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













Base de datos
Intervalo de año de publicación
1.
JAMA Netw Open ; 7(3): e241155, 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38441899

RESUMEN

This secondary analysis of a randomized clinical trial evaluates the effectiveness of ergocalciferol vs placebo in youths with newly diagnosed type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Ergocalciferoles , Humanos , Diabetes Mellitus Tipo 1/tratamiento farmacológico
2.
J Clin Invest ; 133(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37712417

RESUMEN

Expanding ß cell mass is a critical goal in the fight against diabetes. CDK4, an extensively characterized cell cycle activator, is required to establish and maintain ß cell number. ß cell failure in the IRS2-deletion mouse type 2 diabetes model is, in part, due to loss of CDK4 regulator cyclin D2. We set out to determine whether replacement of endogenous CDK4 with the inhibitor-resistant mutant CDK4-R24C rescued the loss of ß cell mass in IRS2-deficient mice. Surprisingly, not only ß cell mass but also ß cell dedifferentiation was effectively rescued, despite no improvement in whole body insulin sensitivity. Ex vivo studies in primary islet cells revealed a mechanism in which CDK4 intervened downstream in the insulin signaling pathway to prevent FOXO1-mediated transcriptional repression of critical ß cell transcription factor Pdx1. FOXO1 inhibition was not related to E2F1 activity, to FOXO1 phosphorylation, or even to FOXO1 subcellular localization, but rather was related to deacetylation and reduced FOXO1 abundance. Taken together, these results demonstrate a differentiation-promoting activity of the classical cell cycle activator CDK4 and support the concept that ß cell mass can be expanded without compromising function.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Islotes Pancreáticos , Animales , Ratones , Diabetes Mellitus Tipo 2/genética , Diferenciación Celular , Desdiferenciación Celular/genética , Modelos Animales de Enfermedad
3.
Nat Commun ; 13(1): 735, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35136059

RESUMEN

Insulin receptor (Insr) protein is present at higher levels in pancreatic ß-cells than in most other tissues, but the consequences of ß-cell insulin resistance remain enigmatic. Here, we use an Ins1cre knock-in allele to delete Insr specifically in ß-cells of both female and male mice. We compare experimental mice to Ins1cre-containing littermate controls at multiple ages and on multiple diets. RNA-seq of purified recombined ß-cells reveals transcriptomic consequences of Insr loss, which differ between female and male mice. Action potential and calcium oscillation frequencies are increased in Insr knockout ß-cells from female, but not male mice, whereas only male ßInsrKO islets have reduced ATP-coupled oxygen consumption rate and reduced expression of genes involved in ATP synthesis. Female ßInsrKO and ßInsrHET mice exhibit elevated insulin release in ex vivo perifusion experiments, during hyperglycemic clamps, and following i.p. glucose challenge. Deletion of Insr does not alter ß-cell area up to 9 months of age, nor does it impair hyperglycemia-induced proliferation. Based on our data, we adapt a mathematical model to include ß-cell insulin resistance, which predicts that ß-cell Insr knockout improves glucose tolerance depending on the degree of whole-body insulin resistance. Indeed, glucose tolerance is significantly improved in female ßInsrKO and ßInsrHET mice compared to controls at 9, 21 and 39 weeks, and also in insulin-sensitive 4-week old males. We observe no improved glucose tolerance in older male mice or in high fat diet-fed mice, corroborating the prediction that global insulin resistance obscures the effects of ß-cell specific insulin resistance. The propensity for hyperinsulinemia is associated with mildly reduced fasting glucose and increased body weight. We further validate our main in vivo findings using an Ins1-CreERT transgenic line and find that male mice have improved glucose tolerance 4 weeks after tamoxifen-mediated Insr deletion. Collectively, our data show that ß-cell insulin resistance in the form of reduced ß-cell Insr contributes to hyperinsulinemia in the context of glucose stimulation, thereby improving glucose homeostasis in otherwise insulin sensitive sex, dietary and age contexts.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Hiperinsulinismo/genética , Resistencia a la Insulina/genética , Células Secretoras de Insulina/metabolismo , Receptor de Insulina/genética , Animales , Conjuntos de Datos como Asunto , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Glucosa/metabolismo , Humanos , Hiperinsulinismo/sangre , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Insulina/sangre , Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Transgénicos , RNA-Seq , Receptor de Insulina/deficiencia , Factores Sexuales
4.
J Endocr Soc ; 6(1): bvab179, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34913020

RESUMEN

CONTEXT: The effect of the anti-inflammatory and immunomodulatory actions of vitamin D on the duration of partial clinical remission (PR) in youth with type 1 diabetes (T1D) is unclear. OBJECTIVE: This work aimed to determine the effect of adjunctive ergocalciferol on residual ß-cell function (RBCF) and PR in youth with newly diagnosed T1D who were maintained on a standardized insulin treatment protocol. The hypothesis was that ergocalciferol supplementation increases RBCF and prolongs PR. METHODS: A 12-month, randomized, double-blind, placebo-controlled trial was conducted of 50 000 IU of ergocalciferol per week for 2 months, and then once every 2 weeks for 10 months, vs placebo in 36 individuals aged 10 to 21 years, with T1D of less than 3 months and a stimulated C-peptide (SCP) level greater than or equal to 0.2 nmol/L (≥ 0.6 ng/mL). The ergocalciferol group had 18 randomly assigned participants (10 male/8 female), mean age 13.3 ±â€…2.8 years, while the control group had 18 participants (14 male/4 female), aged 14.3 ±â€…2.9 years. RESULTS: The ergocalciferol treatment group had statistically significantly higher serum 25-hydroxyvitamin D at 6 months (P = .01) and 9 months (P = .02) than the placebo group. At 12 months, the ergocalciferol group had a statistically significantly lower serum tumor necrosis factor α (TNF-α) concentration (P = .03). There were no statistically significant differences between the groups at each time point from baseline to 12 months for SCP concentration (P = .08), glycated hemoglobin A1c (HbA1c) (P = .09), insulin dose-adjusted A1c (IDAA1c), or total daily dose of insulin. Temporal trends for rising HbA1c (P = .04) and IDAA1c (P = .02) were statistically significantly blunted in the ergocalciferol group. CONCLUSION: Ergocalciferol statistically significantly reduced serum TNF-α concentration and the rates of increase both in A1c and IDAA1c, suggesting a protection of RBCF and PR in youth with newly diagnosed T1D.

5.
Diabetes ; 70(11): 2431-2443, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34711668

RESUMEN

Type 2 diabetes (T2D) is a growing cause of poor health, psychosocial burden, and economic costs worldwide. The pancreatic ß-cell is a cornerstone of metabolic physiology. Insulin deficiency leads to hyperglycemia, which was fatal before the availability of therapeutic insulins; even partial deficiency of insulin leads to diabetes in the context of insulin resistance. Comprising only an estimated 1 g or <1/500th of a percent of the human body mass, pancreatic ß-cells of the islets of Langerhans are a vulnerable link in metabolism. Proinsulin production constitutes a major load on ß-cell endoplasmic reticulum (ER), and decompensated ER stress is a cause of ß-cell failure and loss in both type 1 diabetes (T1D) and T2D. The unfolded protein response (UPR), the principal ER stress response system, is critical for maintenance of ß-cell health. Successful UPR guides expansion of ER protein folding capacity and increased ß-cell number through survival pathways and cell replication. However, in some cases the ER stress response can cause collateral ß-cell damage and may even contribute to diabetes pathogenesis. Here we review the known beneficial and harmful effects of UPR pathways in pancreatic ß-cells. Improved understanding of this stress response tipping point may lead to approaches to maintain ß-cell health and function.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Células Secretoras de Insulina/metabolismo , Humanos , Proinsulina/metabolismo , Respuesta de Proteína Desplegada/fisiología
6.
Front Endocrinol (Lausanne) ; 12: 734079, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34531828

RESUMEN

Aging is associated with loss of proliferation of the insulin-secreting ß-cell, a possible contributing factor to the increased prevalence of type 2 diabetes in the elderly. Our group previously discovered that moderate endoplasmic reticulum (ER) stress occurring during glucose exposure increases the adaptive ß-cell proliferation response. Specifically, the ATF6α arm of the tripartite Unfolded Protein Response (UPR) promotes ß-cell replication in glucose excess conditions. We hypothesized that ß-cells from older mice have reduced proliferation due to aberrant UPR signaling or an impaired proliferative response to ER stress or ATF6α activation. To investigate, young and old mouse islet cells were exposed to high glucose with low-dose thapsigargin or activation of overexpressed ATF6α, and ß-cell proliferation was quantified by BrdU incorporation. UPR pathway activation was compared by qPCR of target genes and semi-quantitative Xbp1 splicing assay. Intriguingly, although old ß-cells had reduced proliferation in high glucose compared to young ß-cells, UPR activation and induction of proliferation in response to low-dose thapsigargin or ATF6α activation in high glucose were largely similar between young and old. These results suggest that loss of UPR-led adaptive proliferation does not explain the reduced cell cycle entry in old ß-cells, and raise the exciting possibility that future therapies that engage adaptive UPR could increase ß-cell number through proliferation even in older individuals.


Asunto(s)
Envejecimiento/fisiología , Proliferación Celular , Estrés del Retículo Endoplásmico/fisiología , Células Secretoras de Insulina/fisiología , Envejecimiento/metabolismo , Animales , Células Cultivadas , Retículo Endoplásmico/metabolismo , Femenino , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
7.
Nat Metab ; 3(5): 682-700, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34031592

RESUMEN

It is known that ß cell proliferation expands the ß cell mass during development and under certain hyperglycemic conditions in the adult, a process that may be used for ß cell regeneration in diabetes. Here, through a new high-throughput screen using a luminescence ubiquitination-based cell cycle indicator (LUCCI) in zebrafish, we identify HG-9-91-01 as a driver of proliferation and confirm this effect in mouse and human ß cells. HG-9-91-01 is an inhibitor of salt-inducible kinases (SIKs), and overexpression of Sik1 specifically in ß cells blocks the effect of HG-9-91-01 on ß cell proliferation. Single-cell transcriptomic analyses of mouse ß cells demonstrate that HG-9-91-01 induces a wave of activating transcription factor (ATF)6-dependent unfolded protein response (UPR) before cell cycle entry. Importantly, the UPR wave is not associated with an increase in insulin expression. Additional mechanistic studies indicate that HG-9-91-01 induces multiple signalling effectors downstream of SIK inhibition, including CRTC1, CRTC2, ATF6, IRE1 and mTOR, which integrate to collectively drive ß cell proliferation.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Respuesta de Proteína Desplegada/efectos de los fármacos , Factor de Transcripción Activador 6/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Endorribonucleasas/metabolismo , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Pez Cebra
8.
J Biol Chem ; 295(41): 14164-14177, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-32788214

RESUMEN

Success or failure of pancreatic beta cell adaptation to ER stress is a determinant of diabetes susceptibility. The ATF6 and IRE1/XBP1 pathways are separate ER stress-response effectors important to beta cell health and function. ATF6α. and XBP1 direct overlapping transcriptional responses in some cell types. However, the signaling dynamics and interdependence of ATF6α and XBP1 in pancreatic beta cells have not been explored. To assess pathway-specific signal onset, we performed timed exposures of primary mouse islet cells to ER stressors and measured the early transcriptional response. Comparing the time course of induction of ATF6 and XBP1 targets suggested that the two pathways have similar response dynamics. The role of ATF6α in target induction was assessed by acute knockdown using islet cells from Atf6α flox/flox mice transduced with adenovirus expressing Cre recombinase. Surprisingly, given the mild impact of chronic deletion in mice, acute ATF6α knockdown markedly reduced ATF6-pathway target gene expression under both basal and stressed conditions. Intriguingly, although ATF6α knockdown did not alter Xbp1 splicing dynamics or intensity, it did reduce induction of XBP1 targets. Inhibition of Xbp1 splicing did not decrease induction of ATF6α targets. Taken together, these data suggest that the XBP1 and ATF6 pathways are simultaneously activated in islet cells in response to acute stress and that ATF6α is required for full activation of XBP1 targets, but XBP1 is not required for activation of ATF6α targets. These observations improve understanding of the ER stress transcriptional response in pancreatic islets.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/metabolismo , Transducción de Señal , Transcripción Genética , Proteína 1 de Unión a la X-Box/metabolismo , Factor de Transcripción Activador 6/genética , Animales , Técnicas de Silenciamiento del Gen , Ratones , Ratones Transgénicos
9.
Arthritis Rheumatol ; 72(2): 359-370, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31464028

RESUMEN

OBJECTIVE: Patients with hypomorphic mutations in DNase II develop a severe and debilitating autoinflammatory disease. This study was undertaken to compare the disease parameters in these patients to those in a murine model of DNase II deficiency, and to evaluate the role of specific nucleic acid sensors and identify the cell types responsible for driving the autoinflammatory response. METHODS: To avoid embryonic death, Dnase2-/- mice were intercrossed with mice that lacked the type I interferon (IFN) receptor (Ifnar-/- ). The hematologic changes and immune status of these mice were evaluated using complete blood cell counts, flow cytometry, serum cytokine enzyme-linked immunosorbent assays, and liver histology. Effector cell activity was determined by transferring T cells from Dnase2-/- × Ifnar-/- double-knockout (DKO) mice into Rag1-/- mice, and 4 weeks after cell transfer, induced changes were assessed in the recipient mice. RESULTS: In Dnase2-/- × Ifnar-/- DKO mice, many of the disease features found in DNase II-deficient patients were recapitulated, including cytopenia, extramedullary hematopoiesis, and liver fibrosis. Dnase2+/+ × Rag1-/- mice (n > 22) developed a hematologic disorder that was attributed to the transfer of an unusual IFNγ-producing T cell subset from the spleens of donor Dnase2-/- × Ifnar-/- DKO mice. Autoinflammation in this murine model did not depend on the stimulator of IFN genes (STING) pathway but was highly dependent on the chaperone protein Unc93B1. CONCLUSION: Dnase2-/- × Ifnar-/- DKO mice may be a valid model for exploring the innate and adaptive immune mechanisms responsible for the autoinflammation similar to that seen in DNASE2-hypomorphic patients. In this murine model, IFNγ is required for T cell activation and the development of clinical manifestations. The role of IFNγ in DNASE2-deficient patient populations remains to be determined, but the ability of Dnase2-/- mouse T cells to transfer disease to Rag1-/- mice suggests that T cells may be a relevant therapeutic target in patients with IFN-related systemic autoinflammatory diseases.


Asunto(s)
Enfermedades Autoinmunes/etiología , Endodesoxirribonucleasas/deficiencia , Inflamación/inmunología , Interferón gamma/biosíntesis , Células TH1/metabolismo , Animales , Modelos Animales de Enfermedad , Interferón Tipo I , Ratones , Ratones Endogámicos C57BL
10.
Mol Metab ; 27S: S69-S80, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31500833

RESUMEN

BACKGROUND: A growing body of literature suggests the cell-intrinsic activity of Atf6α during ER stress responses has implications for tissue cell number during growth and development, as well as in adult biology and tumorigenesis [1]. This concept is important, linking the cellular processes of secretory protein synthesis and endoplasmic reticulum stress response with functional tissue capacity and organ size. However, the field contains conflicting observations, especially notable in secretory cell types like the pancreatic beta cell. SCOPE OF REVIEW: Here we summarize current knowledge of the basic biology of Atf6α, along with the pleiotropic roles Atf6α plays in cell life and death decisions and possible explanations for conflicting observations. We include studies investigating the roles of Atf6α in cell survival, death and proliferation using well-controlled methodology and specific validated outcome measures, with a focus on endocrine and metabolic tissues when information was available. MAJOR CONCLUSIONS: The net outcome of Atf6α on cell survival and cell death depends on cell type and growth conditions, the presence and degree of ER stress, and the duration and intensity of Atf6α activation. It is unquestioned that Atf6α activity influences the cell fate decision between survival and death, although opposite directions of this outcome are reported in different contexts. Atf6α can also trigger cell cycle activity to expand tissue cell number through proliferation. Much work remains to be done to clarify the many gaps in understanding in this important emerging field.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Muerte Celular , Proliferación Celular , Animales , Recuento de Células , Supervivencia Celular , Humanos
11.
Diabetes ; 68(5): 975-987, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30833468

RESUMEN

Pancreatic ß-cell regeneration, the therapeutic expansion of ß-cell number to reverse diabetes, is an important goal. Replication of differentiated insulin-producing cells is the major source of new ß-cells in adult mice and juvenile humans. Nucleoside analogs such as BrdU, which are incorporated into DNA during S-phase, have been widely used to quantify ß-cell proliferation. However, reports of ß-cell nuclei labeling with both BrdU and γ-phosphorylated H2A histone family member X (γH2AX), a DNA damage marker, have raised questions about the fidelity of BrdU to label S-phase, especially during conditions when DNA damage is present. We performed experiments to clarify the causes of BrdU-γH2AX double labeling in mouse and human ß-cells. BrdU-γH2AX colabeling is neither an age-related phenomenon nor limited to human ß-cells. DNA damage suppressed BrdU labeling and BrdU-γH2AX colabeling. In dispersed islet cells, but not in intact islets or in vivo, pro-proliferative conditions promoted both BrdU and γH2AX labeling, which could indicate DNA damage, DNA replication stress, or cell cycle-related intrinsic H2AX phosphorylation. Strategies to increase ß-cell number must not only tackle the difficult challenge of enticing a quiescent cell to enter the cell cycle, but also achieve safe completion of the cell division process.


Asunto(s)
Bromodesoxiuridina/química , Bromodesoxiuridina/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , ADN/metabolismo , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Ratones
12.
Diabetes ; 67(5): 872-884, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29432124

RESUMEN

Genome-wide association studies link the CDKN2A/B locus with type 2 diabetes (T2D) risk, but mechanisms increasing risk remain unknown. The CDKN2A/B locus encodes cell cycle inhibitors p14, p15, and p16; MTAP; and ANRIL, a long noncoding RNA. The goal of this study was to determine whether CDKN2A/B T2D risk SNPs impact locus gene expression, insulin secretion, or ß-cell proliferation in human islets. Islets from donors without diabetes (n = 95) were tested for SNP genotype (rs10811661, rs2383208, rs564398, and rs10757283), gene expression (p14, p15, p16, MTAP, ANRIL, PCNA, KI67, and CCND2), insulin secretion (n = 61), and ß-cell proliferation (n = 47). Intriguingly, locus genes were coregulated in islets in two physically overlapping cassettes: p14-p16-ANRIL, which increased with age, and MTAP-p15, which did not. Risk alleles at rs10811661 and rs2383208 were differentially associated with expression of ANRIL, but not p14, p15, p16, or MTAP, in age-dependent fashion, such that younger homozygous risk donors had higher ANRIL expression, equivalent to older donor levels. We identified several risk SNP combinations that may impact locus gene expression, suggesting possible mechanisms by which SNPs impact locus biology. Risk allele carriers at ANRIL coding SNP rs564398 had reduced ß-cell proliferation index. In conclusion, CDKN2A/B locus SNPs may impact T2D risk by modulating islet gene expression and ß-cell proliferation.


Asunto(s)
Proliferación Celular/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Diabetes Mellitus Tipo 2/genética , Regulación de la Expresión Génica/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Secreción de Insulina , Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Polimorfismo de Nucleótido Simple
13.
Diabetologia ; 59(8): 1579-93, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27155872

RESUMEN

Type 2 diabetes, fuelled by the obesity epidemic, is an escalating worldwide cause of personal hardship and public cost. Diabetes incidence increases with age, and many studies link the classic senescence and ageing protein p16(INK4A) to diabetes pathophysiology via pancreatic islet biology. Genome-wide association studies (GWASs) have unequivocally linked the CDKN2A/B locus, which encodes p16 inhibitor of cyclin-dependent kinase (p16(INK4A)) and three other gene products, p14 alternate reading frame (p14(ARF)), p15(INK4B) and antisense non-coding RNA in the INK4 locus (ANRIL), with human diabetes risk. However, the mechanism by which the CDKN2A/B locus influences diabetes risk remains uncertain. Here, we weigh the evidence that CDKN2A/B polymorphisms impact metabolic health via islet biology vs effects in other tissues. Structured in a bedside-to-bench-to-bedside approach, we begin with a summary of the evidence that the CDKN2A/B locus impacts diabetes risk and a brief review of the basic biology of CDKN2A/B gene products. The main emphasis of this work is an in-depth look at the nuanced roles that CDKN2A/B gene products and related proteins play in the regulation of beta cell mass, proliferation and insulin secretory function, as well as roles in other metabolic tissues. We finish with a synthesis of basic biology and clinical observations, incorporating human physiology data. We conclude that it is likely that the CDKN2A/B locus influences diabetes risk through both islet and non-islet mechanisms.


Asunto(s)
Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Animales , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Diabetes Mellitus Tipo 2/patología , Estudio de Asociación del Genoma Completo , Humanos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Factores de Riesgo
14.
J Biol Chem ; 291(31): 16221-30, 2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27226575

RESUMEN

Previous studies revealed a paradox whereby mitogen-activated protein kinase kinase kinase kinase 4 (Map4k4) acted as a negative regulator of insulin sensitivity in chronically obese mice, yet systemic deletion of Map4k4 did not improve glucose tolerance. Here, we report markedly reduced glucose-responsive plasma insulin and C-peptide levels in whole body Map4k4-depleted mice (M4K4 iKO) as well as an impaired first phase of insulin secretion from islets derived from M4K4 iKO mice ex vivo After long-term high fat diet (HFD), M4K4 iKO mice pancreata also displayed reduced ß cell mass, fewer proliferating ß cells and reduced islet-specific gene mRNA expression compared with controls, although insulin content was normal. Interestingly, the reduced plasma insulin in M4K4 iKO mice exposed to chronic (16 weeks) HFD was not observed in response to acute HFD challenge or short term treatment with the insulin receptor antagonist S961. Furthermore, the improved insulin sensitivity in obese M4K4 iKO mice was abrogated by high exogenous insulin over the course of a euglycemic clamp study, indicating that hypoinsulinemia promotes insulin sensitivity in chronically obese M4K4 iKO mice. These results demonstrate that protein kinase Map4k4 drives obesity-induced hyperinsulinemia and insulin resistance in part by promoting insulin secretion from ß cells in mice.


Asunto(s)
Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Obesidad/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/patología , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , Péptidos/farmacología , Proteínas Serina-Treonina Quinasas/genética , Quinasa de Factor Nuclear kappa B
15.
Mol Ther ; 24(7): 1247-57, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27117222

RESUMEN

Adeno-associated viral (AAV) vectors have shown promise as a platform for gene therapy of neurological disorders. Achieving global gene delivery to the central nervous system (CNS) is key for development of effective therapies for many of these diseases. Here we report the isolation of a novel CNS tropic AAV capsid, AAV-B1, after a single round of in vivo selection from an AAV capsid library. Systemic injection of AAV-B1 vector in adult mice and cat resulted in widespread gene transfer throughout the CNS with transduction of multiple neuronal subpopulations. In addition, AAV-B1 transduces muscle, ß-cells, pulmonary alveoli, and retinal vasculature at high efficiency. This vector is more efficient than AAV9 for gene delivery to mouse brain, spinal cord, muscle, pancreas, and lung. Together with reduced sensitivity to neutralization by antibodies in pooled human sera, the broad transduction profile of AAV-B1 represents an important improvement over AAV9 for CNS gene therapy.


Asunto(s)
Proteínas de la Cápside/genética , Sistema Nervioso Central/metabolismo , Dependovirus/fisiología , Vectores Genéticos/genética , Músculos/metabolismo , Transducción Genética , Tropismo Viral , Animales , Proteínas de la Cápside/química , Dependovirus/clasificación , Expresión Génica , Técnicas de Transferencia de Gen , Genes Reporteros , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Modelos Moleculares , Conformación Proteica , Transgenes
16.
Diabetes ; 65(4): 981-95, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26740601

RESUMEN

An important goal in diabetes research is to understand the processes that trigger endogenous ß-cell proliferation. Hyperglycemia induces ß-cell replication, but the mechanism remains debated. A prime candidate is insulin, which acts locally through the insulin receptor. Having previously developed an in vivo mouse hyperglycemia model, we tested whether glucose induces ß-cell proliferation through insulin signaling. By using mice lacking insulin signaling intermediate insulin receptor substrate 2 (IRS2), we confirmed that hyperglycemia-induced ß-cell proliferation requires IRS2 both in vivo and ex vivo. Of note, insulin receptor activation was not required for glucose-induced proliferation, and insulin itself was not sufficient to drive replication. Glucose and insulin caused similar acute signaling in mouse islets, but chronic signaling differed markedly, with mammalian target of rapamycin (MTOR) and extracellular signal-related kinase (ERK) activation by glucose and AKT activation by insulin. MTOR but not ERK activation was required for glucose-induced proliferation. Cyclin D2 was necessary for glucose-induced ß-cell proliferation. Cyclin D2 expression was reduced when either IRS2 or MTOR signaling was lost, and restoring cyclin D2 expression rescued the proliferation defect. Human islets shared many of these regulatory pathways. Taken together, these results support a model in which IRS2, MTOR, and cyclin D2, but not the insulin receptor, mediate glucose-induced proliferation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Animales , Proliferación Celular/genética , Células Cultivadas , Ciclina D2/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
17.
J Clin Invest ; 125(10): 3831-46, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26389675

RESUMEN

Although stem cell populations mediate regeneration of rapid turnover tissues, such as skin, blood, and gut, a stem cell reservoir has not been identified for some slower turnover tissues, such as the pancreatic islet. Despite lacking identifiable stem cells, murine pancreatic ß cell number expands in response to an increase in insulin demand. Lineage tracing shows that new ß cells are generated from proliferation of mature, differentiated ß cells; however, the mechanism by which these mature cells sense systemic insulin demand and initiate a proliferative response remains unknown. Here, we identified the ß cell unfolded protein response (UPR), which senses insulin production, as a regulator of ß cell proliferation. Using genetic and physiologic models, we determined that among the population of ß cells, those with an active UPR are more likely to proliferate. Moreover, subthreshold endoplasmic reticulum stress (ER stress) drove insulin demand-induced ß cell proliferation, through activation of ATF6. We also confirmed that the UPR regulates proliferation of human ß cells, suggesting that therapeutic UPR modulation has potential to expand ß cell mass in people at risk for diabetes. Together, this work defines a stem cell-independent model of tissue homeostasis, in which differentiated secretory cells use the UPR sensor to adapt organ size to meet demand.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Respuesta de Proteína Desplegada/fisiología , Factor de Transcripción Activador 6/antagonistas & inhibidores , Factor de Transcripción Activador 6/biosíntesis , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/fisiología , Adaptación Fisiológica , Animales , Biomarcadores , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , División Celular , Células Cultivadas , Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico Rugoso/ultraestructura , Regulación de la Expresión Génica , Glicosilación , Humanos , Hiperglucemia/fisiopatología , Insulina/genética , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Modelos Biológicos , Obesidad/genética , Obesidad/fisiopatología , Proinsulina/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Receptores de Leptina/deficiencia , Proteínas Recombinantes de Fusión/metabolismo
18.
Curr Diab Rep ; 14(6): 492, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24740729

RESUMEN

Free fatty acids (FFAs) exert both positive and negative effects on beta cell survival and insulin secretory function, depending on concentration, duration, and glucose abundance. Lipid signals are mediated not only through metabolic pathways, but also through cell surface and nuclear receptors. Toxicity is modulated by positive signals arising from circulating factors such as hormones, growth factors and incretins, as well as negative signals such as inflammatory mediators and cytokines. Intracellular mechanisms of lipotoxicity include metabolic interference and cellular stress responses such as oxidative stress, endoplasmic reticulum (ER) stress, and possibly autophagy. New findings strengthen an old hypothesis that lipids may also impair compensatory beta cell proliferation. Clinical observations continue to support a role for lipid biology in the risk and progression of both type 1 (T1D) and type 2 diabetes (T2D). This review summarizes recent work in this important, rapidly evolving field.


Asunto(s)
Proliferación Celular , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Inflamación/fisiopatología , Células Secretoras de Insulina/metabolismo , Lípidos/sangre , Apoptosis , Supervivencia Celular , Progresión de la Enfermedad , Estrés del Retículo Endoplásmico , Ácidos Grasos no Esterificados , Femenino , Humanos , Peroxidación de Lípido , Masculino , Estrés Oxidativo , Transducción de Señal
19.
Biochem Biophys Res Commun ; 436(4): 625-31, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23764401

RESUMEN

Solid tumors contain a rare population of cancer stem cells (CSCs) that are responsible for relapse and metastasis. The existence of CSC however, remains highly controversial issue. Here we present the evidence for putative CSCs from mammary tumors amplified by vitamin A/retinol signaling. The cells exhibit mammary stem cell specific CD29(hi)/CD49f(hi)/CD24(hi) markers, resistance to radiation and chemo therapeutic agents and form highly metastatic tumors in NOD/SCID mice. The cells exhibit indefinite self renewal as cell lines. Furthermore, the cells exhibit impaired retinol metabolism and do not express enzymes that metabolize retinol into retinoic acid. Vitamin A/retinol also amplified putative CSCs from breast cancer cell lines that form highly aggressive tumors in NOD SCID mice. The studies suggest that high purity putative CSCs can be isolated from solid tumors to establish patient specific cell lines for personalized therapeutics for pre-clinical translational applications. Characterization of CSCs will allow understanding of basic cellular and molecular pathways that are deregulated, mechanisms of tumor metastasis and evasion of therapies that has direct clinical relevance.


Asunto(s)
Neoplasias Mamarias Experimentales/patología , Células Madre Neoplásicas/efectos de los fármacos , Vitamina A/farmacología , Animales , Antígenos CD/inmunología , Línea Celular Tumoral , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología
20.
Am J Physiol Endocrinol Metab ; 305(1): E149-59, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23673159

RESUMEN

Type 2 diabetes (T2D) is caused by relative insulin deficiency, due in part to reduced ß-cell mass (11, 62). Therapies aimed at expanding ß-cell mass may be useful to treat T2D (14). Although feeding rodents a high-fat diet (HFD) for an extended period (3-6 mo) increases ß-cell mass by inducing ß-cell proliferation (16, 20, 53, 54), evidence suggests that adult human ß-cells may not meaningfully proliferate in response to obesity. The timing and identity of the earliest initiators of the rodent compensatory growth response, possible therapeutic targets to drive proliferation in refractory human ß-cells, are not known. To develop a model to identify early drivers of ß-cell proliferation, we studied mice during the first week of HFD exposure, determining the onset of proliferation in the context of diet-related physiological changes. Within the first week of HFD, mice consumed more kilocalories, gained weight and fat mass, and developed hyperglycemia, hyperinsulinemia, and glucose intolerance due to impaired insulin secretion. The ß-cell proliferative response also began within the first week of HFD feeding. Intriguingly, ß-cell proliferation increased before insulin resistance was detected. Cyclin D2 protein expression was increased in islets by day 7, suggesting it may be an early effector driving compensatory ß-cell proliferation in mice. This study defines the time frame and physiology to identify novel upstream regulatory signals driving mouse ß-cell mass expansion, in order to explore their efficacy, or reasons for inefficacy, in initiating human ß-cell proliferation.


Asunto(s)
Ciclina D2/genética , Ciclina D2/metabolismo , Grasas de la Dieta/farmacología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Adaptación Fisiológica/fisiología , Animales , Glucemia/metabolismo , Proliferación Celular , Ingestión de Energía/fisiología , Intolerancia a la Glucosa/metabolismo , Hiperglucemia/metabolismo , Hiperinsulinismo/metabolismo , Insulina/sangre , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Aumento de Peso/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA