Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Curr Diab Rep ; 17(11): 113, 2017 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-28956297

RESUMEN

PURPOSE OF REVIEW: Despite immense research efforts, type 1 diabetes (T1D) remains an autoimmune disease without a known trigger or approved intervention. Over the last three decades, studies have primarily focused on delineating the role of the adaptive immune system in the mechanism of T1D. The discovery of Toll-like receptors in the 1990s has advanced the knowledge on the role of the innate immune system in host defense as well as mechanisms that regulate adaptive immunity including the function of autoreactive T cells. RECENT FINDINGS: Recent investigations suggest that inflammation plays a key role in promoting a large number of autoimmune disorders including T1D. Data from the LEW1.WR1 rat model of virus-induced disease and the RIP-B7.1 mouse model of diabetes suggest that innate immune signaling plays a key role in triggering disease progression. There is also evidence that innate immunity may be involved in the course of T1D in humans; however, a small number of clinical trials have shown that interfering with the function of the innate immune system following disease onset exerts only a modest effect on ß-cell function. The data implying that innate immune pathways are linked with mechanisms of islet autoimmunity hold great promise for the identification of novel disease pathways that may be harnessed for clinical intervention. Nevertheless, more work needs to be done to better understand mechanisms by which innate immunity triggers ß-cell destruction and assess the therapeutic value in blocking innate immunity for diabetes prevention.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Inmunidad Innata , Animales , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Inflamación/patología , Receptores Toll-Like/metabolismo
2.
Curr Diab Rep ; 16(10): 89, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27523648

RESUMEN

The gastrointestinal system represents one of the largest interfaces between the human internal microenvironment and the external world. This system harbors trillions of commensal bacteria that reside in symbiosis with the host. Intestinal bacteria play a crucial role in maintaining systemic and intestinal immune and metabolic homeostasis because of their effect on nutrient absorption and immune development and function. Recently, altered gut bacterial composition (dysbiosis) was hypothesized to be involved in mechanisms through which islet autoimmunity is triggered. Evidence from animal models indicates that alterations in the gut bacterial composition precede disease onset, thus implicating a causal role for the gut microbiome in islet destruction. However, it remains unclear whether dysbiosis is directly linked to the mechanisms of human type 1 diabetes (T1D). In this review, we discuss data implicating the gut microbiota in disease progression with an emphasis on our recent studies performed in humans and in rodent models of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/etiología , Microbioma Gastrointestinal , Intestinos/microbiología , Animales , Autoinmunidad , Humanos , Simbiosis , Receptores Toll-Like/fisiología
3.
J Immunol ; 189(8): 3805-14, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22988033

RESUMEN

Microbes were hypothesized to play a key role in the progression of type 1 diabetes (T1D). We used the LEW1.WR1 rat model of Kilham rat virus (KRV)-induced T1D to test the hypothesis that the intestinal microbiota is involved in the mechanism leading to islet destruction. Treating LEW1.WR1 rats with KRV and a combination of trimethoprim and sulfamethoxazole (Sulfatrim) beginning on the day of infection protected the rats from insulitis and T1D. Pyrosequencing of bacterial 16S rRNA and quantitative RT-PCR indicated that KRV infection resulted in a transient increase in the abundance of Bifidobacterium spp. and Clostridium spp. in fecal samples from day 5- but not day 12-infected versus uninfected animals. Similar alterations in the gut microbiome were observed in the jejunum of infected animals on day 5. Treatment with Sulfatrim restored the level of intestinal Bifidobacterium spp. and Clostridium spp. We also observed that virus infection induced the expression of KRV transcripts and the rapid upregulation of innate immune responses in Peyer's patches and pancreatic lymph nodes. However, antibiotic therapy reduced the virus-induced inflammation as reflected by the presence of lower amounts of proinflammatory molecules in both the Peyer's patches and pancreatic lymph nodes. Finally, Sulfatrim treatment reduced the number of B cells in Peyer's patches and downmodulated adaptive immune responses to KRV, but did not interfere with antiviral Ab responses or viral clearance from the spleen, pancreatic lymph nodes, and serum. The data suggest that gut microbiota may be involved in promoting virus-induced T1D in the LEW1.WR1 rat model.


Asunto(s)
Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Experimental/virología , Diabetes Mellitus Tipo 1/prevención & control , Diabetes Mellitus Tipo 1/virología , Parvovirus/inmunología , Animales , Diabetes Mellitus Experimental/microbiología , Diabetes Mellitus Tipo 1/microbiología , Combinación de Medicamentos , Femenino , Mediadores de Inflamación/administración & dosificación , Islotes Pancreáticos/microbiología , Islotes Pancreáticos/patología , Islotes Pancreáticos/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ganglios Linfáticos Agregados/microbiología , Ganglios Linfáticos Agregados/patología , Ganglios Linfáticos Agregados/virología , Ratas , Ratas Endogámicas Lew , Sulfadoxina/administración & dosificación , Sulfametoxazol/administración & dosificación , Sulfametoxazol/análogos & derivados , Trimetoprim/administración & dosificación
4.
Clin Immunol ; 146(2): 112-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23314185

RESUMEN

The digestive tract hosts trillions of bacteria that interact with the immune system and can influence the balance between pro-inflammatory and regulatory immune responses. Recent studies suggest that alterations in the composition of the intestinal microbiota may be linked with the development of type 1 diabetes (T1D). Data from the biobreeding diabetes prone (BBDP) and the LEW1.WR1 models of T1D support the hypothesis that intestinal bacteria may be involved in early disease mechanisms. The data indicate that cross-talk between the gut microbiota and the innate immune system may be involved in islet destruction. Whether a causal link between intestinal microbiota and T1D exists, the identity of the bacteria and the mechanism whereby they promote the disease remain to be examined. A better understanding of the interplay between microbes and innate immune pathways in early disease stages holds promise for the design of immune interventions and disease prevention in genetically susceptible individuals.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/microbiología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Animales , Diabetes Mellitus Experimental/virología , Diabetes Mellitus Tipo 1/virología , Humanos , Inmunidad Innata , Mucosa Intestinal/virología , Ratas , Ratas Endogámicas
5.
Diabetes Metab Res Rev ; 27(8): 824-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22069267

RESUMEN

BACKGROUND: Rat models of diabetes have emerged as a powerful experimental tool for addressing the role of microbial pathogens in the mechanism of autoimmune diabetes. We have used the biobreeding diabetes resistant and LEW1.WR1 rat models to identify the role of virus-induced innate immunity in the mechanism of type 1 diabetes. METHODS: Groups of rats 21-25 days of age were left untreated, injected i.p. with 1×10(7) PFU of Kilham rat virus (KRV) only, or with 1-3 µg/g body-weight-purified toll-like receptor agonists on three consecutive days and infected with 1×10(7) PFU of KRV on the following day. Spleens and pancreatic lymph nodes were recovered 5 days after infection and used for gene array analysis. To test the role of inflammation in diabetes, rats injected with KRV only or Poly(I:C) plus KRV were also administered with 2 or 0.2 µg/g body weight of dexamethasone and followed for diabetes for 40 days. RESULTS: KRV induced the expression of a vast array of proinflammatory genes in pancreatic lymph nodes on day 5 following infection. Brief dexamethasone therapy downmodulated inflammation and completely blocked diabetes. CONCLUSIONS: Our data suggest a strong association between early virus-induced proinflammatory responses and islet destruction and raise the possibility that targeting innate immune pathways in the early stages of diabetes may be a useful strategy for disease prevention.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Inmunidad Innata , Infecciones por Parvoviridae/inmunología , Animales , Antiinflamatorios/uso terapéutico , Dexametasona/uso terapéutico , Diabetes Mellitus Tipo 1/prevención & control , Diabetes Mellitus Tipo 1/virología , Perfilación de la Expresión Génica , Parvovirus/inmunología , Ratas , Ratas Endogámicas BB , Ratas Endogámicas Lew
6.
Front Immunol ; 12: 702506, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34421908

RESUMEN

Type 1 diabetes (T1D) is a proinflammatory pathology that leads to the specific destruction of insulin producing ß-cells and hyperglycaemia. Much of the knowledge about type 1 diabetes (T1D) has focused on mechanisms of disease progression such as adaptive immune cells and the cytokines that control their function, whereas mechanisms linked with the initiation of the disease remain unknown. It has been hypothesized that in addition to genetics, environmental factors play a pivotal role in triggering ß-cell autoimmunity. The BioBreeding Diabetes Resistant (BBDR) and LEW1.WR1 rats have been used to decipher the mechanisms that lead to virus-induced T1D. Both animals develop ß-cell inflammation and hyperglycemia upon infection with the parvovirus Kilham Rat Virus (KRV). Our earlier in vitro and in vivo studies indicated that KRV-induced innate immune upregulation early in the disease course plays a causal role in triggering ß-cell inflammation and destruction. Furthermore, we recently found for the first time that infection with KRV induces inflammation in visceral adipose tissue (VAT) detectable as early as day 1 post-infection prior to insulitis and hyperglycemia. The proinflammatory response in VAT is associated with macrophage recruitment, proinflammatory cytokine and chemokine upregulation, endoplasmic reticulum (ER) and oxidative stress responses, apoptosis, and downregulation of adipokines and molecules that mediate insulin signaling. Downregulation of inflammation suppresses VAT inflammation and T1D development. These observations are strikingly reminiscent of data from obesity and type 2 diabetes (T2D) in which VAT inflammation is believed to play a causal role in disease mechanisms. We propose that VAT inflammation and dysfunction may be linked with the mechanism of T1D progression.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/virología , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/virología , Infecciones por Parvoviridae/inmunología , Animales , Humanos , Parvovirus/inmunología , Ratas
7.
J Proteomics ; 240: 104219, 2021 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-33831598

RESUMEN

Faecal proteomics studies have focussed on identification of microbial proteins, however; stool represents a valuable resource to interrogate the host interactions with the microbiota without the need for invasive intestinal biopsies. As the widely used enrichment method (differential centrifugation, DC) enriches for microbial proteins, we compared two other methods for enrichment of host proteins, termed 'host enriched' (HE) and ALL (all proteins). The HE and ALL protocols identified 1.8-fold more host proteins than DC while detecting a similar number of microbial proteins, but the methods had limited overlap in the specific microbial proteins detected. To maximize identification of both host and microbial proteins, samples were subjected to HE and the remaining material was used to perform DC. These two fractions displayed large differences in relative taxonomic abundance and cellular compartmentalization, with proteins from Bacteroidales and extracellular vesicles were enriched in the soluble HE component. The combination of data generated from these two fractions may allow identification of more distinct proteins than simply performing samples in duplicate or more complex fractionation techniques, or a single fraction could be chosen to suit the experimental hypothesis. SIGNIFICANCE: We compared how different stool protein preparation methods influenced the taxonomic and functional characteristics of microbial and host proteins identified. Surprisingly, a method designed to enrich for host proteins recovered a similar number of microbial protein groups to the method that specifically enriched intact bacterial cells. However, the taxonomic and subcellular origin of the microbial proteins differed considerably between the methods. By implementing a two-step method, we could maximize recovery of both host and microbial proteins derived from different cellular compartments and taxa.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Heces , Proteínas , Proteómica
8.
Clin Immunol ; 135(3): 401-11, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20167539

RESUMEN

We used the LEW1.WR1 rat to test the hypothesis that Kilham rat virus-induced innate immune activation is involved in the mechanism of autoimmune diabetes. Animals were treated with dexamethasone, an anti-inflammatory glucocorticoid, beginning on the day of infection. Administering dexamethasone on five consecutive days completely blocked the disease. Strikingly, a single dose of dexamethasone was sufficient to prevent islet destruction. Dexamethasone downmodulated inflammation and restored normal ratios between CD8(+) and CD4(+)CD25(+)Foxp3(+) cells in the spleen. Finally, dexamethasone therapy lowered the frequency of splenic anti-virus CD8(+) T cells, but did not interfere with the ability of the host to generate anti-KRV antibodies and eliminate the virus from the spleen. Our data demonstrate a strong association between early virus-induced proinflammatory responses and islet destruction and raise the possibility that targeting innate immune pathways in the early stages of diabetes may be a useful strategy for disease prevention.


Asunto(s)
Antiinflamatorios/uso terapéutico , Dexametasona/uso terapéutico , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/prevención & control , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Separación Celular , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/virología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/virología , Citometría de Flujo , Inmunidad Innata/inmunología , Infecciones por Parvoviridae/complicaciones , Parvovirus , Ratas , Ratas Endogámicas Lew , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
9.
Adv Exp Med Biol ; 654: 585-610, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20217515

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease that results in the progressive loss of insulin producing cells. Studies performed in humans with T1D and animal models of the disease over the past two decades have suggested a key role for the adaptive immune system in disease mechanisms. The role of the innate immune system in triggering T1D was shown only recently. Research in this area was greatly facilitated by the discovery of toll-like receptors (TLRs) that were found to be a key component of the innate immune system that detect microbial infections and initiate antimicrobial host defense responses. New data indicate that in some situations, the innate immune system is associated with mechanisms triggering autoimmune diabetes. In fact, studies preformed in the BioBreeding Diabetes Resistant (BBDR) and LEW1.WR1 rat models of T1D demonstrate that virus infection leads to islet destruction via mechanisms that may involve TLR9-induced innate immune system activation. Data from these studies also show that TLR upregulation can synergize with virus infection to dramatically increase disease penetrance. Reports from murine models of T1D implicate both MyD88-dependent and MyD88-independent pathways in the course of disease. The new knowledge about the role of innate immune pathways in triggering islet destruction could lead to the discovery of new molecules that may be targeted for disease prevention.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Regulación de la Expresión Génica , Receptores Toll-Like/metabolismo , Animales , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Inflamación , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos NOD , Modelos Biológicos , Factor 88 de Diferenciación Mieloide/química , Ratas , Receptor Toll-Like 9/metabolismo
10.
Curr Mol Med ; 9(1): 52-68, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19199942

RESUMEN

Toll-like receptors (TLRs) and the innate immune system play a key role in sensing and eliminating microbial infections. Interactions between TLRs and their ligands expressed by microbial pathogens induce a cascade of intracellular signaling events, culminating in the upregulation of proinflammatory pathways. Over the past two decades, numerous studies have established the role of the acquired immune system in the mechanism triggering type 1 diabetes (T1D). The recent discovery of TLRs has led to the recognition that the innate immune system may act, under some circumstances, as a double-edged sword. In addition to its beneficial role in host defense, it may lead to upregulation of proinflammatory autoimmune responses, islet destruction and diabetes. Indeed, recent observations are consistent with the hypothesis that altered innate functions exist in patients with T1D and could be part of the mechanism leading to disease onset, but the underlying mechanisms and the relevance of these alterations to early events triggering disease remain to be identified. Data obtained from mouse and rat models of T1D implicated TLR pathways in both disease induction and prevention. In both the NOD mouse and diabetes-prone BB (BBDP) rat, TLR upregulation can suppress disease. In the BioBreeding Diabetes Resistant (BBDR) rat, however, diabetes induced by virus infection involves the upregulation of TLR9 pathways, and generic TLR upregulation synergizes with virus infection on diabetes induction. Studies performed in mouse models of T1D with spontaneous or induced T1D implicate TLR1, TLR2, TLR3, and TLR7 in disease mechanisms. The finding that TLR pathways are involved in mediating islet inflammation holds great promise for identifying new molecules that could potentially be targeted to specifically suppress the autoimmune process in individuals at high risk for disease development. The potential link between TLR upregulation and autoimmunity emphasizes the need for caution in using new therapies involving TLR agonists as vaccine adjuvants.


Asunto(s)
Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/inmunología , Inmunidad Innata , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Diabetes Mellitus Tipo 1/genética , Humanos , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos NOD , Factor 88 de Diferenciación Mieloide/inmunología , Ratas , Ratas Endogámicas BB , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Regulación hacia Arriba
11.
Methods Mol Biol ; 2128: 107-114, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32180189

RESUMEN

Studies performed in humans and animal models have implicated the environment in the etiology of type 1 diabetes (T1D), but the nature and timing of the interactions triggering ß cell autoimmunity are poorly understood. Virus infections have been postulated to be involved in disease mechanisms, but the underlying mechanisms are not known. It is exceedingly difficult to establish a cause-and-effect relationship between viral infection and diabetes in humans. Thus, we have used the BioBreeding Diabetes-Resistant (BBDR) and the LEW1.WR1 rat models of virus-induced disease to elucidate how virus infection leads to T1D. The immunophenotype of these strains is normal, and spontaneous diabetes does not occur in a specific pathogen-free environment. However, ß cell inflammation and diabetes with many similarities to the human disease are induced by infection with the parvovirus Kilham rat virus (KRV). KRV-induced diabetes in the BBDR and LEW1.WR1 rat models is limited to young animals and can be induced in both male and female rats. Thus, these animals provide a powerful experimental tool to identify mechanisms underlying virus-induced T1D development.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/virología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/virología , Infecciones por Parvoviridae/complicaciones , Parvovirus/inmunología , Animales , Glucemia/análisis , Técnicas de Cultivo de Célula , Línea Celular , Diabetes Mellitus Experimental/diagnóstico , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/etiología , Femenino , Glucosuria , Inflamación/inmunología , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/virología , Masculino , Ratas , Ratas Endogámicas BB , Ratas Endogámicas Lew , Ratas Endogámicas WF
12.
Clin Immunol ; 131(1): 11-23, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19185542

RESUMEN

There is a consensus among epidemiologists that the worldwide incidence rate of type 1 diabetes has been rising in recent decades. The cause of this rise is unknown, but epidemiological studies suggest the involvement of environmental factors, and viral infections in particular. Data demonstrating a cause-and-effect relationship between microbial infections and type 1 diabetes and how viruses may cause disease in humans are currently lacking. However, new evidence from animal models supports the hypothesis that viruses induce disease via mechanisms linked with innate immune upregulation. In the BioBreeding Diabetes Resistant rat, infection with a parvovirus induces islet destruction via upregulation of the toll-like receptor 9 (TLR9) signaling pathway. Data from mouse models of diabetes implicate TLR2, TLR3, and TLR7 in the disease process. Understanding the link between environmental agents and innate immune pathways involved in early stages of diabetes may advance the design of immune interventions to prevent disease in genetically susceptible individuals.


Asunto(s)
Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/virología , Virosis/epidemiología , Virosis/virología , Animales , Modelos Animales de Enfermedad , Humanos , Incidencia
13.
Clin Immunol ; 132(1): 103-15, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19328037

RESUMEN

We have recently demonstrated that upregulation of the innate immune system plays a key role in KRV-induced autoimmune diabetes in the BBDR rat, but the nature of this proinflammatory reaction has not yet been addressed. Using a DNA microarray approach, we identified 569 genes upregulated in pancreatic lymph nodes following virus infection. Among the most highly activated are IL-1 pathways, IFN-gamma-induced chemokines, and genes associated with interferon production and signaling. Ex vivo and in vitro studies indicate that KRV upregulates proinflammatory cytokines and chemokines in B lymphocytes and Flt-3L-induced plasmacytoid DCs (pDCs). Finally, in contrast to KRV, infection of BBDR rats with the non-diabetogenic KRV homologue H-1 parvovirus fails to induce a robust proinflammatory response in pancreatic lymph nodes. Our findings provide new insights into KRV-induced innate immune pathways that may play a role in early mechanisms leading to islet inflammation and diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Perfilación de la Expresión Génica , Inmunidad Innata/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Varianza , Animales , Análisis por Conglomerados , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/virología , Femenino , Parvovirus H-1/fisiología , Interacciones Huésped-Patógeno , Ganglios Linfáticos/metabolismo , Masculino , Páncreas/inmunología , Páncreas/metabolismo , Parvovirus/fisiología , Ratas , Ratas Endogámicas BB , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
J Endocrinol ; 238(1): 61-75, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29743341

RESUMEN

The etiopathogenesis of type 1 diabetes (T1D) remains poorly understood. We used the LEW1.WR1 rat model of Kilham rat virus (KRV)-induced T1D to better understand the role of the innate immune system in the mechanism of virus-induced disease. We observed that infection with KRV results in cell influx into visceral adipose tissue soon following infection prior to insulitis and hyperglycemia. In sharp contrast, subcutaneous adipose tissue is free of cellular infiltration, whereas ß cell inflammation and diabetes are observed beginning on day 14 post infection. Immunofluorescence studies further demonstrate that KRV triggers CD68+ macrophage recruitment and the expression of KRV transcripts and proinflammatory cytokines and chemokines in visceral adipose tissue. Adipocytes from naive rats cultured in the presence of KRV express virus transcripts and upregulate cytokine and chemokine gene expression. KRV induces apoptosis in visceral adipose tissue in vivo, which is reflected by positive TUNEL staining and the expression of cleaved caspase-3. Moreover, KRV leads to an oxidative stress response and downregulates the expression of adipokines and genes associated with mediating insulin signaling. Activation of innate immunity with Poly I:C in the absence of KRV leads to CD68+ macrophage recruitment to visceral adipose tissue and a decrease in adipokine expression detected 5 days following Poly (I:C) treatment. Finally, proof-of-principle studies show that brief anti-inflammatory steroid therapy suppresses visceral adipose tissue inflammation and protects from virus-induced disease. Our studies provide evidence raising the hypothesis that visceral adipose tissue inflammation and dysfunction may be involved in early mechanisms triggering ß cell autoimmunity.


Asunto(s)
Tejido Adiposo/patología , Tejido Adiposo/fisiopatología , Diabetes Mellitus Tipo 1/virología , Inflamación/complicaciones , Paniculitis/complicaciones , Parvovirus/fisiología , Tejido Adiposo/inmunología , Tejido Adiposo/virología , Animales , Células Cultivadas , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Femenino , Inmunidad Innata/fisiología , Inflamación/patología , Inflamación/virología , Macrófagos/fisiología , Masculino , Paniculitis/inmunología , Paniculitis/patología , Paniculitis/virología , Parvovirus/inmunología , Ratas , Transducción de Señal/inmunología
15.
Diabetes Care ; 41(10): 2178-2186, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30100563

RESUMEN

OBJECTIVE: Dysbiosis of the gut microbiota has been linked to disease pathogenesis in type 1 diabetes, yet the functional consequences to the host of this dysbiosis are unknown. We investigated the functional interactions between the microbiota and the host associated with type 1 diabetes disease risk. RESEARCH DESIGN AND METHODS: We performed a cross-sectional analysis of stool samples from subjects with recent-onset type 1 diabetes (n = 33), islet autoantibody-positive subjects (n = 17), low-risk autoantibody-negative subjects (n = 29), and healthy subjects (n = 22). Metaproteomic analysis was used to identify gut- and pancreas-derived host and microbial proteins, and these data were integrated with sequencing-based microbiota profiling. RESULTS: Both human (host-derived) proteins and microbial-derived proteins could be used to differentiate new-onset and islet autoantibody-positive subjects from low-risk subjects. Significant alterations were identified in the prevalence of host proteins associated with exocrine pancreas output, inflammation, and mucosal function. Integrative analysis showed that microbial taxa associated with host proteins involved in maintaining function of the mucous barrier, microvilli adhesion, and exocrine pancreas were depleted in patients with new-onset type 1 diabetes. CONCLUSIONS: These data support that patients with type 1 diabetes have increased intestinal inflammation and decreased barrier function. They also confirmed that pancreatic exocrine dysfunction occurs in new-onset type 1 diabetes and show for the first time that this dysfunction is present in high-risk individuals before disease onset. The data identify a unique type 1 diabetes-associated signature in stool that may be useful as a means to monitor disease progression or response to therapies aimed at restoring a healthy microbiota.


Asunto(s)
Diabetes Mellitus Tipo 1/microbiología , Diabetes Mellitus Tipo 1/fisiopatología , Disbiosis/microbiología , Disbiosis/fisiopatología , Microbioma Gastrointestinal/fisiología , Interacciones Huésped-Patógeno/fisiología , Adolescente , Adulto , Proteínas Bacterianas/fisiología , Niño , Preescolar , Estudios Transversales , Heces/microbiología , Femenino , Humanos , Inflamación/microbiología , Intestinos/fisiopatología , Masculino , Persona de Mediana Edad , Páncreas/metabolismo , Páncreas/fisiopatología , Proteínas/fisiología , Proteómica , Medición de Riesgo , Factores de Riesgo , Adulto Joven
16.
PLoS One ; 12(9): e0183786, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28886045

RESUMEN

We recently hypothesized that the intestinal microbiota and the innate immune system play key roles in the mechanism of Kilham Rat Virus-induced type 1 diabetes in the LEW1.WR1 rat. We used this animal model to test the hypothesis that maternal therapy with short-chain fatty acids can modulate the intestinal microbiota and reverse virus-induced proinflammatory responses and type 1 diabetes in rat offspring. We observed that administration of short-chain fatty acids to rat breeders via drinking water prior to pregnancy and further treatment of the offspring with short-chain fatty acids after weaning led to disease amelioration. In contrast, rats that were administered short-chain fatty acids beginning at weaning were not protected from type 1 diabetes. Short-chain fatty acid therapy exerted a profound effect on the intestinal microbiome in the offspring reflected by a reduction and an increase in the abundances of Firmicutes and Bacteroidetes taxa, respectively, on day 5 post-infection, and reversed virus-induced alterations in certain bacterial taxa. Principal component analysis and permutation multivariate analysis of variance tests further revealed that short-chain fatty acids induce a distinct intestinal microbiota compared with uninfected animals or rats that receive the virus only. Short-chain fatty acids downregulated Kilham Rat Virus-induced proinflammatory responses in the intestine. Finally, short-chain fatty acids altered the B and T cell compartments in Peyer's patches. These data demonstrate that short-chain fatty acids can reshape the intestinal microbiota and prevent virus-induced islet autoimmunity and may therefore represent a useful therapeutic strategy for disease prevention.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Ácidos Grasos Volátiles/uso terapéutico , Microbioma Gastrointestinal/efectos de los fármacos , Intestinos/inmunología , Intestinos/microbiología , Animales , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Reacción en Cadena de la Polimerasa , Embarazo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
PLoS One ; 12(3): e0173968, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28301545

RESUMEN

Type 1 diabetes (T1D) is an autoimmune proinflammatory disease with no effective intervention. A major obstacle in developing new immunotherapies for T1D is the lack of means for monitoring immune responsiveness to experimental therapies. The LEW1.WR1 rat develops autoimmunity following infection with the parvovirus Kilham rat virus (KRV) via mechanisms linked with activation of proinflammatory pathways and alterations in the gut bacterial composition. We used this animal to test the hypothesis that intervention with agents that block innate immunity and diabetes is associated with a shift in the gut microbiota. We observed that infection with KRV results in the induction of proinflammatory gene activation in both the spleen and pancreatic lymph nodes. Furthermore, administering animals the histone deacetylase inhibitor ITF-2357 and IL-1 receptor antagonist (Anakinra) induced differential STAT-1 and the p40 unit of IL-12/IL-23 gene expression. Sequencing of bacterial 16S rRNA genes demonstrated that both ITF-2357 and Anakinra alter microbial diversity. ITF-2357 and Anakinra modulated the abundance of 23 and 8 bacterial taxa in KRV-infected animals, respectively, of which 5 overlapped between the two agents. Lastly, principal component analysis implied that ITF-2357 and Anakinra induce distinct gut microbiomes compared with those from untreated animals or rats provided KRV only. Together, the data suggest that ITF-2357 and Anakinra differentially influence the innate immune system and the intestinal microbiota and highlight the potential use of the gut microbiome as a surrogate means of assessing anti-inflammatory immune effects in type 1 diabetes.


Asunto(s)
Biomarcadores/metabolismo , Diabetes Mellitus Tipo 1/terapia , Intestinos/microbiología , Microbiota , Animales , Biodiversidad , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/microbiología , Heces/microbiología , Secuenciación de Nucleótidos de Alto Rendimiento , Ácidos Hidroxámicos/farmacología , Inmunidad Innata , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Microbiota/efectos de los fármacos , Pancrelipasa/efectos de los fármacos , Pancrelipasa/inmunología , Parvovirus/patogenicidad , Análisis de Componente Principal , ARN Ribosómico 16S/genética , Ratas , Ratas Endogámicas Lew , Bazo/efectos de los fármacos , Bazo/inmunología
18.
Diabetes ; 64(10): 3510-20, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26068542

RESUMEN

We tested the hypothesis that alterations in the intestinal microbiota are linked with the progression of type 1 diabetes (T1D). Herein, we present results from a study performed in subjects with islet autoimmunity living in the U.S. High-throughput sequencing of bacterial 16S rRNA genes and adjustment for sex, age, autoantibody presence, and HLA indicated that the gut microbiomes of seropositive subjects differed from those of autoantibody-free first-degree relatives (FDRs) in the abundance of four taxa. Furthermore, subjects with autoantibodies, seronegative FDRs, and new-onset patients had different levels of the Firmicutes genera Lactobacillus and Staphylococcus compared with healthy control subjects with no family history of autoimmunity. Further analysis revealed trends toward increased and reduced abundances of the Bacteroidetes genera Bacteroides and Prevotella, respectively, in seropositive subjects with multiple versus one autoantibody. Canonical discriminant analysis suggested that the gut microbiomes of autoantibody-positive individuals and seronegative FDRs clustered together but separate from those of new-onset patients and unrelated healthy control subjects. Finally, no differences in biodiversity were evident in seropositive versus seronegative FDRs. These observations suggest that altered intestinal microbiota may be associated with disease susceptibility.


Asunto(s)
Bacterias/clasificación , Diabetes Mellitus Tipo 1/etiología , Microbioma Gastrointestinal/fisiología , Islotes Pancreáticos/inmunología , Adolescente , Adulto , Autoanticuerpos/sangre , Autoinmunidad , Estudios de Casos y Controles , Niño , Preescolar , Estudios de Cohortes , Susceptibilidad a Enfermedades , Heces/microbiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , ARN Ribosómico 16S/química , ARN Ribosómico 16S/genética , Estados Unidos , Adulto Joven
19.
JOP ; 3(3): 76-82, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12004164

RESUMEN

CONTEXT: Type 1 diabetes mellitus is a T cell mediated autoimmune disease with no known methods of prevention. The BioBreeding rat is used as an animal model for the study of human Type 1 diabetes. In spite of a severe lymphopenia, these animals develop spontaneous diabetes at the age of 10-12 weeks. OBJECTIVE: To examine whether anti-inflammatory gene therapy could be used to prevent autoimmune diabetes in the BioBreeding rat. DESIGN: A retroviral DNA vector, MSCVneo.IL-4, carrying the DNA sequence encoding the rat interleukin-4, was designed to transfer interleukin-4 to BioBreeding rats. Spleen cells of prediabetic animals were activated and transduced in vitro with replication-defective retroviruses expressing the MSCVneo.IL-4 vector. These lymphocytes were subsequently administered intraperitoneally to 3-4 week old prediabetic BioBreeding rats. Control animals were reconstituted with spleen cells transduced with MSCVneo vector. RESULTS: The neo gene marker was detectable by RT-PCR in rat spleen cells of up to 6 to 12 months after treatment. Fifty percent (6 out of 12) of the animals treated were protected from autoimmune disease development. CONCLUSION: Our results suggest that the BioBreeding rat can be used as a useful model to develop gene therapy regimens for diabetes. These studies provide further support for the hypothesis that interleukin-4 based gene therapy may have potential clinical value for preventing autoimmune diabetes in humans.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Terapia Genética/métodos , Interleucina-4/administración & dosificación , Linfocitos/virología , Retroviridae , Células 3T3 , Animales , Línea Celular , Transformación Celular Viral/genética , Esquema de Medicación , Femenino , Regulación Viral de la Expresión Génica , Vectores Genéticos/genética , Humanos , Inyecciones Intraperitoneales , Interleucina-4/biosíntesis , Interleucina-4/genética , Transfusión de Linfocitos , Masculino , Ratones , Ratas , Ratas Endogámicas BB , Ratas Endogámicas WF , Retroviridae/genética , Bazo/citología , Bazo/trasplante , Bazo/virología
20.
Virology ; 468-470: 19-27, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25129435

RESUMEN

We used the LEW1.WR1 rat model of Kilham Rat Virus (KRV)-induced type 1 diabetes (T1D) to test the hypothesis that disease mechanisms are linked with beta cell infection and intra-islet immune activation prior to insulitis. KRV induces genes involved in type I and type II interferon pathways in islet cell lines in vitro and in islets from day-5-infected animals in vivo via mechanisms that do not involve insulitis, beta cell apoptosis, or impaired insulin expression. Immunohistochemistry studies indicated that KRV protein is expressed in beta cells 5 days following infection. KRV induces the phosphorylation of Janus Kinase 1/2 (JAK1/2) and signal transducer and activator of transcription 1 (STAT-1) in islet cells via a mechanism that could involve TLR9 and NF-κB pathways. These data demonstrate for the first time that KRV-induced islet destruction is associated with beta cell infection and intra-islet innate immune upregulation early in the disease process.


Asunto(s)
Diabetes Mellitus Tipo 1/virología , Islotes Pancreáticos/fisiología , Janus Quinasa 1/metabolismo , Janus Quinasa 2/metabolismo , Parvovirus/fisiología , Factor de Transcripción STAT1/metabolismo , Animales , Línea Celular , Diabetes Mellitus Tipo 1/patología , Inflamación/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/virología , Janus Quinasa 1/genética , Janus Quinasa 2/genética , Ratas , Ratas Endogámicas , Factor de Transcripción STAT1/genética , Transducción de Señal , Organismos Libres de Patógenos Específicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA