Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 195(7): 3011-9, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26283479

RESUMEN

NOD-scid.Il2rg(null) (NSG) mice are currently being used as recipients to screen for pathogenic autoreactive T cells in type 1 diabetes (T1D) patients. We questioned whether the restriction of IL-2R γ-chain (Il-2rγ)-dependent cytokine signaling only to donor cells in NSG recipients differently influenced the activities of transferred diabetogenic T cells when they were introduced as a monoclonal/oligoclonal population versus being part of a polyclonal repertoire. Unexpectedly, a significantly decreased T1D transfer by splenocytes from prediabetic NOD donors was observed in Il-2rγ(null)-NSG versus Il-2rγ-intact standard NOD-scid recipients. In contrast, NOD-derived monoclonal/oligoclonal TCR transgenic ß cell-autoreactive T cells in either the CD8 (AI4, NY8.3) or CD4 (BDC2.5) compartments transferred disease significantly more rapidly to NSG than to NOD-scid recipients. The reduced diabetes transfer efficiency by polyclonal T cells in NSG recipients was associated with enhanced activation of regulatory T cells (Tregs) mediated by NSG myeloid APC. This enhanced suppressor activity was associated with higher levels of Treg GITR expression in the presence of NSG than NOD-scid APC. These collective results indicate NSG recipients might be efficiently employed to test the activity of T1D patient-derived ß cell-autoreactive T cell clones and lines, but, when screening for pathogenic effectors within polyclonal populations, Tregs should be removed from the transfer inoculum to avoid false-negative results.


Asunto(s)
Traslado Adoptivo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Receptores de Interleucina-2/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Proteína Relacionada con TNFR Inducida por Glucocorticoide/biosíntesis , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores de Interleucina-2/genética , Transducción de Señal/inmunología , Bazo/citología , Bazo/inmunología , Linfocitos T Reguladores/trasplante
2.
J Immunol ; 186(7): 4278-84, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21357538

RESUMEN

Although P2rx7 has been proposed as a type 1 diabetes (T1D) susceptibility gene in NOD mice, its potential pathogenic role has not been directly determined. To test this possibility, we generated a new NOD stock deficient in P2X(7) receptors. T1D development was not altered by P2X(7) ablation. Previous studies found CD38 knockout (KO) NOD mice developed accelerated T1D partly because of a loss of CD4(+) invariant NKT (iNKT) cells and Foxp3(+) regulatory T cells (Tregs). These immunoregulatory T cell populations are highly sensitive to NAD-induced cell death activated by ADP ribosyltransferase-2 (ART2)-mediated ADP ribosylation of P2X(7) receptors. Therefore, we asked whether T1D acceleration was suppressed in a double-KO NOD stock lacking both P2X(7) and CD38 by rescuing CD4(+) iNKT cells and Tregs from NAD-induced cell death. We demonstrated that P2X(7) was required for T1D acceleration induced by CD38 deficiency. The CD38 KO-induced defects in homeostasis of CD4(+) iNKT cells and Tregs were corrected by coablation of P2X(7). T1D acceleration in CD38-deficient NOD mice also requires ART2 expression. If increased ADP ribosylation of P2X(7) in CD38-deficient NOD mice underlies disease acceleration, then a comparable T1D incidence should be induced by coablation of both CD38 and ART2, or CD38 and P2X(7). However, a previously established NOD stock deficient in both CD38 and ART2 expression is T1D resistant. This study demonstrated the presence of a T1D resistance gene closely linked to the ablated Cd38 allele in the previously reported NOD stock also lacking ART2, but not in the newly generated CD38/P2X(7) double-KO line.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Receptores Purinérgicos P2X7/fisiología , ADP-Ribosil Ciclasa 1/deficiencia , ADP-Ribosil Ciclasa 1/genética , ADP-Ribosil Ciclasa 1/fisiología , Animales , Antígenos CD4/biosíntesis , Células Cultivadas , Técnicas de Cocultivo , Diabetes Mellitus Tipo 1/patología , Femenino , Predisposición Genética a la Enfermedad , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Células T Asesinas Naturales/patología , Receptores Purinérgicos P2X7/deficiencia , Receptores Purinérgicos P2X7/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
3.
Front Genet ; 13: 994501, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36276935

RESUMEN

Alloxan (AL)-generated Reactive Oxygen Species (ROS) selectively destroy insulin-producing pancreatic ß-cells. A previous genome-wide scan (GWS) using a cohort of 296 F2 hybrids between NOD (AL-sensitive) and ALR (AL-resistant) mice identified linkages contributing to ß-cell susceptibility or resistance to AL-induced diabetes on Chromosomes (Chr) 2, 3, 8, and a single nucleotide polymorphism in mt-Nd2 of the mitochondrial genome (mtDNA). AL treatment of congenic and consomic NOD mouse stocks confirmed resistance linked to both the mtDNA and the Chr 8 locus from ALR [NOD.mtALR.ALR-(D8Mit293-D8Mit137)]. To identify possible epistatic interactions, the GWS analysis was expanded to 678 F2 mice. ALR-derived diabetes-resistance linkages on Chr 8 as well as the mt-Nd2 a allele were confirmed and novel additional linkages on Chr 4, 5, 6, 7, and 13 were identified. Epistasis was observed between the linkages on Chr 8 and 2 and Chr 8 and 6. Furthermore, the mt-Nd2 genotype affected the epistatic interactions between Chr 8 and 2. These results demonstrate that a combination of nuclear-cytoplasmic genome interactions regulates ß-cell sensitivity to ROS-mediated ALD.

4.
J Am Soc Nephrol ; 20(12): 2503-12, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19729434

RESUMEN

Diabetic nephropathy is a major cause of ESRD worldwide. Despite its prevalence, a lack of reliable animal models that mimic human disease has delayed the identification of specific factors that cause or predict diabetic nephropathy. The Animal Models of Diabetic Complications Consortium (AMDCC) was created in 2001 by the National Institutes of Health to develop and characterize models of diabetic nephropathy and other complications. This interim report and our online supplement detail the progress made toward that goal, specifically in the development and testing of murine models. Updates are provided on validation criteria for early and advanced diabetic nephropathy, phenotyping methods, the effect of background strain on nephropathy, current best models of diabetic nephropathy, negative models, and views of future directions. AMDCC investigators and other investigators in the field have yet to validate a complete murine model of human diabetic kidney disease. Nonetheless, the critical analysis of existing murine models substantially enhances our understanding of this disease process.


Asunto(s)
Nefropatías Diabéticas/etiología , Animales , Decorina , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/deficiencia , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo III/deficiencia , Fenotipo , Proteoglicanos/deficiencia , Receptor de Bradiquinina B2/deficiencia , Renina/genética , Especificidad de la Especie
5.
J Lipid Res ; 50(10): 2083-94, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19436067

RESUMEN

To identify genes controlling plasma HDL and triglyceride levels, quantitative trait locus (QTL) analysis was performed in one backcross, (NZO/H1Lt x NON/LtJ) x NON/LtJ, and three intercrosses, C57BL/6J x DBA/2J, C57BL/6J x C3H/HeJ, and NZB/B1NJ x NZW/LacJ. HDL concentrations were affected by 25 QTL distributed on most chromosomes (Chrs); those on Chrs 1, 8, 12, and 16 were newly identified, and the remainder were replications of previously identified QTL. Triglyceride concentrations were controlled by nine loci; those on Chrs 1, 2, 3, 7, 16, and 18 were newly identified QTL, and the remainder were replications. Combining mouse crosses with haplotype analysis for the HDL QTL on Chr 18 reduced the list of candidates to six genes. Further expression analysis, sequencing, and quantitative complementation testing of these six genes identified Lipg as the HDL QTL gene on distal Chr 18. The data from these crosses further increase the ability to perform haplotype analyses that can lead to the identification of causal lipid genes.


Asunto(s)
Sitios de Carácter Cuantitativo/genética , Animales , Prueba de Complementación Genética , Genotipo , Haplotipos , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Ratones , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple/genética , Triglicéridos/metabolismo
6.
J Urol ; 182(6 Suppl): S8-13, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19846143

RESUMEN

PURPOSE: Diabetes mellitus is a group of debilitating and costly diseases with multiple serious complications. Lower urinary tract complications or diabetic uropathy are among the most common complications of diabetes mellitus, surpassing widely recognized complications such as neuropathy and nephropathy. Diabetic uropathy develops in individuals with types 1 and 2 diabetes, and little is known about the natural history of these common and troublesome complications. Animal models have the potential to reveal mechanisms and aid in the development of treatment strategies. MATERIALS AND METHODS: We present a review of available animal models of diabetes mellitus relative to their use in the study of diabetic uropathy. RESULTS: Large and small animal models of diabetes mellitus are available. While large animals such as dogs and swine may closely mirror the human disease in size and phenotype, the time between diabetic complication onset and development, and associated husbandry expenditures can make acquiring data on statistically valid sample sizes prohibitively expensive. In contrast, small animal models (rats and mice) have much lower expenditures for a larger number of animals and compressed observation time due to a shorter life span. Also, mice are readily manipulated genetically to facilitate the isolation of the effect of single genes (transgenic and knockout mice). Type 1 diabetes mellitus can be induced chemically with streptozotocin, which is selectively toxic to pancreatic beta cells. Type 2 diabetes mellitus models have been developed by selective breeding for hyperglycemia with or without associated obesity. Diabetic uropathy has been noted in several well characterized, predictable animal models of diabetes mellitus. CONCLUSIONS: Diabetic uropathy, including diabetic bladder dysfunction, has been more frequently studied in small animals with type I diabetes. The recent availability of transgenic models provides a new opportunity for further studies of diabetic uropathy in mouse models of types I and II diabetes mellitus.


Asunto(s)
Complicaciones de la Diabetes , Diabetes Mellitus , Modelos Animales de Enfermedad , Enfermedades Urológicas , Animales , Obesidad/complicaciones
7.
Free Radic Biol Med ; 45(9): 1263-70, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18718526

RESUMEN

ALR/Lt, a NOD-related mouse strain, was selected for resistance to alloxan free radical-mediated diabetes (ALD). Despite extensive genomic identity with NOD (>70%), ALR mice display strong resistance to autoimmune type 1 diabetes (T1D) due to both an unusual elevation in systemic antioxidant defenses and a reduction in cellular ROS production that extends to the beta cell level. Reciprocal backcross to NOD previously linked the ALR-derived T1D resistance to Chr. 3, 8, and 17 as well as to the ALR mt-Nd2(a) allele encoded by the mitochondrial genome (mtDNA). To determine whether any of the ALR-derived loci protecting against T1D also protected against ALD, 296 six-week-old F2 mice from reciprocal outcrosses were alloxan-treated and assessed for diabetes onset, and a genome-wide scan (GWS) was conducted. GWS linked mt-Nd2 as well as three nuclear loci with alloxan-induced diabetes. A dominant ALR-derived ALD resistance locus on Chr. 8 colocalized with the ALR-derived T1D resistance locus identified in the previous backcross analysis. In contrast, whereas ALR contributed a novel T1D resistance locus on Chr. 3 marked by Susp, a more proximal ALR-derived region marked by Il-2 contributed ALD susceptibility, not resistance. In addition, a locus was mapped on Chr. 2, where heterozygosity provided heightened susceptibility. Tests for alloxan sensitivity in ALR conplastic mice encoding the NOD mt-Nd2(c) allele and NOD mice congenic for the protective Chr. 8 locus supported our mapping results. Alloxan sensitivity was increased in ALR.mt(NOD) mice, whereas it was decreased by congenic introduction of ALR genome on Chr. 8 into NOD. These data demonstrate both similarities and differences in the genetic control of T1D versus ROS-induced diabetes.


Asunto(s)
Autoinmunidad , Diabetes Mellitus/metabolismo , Radicales Libres , Aloxano/metabolismo , Animales , Glucemia/metabolismo , Cromosomas/ultraestructura , Cruzamientos Genéticos , ADN Mitocondrial/metabolismo , Ligamiento Genético , Genoma , Genotipo , Ratones , Ratones Endogámicos NOD , Modelos Biológicos
8.
Novartis Found Symp ; 292: 32-46; discussion 46-9, 122-9, 202-3, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19203091

RESUMEN

The major histocompatibility complex (MHC) of the type 1 diabetes-prone NOD mouse lacks a functional class II H2-Ea gene such that antigen presenting cells (APCs) are I-E null. Transgenic expression of Ea in NOD mice both restores I-E expression and confers complete protection from diabetes progression. Non-myeloablative neonatal transplantation of bone marrow cells from such I-E+ transgenic donors into NOD recipients resulted in low-level but long-term haematopoietic stem cell (HSC) engraftment. Despite low levels of I-E antigen expression in blood (averaging 0.4-3.8% of total MHC class II-positive population), chimeric recipients were protected from overt diabetes, although not insulitis development. Adoptive transfer of diabetes into immunodeficient NOD-Rag recipients that received chimeric splenocytes from primary recipients confirmed the presence of an autoreactive T cell repertoire. The demonstration that purified T cells from these weak chimeras were not tolerant to irradiated transgenic I-E+ splenocytes indicated that I-E+ donor cells provide a constant, low-level immune stimulation capable of up-regulating nominally deficient immunoregulatory networks. This study raises the possibility that cord blood HSCs from infants with high risk HLA haplotypes and a family history of type 1 diabetes might be re-introduced without myoablative treatments following transfection with a single HLA class II allele associated with diabetes resistance.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Alelos , Animales , Trasplante de Médula Ósea , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Factores Sexuales
9.
Arch Immunol Ther Exp (Warsz) ; 66(1): 11-19, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28733878

RESUMEN

Transforming growth factor (TGF)-ß has been implicated in regulation of the immune system, including autoimmunity. We have found that TGF-ß is readily produced by T cells following immunization with self-peptide epitopes that downregulate autoimmune responses in type 1 diabetes (T1D) prone nonobese diabetic (NOD) mice. These include multiple peptide epitopes derived from the islet ß-cell antigens GAD65 (GAD65 p202-221, GAD65 p217-236), GAD67 (GAD67 p210-229, GAD67 p225-244), IGRP (IGRP p123-145, IGRP p195-214) and insulin B-chain (Ins. B:9-23) that protected NOD mice from T1D. Immunization of NOD mice with the self-MHC class II I-Ag7 ß-chain-derived peptide, I-Aßg7 p54-76 also induced large amounts of TGF-ß and also protected these mice from diabetes development. These results indicate that peptides derived from disease related self-antigens and MHC class II molecules primarily induce TGF-ß producing regulatory Th3 and Tr1-like cells. TGF-ß produced by these cells could enhance the differentiation of induced regulatory iTreg and iTreg17 cells to prevent induction and progression of autoimmune diseases. We therefore suggest that peripheral immune tolerance could be induced and maintained by immunization with self-peptides that induce TGF-ß producing T cells.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Autoantígenos/inmunología , Autoinmunidad , Humanos , Tolerancia Inmunológica , Inmunización , Ratones , Ratones Endogámicos NOD , Péptidos/inmunología
10.
Diabetes ; 55(1): 171-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16380490

RESUMEN

Recently, we identified in normally type 1 diabetes-prone NOD/LtJ mice a spontaneous new leptin receptor (LEPR) mutation (designated Lepr(db-5J)) producing juvenile obesity, hyperglycemia, hyperinsulinemia, and hyperleptinemia. This early type 2 diabetes syndrome suppressed intra-islet insulitis and permitted spontaneous diabetes remission. No significant differences in plasma corticosterone, splenic CD4(+) or CD8(+) T-cell percentages, or functions of CD3(+) T-cells in vitro distinguished NOD wild-type from mutant mice. Yet splenocytes from hyperglycemic mutant donors failed to transfer type 1 diabetes into NOD.Rag1(-/-) recipients over a 13-week period, whereas wild-type donor cells did so. This correlated with significantly reduced (P < 0.01) frequencies of insulin and islet-specific glucose-6-phosphatase catalytic subunit-related protein-reactive CD8(+) T-effector clonotypes in mutant mice. Intra-islet insulitis was also significantly suppressed in lethally irradiated NOD-Lepr(db-5J)/Lt recipients reconstituted with wild-type bone marrow (P < 0.001). In contrast, type 1 diabetes eventually developed when mutant marrow was transplanted into irradiated wild-type recipients. Mitogen-induced T-cell blastogenesis was significantly suppressed when splenic T-cells from both NOD/Lt and NOD-Lepr(db-5J)/Lt donors were incubated with irradiated mutant peritoneal exudate cells (P < 0.005). In conclusion, metabolic disturbances elicited by a type 2 diabetes syndrome (insulin and/or leptin resistance, but not hypercorticism) appear to suppress type 1 diabetes development in NOD-Lepr(db-5J)/Lt by inhibiting activation of T-effector cells.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatología , Mutación Puntual , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Traslado Adoptivo , Secuencia de Aminoácidos , Animales , Glucemia , Concanavalina A , Corticosterona/sangre , Diabetes Mellitus Tipo 1/inmunología , Progresión de la Enfermedad , Femenino , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos NOD , Fenotipo , Quimera por Radiación , Receptores de Leptina , Bazo/citología , Subgrupos de Linfocitos T/metabolismo
11.
J Clin Invest ; 112(6): 826-8, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12975466

RESUMEN

Insulin-dependent diabetes mellitus is usually caused by the autoimmune destruction of pancreatic beta cells by T cells. Methodologies to track the development, migration, and functional activation of one class of such T cells (CD4 T cells) have been limited. However, it now appears that this limitation has been overcome.


Asunto(s)
Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Complejo Mayor de Histocompatibilidad/genética , Animales , Linfocitos T CD4-Positivos/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos NOD , Modelos Moleculares , Imitación Molecular , Biblioteca de Péptidos , Péptidos/química , Péptidos/inmunología
12.
Cancer Cell Int ; 7: 18, 2007 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-18045478

RESUMEN

BACKGROUND: Glucocorticoids (GC's) play an integral role in treatment strategies designed to combat various forms of hematological malignancies. GCs also are powerful inhibitors of the immune system, through regulation of appropriate cytokines and by causing apoptosis of immature thymocytes. By activating the glucocorticoid receptor (GR), GCs evoke apoptosis through transcriptional regulation of a complex, interactive gene network over a period of time preceding activation of the apoptotic enzymes. In this study we used microarray technology to determine whether several disparate types of hematologic cells, all sensitive to GC-evoked apoptosis, would identify a common set of regulated genes. We compared gene expression signatures after treatment with two potent synthetic GCs, dexamethasone (Dex) and cortivazol (CVZ) using a panel of hematologic cells. Pediatric CD4+/CD8+ T-cell leukemia was represented by 3 CEM clones: two sensitive, CEM-C7-14 and CEM-C1-6, and one resistant, CEM-C1-15, to Dex. CEM-C1-15 was also tested when rendered GC-sensitive by several treatments. GC-sensitive pediatric B-cell leukemia was represented by the SUP-B15 line and adult B-cell leukemia by RS4;11 cells. Kasumi-1 cells gave an example of the rare Dex-sensitive acute myeloblastic leukemia (AML). To test the generality of the correlations in malignant cell gene sets, we compared with GC effects on mouse non-transformed thymocytes. RESULTS: We identified a set of genes regulated by GCs in all GC-sensitive malignant cells. A portion of these were also regulated in the thymocytes. Because we knew that the highly Dex-resistant CEM-C1-15 cells could be killed by CVZ, we tested these cells with the latter steroid and again found that many of the same genes were now regulated as in the inherently GC-sensitive cells. The same result was obtained when we converted the Dex-resistant clone to Dex-sensitive by treatment with forskolin (FSK), to activate the adenyl cyclase/protein kinase A pathway (PKA). CONCLUSION: Our results have identified small sets of genes that correlate with GC-sensitivity in cells from several hematologic malignancies. Some of these are also regulated in normal mouse thymocytes.

13.
Endocrinology ; 147(2): 919-26, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16254032

RESUMEN

Polygenic mouse models for obesity-induced type 2 diabetes (T2D) more accurately reflect the most common manifestations of the human disease. Two inbred mouse strains (NON/Lt and NZO/HlLt) separately contributed T2D susceptibility- conferring quantitative trait loci to F1 males. Although chronic administration of rosiglitazone (Rosi) in diet (50 mg/kg) effectively suppressed F1 diabetes, hepatosteatosis was an undesired side effect. Three recombinant congenic strains (designated RCS1, -2, and -10) developed on the NON/Lt background carry variable numbers of these quantitative trait loci that elicit differential weight gain and male glucose intolerance syndromes of variable severity. We previously showed that RCS1 and -2 mice responded to chronic Rosi therapy without severe steatosis, whereas RCS10 males were moderately sensitive. In contrast, another recombinant congenic strain, RCS8, responded to Rosi therapy with the extreme hepatosteatosis observed in the F1. Longitudinal changes in multiple plasma analytes, including insulin, the adipokines leptin, resistin, and adiponectin, and plasminogen activator inhibitor-1 (PAI-1) allowed profiling of the differential Rosi responses in steatosis-exacerbated F1 and RCS8 males vs. the resistant RCS1 and RCS2 or moderately sensitive RCS10. Of these biomarkers, PAI-1 most effectively predicted adverse drug responses. Unexpectedly, mean resistin concentrations were higher in Rosi-treated RCS8 and RCS10. In summary, longitudinal profiling of multiple plasma analytes identified PAI-1 as a useful biomarker to monitor for differential pharmacogenetic responses to Rosi in these new mouse models of T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Obesidad/sangre , Inhibidor 1 de Activador Plasminogénico/sangre , Resistina/sangre , Tiazolidinedionas/farmacología , Adiponectina/sangre , Análisis de Varianza , Animales , Biomarcadores/sangre , Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Hígado Graso/sangre , Hígado Graso/inducido químicamente , Femenino , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/tratamiento farmacológico , Intolerancia a la Glucosa/genética , Hipoglucemiantes/efectos adversos , Insulina/sangre , Leptina/sangre , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos NOD , Ratones Obesos , Obesidad/complicaciones , Fenotipo , Carácter Cuantitativo Heredable , Recombinación Genética , Rosiglitazona , Especificidad de la Especie , Síndrome , Tiazolidinedionas/efectos adversos
14.
Diabetes ; 54 Suppl 2: S151-8, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16306333

RESUMEN

In humans, both type 1 and type 2 diabetes exemplify genetically heterogeneous complex diseases in which epigenetic factors contribute to underlying genetic susceptibility. Extended human pedigrees often show inheritance of both diabetes types. A common pathophysiological denominator in both disease forms is pancreatic beta-cell exposure to proinflammatory cytokines. Hence, it is intuitive that systemically expressed genes regulating beta-cell ability to withstand chronic diabetogenic stress may represent a component of shared susceptibility to both major disease forms. In this review, the authors assemble evidence from genetic experiments using animal models developing clearly distinct diabetes syndromes to inquire whether some degree of overlap in genes contributing susceptibility can be demonstrated. The conclusion is that although overlap exists in the pathophysiological insults leading to beta-cell destruction in the currently studied rodent models, the genetic bases seem quite distinct.


Asunto(s)
Diabetes Mellitus Tipo 1/clasificación , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/clasificación , Diabetes Mellitus Tipo 2/genética , Animales , Antioxidantes/metabolismo , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Ratones/genética , Ratones Endogámicos C57BL , Obesidad/genética
15.
Diabetes ; 54(6): 1854-62, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15919809

RESUMEN

Although thiazolidinediones suppress hyperglycemia in diabetic (NON x NZO)F1 males, these mice exhibit unusual sensitivity to drug-induced exacerbation of an underlying hepatosteatosis only rarely experienced in human patients. To establish the pharmacogenetic basis for this sensitivity, a panel of recombinant congenic strains (RCSs) with varying degrees of obesity and diabetes was generated by fixing selected NZO HlLt alleles on the diabetes- and hepatosteatosis-resistant NON/Lt background. Four new strains in this panel were exposed to chronic rosiglitazone treatment. Only one, NONcNZO8 (designated RCS8), exhibited an F1-like hepatosteatotic response. In both the F1 and RCS8 males, this adverse effect correlated with rosiglitazone suppression of already impaired hepatic phosphatidylcholine biosynthetic enzymes in both arms of the biosynthetic pathway, the phosphatidylethanolamine methyl- transferase pathway, and the CDP-choline pathway, including choline kinase and CTP-cholinephosphate cytidylyltransferase. This adverse response was not reproduced by CL316,243, a beta3-adrenergic receptor agonist with potent antihyperlipemic effects. Genome comparison showed that RCS8 differed from the other strains in carrying NZO-derived genome on virtually all of chromosome 16 and in smaller segments on chromosomes 6, 14, and 17. Thus, these RCSs present a panel of new mouse models exhibiting differential levels of obesity and diabetes as well as different drug responses. This panel can be used to screen for treatments for type 2 diabetes and its complications.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Hígado Graso/inducido químicamente , Hipoglucemiantes/toxicidad , Tiazolidinedionas/toxicidad , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Modelos Animales de Enfermedad , Hígado Graso/genética , Femenino , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratones , Ratones Congénicos , Farmacogenética , Fenotipo , Fosfatidilcolinas/biosíntesis , Rosiglitazona
16.
Diabetes ; 54(5): 1603-6, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15855353

RESUMEN

The diabetogenic major histocompatibility complex (MHC) (H2(g7)) of NOD mice comprises contributions from several class II loci collectively designated as Idd1. Introduction of the H2(gx) haplotype from the related but diabetes-resistant cataract Shionogi (CTS) strain demonstrated an additional MHC-linked locus designated Idd16. The NOD-related alloxan resistant (ALR)/Lt strain is also characterized by the H2(gx) haplotype, which does not differ from H2(g7) from the class I H2-K(d) gene distally through the class II and into the class III region. Polymorphisms distal to the heat shock protein 70 locus (Hspa1b) include a rare H2-D(dx) rather than the H2(g7) encoded D(b) allele. Two differential-length NOD.ALR-H2(gx) congenic stocks (D.R1 and D.R2), both containing H2-D(dx), significantly suppressed diabetogenesis. This protection was lost when ALR alleles between the class III region and H2-D were removed in a shorter interval congenic (D.R3). Because no differences were observed in the ALR-derived interval extending 0.41 mB proximal to H2-K in any of these congenic stocks, a component of what was originally designated "Idd16" was sited to an interval shorter than 7.33 mB, distinguishing D.R2 from D.R3. Evidence supporting the candidacy of the ALR/CTS-shared H2-D(dx) MHC class I variant present in both diabetes-resistant stocks, but not the susceptible stock, is discussed.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Predisposición Genética a la Enfermedad , Complejo Mayor de Histocompatibilidad , Envejecimiento , Animales , Biomarcadores , Diabetes Mellitus Tipo 1/epidemiología , Femenino , Incidencia , Masculino , Ratones , Ratones Endogámicos NOD , Caracteres Sexuales
17.
Diabetes ; 54(9): 2525-32, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16123339

RESUMEN

A spontaneous single-base mutation in the leptin receptor of type 1 diabetes-prone NOD/LtJ mice (designated as Lepr(db-5J)) produced a glycine640valine transversion in the extracellular domain. All mutant mice became obese and hyperinsulinemic at weaning, with 70-80% developing early-onset hyperglycemia. However, these obese diabetic mice continued to gain weight without insulin therapy. Spontaneous diabetes remission was observed in all obese females and a subset of obese males. Insulitis was largely limited to islet perimeters, with intraislet insulitis infrequently observed. In 17 obese males (age 39 weeks), we observed phenotypic heterogeneity, including full remission from hyperglycemia (24%), intermediate hyperglycemia with elevated body weight (41%), and severe hyperglycemia and weight loss (35%). The remitting normoglycemic and intermediate hyperglycemic phenotypes were associated with extensive beta-cell hyperplasia. Unlike the extensive intraislet insulitis present in diabetic lean NOD/Lt mice, the severe obese diabetic phenotype was associated with islet atrophy without extensive intraislet insulitis. These results indicated that the manipulation of the leptin/leptin receptor axis may provide a novel means of downregulating autoimmunity in type 1 diabetes and confirmed a role for leptin as a mediator in the development of this disease in NOD mice.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatología , Mutación Puntual , Receptores de Superficie Celular/genética , Secuencia de Aminoácidos , Animales , Glucemia , Femenino , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos NOD , Obesidad , Fenotipo , Receptores de Leptina , Factores de Tiempo
18.
FEBS Lett ; 580(25): 5953-8, 2006 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-17046758

RESUMEN

New Zealand obese (NZO/HlLt) male mice develop polygenic diabetes and altered phosphatidylcholine metabolism. The gene encoding phosphatidylcholine transfer protein (PC-TP) is sited within the support interval for Nidd3, a recessive NZO-derived locus on Chromosome 11 identified by prior segregation analysis between NZO/HlLt and NON/Lt. Sequence analysis revealed that the NZO-derived PC-TP contained a non-synonymous point mutation that resulted in an Arg120His substitution, which was shared by the related NZB/BlNJ and NZW/LacJ mouse strains. Consistent with the structure-based predictions, functional studies demonstrated that Arg120His PC-TP was inactive, suggesting that this mutation contributes to the deficiencies in phosphatidylcholine metabolism observed in NZO mice.


Asunto(s)
Ratones Endogámicos NZB/genética , Ratones Obesos/genética , Proteínas de Transferencia de Fosfolípidos/antagonistas & inhibidores , Proteínas de Transferencia de Fosfolípidos/genética , Polimorfismo Genético , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , ADN Complementario/genética , Masculino , Ratones , Ratones Endogámicos , Ratones Obesos/metabolismo , Modelos Moleculares , Fosfatidilcolinas/metabolismo , Proteínas de Transferencia de Fosfolípidos/química , Mutación Puntual , Conformación Proteica , Especificidad de la Especie
19.
Vascul Pharmacol ; 45(1): 65-71, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16750656

RESUMEN

Given the heterogeneous nature of metabolic dysfunctions associated with insulin resistance and type 2 diabetes (T2D), a single pharmaceutical cannot be expected to provide complication-free therapy in all patients. Thiazolidinediones (TZD) increase insulin sensitivity, reduce blood glucose and improve cardiovascular parameters. However, in addition to increasing fat mass, TZD have the potential in certain individuals to exacerbate underlying hepatosteatosis and diabetic cardiomyopathy. Pharmacogenetics should allow patient selection to maximize therapy and minimize risk. To this end, we have combined two genetically diverse inbred strains, NON/Lt and NZO/Lt, to produce a "negative heterosis" increasing the frequency of T2D in F1 males. As in humans with T2D, treatment of diabetic and hyperlipemic F1 males with rosiglitazone (Rosi), an agonist of peroxisome proliferator-activated gamma receptor (PPARgamma), reverses these disease phenotypes. However, the hybrid genome perturbed both major pathways for phosphatidylcholine (PC) biosynthesis in the liver, and effected remarkable alterations in the composition of cardiolipin in heart mitochondria. These metabolic defects severely exacerbated an underlying hepatosteatosis and increased levels of the adipokine, plasminogen activator inhibitor-1 (PAI-1), a risk factor for cardiovascular events. This model system demonstrates how the power of mouse genetics can be used to identify the metabolic signatures of individuals who may be prone to drug side effects.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Hipoglucemiantes/efectos adversos , Hígado/efectos de los fármacos , Fosfatidilcolinas/metabolismo , Tiazolidinedionas/efectos adversos , Animales , Cardiolipinas/metabolismo , Sistema Cardiovascular/metabolismo , Citidililtransferasa de Colina-Fosfato/antagonistas & inhibidores , Citidililtransferasa de Colina-Fosfato/metabolismo , Cruzamientos Genéticos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Obesos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , PPAR gamma/agonistas , PPAR gamma/metabolismo , Fosfatidiletanolamina N-Metiltransferasa/antagonistas & inhibidores , Fosfatidiletanolamina N-Metiltransferasa/metabolismo , Rosiglitazona
20.
Arch Physiol Biochem ; 112(1): 49-58, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16754203

RESUMEN

New Zealand Obese (NZO) male mice develop a polygenic juvenile-onset obesity and maturity onset hyperinsulinemia. Approximately 50% transit to chronic hyperglycemia. Here we report on the proliferation of beta cells in relation to both the individual's metabolic status and structural parameters of the endocrine pancreas. Proliferating beta cells were quantified in pancreas sections by immunoenzymatic double staining of Ki-67 protein, as a marker for proliferating cells, and endocrine non-beta cells in order to distinguish them from beta cells. In normoglycemic NZO/Hl males Ki-67 labelling indices (IKi-67) of beta cells varied between 0.14 and 1.5%, and correlated significantly with both serum insulin levels and beta cell size. There was no correlation with the glycemic status. In diabetic males, beta cell size was increased. IKi-67 varied between 1 and 3%. The data suggest that the secretory activity of beta cells triggered by glucose, entailed changes in both beta cell hypertrophy and proliferation. As shown by morphometric measurements, beta cell expansion in diabetic mice was limited, in spite of high IKi-67 values. This suggested increased death rates of beta cells.


Asunto(s)
Hiperglucemia/patología , Hiperinsulinismo/patología , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Obesidad/patología , Animales , Apoptosis , Proliferación Celular , Diabetes Mellitus Tipo 1/patología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Insulina/análisis , Células Secretoras de Insulina/química , Antígeno Ki-67/análisis , Masculino , Ratones , Ratones Mutantes , Ratones Obesos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA