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.
Proc Natl Acad Sci U S A ; 117(28): 16465-16474, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32601220

RESUMEN

Under steady-state conditions, the immune system is poised to sense and respond to the microbiota. As such, immunity to the microbiota, including T cell responses, is expected to precede any inflammatory trigger. How this pool of preformed microbiota-specific T cells contributes to tissue pathologies remains unclear. Here, using an experimental model of psoriasis, we show that recall responses to commensal skin fungi can significantly aggravate tissue inflammation. Enhanced pathology caused by fungi preexposure depends on Th17 responses and neutrophil extracellular traps and recapitulates features of the transcriptional landscape of human lesional psoriatic skin. Together, our results propose that recall responses directed to skin fungi can directly promote skin inflammation and that exploration of tissue inflammation should be assessed in the context of recall responses to the microbiota.


Asunto(s)
Arthrodermataceae/fisiología , Microbiota , Psoriasis/inmunología , Piel/microbiología , Animales , Arthrodermataceae/clasificación , Arthrodermataceae/genética , Arthrodermataceae/aislamiento & purificación , Trampas Extracelulares/inmunología , Trampas Extracelulares/microbiología , Femenino , Humanos , Inmunidad , Masculino , Ratones , Ratones Endogámicos C57BL , Psoriasis/microbiología , Psoriasis/patología , Piel/inmunología , Piel/patología , Simbiosis , Células Th17/inmunología
2.
Arthritis Rheumatol ; 72(3): 454-464, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31566908

RESUMEN

OBJECTIVE: A role for mitochondrial dysfunction has been proposed in the immune dysregulation and organ damage characteristic of systemic lupus erythematosus (SLE). Idebenone is a coenzyme Q10 synthetic quinone analog and an antioxidant that has been used in humans to treat diverse diseases in which mitochondrial function is impaired. This study was undertaken to assess whether idebenone ameliorates lupus in murine models. METHODS: Idebenone was administered orally to MRL/lpr mice at 2 different doses (1 gm/kg or 1.5 gm/kg idebenone-containing diet) for 8 weeks. At peak disease activity, clinical, immunologic, and metabolic parameters were analyzed and compared to those in untreated mice (n = 10 per treatment group). Results were confirmed in the lupus-prone NZM2328 mouse model. RESULTS: In MRL/lpr mice, idebenone-treated mice showed a significant reduction in mortality incidence (P < 0.01 versus untreated mice), and the treatment attenuated several disease features, including glomerular inflammation and fibrosis (each P < 0.05 versus untreated mice), and improved renal function in association with decreased renal expression of interleukin-17A (IL-17A) and mature IL-18. Levels of splenic proinflammatory cytokines and inflammasome-related genes were significantly decreased (at least P < 0.05 and some with higher significance) in mice treated with idebenone, while no obvious drug toxicity was observed. Idebenone inhibited neutrophil extracellular trap formation in neutrophils from lupus-prone mice (P < 0.05) and human patients with SLE. Idebenone also improved mitochondrial metabolism (30% increase in basal respiration and ATP production), reduced the extent of heart lipid peroxidation (by one-half that of untreated mice), and significantly improved endothelium-dependent vasorelaxation (P < 0.001). NZM2328 mice exposed to idebenone also displayed improvements in renal and systemic inflammation, reducing the kidney pathology score (P < 0.05), IgG/C3 deposition (P < 0.05), and the gene expression of interferon, proinflammatory, and inflammasome-related genes (at least P < 0.05 and some with higher significance). CONCLUSION: Idebenone ameliorates murine lupus disease activity and the severity of organ damage, supporting the hypothesis that agents that modulate mitochondrial biologic processes may have a therapeutic role in human SLE.


Asunto(s)
Antioxidantes/administración & dosificación , Lupus Eritematoso Sistémico/terapia , Ubiquinona/análogos & derivados , Animales , Modelos Animales de Enfermedad , Trampas Extracelulares/efectos de los fármacos , Inflamación , Interleucina-17/metabolismo , Interleucina-18/metabolismo , Riñón/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Ratones , Ratones Endogámicos MRL lpr , Mitocondrias/metabolismo , Ubiquinona/administración & dosificación
3.
Ann Rheum Dis ; 78(7): 957-966, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31040119

RESUMEN

OBJECTIVES: The presence of proinflammatory low-density granulocytes (LDG) has been demonstrated in autoimmune and infectious diseases. Recently, regulatory neutrophilic polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) were identified in systemic lupus erythematosus (SLE). Because LDG and PMN-MDSC share a similar phenotype with contrasting functional effects, we explored these cells in a cohort of patients with SLE. METHODS: LDG and normal-density granulocytes (NDG) were isolated from fresh blood of healthy donors (HD) and patients with SLE. Associations between LDG and clinical manifestations were analysed. Multicolor flow cytometry and confocal imaging were performed to immunophenotype the cells. The ability of LDG and NDG to suppress T cell function and induce cytokine production was quantified. RESULTS: LDG prevalence was elevated in SLE versus HD, associated with the interferon (IFN) 21-gene signature and disease activity. Also, the LDG-to-lymphocyte ratio associated better with SLE disease activity index than neutrophil-to-lymphocyte ratio. SLE LDG exhibited significantly heightened surface expression of various activation markers and also of lectin-like oxidised low-density lipoprotein receptor-1, previously described to be associated with PMN-MDSC. Supernatants from SLE LDG did not restrict HD CD4+ T cell proliferation in an arginase-dependent manner, suggesting LDG are not immunosuppressive. SLE LDG supernatants induced proinflammatory cytokine production (IFN gamma, tumour necrosis factor alpha and lymphotoxin alpha) from CD4+ T cells. CONCLUSIONS: Based on our results, SLE LDG display an activated phenotype, exert proinflammatory effects on T cells and do not exhibit MDSC function. These results support the concept that LDG represent a distinct proinflammatory subset in SLE with pathogenic potential, at least in part, through their ability to activate type 1 helper responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Granulocitos/inmunología , Lupus Eritematoso Sistémico/inmunología , Neutrófilos/inmunología , Adolescente , Adulto , Anciano , Estudios de Casos y Controles , Proliferación Celular , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación , Lupus Eritematoso Sistémico/sangre , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Fenotipo , Adulto Joven
4.
JCI Insight ; 3(23)2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30518690

RESUMEN

The peptidylarginine deiminases PAD2 and PAD4 are implicated in the pathogenesis of several autoimmune diseases. PAD4 may be pathogenic in systemic lupus erythematosus (SLE) through its role in neutrophil extracellular trap (NET) formation that promotes autoantigen externalization, immune dysregulation, and organ damage. The role of this enzyme in mouse models of autoimmunity remains unclear, as pan-PAD chemical inhibitors improve clinical phenotype, whereas PAD4-KO models have given conflicting results. The role of PAD2 in SLE has not been investigated. The differential roles of PAD2 and PAD4 in TLR-7-dependent lupus autoimmunity were examined. Padi4-/- displayed decreased autoantibodies, type I IFN responses, immune cell activation, vascular dysfunction, and NET immunogenicity. Padi2-/- mice showed abrogation of Th subset polarization, with some disease manifestations reduced compared with WT but to a lesser extent than Padi4-/- mice. RNA sequencing analysis revealed distinct modulation of immune-related pathways in PAD-KO lymphoid organs. Human T cells express both PADs and, when exposed to either PAD2 or PAD4 inhibitors, displayed abrogation of Th1 polarization. These results suggest that targeting PAD2 and/or PAD4 activity modulates dysregulated TLR-7-dependent immune responses in lupus through differential effects of innate and adaptive immunity. Compounds that target PADs may have potential therapeutic roles in T cell-mediated diseases.


Asunto(s)
Inmunidad Adaptativa/inmunología , Inmunidad Innata/inmunología , Lupus Eritematoso Sistémico/inmunología , Desiminasas de la Arginina Proteica/inmunología , Desiminasas de la Arginina Proteica/metabolismo , Receptor Toll-Like 7/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Trampas Extracelulares , Femenino , Regulación de la Expresión Génica , Histonas , Humanos , Hidrolasas/genética , Hidrolasas/inmunología , Hidrolasas/metabolismo , Inflamación , Interferón Tipo I , Ratones , Ratones Noqueados , Arginina Deiminasa Proteína-Tipo 2 , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células TH1 , Células Th17 , Transcriptoma
5.
Ann Rheum Dis ; 77(8): 1226-1233, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29724730

RESUMEN

OBJECTIVES: To characterise renal tissue metabolic pathway gene expression in different forms of glomerulonephritis. METHODS: Patients with nephrotic syndrome (NS), antineutrophil cytoplasmic antibody-associated vasculitis (AAV), systemic lupus erythematosus (SLE) and healthy living donors (LD) were studied. Clinically indicated renal biopsies were obtained at time of diagnosis and microdissected into glomerular and tubulointerstitial compartments. Microarray-derived differential gene expression of 88 genes representing critical enzymes of metabolic pathways and 25 genes related to immune cell markers was compared between disease groups. Correlation analyses measured relationships between metabolic pathways, kidney function and cytokine production. RESULTS: Reduced steady state levels of mRNA species were enriched in pathways of oxidative phosphorylation and increased in the pentose phosphate pathway (PPP) with maximal perturbation in AAV and SLE followed by NS, and least in LD. Transcript regulation was isozymes specific with robust regulation in hexokinases, enolases and glucose transporters. Intercorrelation networks were observed between enzymes of the PPP (eg, transketolase) and macrophage markers (eg, CD68) (r=0.49, p<0.01). Increased PPP transcript levels were associated with reduced glomerular filtration rate in the glomerular (r=-0.49, p<0.01) and tubulointerstitial (r=-0.41, p<0.01) compartments. PPP expression and tumour necrosis factor activation were tightly co-expressed (r=0.70, p<0.01). CONCLUSION: This study demonstrated concordant alterations of the renal transcriptome consistent with metabolic reprogramming across different forms of glomerulonephritis. Activation of the PPP was tightly linked with intrarenal macrophage marker expression, reduced kidney function and increased production of cytokines. Modulation of glucose metabolism may offer novel immune-modulatory therapeutic approaches in rare kidney diseases.


Asunto(s)
Glomerulonefritis/metabolismo , Redes y Vías Metabólicas/genética , Adulto , Anciano , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/genética , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/metabolismo , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/patología , Biopsia , Citocinas/biosíntesis , Femenino , Regulación de la Expresión Génica , Glomerulonefritis/genética , Glomerulonefritis/patología , Humanos , Isoenzimas/metabolismo , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/patología , Masculino , Redes y Vías Metabólicas/inmunología , Persona de Mediana Edad , Síndrome Nefrótico/genética , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Vía de Pentosa Fosfato/genética , ARN Mensajero/genética , Transcriptoma , Adulto Joven
6.
Curr Opin Rheumatol ; 29(5): 442-449, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28639951

RESUMEN

PURPOSE OF REVIEW: Upon antigen exposure, immune cells rely on cell-specific metabolic pathways to mount an efficient immune response. In autoimmunity, failure in critical metabolic checkpoints may lead to immune cell hyperactivation and tissue damage. Oxidative stress in autoimmune patients can also contribute to immune dysregulation and injury to the host. Recent insights into the immune cell metabolism signatures, specifically associated with systemic lupus erythematosus (SLE) and the consequences of heightened oxidative stress in patients, are discussed herein. RECENT FINDINGS: Glucose metabolism inhibitors, mechanistic target of rapamycin pathway modulators, and peroxisome proliferator-activated receptor gamma-activating compounds demonstrate therapeutic benefit in experimental models of lupus. Mitochondrial-derived reactive oxygen species (ROS) and molecular modifications induced by oxidative stress appear to be detrimental in lupus. Effective therapies tailored toward the reconfiguration of metabolic imbalances in lupus immune cells and the reduction of mitochondrial ROS production/availability are currently being tested. SUMMARY: A paucity of knowledge exists regarding the metabolic needs of a number of immune cells involved in the pathogenesis of SLE, including myeloid cells and B cells. Nonetheless, SLE-specific metabolic signatures have been identified and their specific targeting, along with mitochondrial ROS inhibitors/scavengers, could show therapeutic advantage in lupus patients.


Asunto(s)
Autoinmunidad , Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Humanos , Neutrófilos/patología
7.
Curr Opin Rheumatol ; 29(1): 65-70, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27755122

RESUMEN

PURPOSE OF REVIEW: A breakdown of immune tolerance to self-antigens in a genetically predisposing background, precipitated by environmental triggers, contributes to the development of systemic autoimmune diseases. Renewed interest in the immunomodulatory capabilities of neutrophils in systemic autoimmunity has identified neutrophil extracellular trap (NET) formation as a distinguishing action of neutrophils in afflicted hosts. RECENT FINDINGS: Oxidation of nucleic acids and posttranslational modifications of proteins distinctly occur during NET formation and may promote enhanced immunogenicity. Various autoantibodies, immune complexes, and other inflammatory stimuli have been recently reported to promote NET formation in individuals with autoimmune diseases. Associations between level of NETosis and adverse outcomes in systemic autoimmune diseases, including thrombosis, adverse pregnancy outcomes, and renal disease, continue to be investigated. SUMMARY: Understanding the putative pathogenic role and sequelae of NETosis in rheumatic diseases is a major focus of ongoing research efforts. Mechanisms elucidated by these discoveries may provide novel therapeutic targets to inhibit NET formation and/or promote the clearance of immunogenic NET material.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Trampas Extracelulares/inmunología , Neutrófilos/inmunología , Enfermedades Reumáticas/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Autoinmunidad/inmunología , Humanos , Procesamiento Proteico-Postraduccional/inmunología
8.
Diabetes ; 64(11): 3885-90, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26216853

RESUMEN

Most natural history models for type 1 diabetes (T1D) propose that overt hyperglycemia results after a progressive loss of insulin-secreting ß-cell mass and/or function. To experimentally address this concept, we prospectively determined morning blood glucose measurements every other day in multiple cohorts (total n = 660) of female NOD/ShiLtJ mice starting at 8 weeks of age until diabetes onset or 26 weeks of age. Consistent with this notion, a majority of mice that developed diabetes (354 of 489 [72%]) displayed a progressive increase in blood glucose with transient excursions >200 mg/dL, followed by acute and persistent hyperglycemia at diabetes onset. However, 135 of the 489 (28%) diabetic animals demonstrated normal glucose values followed by acute (i.e., sudden) hyperglycemia. Interestingly, diabetes onset occurred earlier in mice with acute versus progressive disease onset (15.37 ± 0.3207 vs. 17.44 ± 0.2073 weeks of age, P < 0.0001). Moreover, the pattern of onset (i.e., progressive vs. acute) dramatically influenced the ability to achieve reversal of T1D by immunotherapeutic intervention, with increased effectiveness observed in situations of a progressive deterioration in euglycemia. These studies highlight a novel natural history aspect in this animal model, one that may provide important guidance for the selection of subjects participating in human trials seeking disease reversal.


Asunto(s)
Glucemia/análisis , Diabetes Mellitus Tipo 1/sangre , Hiperglucemia/sangre , Hipoglucemiantes/uso terapéutico , Animales , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Progresión de la Enfermedad , Femenino , Hiperglucemia/tratamiento farmacológico , Ratones , Ratones Endogámicos NOD
9.
EMBO J ; 34(7): 881-95, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25666591

RESUMEN

Intestinal immune regulatory signals govern gut homeostasis. Breakdown of such regulatory mechanisms may result in inflammatory bowel disease (IBD). Lactobacillus acidophilus contains unique surface layer proteins (Slps), including SlpA, SlpB, SlpX, and lipoteichoic acid (LTA), which interact with pattern recognition receptors to mobilize immune responses. Here, to elucidate the role of SlpA in protective immune regulation, the NCK2187 strain, which solely expresses SlpA, was generated. NCK2187 and its purified SlpA bind to the C-type lectin SIGNR3 to exert regulatory signals that result in mitigation of colitis, maintenance of healthy gastrointestinal microbiota, and protected gut mucosal barrier function. However, such protection was not observed in Signr3(-/-) mice, suggesting that the SlpA/SIGNR3 interaction plays a key regulatory role in colitis. Our work presents critical insights into SlpA/SIGNR3-induced responses that are integral to the potential development of novel biological therapies for autoinflammatory diseases, including IBD.


Asunto(s)
Antígenos CD/inmunología , Proteínas Bacterianas/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Lactobacillus acidophilus/inmunología , Lectinas Tipo C/inmunología , Animales , Antígenos CD/genética , Proteínas Bacterianas/genética , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Lactobacillus acidophilus/genética , Lectinas Tipo C/genética , Lipopolisacáridos/genética , Lipopolisacáridos/inmunología , Ratones , Ratones Noqueados , Unión Proteica/genética , Unión Proteica/inmunología , Ácidos Teicoicos/genética , Ácidos Teicoicos/inmunología
10.
PLoS One ; 9(6): e100532, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24945934

RESUMEN

Gastrointestinal (GI) anthrax results from the ingestion of Bacillus anthracis. Herein, we investigated the pathogenesis of GI anthrax in animals orally infected with toxigenic non-encapsulated B. anthracis Sterne strain (pXO1+ pXO2-) spores that resulted in rapid animal death. B. anthracis Sterne induced significant breakdown of intestinal barrier function and led to gut dysbiosis, resulting in systemic dissemination of not only B. anthracis, but also of commensals. Disease progression significantly correlated with the deterioration of innate and T cell functions. Our studies provide critical immunologic and physiologic insights into the pathogenesis of GI anthrax infection, whereupon cleavage of mitogen-activated protein kinases (MAPKs) in immune cells may play a central role in promoting dysfunctional immune responses against this deadly pathogen.


Asunto(s)
Carbunco/inmunología , Carbunco/microbiología , Bacillus anthracis/inmunología , Colon/inmunología , Disbiosis/inmunología , Disbiosis/microbiología , Enfermedades Gastrointestinales/inmunología , Enfermedades Gastrointestinales/microbiología , Tolerancia Inmunológica , Animales , Carbunco/enzimología , Carbunco/patología , Colon/microbiología , Colon/patología , Disbiosis/patología , Epitelio/inmunología , Epitelio/microbiología , Epitelio/patología , Enfermedades Gastrointestinales/enzimología , Enfermedades Gastrointestinales/patología , Inmunidad Innata , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Linfocitos T/inmunología
11.
J Infect Dis ; 210(9): 1499-507, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24829464

RESUMEN

Ingestion of Bacillus anthracis spores causes gastrointestinal (GI) anthrax. Humoral immune responses, particularly immunoglobulin A (IgA)-secreting B-1 cells, play a critical role in the clearance of GI pathogens. Here, we investigated whether B. anthracis impacts the function of colonic B-1 cells to establish active infection. GI anthrax led to significant inhibition of immunoglobulins (eg, IgA) and increased expression of program death 1 on B-1 cells. Furthermore, infection also diminished type 2 innate lymphoid cells (ILC2) and their ability to enhance differentiation and immunoglobulin production by secreting interleukin 5 (IL-5). Such B-1-cell and ILC2 dysfunction is potentially due to cleavage of p38 and Erk1/2 mitogen-activated protein kinases in these cells. Conversely, mice that survived infection generated neutralizing antibodies via the formation of robust germinal center B cells in Peyer's patches and had restored B-1-cell and ILC2 function. These data may provide additional insight for designing efficacious vaccines and therapeutics against this deadly pathogen.


Asunto(s)
Carbunco/inmunología , Linfocitos B/fisiología , Bacillus anthracis/fisiología , Enfermedades Gastrointestinales/inmunología , Animales , Bacillus anthracis/inmunología , Colon/inmunología , Colon/microbiología , Citometría de Flujo , Inmunidad Celular/inmunología , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
FEBS Lett ; 588(22): 4158-66, 2014 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-24842610

RESUMEN

The intestinal epithelium is equipped with sensing receptor mechanisms that interact with luminal microorganisms and nutrients to regulate barrier function and gut immune responses, thereby maintaining intestinal homeostasis. Herein, we clarify the role of the extracellular calcium-sensing receptor (CaSR) using intestinal epithelium-specific Casr(-/-) mice. Epithelial CaSR deficiency diminished intestinal barrier function, altered microbiota composition, and skewed immune responses towards proinflammatory. Consequently, Casr(-/-) mice were significantly more prone to chemically induced intestinal inflammation resulting in colitis. Accordingly, CaSR represents a potential therapeutic target for autoinflammatory disorders, including inflammatory bowel diseases.


Asunto(s)
Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Receptores Sensibles al Calcio/deficiencia , Animales , Colitis/inducido químicamente , Colitis/inmunología , Colitis/metabolismo , Colitis/microbiología , Sulfato de Dextran/efectos adversos , Técnicas de Inactivación de Genes , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Receptores Sensibles al Calcio/genética , Receptores Sensibles al Calcio/metabolismo , Transducción de Señal/efectos de los fármacos
13.
Curr Pharm Biotechnol ; 14(10): 867-77, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24372255

RESUMEN

Production of long-lived, high affinity humoral immunity is an essential characteristic of successful vaccination and requires cognate interactions between T and B cells in germinal centers. Within germinal centers, specialized T follicular helper cells assist B cells and regulate the antibody response by mediating the differentiation of B cells into memory or plasma cells after exposure to T cell-dependent antigens. It is now appreciated that local immune responses are also essential for protection against infectious diseases that gain entry to the host by the mucosal route; therefore, targeting the mucosal compartments is the optimum strategy to induce protective immunity. However, because the gastrointestinal mucosae are exposed to large amounts of environmental and dietary antigens on a daily basis, immune regulatory mechanisms exist to favor tolerance and discourage autoimmunity at these sites. Thus, mucosal vaccination strategies must ensure that the immunogen is efficiently taken up by the antigen presenting cells, and that the vaccine is capable of activating humoral and cellular immunity, while avoiding the induction of tolerance. Despite significant progress in mucosal vaccination, this potent platform for immunotherapy and disease prevention must be further explored and refined. Here we discuss recent progress in the understanding of the role of different phenotypes of B cells in the development of an efficacious mucosal vaccine against infectious disease.


Asunto(s)
Linfocitos B/inmunología , Células Dendríticas/inmunología , Inmunidad Mucosa , Activación de Linfocitos/inmunología , Vacunas , Administración a través de la Mucosa , Administración Oral , Animales , Enfermedades Transmisibles/inmunología , Sistemas de Liberación de Medicamentos , Humanos , Tolerancia Inmunológica , Inmunidad Celular , Membrana Mucosa/inmunología , Vacunas/administración & dosificación , Vacunas/inmunología
14.
Trends Mol Med ; 19(12): 714-25, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24051204

RESUMEN

Mechanisms of colorectal cancer (CRC) development can be generally divided into three categories: genetic, epigenetic, and aberrant immunologic signaling pathways, all of which may be triggered by an imbalanced intestinal microbiota. Aberrant gut microbial composition, termed 'dysbiosis', has been reported in inflammatory bowel disease patients who are at increased risk for CRC development. Recent studies indicate that it is feasible to rescue experimental models of colonic cancer by oral treatment with genetically engineered beneficial bacteria and/or their immune-regulating gene products. Here, we review the mechanisms of epigenetic modulation implicated in the development and progression of CRC, which may be the result of dysbiosis, and therefore may be amenable to therapeutic intervention.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/microbiología , Epigénesis Genética , Tracto Gastrointestinal/microbiología , Microbiota , Animales , Bacterias/genética , Bacterias/inmunología , Neoplasias Colorrectales/inmunología , Humanos
15.
Front Immunol ; 4: 25, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23390423

RESUMEN

As highlighted by the development of intestinal autoinflammatory disorders when tolerance is lost, homeostatic interactions between gut microbiota, resident immune cells, and the gut epithelium are key in the maintenance of gastrointestinal health. Gut immune responses, whether stimulatory or regulatory, are dictated by the activated dendritic cells (DCs) that first interact with microorganisms and their gene products to then elicit T and B cell responses. Previously, we have demonstrated that treatment with genetically modified Lactobacillus acidophilus is sufficient to tilt the immune balance from proinflammatory to regulatory in experimental models of colitis and colon cancer. Given the significant role of DCs in efficiently orchestrating intestinal immune responses, characterization of the signals induced within these cells by the surface layer molecules, such as lipoteichoic acid (LTA), and proteins of L. acidophilus is critical for future treatment and prevention of gastrointestinal diseases. Here, we discuss the potential regulatory pathways involved in the downregulation of pathogenic inflammation in the gut, and explore questions regarding the immune responses to LTA-deficient L. acidophilus that require future studies.

16.
PLoS One ; 8(1): e55143, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23383086

RESUMEN

BACKGROUND: Currently, sufficient data exist to support the use of lactobacilli as candidates for the development of new oral targeted vaccines. To this end, we have previously shown that Lactobacillus gasseri expressing the protective antigen (PA) component of anthrax toxin genetically fused to a dendritic cell (DC)-binding peptide (DCpep) induced efficacious humoral and T cell-mediated immune responses against Bacillus anthracis Sterne challenge. METHODOLOGY/PRINCIPAL FINDING: In the present study, we investigated the effects of a dose dependent treatment of mice with L. gasseri expressing the PA-DCpep fusion protein on intestinal and systemic immune responses and confirmed its safety. Treatment of mice with different doses of L. gasseri expressing PA-DCpep stimulated colonic immune responses, resulting in the activation of innate immune cells, including dendritic cells, which induced robust Th1, Th17, CD4(+)Foxp3(+) and CD8(+)Foxp3(+) T cell immune responses. Notably, high doses of L. gasseri expressing PA-DCpep (10(12) CFU) were not toxic to the mice. Treatment of mice with L. gasseri expressing PA-DCpep triggered phenotypic maturation and the release of proinflammatory cytokines by dendritic cells and macrophages. Moreover, treatment of mice with L. gasseri expressing PA-DCpep enhanced antibody immune responses, including IgA, IgG(1), IgG(2b), IgG(2c) and IgG(3). L. gasseri expressing PA-DCpep also increased the gene expression of numerous pattern recognition receptors, including Toll-like receptors, C-type lectin receptors and NOD-like receptors. CONCLUSION/SIGNIFICANCE: These findings suggest that L. gasseri expressing PA-DCpep has substantial immunopotentiating properties, as it can induce humoral and T cell-mediated immune responses upon oral administration and may be used as a safe oral vaccine against anthrax challenge.


Asunto(s)
Colon/inmunología , Vacunación/métodos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Administración Oral , Animales , Antígenos Bacterianos/genética , Toxinas Bacterianas/genética , Colon/metabolismo , Citocinas/biosíntesis , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunidad Humoral/inmunología , Lactobacillus/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores de Reconocimiento de Patrones/metabolismo , Seguridad , Subgrupos de Linfocitos T/inmunología , Vacunas de Subunidad/efectos adversos , Vacunas de Subunidad/genética
17.
Gut Microbes ; 4(1): 84-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23137966

RESUMEN

Pathogenic autoinflammatory responses triggered by dysregulated microbial interactions may lead to intestinal disorders and malignancies. Previously, we demonstrated that a lipoteichoic acid (LTA)-deficient Lactobacillus acidophilus strain, NCK2025, ameliorated inflammation-induced colitis, significantly reduced the number of polyps in a colonic polyposis cancer model and restored physiological homeostasis in both cases. Nonetheless, the regulatory signals delivered by NCK2025 to reprogram the gastrointestinal microenvironment, and thus resist colonic cancer progression, remain unknown. Accumulating evidence suggest that epigenetic changes, in the presence and absence of pathogenic inflammation, can result in colorectal cancer (CRC). To test possible epigenetic modifications induced by NCK2025, the expression of epigenetically regulated, CRC-associated genes was measured with and without bacterial treatment. In vivo and in vitro, NCK2025 enhanced the expression of tumor suppressor genes that may regulate CRC development. Therefore, differential epigenetic regulation of CRC-related genes by NCK2025 represents a potential therapy against colitis-associated and sporadic CRC.


Asunto(s)
Neoplasias del Colon/patología , Metilación de ADN , Genes Supresores de Tumor , Lactobacillus acidophilus/metabolismo , Lipopolisacáridos/deficiencia , Animales , Terapia Biológica/métodos , Neoplasias del Colon/terapia , Epigénesis Genética , Perfilación de la Expresión Génica , Humanos , Ácidos Teicoicos
19.
Methods Mol Biol ; 900: 347-62, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22933078

RESUMEN

Autoimmune Type 1 A Diabetes (T1D) is characterized by dependence on exogenous insulin consequential to the autoimmune attack and destruction of insulin-producing islet beta cells. Pancreatic islet cell inflammation, or insulitis, precedes beta cell death and T1D onset. In the insulitic lesion, innate immune cells produce chemokines and cytokines that recruit and activate adaptive immune cells (Eizirik D et al., Nat Rev Endocrinol 5:219-226, 2009). Locally produced cytokines not only increase immune surveillance of beta cells (Hanafusa T and Imagawa A, Ann NY Acad Sci 1150:297-299, 2008), but also cause beta cell dysfunction and decreased insulin secretion due to the generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) by the beta cells. This, coupled to the high levels of ROS and RNS secreted by activated macrophages and the low antioxidant capacities of beta cells (Huurman VA, PLoS One 3:e2435, 2008; Schatz D, Pediatr Diabetes 5:72-79, 2004; Verge CF, Diabetes 44:1176-1179, 1995), implicates free radicals as important effectors in T1D pathogenesis (Eizirik D et al., Nat Rev Endocrinol 5:219-226, 2009; Hanafusa T and Imagawa A, Ann NY Acad Sci 1150:297-299, 2008; Eisenbarth GS and Jeffrey J, Arq Bras Endocrinol Metabol 52:146-155, 2008; Pietropaolo M et al., Pediatr Diabetes 6:184-192, 2005).


Asunto(s)
Células Secretoras de Insulina/patología , Biología Molecular/métodos , Estrés Oxidativo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Caspasas/metabolismo , Separación Celular , Supervivencia Celular , Células Cultivadas , Pruebas de Enzimas , Glutatión/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Espacio Intracelular/metabolismo , Ratones , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo
20.
Eur J Clin Invest ; 42(11): 1244-51, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22924552

RESUMEN

Type 1 diabetes (T1D) is a chronic, multifactorial disorder that results from a contretemps of genetic and environmental factors. Autoimmune attack and functional inhibition of the insulin-producing ß cells in the pancreas lead to the inability of ß cells to metabolize glucose, and thus results the hallmark clinical symptom of diabetes: abnormally high blood glucose levels. Treatment and protection from T1D require a detailed knowledge of the molecular effectors and the mechanism(s) of cell death leading to ß-cell demise. Primary islets and surrogate ß cells have been utilized in vitro to investigate in isolation-specific mechanisms associated with progression to T1D in vivo. This review focuses on the data obtained from these experiments. Studies using transformed ß cells of human sources are described.


Asunto(s)
Muerte Celular/inmunología , Diabetes Mellitus Tipo 1/inmunología , Células Secretoras de Insulina/metabolismo , Páncreas/metabolismo , Autoinmunidad/fisiología , Línea Celular , Humanos , Células Secretoras de Insulina/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA