Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Am J Physiol Endocrinol Metab ; 315(5): E758-E770, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30086648

RESUMEN

Accumulation of myeloid cells in the liver, notably dendritic cells (DCs) and monocytes/macrophages (MCs), is a major component of the metainflammation of obesity. However, the mechanism(s) stimulating hepatic DC/MC infiltration remain ill defined. Herein, we addressed the hypothesis that adipose tissue (AT) free fatty acids (FFAs) play a central role in the initiation of hepatic DC/MC accumulation, using a number of mouse models of altered FFA supply to the liver. In two models of acute FFA elevation (lipid infusion and fasting) hepatic DC/MC and triglycerides (TGs) but not AT DC/MC were increased without altering plasma cytokines (PCs; TNFα and monocyte chemoattractant protein 1) and with variable effects on oxidative stress (OxS) markers. However, fasting in mice with profoundly reduced AT lipolysis (AT-specific deletion of adipose TG lipase; AAKO) failed to elevate liver DC/MC, TG, or PC, but liver OxS increased. Livers of obese AAKO mice that are known to be resistant to steatosis were similarly protected from inflammation. In high-fat feeding studies of 1, 3, 6, or 20-wk duration, liver DC/MC accumulation dissociated from PC and OxS but tracked with liver TGs. Furthermore, decreasing OxS by ~80% in obese mice failed to decrease liver DC/MC. Therefore, FFA and more specifically AT-derived FFA stimulate hepatic DC/MC accumulation, thus recapitulating the pathology of the obese liver. In a number of cases the effects of FFA can be dissociated from OxS and PC but match well with liver TG, a marker of FFA oversupply.


Asunto(s)
Tejido Adiposo/metabolismo , Ayuno/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Hígado/metabolismo , Células Mieloides/metabolismo , Animales , Citocinas/sangre , Dieta Alta en Grasa , Ácidos Grasos no Esterificados/farmacología , Lipasa/genética , Lipasa/metabolismo , Lipólisis/fisiología , Hígado/efectos de los fármacos , Ratones , Ratones Noqueados , Obesidad/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Triglicéridos/metabolismo
3.
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
4.
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
5.
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
6.
Endocr Pract ; 21(3): 237-46, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25100376

RESUMEN

OBJECTIVE: To describe the evaluation and treatment of hyperinsulinemic hypoglycemia in adults who had undergone gastric bypass surgery. A small number of patients who undergo Roux-en-Y bypass surgery develop postprandial hypoglycemia in the absence of dumping. In some cases, such patients have been treated with pancreatectomy. METHODS: We report the demographics, diagnostic results, response to medical therapy, and subsequent course of 6 referral patients with post-Roux-en-Y gastric bypass hypoglycemia. RESULTS: Characteristic clinical and metabolic parameters consistent with hyperinsulinemic hypoglycemia were identified. Parameters were similar for both spontaneous and glucose-challenge-induced hypoglycemia. In the context of exclusively postprandial symptoms, simultaneous glucose ≤55 mg/dL, insulin ≥17 µU/mL, C peptide ≥3.0 ng/mL, and insulin to glucose ratio >0.3 were associated with Roux-en-Y gastric bypass hyperinsulinemic hypoglycemia. Five of 6 patients improved on therapy consisting of dietary modification plus either calcium channel blockade, acarbose, or both. Two patients have remained on therapy for 12 to 15 months. The nonresponder was atypical and had had hypoglycemic events for several decades. Three treated patients were subsequently observed to have undergone partial or complete remission from hypoglycemic episodes after 2 to 37 months of therapy. None of the 6 have undergone pancreatectomy, and none have evidence of insulinoma. Invasive diagnostic procedures were of limited utility. CONCLUSION: In a subset of patients with post-Roux-en-Y gastric bypass hyperinsulinemic hypoglycemia, medical management can be efficacious and an alternative to partial pancreatectomy. In some cases, the disorder remits spontaneously.


Asunto(s)
Derivación Gástrica/efectos adversos , Hiperinsulinismo/complicaciones , Hipoglucemia/tratamiento farmacológico , Adolescente , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad
7.
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
8.
J Diabetes Investig ; 15(3): 263-274, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38193815

RESUMEN

Overconsumption of energy-rich foods that disrupt caloric balance is a fundamental cause of overweight, obesity and diabetes. Dysglycemia and the resulting cardiovascular disease cause substantial morbidity and mortality worldwide, as well as high societal cost. The prevalence of obesity in childhood and adolescence is increasing, leading to younger diabetes diagnosis, and higher severity of microvascular and macrovascular complications. An important goal is to identify early life conditions that increase future metabolic risk, toward the goal of preventing diabetes and cardiovascular disease. An ample body of evidence implicates prenatal and postnatal childhood growth trajectories in the programming of adult metabolic disease. Human epidemiological data show that accelerated childhood growth increases risk of type 2 diabetes in adulthood. Type 2 diabetes results from the combination of insulin resistance and pancreatic ß-cell failure, but specific mechanisms by which accelerated postnatal growth impact one or both of these processes remain uncertain. This review explores the metabolic impact of overnutrition during postnatal life in humans and in rodent models, with specific attention to the connection between accelerated childhood growth and future adiposity, insulin resistance, ß-cell mass and ß-cell dysfunction. With improved knowledge in this area, we might one day be able to modulate nutrition and growth in the critical postnatal window to maximize lifelong metabolic health.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Hipernutrición , Obesidad Infantil , Adolescente , Adulto , Femenino , Embarazo , Humanos , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/etiología , Hipernutrición/complicaciones
9.
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
10.
Eur J Appl Physiol ; 113(2): 467-78, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22801715

RESUMEN

Hypoxia manifests in many forms including the short repetitive intermittent hypoxia (IH) of sleep apnoea and the continuous hypoxia (CH) of altitude, both of which may impact metabolic function. Based on our own previous studies and the available literature, we hypothesized that whereas acute exposure to IH and CH would lead to comparable metabolic dysfunction, with longer-term exposure, metabolism would normalize to a greater extent with CH than IH. Studies were conducted in lean C57BL/6J mice exposed to either IH or CH for 1 day or 4 weeks and compared to either intermittent air (IA) or unhandled (UN) controls, respectively. We utilized the frequently sampled intravenous glucose tolerance test and minimal model analyses to determine insulin-dependent (insulin sensitivity; S (I)) and insulin-independent (glucose effectiveness; S (g)) glucose disposal, as well as the insulin response to glucose (acute insulin response to glucose; AIR(g)). Our data show that 1-day exposure impaired the glucose tolerance and caused reductions in S (g) and AIR(g) in both the IH and CH groups, but only IH caused a significant decrease in S (I) (7.5 ± 2.7 vs. 17.0 ± 5.3 µU ml(-1) min(-1); p < 0.05). After 4-week exposure, there was evidence of metabolic adaptation in both hypoxic groups, however, in the CH group, there was a supranormal increase in S (I) relative to both UN and IH groups. We conclude that in lean mice, the marked metabolic dysfunction that occurs with acute exposure to hypoxia is reversed to a greater extent with chronic CH exposure than chronic IH exposure.


Asunto(s)
Glucemia/análisis , Insulina/sangre , Enfermedad Aguda , Adaptación Fisiológica , Animales , Enfermedad Crónica , Hipoxia , Masculino , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL
11.
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
12.
Front Med (Lausanne) ; 10: 1198096, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37538312

RESUMEN

Background: Telemedicine is now common practice for many fields of medicine, but questions remain as to whether telemedicine will continue as an important patient care modality once COVID-19 becomes endemic. We explored provider and patients' perspectives on telemedicine implementation. Methods: Physicians from three specialties within the Department of Medicine of a single institution were electronically surveyed regarding their perceptions of satisfaction, benefits, and challenges of video visits, as well as the quality of interactions with patients. Patients were surveyed via telephone by the Survey Research Group at Cornell about participation in video visits, challenges encountered, perceived benefits, preferences for care, and overall satisfaction. Results: Providers reported an overwhelmingly positive experience with video visits, with the vast majority agreeing that they were comfortable with the modality (98%) and that it was easy to interact with patients (92%). Most providers (72%) wanted to have more telemedicine encounters in the future. Key factors interfering with successful telemedicine encounters were technical challenges and insufficient technical support. Overall, patients also perceived video visits very positively regarding ease of communication and care received and had few privacy concerns. Some (10%-15%) patients expressed interest in receiving more technical support and training. There was a gradient of satisfaction with telemedicine across specialties with patients receiving weight management reporting more favorable responses while patients with lymphoma expressed more mixed responses. Conclusion: Both providers and patients found telemedicine to be an acceptable and useful modality to provide or receive medical care. The principal barrier to successful encounters was technical challenges.

13.
Sci Rep ; 12(1): 7745, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35546161

RESUMEN

The antisense non-coding RNA in the INK locus (ANRIL) is a hotspot for genetic variants associated with cardiometabolic disease. We recently found increased ANRIL abundance in human pancreatic islets from donors with certain Type II Diabetes (T2D) risk-SNPs, including a T2D risk-SNP located within ANRIL exon 2 associated with beta cell proliferation. Recent studies have found that expression of circular species of ANRIL is linked to the regulation of cardiovascular phenotypes. Less is known about how the abundance of circular ANRIL may influence T2D phenotypes. Herein, we sequence circular RNA in pancreatic islets to characterize circular isoforms of ANRIL. We identify several consistently expressed circular ANRIL isoforms whose expression is correlated across dozens of individuals and characterize ANRIL splice sites that are commonly involved in back-splicing. We find that samples with the T2D risk allele in ANRIL exon 2 had higher ratios of circular to linear ANRIL compared to protective-allele carriers, and that higher circular:linear ANRIL was associated with decreased beta cell proliferation. Our study points to a combined involvement of both linear and circular ANRIL species in T2D phenotypes and opens the door for future studies of the molecular mechanisms by which ANRIL impacts cellular function in pancreatic islets.


Asunto(s)
Diabetes Mellitus Tipo 2 , Islotes Pancreáticos , ARN Largo no Codificante , Proliferación Celular/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Islotes Pancreáticos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Circular , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
14.
Front Endocrinol (Lausanne) ; 13: 793290, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35360066

RESUMEN

Background: Amidst the COVID-19 pandemic, telemedicine was rapidly implemented to maintain patient care during quarantine. However, there is little data on how this transition may have impacted weight loss outcomes and interventions among patients with overweight or obesity. Methods: This was a retrospective observational study of adults who established care for medically managed obesity at the Weill Cornell Comprehensive Weight Control Center during September-November 2019 and May-July 2020 and who completed 6 months of follow-up. Weight loss outcomes and weight management interventions were explored and stratified by patient-provider interaction: in-person visits only, in-person and video visits, and video visits only. Results: Of 499 charts eligible for review, 245 (49%) returned for their 6-month follow-up visit and were included for analysis. Of 245 patients, 69 had in-person visits only ("in-person"), 85 started in-person and later switched to video visits ("hybrid"), and 91 had video visits only ("video"). All cohorts were predominantly white and female. Median ages were 56, 49, and 49 years; baseline median weights were 98.9, 96.8, and 93.0 kg; and baseline median BMIs were 35.3, 34.4, and 34.0 kg/m2 for in-person, hybrid, and video cohorts, respectively. The median percent weight changes over 6 months were not significantly different among cohorts: -4.3% [-8.5, -1.5] in the in-person cohort, -5.6% [-8.7, -2.2] in the hybrid group, and -5.8% [-9.7, -2.4] in the video cohort. The percent of patients who achieved ≥5% weight loss were also similar: 46.4%, 55.3%, and 59.3%, respectively. The median number of visits in the video cohort was more than in the in-person or hybrid groups (5 vs. 4). Median number of anti-obesity medications (AOMs) prescribed was similar among groups. The most common AOMs were metformin (all cohorts) followed by semaglutide 1.0 mg (in-person and video) or topiramate (hybrid). Conclusion: Patients on anti-obesity medications who were followed for 6 months via video or video plus in-person visits (hybrid) experienced clinically significant weight loss. Median number of AOMs were similar among groups, and the most common AOMs were metformin, semaglutide 1.0 mg, and topiramate. More investigation is required to compare telemedicine models with in-person care.


Asunto(s)
Fármacos Antiobesidad , COVID-19 , Telemedicina , Adulto , Fármacos Antiobesidad/uso terapéutico , COVID-19/epidemiología , COVID-19/terapia , Femenino , Humanos , Pandemias , Pérdida de Peso
15.
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.

16.
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
17.
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
18.
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
19.
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
20.
J Diabetes Complications ; 35(8): 107967, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34099384

RESUMEN

OBJECTIVE: To explore predictors of severe COVID-19 disease in patients with diabetes hospitalized for COVID-19. METHODS: This is a retrospective observational study of adults with diabetes admitted for COVID-19. Bivariate tests and multivariable Cox regression were used to identify risk factors for severe COVID-19, defined as a composite endpoint of intensive care unit admission/intubation or in-hospital death. RESULTS: In 1134 patients with diabetes admitted for COVID-19, more severe disease was associated with older age (HR 1.02, p<0.001), male sex (HR 1.28, p=0.017), Asian race (HR 1.34, p=0.029 [reference: white]), and greater obesity (moderate obesity HR 1.59, p=0.015; severe obesity HR 2.07, p=0.002 [reference: normal body mass index]). Outpatient diabetes medications were not associated with outcomes. CONCLUSIONS: Age, male sex, Asian race, and obesity were associated with increased risk of severe COVID-19 disease in adults with type 2 diabetes hospitalized for COVID-19. SUMMARY: In patients with type 2 diabetes hospitalized for COVID-19 disease, we observed that age, male sex, Asian race, and obesity predicted severe COVID-19 outcomes of intensive care unit admission, intubation, or in-hospital death. The risk conferred by obesity increased with worsening obesity. Outpatient diabetes medications were not observed to be significant predictors of study outcomes.


Asunto(s)
COVID-19/diagnóstico , COVID-19/epidemiología , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/epidemiología , Admisión del Paciente/estadística & datos numéricos , Adulto , Anciano , Anciano de 80 o más Años , Índice de Masa Corporal , COVID-19/patología , COVID-19/terapia , Comorbilidad , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/terapia , Femenino , Humanos , Unidades de Cuidados Intensivos/estadística & datos numéricos , Masculino , Persona de Mediana Edad , New York/epidemiología , Obesidad/complicaciones , Obesidad/diagnóstico , Obesidad/epidemiología , Obesidad/terapia , Pronóstico , Grupos Raciales/estadística & datos numéricos , Estudios Retrospectivos , Factores de Riesgo , SARS-CoV-2/fisiología , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA