that DGR presents genes related to glucose metabolism, oxidative damage and neurodegenerative diseases. Young SLA16 presented higher cholesterol, triglycerides, glucose and locomotion in the open field than SHR rats. In adulthood, SLA16 rats presented high triglycerides and locomotion in the open field and impairment on spatial learning and memory. Finally, the treatment with metformin decreased the glucose tolerance curve and also improved long-term memory in SLA16 rats. These results indicate that DGR presents genes associated with metabolic pathways and neurobiological processes that may produce alterations in glucose metabolism and spatial learning/memory. Therefore, we suggest that SHR and SLA16 strains could be important for the study of genes and subsequent mechanisms that produce metabolic glucose alterations and age-related cognitive deficits.


Asunto(s)
Ratas Endogámicas SHR/genética , Memoria Espacial/fisiología , Animales , Conducta Animal , Cromosomas Humanos Par 4/genética , Cromosomas de los Mamíferos/genética , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Genoma/genética , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Masculino , Enfermedades Metabólicas/genética , Ratas/genética
11.
Nat Commun ; 10(1): 3273, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-31332169

RESUMEN

Severe respiratory syncytial virus (RSV) infection is a major cause of morbidity and mortality in infants <2 years-old. Here we describe that high-fiber diet protects mice from RSV infection. This effect was dependent on intestinal microbiota and production of acetate. Oral administration of acetate mediated interferon-ß (IFN-ß) response by increasing expression of interferon-stimulated genes in the lung. These effects were associated with reduction of viral load and pulmonary inflammation in RSV-infected mice. Type 1 IFN signaling via the IFN-1 receptor (IFNAR) was essential for acetate antiviral activity in pulmonary epithelial cell lines and for the acetate protective effect in RSV-infected mice. Activation of Gpr43 in pulmonary epithelial cells reduced virus-induced cytotoxicity and promoted antiviral effects through IFN-ß response. The effect of acetate on RSV infection was abolished in Gpr43-/- mice. Our findings reveal antiviral effects of acetate involving IFN-ß in lung epithelial cells and engagement of GPR43 and IFNAR.


Asunto(s)
Acetatos/farmacología , Interferón Tipo I/metabolismo , Microbiota , Receptores Acoplados a Proteínas G/metabolismo , Infecciones por Virus Sincitial Respiratorio/prevención & control , Células A549 , Acetatos/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/virología , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Sustancias Protectoras/metabolismo , Sustancias Protectoras/farmacología , Receptor de Interferón alfa y beta/genética , Receptores Acoplados a Proteínas G/genética , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/virología , Células Vero , Carga Viral/efectos de los fármacos , Carga Viral/genética
12.
BMC Immunol ; 9: 38, 2008 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-18647414

RESUMEN

BACKGROUND: The greatest challenges in vaccine development include optimization of DNA vaccines for use in humans, creation of effective single-dose vaccines, development of delivery systems that do not involve live viruses, and the identification of effective new adjuvants. Herein, we describe a novel, simple technique for efficiently vaccinating mice against tuberculosis (TB). Our technique consists of a single-dose, genetic vaccine formulation of DNA-hsp65 complexed with cationic liposomes and administered intranasally. RESULTS: We developed a novel and non-toxic formulation of cationic liposomes, in which the DNA-hsp65 vaccine was entrapped (ENTR-hsp65) or complexed (COMP-hsp65), and used to immunize mice by intramuscular or intranasal routes. Although both liposome formulations induced a typical Th1 pattern of immune response, the intramuscular route of delivery did not reduce the number of bacilli. However, a single intranasal immunization with COMP-hsp65, carrying as few as 25 microg of plasmid DNA, leads to a remarkable reduction of the amount of bacilli in lungs. These effects were accompanied by increasing levels of IFN-gamma and lung parenchyma preservation, results similar to those found in mice vaccinated intramuscularly four times with naked DNA-hsp65 (total of 400 microg). CONCLUSION: Our objective was to overcome the significant obstacles currently facing DNA vaccine development. Our results in the mouse TB model showed that a single intranasal dose of COMP-hsp65 elicited a cellular immune response that was as strong as that induced by four intramuscular doses of naked-DNA. This formulation allowed a 16-fold reduction in the amount of DNA administered. Moreover, we demonstrated that this vaccine is safe, biocompatible, stable, and easily manufactured at a low cost. We believe that this strategy can be applied to human vaccines to TB in a single dose or in prime-boost protocols, leading to a tremendous impact on the control of this infectious disease.


Asunto(s)
Proteínas Bacterianas/administración & dosificación , Chaperoninas/administración & dosificación , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis Pulmonar/inmunología , Vacunas de ADN/administración & dosificación , Administración Intranasal , Animales , Proteínas Bacterianas/inmunología , Chaperonina 60 , Chaperoninas/inmunología , Femenino , Inmunidad Activa/efectos de los fármacos , Inmunización Secundaria , Liposomas , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Células TH1/efectos de los fármacos , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/prevención & control
13.
Mol Immunol ; 43(5): 464-72, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16337489

RESUMEN

In this study, we observed the occurrence of TRBV8.1-DB2.1 V(D)J recombination in murine fetal thymus organ culture (FTOC), in which the thymic microenvironment is mimicked. Since ionizing radiation affects T-cell development, we irradiated FTOCs with gamma rays to evaluate the modulation of genes implicated in TRBV8.1-BD2.1 rearrangements. The nylon cDNA microarray method was employed to monitor the expression of 9216 genes, which were organized in coexpression clusters. Clustering analysis showed similar expression profiling of genes implicated in the V(D)J recombination and DNA double strand break (DSB) repair processes such as XRCC4, RAG-2, Artemis and DNA-PK-cs, thus suggesting overlap between the two processes. The RUNX3 gene, whose coded protein binds to the enhancers of TR genes, was also modulated and the DNA cross-linking LR1 gene, which plays a role in the opening of hairpin DNA structures and whose expression pattern is similar to Artemis, may play a role in the control of V(D)J recombination. Furthermore, our data demonstrate that the FTOC model system and cDNA microarray method are useful tools to evidentiate genes that may play a role in both processes V(D)J recombination and DNA repair.


Asunto(s)
Reparación del ADN/genética , Perfilación de la Expresión Génica , Timo/efectos de la radiación , VDJ Recombinasas/metabolismo , Animales , Diferenciación Celular , Análisis por Conglomerados , ADN Complementario/genética , Rayos gamma , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T , Ratones , Ratones Endogámicos BALB C , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Técnicas de Cultivo de Órganos , Reacción en Cadena de la Polimerasa/métodos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T/citología , Linfocitos T/efectos de la radiación , Timo/embriología , Timo/metabolismo
14.
Gut Microbes ; 8(1): 59-66, 2017 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-28045579

RESUMEN

Recent discoveries on the role of commensal microbiota have significantly changed our understanding of human physiology. The host-microbiota interplay is now an important aspect to take into account to understand immune responses and immunological diseases. Autoimmune uveitis is a sight-threatening disease that arises without a known infectious etiology. It is unknown where and how autoreactive T cells become primed to trigger disease in the eye, which is an immune privileged site. We recently reported data supporting the notion that retina-specific T cells receive a signal in the gut from commensal microbiota-derived cross-reactive antigen(s) and trigger autoimmune uveitis in the R161H mouse model. Here we discuss our published findings, as well as our recent attempts to identify the responsible microbe(s) by using different antibiotic treatments, 16S rDNA sequencing and homology searches for candidate antigenic mimic(s) of the retinal antigen.


Asunto(s)
Antígenos/inmunología , Enfermedades Autoinmunes/microbiología , Microbioma Gastrointestinal , Uveítis/inmunología , Uveítis/microbiología , Animales , Enfermedades Autoinmunes/inmunología , Autoinmunidad , Humanos , Retina/inmunología , Linfocitos T/inmunología
15.
DNA Cell Biol ; 35(9): 455-8, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27463238

RESUMEN

Intestinal microbes have profound effects on inflammatory autoimmunity in sites distant from the gut. The mechanisms whereby this happens are only now beginning to be understood and may include such diverse effects as innate stimulation of migrating immune cells and effects of circulating bacterial metabolites. Our studies add to this the demonstration that microbiota may provide a source of cross-reactive antigenic material that activates autoreactive lymphocytes within the gut environment. In a spontaneous model of autoimmune uveitis, T lymphocytes specific to a retinal autoantigen are activated through their specific antigen receptor in the gut and acquire the ability to fuel inflammatory autoimmunity in the eye. In view of the huge diversity of commensals, it is conceivable that they may provide surrogate antigens for activation of autoreactive lymphocytes(s) of other tissue specificities, and might therefore be involved in the etiology of autoimmune diseases more frequently than is currently appreciated.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad , Microbioma Gastrointestinal/inmunología , Linfocitos T/inmunología , Uveítis/inmunología , Animales , Autoantígenos/biosíntesis , Enfermedades Autoinmunes/microbiología , Enfermedades Autoinmunes/patología , Movimiento Celular/inmunología , Reacciones Cruzadas , Humanos , Inmunidad Innata , Activación de Linfocitos , Ratones , Retina/inmunología , Retina/patología , Linfocitos T/microbiología , Uveítis/microbiología , Uveítis/patología
16.
Bio Protoc ; 6(18)2016 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-28191487

RESUMEN

The contribution of microbiota in regulating multiple physiological and pathological host responses has been studied intensively in recent years. Evidence suggests that commensal microbiota can directly modulate different populations of cells of the immune system (e.g., Ivanov et al., 2008; Atarashi et al., 2011). Recently, we showed that protein extracts from gut commensal microbiota can activate retina-specific T cells, allowing these autoreactive T cells to then break through the blood-retinal barrier and trigger autoimmune uveitis in the recipient (Horai et al., 2015). The protocol below describes the method to prepare intestinal protein-rich extracts that can be used in various in vitro andin vivo immunological studies.

17.
Hum Vaccin Immunother ; 10(5): 1238-43, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24607935

RESUMEN

Despite substantial efforts in recent years toward the development of new vaccines and drugs against tuberculosis (TB), success has remained elusive. Immunotherapy of TB with mycobacterial Hsp65 as a DNA vaccine (DNA-hsp65) results in a reduction of systemic bacterial loads and lung tissue damage, but the high homology of Hsp65 with the mammalian protein raises concern that pathological autoimmune responses may also be triggered. We searched for autoimmune responses elicited by DNA-hsp65 immunotherapy in mice chronically infected with TB by evaluating the humoral immune response and comprehensive histopathology using stereology. Cross-reactive antibodies between mycobacterial and mammalian Hsp60/65 were detected; however, no signs of pathological autoimmunity were found up to 60 days after the end of the therapy.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Autoinmunidad/inmunología , Proteínas Bacterianas/inmunología , Chaperonina 60/inmunología , Proteínas Mitocondriales/inmunología , Mycobacterium leprae/inmunología , Vacunas de ADN/inmunología , Animales , Autoinmunidad/efectos de los fármacos , Proteínas Bacterianas/administración & dosificación , Chaperonina 60/administración & dosificación , Chaperonina 60/antagonistas & inhibidores , Reacciones Cruzadas/efectos de los fármacos , Reacciones Cruzadas/inmunología , Inmunidad Humoral/efectos de los fármacos , Inmunidad Humoral/inmunología , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C , Proteínas Mitocondriales/antagonistas & inhibidores , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/inmunología , Vacunas de ADN/administración & dosificación
18.
Hum Vaccin Immunother ; 9(5): 1093-103, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23324590

RESUMEN

Despite the enormous efforts displayed globally in the fight against tuberculosis, the disease incidence has modified slightly, which has led to a renewed interest in immunotherapy. In general, successful immunotherapeutic candidates against tuberculosis are agents that can trigger strong, specific pro-inflammatory responses, especially of the T-helper (Th) 1 pattern. However, how these pro-inflammatory agents effectively kill the bacteria without eliciting immunopathology is not well understood. We reasoned that, in addition to the specific immune response elicited by immunotherapy, the evaluation of the overall pro-inflammatory responses should provide additional and valuable information that will be useful in avoiding immunopathology. We evaluated the overall IFN-γ and IL-17 pro-inflammatory responses among CD4(+), CD8(+) and γδ T cells in the lungs of mice that were infected with M. tuberculosis and treated with a DNA vaccine in an immunotherapeutic regimen. Our results demonstrate that mice that effectively combat the pathogen develop a strong, specific Th1 immune response against the therapeutic antigen and have reduced lung inflammation, present in parallel a fine-tuning in the total IFN-γ- and IL-17-mediated immunity in the lungs. This modulation of the total immune response involves reducing the Th17 cell population, augmenting CD8(+) T cells that produce IFN-γ and increasing the total γδ T cell frequency. These results stress the importance of a broad evaluation of not only the specific immune response at the time to evaluate new immune interventional strategies against tuberculosis but also non-conventional T cells, such as γδ T lymphocytes.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Terapia Genética/métodos , Inflamación , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Tuberculosis/terapia , Animales , Modelos Animales de Enfermedad , Femenino , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología
19.
PLoS One ; 5(11): e14140, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21152440

RESUMEN

BACKGROUND: Detailed analysis of the dynamic interactions among biological, environmental, social, and economic factors that favour the spread of certain diseases is extremely useful for designing effective control strategies. Diseases like tuberculosis that kills somebody every 15 seconds in the world, require methods that take into account the disease dynamics to design truly efficient control and surveillance strategies. The usual and well established statistical approaches provide insights into the cause-effect relationships that favour disease transmission but they only estimate risk areas, spatial or temporal trends. Here we introduce a novel approach that allows figuring out the dynamical behaviour of the disease spreading. This information can subsequently be used to validate mathematical models of the dissemination process from which the underlying mechanisms that are responsible for this spreading could be inferred. METHODOLOGY/PRINCIPAL FINDINGS: The method presented here is based on the analysis of the spread of tuberculosis in a Brazilian endemic city during five consecutive years. The detailed analysis of the spatio-temporal correlation of the yearly geo-referenced data, using different characteristic times of the disease evolution, allowed us to trace the temporal path of the aetiological agent, to locate the sources of infection, and to characterize the dynamics of disease spreading. Consequently, the method also allowed for the identification of socio-economic factors that influence the process. CONCLUSIONS/SIGNIFICANCE: The information obtained can contribute to more effective budget allocation, drug distribution and recruitment of human skilled resources, as well as guiding the design of vaccination programs. We propose that this novel strategy can also be applied to the evaluation of other diseases as well as other social processes.


Asunto(s)
Vigilancia de la Población/métodos , Tuberculosis/prevención & control , Tuberculosis/transmisión , Brasil/epidemiología , Geografía , Humanos , Incidencia , Densidad de Población , Dinámica Poblacional , Factores Socioeconómicos , Tuberculosis/epidemiología
20.
PLoS One ; 5(1): e8721, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20090941

RESUMEN

BACKGROUND: We have previously explored a therapeutic strategy for specifically targeting the profibrotic activity of IL-13 during experimental pulmonary fibrosis using a fusion protein comprised of human IL-13 and a mutated form of Pseudomonas aeruginosa exotoxin A (IL13-PE) and observed that the intranasal delivery of IL13-PE reduced bleomycin-induced pulmonary fibrosis through its elimination of IL-13-responsive cells in the lung. The aim of the present study was to determine whether the presence of an immune response to P. aeruginosa and/or its exotoxin A (PE) would diminish the anti-fibrotic properties of IL13-PE. METHODOLOGY/PRINCIPAL FINDINGS: Fourteen days after P. aeruginosa infection, C57BL/6 mice were injected with bleomycin via the intratracheal route. Other groups of mice received 4 doses of saline or IL13-PE by either intranasal or intraperitoneal application, and were challenged i.t. with bleomycin 28 days later. At day 21 after bleomycin, all mice received either saline vehicle or IL13-PE by the intranasal route and histopatological analyses of whole lung samples were performed at day 28 after bleomycin. Intrapulmonary P. aeruginosa infection promoted a neutralizing IgG2A and IgA antibody response in BALF and serum. Surprisingly, histological analysis showed that a prior P. aeruginosa infection attenuated the development of bleomycin-induced pulmonary fibrosis, which was modestly further attenuated by the intranasal administration of IL13-PE. Although prior intranasal administration of IL13-PE failed to elicit an antibody response, the systemic administration of IL13-PE induced a strong neutralizing antibody response. However, the prior systemic sensitization of mice with IL13-PE did not inhibit the anti-fibrotic effect of IL13-PE in fibrotic mice. CONCLUSIONS: Thus, IL13-PE therapy in pulmonary fibrosis works regardless of the presence of a humoral immune response to Pseudomonas exotoxin A. Interestingly, a prior infection with P. aeruginosa markedly attenuated the pulmonary fibrotic response suggesting that the immune elicitation by this pathogen exerts anti-fibrotic effects.


Asunto(s)
ADP Ribosa Transferasas/farmacología , Toxinas Bacterianas/farmacología , Exotoxinas/farmacología , Exotoxinas/uso terapéutico , Inmunotoxinas/uso terapéutico , Interleucina-13/uso terapéutico , Fibrosis Pulmonar/tratamiento farmacológico , Factores de Virulencia/farmacología , ADP Ribosa Transferasas/inmunología , Animales , Toxinas Bacterianas/inmunología , Bleomicina/toxicidad , Exotoxinas/inmunología , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Proteínas Recombinantes de Fusión , Factores de Virulencia/inmunología , Exotoxina A de Pseudomonas aeruginosa
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda