Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 149(17)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36017799

RESUMEN

Signals from the endothelium play a pivotal role in pancreatic lineage commitment. As such, the fate of the epithelial cells relies heavily on the spatiotemporal recruitment of the endothelial cells to the embryonic pancreas. Although it is known that VEGFA secreted by the epithelium recruits the endothelial cells to the specific domains within the developing pancreas, the mechanism that controls the timing of such recruitment is poorly understood. Here, we have assessed the role of focal adhesion kinase (FAK) in mouse pancreatic development based on our observation that the presence of the enzymatically active form of FAK (pFAK) in the epithelial cells is inversely correlated with vessel recruitment. To study the role of FAK in the pancreas, we conditionally deleted the gene encoding focal adhesion kinase in the developing mouse pancreas. We found that homozygous deletion of Fak (Ptk2) during embryogenesis resulted in ectopic epithelial expression of VEGFA, abnormal endothelial recruitment and a delay in endocrine and acinar cell differentiation. The heterozygous mutants were born with no pancreatic phenotype but displayed gradual acinar atrophy due to cell polarity defects in exocrine cells. Together, our findings imply a role for FAK in controlling the timing of pancreatic lineage commitment and/or differentiation in the embryonic pancreas by preventing endothelial recruitment to the embryonic pancreatic epithelium.


Asunto(s)
Células Endoteliales , Animales , Diferenciación Celular/genética , Proteína-Tirosina Quinasas de Adhesión Focal , Homocigoto , Ratones , Eliminación de Secuencia
2.
Nat Immunol ; 11(3): 225-31, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20139986

RESUMEN

Autoreactive CD4(+) T cells are involved in the pathogenesis of many autoimmune diseases, but the antigens that stimulate their responses have been difficult to identify and in most cases are not well defined. In the nonobese diabetic (NOD) mouse model of type 1 diabetes, we have identified the peptide WE14 from chromogranin A (ChgA) as the antigen for highly diabetogenic CD4(+) T cell clones. Peptide truncation and extension analysis shows that WE14 bound to the NOD mouse major histocompatibility complex class II molecule I-A(g7) in an atypical manner, occupying only the carboxy-terminal half of the I-A(g7) peptide-binding groove. This finding extends the list of T cell antigens in type 1 diabetes and supports the idea that autoreactive T cells respond to unusually presented self peptides.


Asunto(s)
Autoantígenos/inmunología , Cromogranina A/inmunología , Diabetes Mellitus Tipo 1/inmunología , Células Secretoras de Insulina/inmunología , Fragmentos de Péptidos/inmunología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Epítopos/inmunología , Antígenos HLA-A , Espectrometría de Masas , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Datos de Secuencia Molecular
3.
Curr Diab Rep ; 21(3): 9, 2021 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-33547977

RESUMEN

PURPOSE OF REVIEW: Type 1 diabetes (T1D) can be managed by insulin replacement, but it is still associated with an increased risk of microvascular/cardiovascular complications. There is considerable interest in antigen-specific approaches for treating T1D due to their potential for a favorable risk-benefit ratio relative to non-specific immune-based treatments. Here we review recent antigen-specific tolerance approaches using auto-antigen and/or immunomodulatory agents in NOD mice and provide insight into seemingly contradictory findings. RECENT FINDINGS: Although delivery of auto-antigen alone can prevent T1D in NOD mice, this approach may be prone to inconsistent results and has not demonstrated an ability to reverse established T1D. Conversely, several approaches that promote presentation of auto-antigen in a tolerogenic context through cell/tissue targeting, delivery system properties, or the delivery of immunomodulatory agents have had success in reversing recent-onset T1D in NOD mice. While initial auto-antigen based approaches were unable to substantially influence T1D progression clinically, recent antigen-specific approaches have promising potential.


Asunto(s)
Diabetes Mellitus Tipo 1 , Tolerancia Inmunológica , Animales , Antígenos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Humanos , Insulina , Ratones , Ratones Endogámicos NOD , Linfocitos T Reguladores
4.
Am J Pathol ; 189(7): 1413-1422, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31054988

RESUMEN

Obesity is a major risk factor for type 2 diabetes because of chronic hepatic inflammation and resultant insulin resistance. Hepatocyte growth factor (HGF) is responsible for resetting hepatic homeostasis after injury following activation by urokinase-type plasminogen activator (u-PA; encoded by the PLAU gene). Plasminogen activator inhibitor type-1 (PAI-1; encoded by the SERPINE1 gene), a u-PA inhibitor and antifibrinolytic agent, is often elevated in obesity and is linked to cardiovascular events. We hypothesized that, in addition to its role in preventing fibrinolysis, elevated PAI-1 inhibits HGF's activation by u-PA and the resultant anti-inflammatory and hepatoprotective properties. Wild-type and PAI-1 knockout (KO) mice on a high-fat diet both became significantly heavier than lean controls; however, the obese KO mice demonstrated improved glucose metabolism compared with wild-type mice. Obese KO mice also exhibited an increase in conversion of latent single-chain HGF to active two-chain HGF, coinciding with an increase in the phosphorylation of the HGF receptor (HGFR or MET, encoded by the MET gene), as well as dampened inflammation. These results strongly suggest that, in addition to its other functions, PAI-mediated inhibition of HGF activation prohibits the resolution of inflammation in the context of obesity-induced type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Obesidad/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Animales , Diabetes Mellitus Tipo 2/inducido químicamente , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , Inhibidor 1 de Activador Plasminogénico/genética , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo
5.
FASEB J ; 33(1): 1330-1346, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30113881

RESUMEN

The incidence and prevalence of inflammatory bowel disease (IBD) are increasing worldwide. IBD is known to be multifactorial, but inflammatory signaling within the intestinal epithelium and a subsequent failure of the intestinal epithelial barrier have been shown to play essential roles in disease pathogenesis. CaMKIV is a multifunctional protein kinase associated with inflammation and cell cycle regulation. CaMKIV has been extensively studied in autoimmune diseases, but a role in idiopathic intestinal inflammation has not been described. In this study, active CaMKIV was highly expressed within the intestinal epithelium of humans with ulcerative colitis and wild-type (WT) mice with experimental induced colitis. Clinical disease severity directly correlates with CaMKIV activation, as does expression of proinflammatory cytokines and histologic features of colitis. In WT mice, CaMKIV activation is associated with increases in expression of 2 cell cycle proarrest signals: p53 and p21. Cell cycle arrest inhibits proliferation of the intestinal epithelium and ultimately results in compromised intestinal epithelial barrier integrity, further perpetuating intestinal inflammation during experimental colitis. Using a CaMKIV null mutant mouse, we demonstrate that a loss of CaMKIV protects against murine DSS colitis. Small molecules targeting CaMKIV activation may provide therapeutic benefit for patients with IBD.-Cunningham, K. E., Novak, E. A., Vincent, G., Siow, V. S., Griffith, B. D., Ranganathan, S., Rosengart, M. R., Piganelli, J. D., Mollen, K. P. Calcium/calmodulin-dependent protein kinase IV (CaMKIV) activation contributes to the pathogenesis of experimental colitis via inhibition of intestinal epithelial cell proliferation.


Asunto(s)
Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/metabolismo , Proliferación Celular , Colitis/enzimología , Colitis/patología , Mucosa Intestinal/patología , Animales , Calcio/metabolismo , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/genética , Colitis/inducido químicamente , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/patología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Sulfato de Dextran/toxicidad , Activación Enzimática , Humanos , Mucosa Intestinal/enzimología , Ratones , Ratones Noqueados , Transducción de Señal
6.
Am J Transplant ; 18(8): 1879-1889, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29464912

RESUMEN

Islet transplantation has become a well-established therapy for select patients with type 1 diabetes. Viability and engraftment can be compromised by the generation of oxidative stress encountered during isolation and culture. We evaluated whether the administration of BMX-001 (MnTnBuOE-2-PyP5+ [Mn(III) meso-tetrakis-(N-b-butoxyethylpyridinium-2-yl)porphyrin]) and its earlier derivative, BMX-010 (MnTE-2-PyP [Mn(III) meso-tetrakis-(N-methylpyridinium-2-yl)porphyrin]) could improve islet function and engraftment outcomes. Long-term culture of human islets with BMX-001, but not BMX-010, exhibited preserved in vitro viability. Murine islets isolated and cultured for 24 hours with 34 µmol/L BMX-001 exhibited improved insulin secretion (n = 3 isolations, P < .05) in response to glucose relative to control islets. In addition, 34 µmol/L BMX-001-supplemented murine islets exhibited significantly reduced apoptosis as indicated by terminal deoxynucleotidyl transferase dUTP nick end labeling, compared with nontreated control islets (P < .05). Murine syngeneic islets transplanted under the kidney capsule at a marginal dose of 150 islets revealed 58% of 34 µmol/L BMX-001-treated islet recipients became euglycemic (n = 11 of 19) compared with 19% of nontreated control islet recipients (n = 3 of 19, P < .05). Of murine recipients receiving a marginal dose of human islets cultured with 34 µmol/L BMX-001, 92% (n = 12 of 13) achieved euglycemia compared with 57% of control recipients (n = 8 of 14, P = .11). These results demonstrate that the administration of BMX-001 enhances in vitro viability and augments murine marginal islet mass engraftment.


Asunto(s)
Apoptosis/efectos de los fármacos , Materiales Biomiméticos/farmacología , Diabetes Mellitus Experimental/prevención & control , Islotes Pancreáticos/efectos de los fármacos , Metaloporfirinas/farmacología , Animales , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Glucosa/farmacología , Supervivencia de Injerto , Humanos , Insulina/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Masculino , Ratones , Ratones Endogámicos BALB C , Oxidación-Reducción , Superóxido Dismutasa/metabolismo
7.
J Biol Chem ; 290(25): 15581-15594, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-25944898

RESUMEN

Excessive endogenous glucose production contributes to fasting hyperglycemia in diabetes. FoxO6 is a distinct member of the FoxO subfamily. To elucidate the role of FoxO6 in hepatic gluconeogenesis and assess its contribution to the pathogenesis of fasting hyperglycemia in diabetes, we generated FoxO6 knock-out (FoxO6-KO) mice followed by determining the effect of FoxO6 loss-of-function on hepatic gluconeogenesis under physiological and pathological conditions. FoxO6 depletion attenuated hepatic gluconeogenesis and lowered fasting glycemia in FoxO6-KO mice. FoxO6-deficient primary hepatocytes were associated with reduced capacities to produce glucose in response to glucagon. When fed a high fat diet, FoxO6-KO mice exhibited significantly enhanced glucose tolerance and reduced blood glucose levels accompanied by improved insulin sensitivity. These effects correlated with attenuated hepatic gluconeogenesis in FoxO6-KO mice. In contrast, wild-type littermates developed fat-induced glucose intolerance with a concomitant induction of fasting hyperinsulinemia and hyperglycemia. Furthermore, FoxO6-KO mice displayed significantly diminished macrophage infiltration into liver and adipose tissues, correlating with the reduction of macrophage expression of C-C chemokine receptor 2 (CCR2), a factor that is critical for regulating macrophage recruitment in peripheral tissues. Our data indicate that FoxO6 depletion protected against diet-induced glucose intolerance and insulin resistance by attenuating hepatic gluconeogenesis and curbing macrophage infiltration in liver and adipose tissues in mice.


Asunto(s)
Grasas de la Dieta/efectos adversos , Factores de Transcripción Forkhead/metabolismo , Gluconeogénesis/efectos de los fármacos , Hiperglucemia/metabolismo , Hiperinsulinismo/metabolismo , Hígado/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Células Cultivadas , Grasas de la Dieta/farmacología , Factores de Transcripción Forkhead/genética , Gluconeogénesis/genética , Hiperglucemia/inducido químicamente , Hiperglucemia/genética , Hiperglucemia/patología , Hiperglucemia/prevención & control , Hiperinsulinismo/inducido químicamente , Hiperinsulinismo/genética , Hiperinsulinismo/patología , Hiperinsulinismo/prevención & control , Hígado/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Receptores CCR2/genética , Receptores CCR2/metabolismo
8.
J Autoimmun ; 72: 33-46, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27173406

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease characterized by pancreatic ß cell destruction induced by islet reactive T cells that have escaped central tolerance. Many physiological and environmental triggers associated with T1D result in ß cell endoplasmic reticulum (ER) stress and dysfunction, increasing the potential for abnormal post-translational modification (PTM) of proteins. We hypothesized that ß cell ER stress induced by environmental and physiological conditions generates abnormally-modified proteins for the T1D autoimmune response. To test this hypothesis we exposed the murine CD4(+) diabetogenic BDC2.5 T cell clone to murine islets in which ER stress had been induced chemically (Thapsigargin). The BDC2.5 T cell IFNγ response to these cells was significantly increased compared to non-treated islets. This ß cell ER stress increased activity of the calcium (Ca(2+))-dependent PTM enzyme tissue transglutaminase 2 (Tgase2), which was necessary for full stress-dependent immunogenicity. Indeed, BDC2.5 T cells responded more strongly to their antigen after its modification by Tgase2. Finally, exposure of non-antigenic murine insulinomas to chemical ER stress in vitro or physiological ER stress in vivo caused increased ER stress and Tgase2 activity, culminating in higher BDC2.5 responses. Thus, ß cell ER stress induced by chemical and physiological triggers leads to ß cell immunogenicity through Ca(2+)-dependent PTM. These findings elucidate a mechanism of how ß cell proteins are modified and become immunogenic, and reveal a novel opportunity for preventing ß cell recognition by autoreactive T cells.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Estrés del Retículo Endoplásmico/inmunología , Células Secretoras de Insulina/inmunología , Secuencia de Aminoácidos , Animales , Autoantígenos/genética , Autoantígenos/inmunología , Autoinmunidad/genética , Western Blotting , Linfocitos T CD4-Positivos/metabolismo , Calcio/inmunología , Calcio/metabolismo , Línea Celular , Células Cultivadas , Cromogranina A/genética , Cromogranina A/inmunología , Cromogranina A/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Estrés del Retículo Endoplásmico/genética , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/inmunología , Proteínas de Unión al GTP/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Modelos Inmunológicos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Procesamiento Proteico-Postraduccional/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masas en Tándem , Transglutaminasas/genética , Transglutaminasas/inmunología , Transglutaminasas/metabolismo
9.
Gastroenterology ; 147(5): 1106-18.e11, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25128759

RESUMEN

BACKGROUND & AIMS: Although the cells that contribute to pancreatic regeneration have been widely studied, little is known about the mediators of this process. During tissue regeneration, infiltrating macrophages debride the site of injury and coordinate the repair response. We investigated the role of macrophages in pancreatic regeneration in mice. METHODS: We used a saporin-conjugated antibody against CD11b to reduce the number of macrophages in mice following diphtheria toxin receptor-mediated cell ablation of pancreatic cells, and evaluated the effects on pancreatic regeneration. We analyzed expression patterns of infiltrating macrophages after cell-specific injury or from the pancreas of nonobese diabetic mice. We developed an in vitro culture system to study the ability of macrophages to induce cell-specific regeneration. RESULTS: Depletion of macrophages impaired pancreatic regeneration. Macrophage polarization, as assessed by expression of tumor necrosis factor-α, interleukin 6, interleukin 10, and CD206, depended on the type of injury. The signals provided by polarized macrophages promoted lineage-specific generation of acinar or endocrine cells. Macrophage from nonobese diabetic mice failed to provide signals necessary for ß-cell generation. CONCLUSIONS: Macrophages produce cell type-specific signals required for pancreatic regeneration in mice. Additional study of these processes and signals might lead to new approaches for treating type 1 diabetes or pancreatitis.


Asunto(s)
Células Acinares/metabolismo , Comunicación Celular , Linaje de la Célula , Proliferación Celular , Microambiente Celular , Células Secretoras de Insulina/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Regeneración , Células Acinares/patología , Factores de Edad , Animales , Anticuerpos/toxicidad , Biomarcadores/metabolismo , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Comunicación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Toxina Diftérica/toxicidad , Modelos Animales de Enfermedad , Inmunoconjugados/toxicidad , Células Secretoras de Insulina/patología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Fenotipo , Proteínas Inactivadoras de Ribosomas Tipo 1/toxicidad , Saporinas , Transducción de Señal
10.
Curr Diab Rep ; 15(11): 90, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26370701

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease in which progressive loss of self-tolerance, evidenced by accumulation of auto-antibodies and auto-reactive T cells that recognize diverse self-proteins, leads to immune-mediated destruction of pancreatic beta cells and loss of insulin secretion. In this review, we discuss antigens and epitopes in T1D and the role that post-translational modifications play in circumventing tolerance mechanisms and increasing antigenic diversity. Emerging data suggest that, analogous to other autoimmune diseases such as rheumatoid arthritis and celiac disease, enzymatically modified epitopes are preferentially recognized in T1D. Modifying enzymes such as peptidyl deiminases and tissue transglutaminase are activated in response to beta cell stress, providing a mechanistic link between post-translational modification and interactions with the environment. Although studies of such responses in the at-risk population have been limited, current data suggests that breakdown in tolerance through post-translational modification represents an important checkpoint in the development of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Epítopos de Linfocito T/inmunología , Animales , Autoinmunidad , Antígenos HLA/inmunología , Humanos , Procesamiento Proteico-Postraduccional , Linfocitos T/inmunología
11.
Nat Commun ; 15(1): 3740, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702347

RESUMEN

Insufficient functional ß-cell mass causes diabetes; however, an effective cell replacement therapy for curing diabetes is currently not available. Reprogramming of acinar cells toward functional insulin-producing cells would offer an abundant and autologous source of insulin-producing cells. Our lineage tracing studies along with transcriptomic characterization demonstrate that treatment of adult mice with a small molecule that specifically inhibits kinase activity of focal adhesion kinase results in trans-differentiation of a subset of peri-islet acinar cells into insulin producing ß-like cells. The acinar-derived insulin-producing cells infiltrate the pre-existing endocrine islets, partially restore ß-cell mass, and significantly improve glucose homeostasis in diabetic mice. These findings provide evidence that inhibition of the kinase activity of focal adhesion kinase can convert acinar cells into insulin-producing cells and could offer a promising strategy for treating diabetes.


Asunto(s)
Células Acinares , Diabetes Mellitus Experimental , Células Secretoras de Insulina , Animales , Células Secretoras de Insulina/metabolismo , Ratones , Células Acinares/metabolismo , Masculino , Insulina/metabolismo , Transdiferenciación Celular , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/farmacología , Islotes Pancreáticos/metabolismo
12.
Front Immunol ; 14: 1290058, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38164129

RESUMEN

Type 1 diabetes (T1D) affects three million Americans, with 80 new people diagnosed each day. T1D is currently uncurable and there is an urgent need to develop additional drug candidates to achieve the prevention of T1D. We propose AZD6738 (ATRi), an orally available drug currently in phases I and II of clinical trials for various cancers, as a novel candidate to prevent T1D. Based on previously reported findings of ATRi inducing cell death in rapidly proliferating T cells, we hypothesized that this drug would specifically affect self-antigen activated diabetogenic T cells. These cells, if left unchecked, could otherwise lead to the destruction of pancreatic ß cells, contributing to the development of T1D. This work demonstrates that increasing the duration of ATRi treatment provides extended protection against T1D onset. Remarkably, 5-week ATRi treatment prevented T1D in a robust adoptive transfer mouse model. Furthermore, the splenocytes of animals that received 5-week ATRi treatment did not transfer immune-mediated diabetes, while the splenocytes from control animal transferred the disease in 10 days. This work shows that ATRi prevents T1D by specifically inducing cell death in self-antigen activated, highly proliferative diabetogenic T cells through the induction of DNA damage, resulting in the inhibition of IFNγ production and proliferation. These findings support the consideration of repurposing ATRi for T1D prevention.


Asunto(s)
Antineoplásicos , Diabetes Mellitus Tipo 1 , Neoplasias , Animales , Ratones , Humanos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/prevención & control , Indoles , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Autoantígenos
13.
Front Immunol ; 14: 1231700, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37744380

RESUMEN

Introduction: We have previously demonstrated that a pathologic downregulation of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC1α) within the intestinal epithelium contributes to the pathogenesis of inflammatory bowel disease (IBD). However, the mechanism underlying downregulation of PGC1α expression and activity during IBD is not yet clear. Methods: Mice (male; C57Bl/6, Villincre/+;Pgc1afl/fl mice, and Pgc1afl/fl) were subjected to experimental colitis and treated with nicotinamide riboside. Western blot, high-resolution respirometry, nicotinamide adenine dinucleotide (NAD+) quantification, and immunoprecipitation were used to in this study. Results: We demonstrate a significant depletion in the NAD+ levels within the intestinal epithelium of mice undergoing experimental colitis, as well as humans with ulcerative colitis. While we found no decrease in the levels of NAD+-synthesizing enzymes within the intestinal epithelium of mice undergoing experimental colitis, we did find an increase in the mRNA level, as well as the enzymatic activity, of the NAD+-consuming enzyme poly(ADP-ribose) polymerase-1 (PARP1). Treatment of mice undergoing experimental colitis with an NAD+ precursor reduced the severity of colitis, restored mitochondrial function, and increased active PGC1α levels; however, NAD+ repletion did not benefit transgenic mice that lack PGC1α within the intestinal epithelium, suggesting that the therapeutic effects require an intact PGC1α axis. Discussion: Our results emphasize the importance of PGC1α expression to both mitochondrial health and homeostasis within the intestinal epithelium and suggest a novel therapeutic approach for disease management. These findings also provide a mechanistic basis for clinical trials of nicotinamide riboside in IBD patients.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Humanos , Masculino , Animales , Ratones , NAD , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Ratones Transgénicos , Mitocondrias , Inflamación
14.
Am J Physiol Lung Cell Mol Physiol ; 302(1): L160-6, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22003088

RESUMEN

The antioxidant enzyme extracellular superoxide dismutase (EC-SOD) is abundant in the lung and is known to limit inflammation and fibrosis following numerous pulmonary insults. Previous studies have reported a loss of full-length EC-SOD from the pulmonary parenchyma with accumulation of proteolyzed EC-SOD in the airspace after an interstitial lung injury. However, following airspace only inflammation, EC-SOD accumulates in the airspace without a loss from the interstitium, suggesting this antioxidant may be released from an extrapulmonary source. Because leukocytes are known to express EC-SOD and are prevalent in the bronchoalveolar lavage fluid (BALF) after injury, it was hypothesized that these cells may transport and release EC-SOD into airspaces. To test this hypothesis, C57BL/6 wild-type and EC-SOD knockout mice were irradiated and transplanted with bone marrow from either wild-type mice or EC-SOD knockout mice. Bone marrow chimeric mice were then intratracheally treated with asbestos and killed 3 and 7 days later. At both 3 and 7 days following asbestos injury, mice without pulmonary EC-SOD expression but with EC-SOD in infiltrating and resident leukocytes did not have detectable levels of EC-SOD in the airspaces. In addition, leukocyte-derived EC-SOD did not significantly lessen inflammation or early stage fibrosis that resulted from asbestos injury in the lungs. Although it is not influential in the asbestos-induced interstitial lung injury model, EC-SOD is still known to be present in leukocytes and may play an influential role in attenuating pneumonias and other inflammatory diseases.


Asunto(s)
Amianto/administración & dosificación , Leucocitos/enzimología , Lesión Pulmonar/enzimología , Pulmón/enzimología , Superóxido Dismutasa/metabolismo , Animales , Amianto/efectos adversos , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Modelos Animales de Enfermedad , Espacio Extracelular/enzimología , Depuradores de Radicales Libres/metabolismo , Inflamación/enzimología , Pulmón/patología , Lesión Pulmonar/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/fisiopatología
15.
Am J Pathol ; 178(6): 2752-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21641397

RESUMEN

Extracellular superoxide dismutase (EC-SOD) is abundant in the lung and limits inflammation and injury in response to many pulmonary insults. To test the hypothesis that EC-SOD has an important role in bacterial infections, wild-type and EC-SOD knockout (KO) mice were infected with Escherichia coli to induce pneumonia. Although mice in the EC-SOD KO group demonstrated greater pulmonary inflammation than did wild-type mice, there was less clearance of bacteria from their lungs after infection. Macrophages and neutrophils express EC-SOD; however, its function and subcellular localization in these inflammatory cells is unclear. In the present study, immunogold electron microscopy revealed EC-SOD in membrane-bound vesicles of phagocytes. These findings suggest that inflammatory cell EC-SOD may have a role in antibacterial defense. To test this hypothesis, phagocytes from wild-type and EC-SOD KO mice were evaluated. Although macrophages lacking EC-SOD produced more reactive oxygen species than did cells expressing EC-SOD after stimulation, they demonstrated significantly impaired phagocytosis and killing of bacteria. Overall, this suggests that EC-SOD facilitates clearance of bacteria and limits inflammation in response to infection by promoting bacterial phagocytosis.


Asunto(s)
Escherichia coli/citología , Espacio Extracelular/enzimología , Macrófagos/citología , Macrófagos/enzimología , Viabilidad Microbiana , Fagocitosis , Superóxido Dismutasa/metabolismo , Animales , Humanos , Inflamación/microbiología , Inflamación/patología , Espacio Intracelular/metabolismo , Pulmón/microbiología , Pulmón/patología , Macrófagos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidantes/metabolismo , Neumonía/microbiología , Neumonía/patología , Superóxido Dismutasa/ultraestructura
16.
Amino Acids ; 42(1): 95-113, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20473774

RESUMEN

The most efficacious Mn(III) porphyrinic (MnPs) scavengers of reactive species have positive charges close to the Mn site, whereby they afford thermodynamic and electrostatic facilitation for the reaction with negatively charged species such as O (2) (•-) and ONOO(-). Those are Mn(III) meso tetrakis(N-alkylpyridinium-2-yl)porphyrins, more specifically MnTE-2-PyP(5+) (AEOL10113) and MnTnHex-2-PyP(5+) (where alkyls are ethyl and n-hexyl, respectively), and their imidazolium analog, MnTDE-2-ImP(5+) (AEOL10150, Mn(III) meso tetrakis(N,N'-diethylimidazolium-2-yl) porphyrin). The efficacy of MnPs in vivo is determined not only by the compound antioxidant potency, but also by its bioavailability. The former is greatly affected by the lipophilicity, size, structure, and overall shape of the compound. These porphyrins have the ability to both eliminate reactive oxygen species and impact the progression of oxidative stress-dependent signaling events. This will effectively lead to the regulation of redox-dependent transcription factors and the suppression of secondary inflammatory- and oxidative stress-mediated immune responses. We have reported on the inhibition of major transcription factors HIF-1α, AP-1, SP-1, and NF-κB by Mn porphyrins. While the prevailing mechanistic view of the suppression of transcription factors activation is via antioxidative action (presumably in cytosol), the pro-oxidative action of MnPs in suppressing NF-κB activation in nucleus has been substantiated. The magnitude of the effect is dependent upon the electrostatic (porphyrin charges) and thermodynamic factors (porphyrin redox ability). The pro-oxidative action of MnPs has been suggested to contribute at least in part to the in vitro anticancer action of MnTE-2-PyP(5+) in the presence of ascorbate, and in vivo when combined with chemotherapy of lymphoma. Given the remarkable therapeutic potential of metalloporphyrins, future studies are warranted to further our understanding of in vivo action/s of Mn porphyrins, particularly with respect to their subcellular distribution.


Asunto(s)
Manganeso/metabolismo , Metaloporfirinas/metabolismo , Metaloporfirinas/farmacología , Estrés Oxidativo/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Animales , Humanos , Manganeso/química , Metaloporfirinas/química , Oxidación-Reducción/efectos de los fármacos
17.
J Inherit Metab Dis ; 35(1): 159-68, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21607759

RESUMEN

OBJECTIVE: We sought to determine the activation status and proliferative capacities of splenic lymphocyte populations from a mevalonate kinase-deficient mouse model of hyper-IgD syndrome (HIDS). We previously reported that murine mevalonate kinase gene ablation was embryonic lethal for homozygous mutants while heterozygotes (Mvk (+/-)) demonstrated several phenotypic features of human HIDS including increased serum levels of IgD, IgA, and TNFα, temperature dysregulation, hematological abnormalities, and splenomegaly. METHODS AND RESULTS: Flow cytometric analysis of cell surface activation markers on T and B lymphocytes, and macrophage populations, demonstrated aberrant expression of B7 glycoproteins in all splenic cell types studied. Differences in expression levels between Mvk (+/-) and Mvk (+/+) littermate controls were observed in both the basal state (unstimulated) and after Concanavalin A (Con-A) stimulation in vitro of whole splenocyte cultures. In Mvk (+/-) CD4 and CD8 T cells, alterations in expression of CD25, CD80, CD152, and CD28 were observed. Mvk (+/-) splenic macrophages expressed altered levels of CD80, CD86, CD40, and CD11c while Mvk (+/-) B lymphocytes had differential expression of CD40, CD80, and CD86. Mvk (+/-) splenocyte subpopulations also exhibited altered proliferative capacities in response to in vitro stimulation. CONCLUSION: We postulate that imbalances in the expression of cell surface proteins necessary for activation, proliferation, and regulation of the intensity and duration of an immune response may result in defective T cell activation, proliferation, and effector functions in our model and potentially in human HIDS.


Asunto(s)
Deficiencia de Mevalonato Quinasa/genética , Deficiencia de Mevalonato Quinasa/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Bazo/citología , Animales , Linfocitos T CD8-positivos/citología , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo/métodos , Heterocigoto , Humanos , Inmunoglobulina A/metabolismo , Inmunoglobulina D/metabolismo , Masculino , Deficiencia de Mevalonato Quinasa/diagnóstico , Ratones , Mutación , Factores Sexuales , Factor de Necrosis Tumoral alfa/metabolismo
18.
J Immunol ; 185(9): 5247-58, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20881184

RESUMEN

Reactive oxygen species are used by the immune system to eliminate infections; however, they may also serve as signaling intermediates to coordinate the efforts of the innate and adaptive immune systems. In this study, we show that by eliminating macrophage and T cell superoxide production through the NADPH oxidase (NOX), T cell polarization was altered. After stimulation with immobilized anti-CD3 and anti-CD28 or priming recall, T cells from NOX-deficient mice exhibited a skewed Th17 phenotype, whereas NOX-intact cells produced cytokines indicative of a Th1 response. These findings were corroborated in vivo by studying two different autoimmune diseases mediated by Th17 or Th1 pathogenic T cell responses. NOX-deficient NOD mice were Th17 prone with a concomitant susceptibility to experimental allergic encephalomyelitis and significant protection against type 1 diabetes. These data validate the role of superoxide in shaping Th responses and as a signaling intermediate to modulate Th17 and Th1 T cell responses.


Asunto(s)
Autoinmunidad/inmunología , Linaje de la Célula/inmunología , NADPH Oxidasas/deficiencia , Superóxidos/inmunología , Linfocitos T Colaboradores-Inductores/citología , Animales , Western Blotting , Separación Celular , Citocinas/biosíntesis , Citocinas/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Interleucina-17/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , NADPH Oxidasas/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/enzimología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/enzimología , Linfocitos T Colaboradores-Inductores/inmunología
19.
J Pharmacol Exp Ther ; 338(3): 906-16, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21652782

RESUMEN

Intracerebroventricular treatment with redox-regulating Mn(III) N-hexylpyridylporphyrin (MnPorphyrin) is remarkably efficacious in experimental central nervous system (CNS) injury. Clinical development has been arrested because of poor blood-brain barrier penetration. Mn(III) meso-tetrakis (N-hexylpyridinium-2-yl) porphyrin (MnTnHex-2-PyP) was synthesized to include four six-carbon (hexyl) side chains on the core MnPorphyrin structure. This has been shown to increase in vitro lipophilicity 13,500-fold relative to the hydrophilic ethyl analog Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin (MnTE-2-PyP). In normal mice, we found brain MnTnHex-2-PyP accumulation to be ∼9-fold greater than MnTE-2-PyP 24 h after a single intraperitoneal dose. We then evaluated MnTnHex-2-PyP efficacy in outcome-oriented models of focal cerebral ischemia and subarachnoid hemorrhage. For focal ischemia, rats underwent 90-min middle cerebral artery occlusion. Parenteral MnTnHex-2-PyP treatment began 5 min or 6 h after reperfusion onset and continued for 7 days. Neurologic function was improved with both early (P = 0.002) and delayed (P = 0.002) treatment onset. Total infarct size was decreased with both early (P = 0.03) and delayed (P = 0.01) treatment. MnTnHex-2-PyP attenuated nuclear factor κB nuclear DNA binding activity and suppressed tumor necrosis factor-α and interleukin-6 expression. For subarachnoid hemorrhage, mice underwent perforation of the anterior cerebral artery and were treated with intraperitoneal MnTnHex-2-PyP or vehicle for 3 days. Neurologic function was improved (P = 0.02), and vasoconstriction of the anterior cerebral (P = 0.0005), middle cerebral (P = 0.003), and internal carotid (P = 0.015) arteries was decreased by MnTnHex-2-PyP. Side-chain elongation preserved MnPorphyrin redox activity, but improved CNS bioavailability sufficient to cause improved outcome from acute CNS injury, despite delay in parenteral treatment onset of up to 6 h. This advance now allows consideration of MnPorphyrins for treatment of cerebrovascular disease.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Metaloporfirinas/farmacología , Fármacos Neuroprotectores , Accidente Cerebrovascular/tratamiento farmacológico , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/complicaciones , Citocinas/biosíntesis , ADN/metabolismo , Relación Dosis-Respuesta a Droga , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Interleucina-6/metabolismo , Flujometría por Láser-Doppler , Masculino , Metaloporfirinas/química , Metaloporfirinas/farmacocinética , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Ratas , Ratas Wistar , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología , Accidente Cerebrovascular/etiología , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/metabolismo
20.
Clin Dev Immunol ; 2011: 593863, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21647409

RESUMEN

Redox reactions are imperative to preserving cellular metabolism yet must be strictly regulated. Imbalances between reactive oxygen species (ROS) and antioxidants can initiate oxidative stress, which without proper resolve, can manifest into disease. In type 1 diabetes (T1D), T-cell-mediated autoimmune destruction of pancreatic ß-cells is secondary to the primary invasion of macrophages and dendritic cells (DCs) into the islets. Macrophages/DCs, however, are activated by intercellular ROS from resident pancreatic phagocytes and intracellular ROS formed after receptor-ligand interactions via redox-dependent transcription factors such as NF-κB. Activated macrophages/DCs ferry ß-cell antigens specifically to pancreatic lymph nodes, where they trigger reactive T cells through synapse formation and secretion of proinflammatory cytokines and more ROS. ROS generation, therefore, is pivotal in formulating both innate and adaptive immune responses accountable for islet cell autoimmunity. The importance of ROS/oxidative stress as well as potential for redox modulation in the context of T1D will be discussed.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Estrés Oxidativo/inmunología , Animales , Autoinmunidad/inmunología , Humanos , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA