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

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Calcif Tissue Int ; 112(4): 518-523, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36575358

RESUMO

INTRODUCTION: Progressive osseous heteroplasia (POH) is a rare, debilitating disorder characterized by heterotopic ossification in the skin and muscles, resulting in contractures of the joints and progressive loss of function. While 60-70% of the POH patients have paternally inherited, inactivating pathogenic variants in GNAS, the remaining 30-40% have no known etiology. FAM111B pathogenic variants, located on chromosome 11q12.1, cause POIKTMP (hereditary fibrosing poikiloderma with tendon contractures, myopathy, and pulmonary fibrosis), a very rare, autosomal-dominant disorder with high frequency of de novo missense pathogenic variants, which affects multiple tissues and organs, causing extensive fibrosis and muscle adiposis, though the exact mechanism is unknown. To our knowledge, there are no reports of FAM111B associated with POH. We describe the first case of POH phenotype associated with a novel de novo frameshift pathogenic variant in the FAM111B and present an analysis of the protein structure and function caused by this genomic disruption. CASE: A 15-year-old African-American male presented with generalized calcific nodules, progressive contractures, and muscle weakness leading to immobility, beginning at 6 years of age. Cutaneous examination showed generalized hard nodules varying from small to plaque-like ulcerated erupted skin lesions. Biochemical evaluation revealed 25(OH) vitamin D insufficiency (20 ng/mL), and normal levels of parathyroid hormone, FGF-23, alkaline phosphatase, calcium, and phosphorus. Skeletal survey radiographs and computed tomography (CT) of the chest, abdomen, and pelvis showed extensive soft tissue and muscle heterotopic ossifications involving shoulders, axillae, trunk, abdomen, pelvis, upper and lower extremities, in a clumped, conglomerate distribution within muscle, subcutaneous fat, and in some areas extending to the skin. There was no pulmonary fibrosis on the chest CT. The clinical and radiographic findings were most consistent with POH. A trio-clinical exome sequencing revealed a de novo heterozygous likely pathogenic variant in the FAM111B (OMIM # 615584) (c.1462delT [p.Cys488Valfs*21]). The resulted frameshift change in exon 4 replaced C-terminal region with 21 alternative amino acids. Multiple, previously reported disease-associated variants appear to localize within the trypsin-like cysteine/serine peptidase domain in which this variant occurs, supporting the functional significance of this region, though none have been previously reported to be associated with POH phenotype. Our 3D protein modeling showed obliteration of predicted protein folding and structure, and elimination of the zinc-binding domain, likely severely affecting protein function. CONCLUSION: This is the first case of POH phenotype associated with a novel de novo pathogenic frameshift variant in FAM111B. Whether the frameshift change in FAM111B predicts POH remains unclear. Further evaluations are necessary to fully elucidate this finding and the potential role and mechanism by which the FAM111B variants contributes to POH phenotype.


Assuntos
Contratura , Ossificação Heterotópica , Masculino , Humanos , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Fenótipo , Contratura/complicações , Contratura/genética , Fibrose , Proteínas de Ciclo Celular/genética
2.
J Theor Biol ; 558: 111341, 2023 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-36335999

RESUMO

Bayesian inference produces a posterior distribution for the parameters of a mathematical model that can be used to guide the formation of hypotheses; specifically, the posterior may be searched for evidence of alternative model hypotheses, which serves as a starting point for hypothesis formation and model refinement. Previous approaches to search for this evidence are largely qualitative and unsystematic; further, demonstrations of these approaches typically stop at hypothesis formation, leaving the questions they raise unanswered. Here, we introduce a Kullback-Leibler (KL) divergence-based ranking to expedite Bayesian hypothesis formation and investigate the hypotheses it generates, ultimately generating novel, biologically significant insights. Our approach uses KL divergence to rank parameters by how much information they gain from experimental data. Subsequently, rather than searching all model parameters at random, we use this ranking to prioritize examining the posteriors of the parameters that gained the most information from the data for evidence of alternative model hypotheses. We test our approach with two examples, which showcase the ability of our approach to systematically uncover different types of alternative hypothesis evidence. First, we test our KL divergence ranking on an established example of Bayesian hypothesis formation. Our top-ranked parameter matches the one previously identified to produce alternative hypotheses. In the second example, we apply our ranking in a novel study of a computational model of prolactin-induced JAK2-STAT5 signaling, a pathway that mediates beta cell proliferation. Within the top 3 ranked parameters (out of 33), we find a bimodal posterior revealing two possible ranges for the prolactin receptor degradation rate. We go on to refine the model, incorporating new data and determining which degradation rate is most plausible. Overall, while the effectiveness of our approach depends on having a properly formulated prior and on the form of the posterior distribution, we demonstrate that our approach offers a novel and generalizable quantitative framework for Bayesian hypothesis formation and use it to produce a novel, biologically-significant insight into beta cell signaling.


Assuntos
Janus Quinase 2 , Modelos Teóricos , Teorema de Bayes
3.
Genes Dev ; 27(4): 372-7, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23431054

RESUMO

In the developing pancreas, self-renewal of progenitors and patterning of cell fates are coordinated to ensure the correct size and cellular makeup of the organ. How this coordination is achieved, however, is not clear. We report that deletion of DNA methyltransferase 1 (Dnmt1) in pancreatic progenitors results in agenesis of the pancreas due to apoptosis of progenitor cells. We show that DNMT1 is bound to the p53 regulatory region and that loss of Dnmt1 results in derepression of the p53 locus. Haploinsufficiency of p53 rescues progenitor cell survival and cellular makeup of the Dnmt1-deleted pancreas.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Organogênese/fisiologia , Pâncreas/embriologia , Pâncreas/enzimologia , Células-Tronco/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/genética , Sobrevivência Celular , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Haploinsuficiência , Camundongos , Pâncreas/citologia , Células-Tronco/citologia , Proteína Supressora de Tumor p53/genética
4.
J Biol Chem ; 294(41): 15182-15192, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31341016

RESUMO

Neurogenin-3 (NEUROG3) is a helix-loop-helix (HLH) transcription factor involved in the production of endocrine cells in the intestine and pancreas of humans and mice. However, the human NEUROG3 loss-of-function phenotype differs subtly from that in mice, but the reason for this difference remains poorly understood. Because NEUROG3 expression precedes exit of the cell cycle and the expression of endocrine cell markers during differentiation, we investigated the effect of lentivirus-mediated overexpression of the human NEUROG3 gene on the cell cycle of BON4 cells and various human nonendocrine cell lines. NEUROG3 overexpression induced a reversible cell cycle exit, whereas expression of a neuronal lineage homolog, NEUROG1, had no such effect. In endocrine lineage cells, the cellular quiescence induced by short-term NEUROG3 expression required cyclin-dependent kinase inhibitor 1A (CDKN1A)/p21CIP1 expression. Expression of endocrine differentiation markers required sustained NEUROG3 expression in the quiescent, but not in the senescent, state. Inhibition of the phosphatase and tensin homolog (PTEN) pathway reversed quiescence by inducing cyclin-dependent kinase 2 (CDK2) and reducing p21CIP1 and NEUROG3 protein levels in BON4 cells and human enteroids. We discovered that NEUROG3 expression stimulates expression of CDKN2a/p16INK4a and BMI1 proto-oncogene polycomb ring finger (BMI1), with the latter limiting expression of the former, delaying the onset of CDKN2a/p16INK4a -driven cellular senescence. Furthermore, NEUROG3 bound to the promoters of both CDKN1a/p21CIP1 and BMI1 genes, and BMI1 attenuated NEUROG3 binding to the CDKN1a/p21CIP1 promoter. Our findings reveal how human NEUROG3 integrates inputs from multiple signaling pathways and thereby mediates cell cycle exit at the onset of differentiation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Pontos de Checagem do Ciclo Celular , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Proteínas do Tecido Nervoso/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Linhagem Celular , Senescência Celular , Regulação da Expressão Gênica , Genes p16 , Humanos , Proto-Oncogene Mas
5.
J Biol Chem ; 293(13): 4735-4751, 2018 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-29378845

RESUMO

Estrogen receptor α (ERα) action plays an important role in pancreatic ß-cell function and survival; thus, it is considered a potential therapeutic target for the treatment of type 2 diabetes in women. However, the mechanisms underlying the protective effects of ERα remain unclear. Because ERα regulates mitochondrial metabolism in other cell types, we hypothesized that ERα may act to preserve insulin secretion and promote ß-cell survival by regulating mitochondrial-endoplasmic reticulum (EndoRetic) function. We tested this hypothesis using pancreatic islet-specific ERα knockout (PERαKO) mice and Min6 ß-cells in culture with Esr1 knockdown (KD). We found that Esr1-KD promoted reactive oxygen species production that associated with reduced fission/fusion dynamics and impaired mitophagy. Electron microscopy showed mitochondrial enlargement and a pro-fusion phenotype. Mitochondrial cristae and endoplasmic reticulum were dilated in Esr1-KD compared with ERα replete Min6 ß-cells. Increased expression of Oma1 and Chop was paralleled by increased oxygen consumption and apoptosis susceptibility in ERα-KD cells. In contrast, ERα overexpression and ligand activation reduced both Chop and Oma1 expression, likely by ERα binding to consensus estrogen-response element sites in the Oma1 and Chop promoters. Together, our findings suggest that ERα promotes ß-cell survival and insulin secretion through maintenance of mitochondrial fission/fusion-mitophagy dynamics and EndoRetic function, in part by Oma1 and Chop repression.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Receptor alfa de Estrogênio/metabolismo , Células Secretoras de Insulina/metabolismo , Mitocôndrias/metabolismo , Mitofagia , Animais , Sobrevivência Celular , Receptor alfa de Estrogênio/genética , Feminino , Insulina/genética , Insulina/metabolismo , Metaloproteases/biossíntese , Metaloproteases/genética , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Proteínas Mitocondriais/biossíntese , Proteínas Mitocondriais/genética , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição CHOP/biossíntese , Fator de Transcrição CHOP/genética
6.
Curr Diab Rep ; 17(11): 116, 2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28980194

RESUMO

PURPOSE OF REVIEW: This review will focus on the multiple approaches to gene editing and address the potential use of genetically modified human pluripotent stem cell-derived beta cells (SC-ß) as a tool to study human beta-cell development and model their function in diabetes. We will explore how new variations of CRISPR/Cas9 gene editing may accelerate our understanding of beta-cell developmental biology, elucidate novel mechanisms that establish and regulate beta-cell function, and assist in pioneering new therapeutic modalities for treating diabetes. RECENT FINDINGS: Improvements in CRISPR/Cas9 target specificity and homology-directed recombination continue to advance its use in engineering stem cells to model and potentially treat disease. We will review how CRISPR/Cas9 gene editing is informing our understanding of beta-cell development and expanding the therapeutic possibilities for treating diabetes and other diseases. Here we focus on the emerging use of gene editing technology, specifically CRISPR/Cas9, as a means of manipulating human gene expression to gain novel insights into the roles of key factors in beta-cell development and function. Taken together, the combined use of SC-ß cells and CRISPR/Cas9 gene editing will shed new light on human beta-cell development and function and accelerate our progress towards developing new therapies for patients with diabetes.


Assuntos
Diabetes Mellitus/genética , Diabetes Mellitus/patologia , Edição de Genes , Células Secretoras de Insulina/patologia , Células-Tronco Pluripotentes/metabolismo , Sistemas CRISPR-Cas/genética , Diabetes Mellitus/imunologia , Humanos , Sistema Imunitário/patologia
7.
Am J Physiol Gastrointest Liver Physiol ; 308(8): G678-90, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25721301

RESUMO

Intestinal epithelial cell renewal relies on the right balance of epithelial cell migration, proliferation, differentiation, and apoptosis. Intestinal epithelial cells consist of absorptive and secretory lineage. The latter is comprised of goblet, Paneth, and enteroendocrine cells. Fibroblast growth factor 10 (FGF10) plays a central role in epithelial cell proliferation, survival, and differentiation in several organs. The expression pattern of FGF10 and its receptors in both human and mouse intestine and their role in small intestine have yet to be investigated. First, we analyzed the expression of FGF10, FGFR1, and FGFR2, in the human ileum and throughout the adult mouse small intestine. We found that FGF10, FGFR1b, and FGFR2b are expressed in the human ileum as well as in the mouse small intestine. We then used transgenic mouse models to overexpress Fgf10 and a soluble form of Fgfr2b, to study the impact of gain or loss of Fgf signaling in the adult small intestine. We demonstrated that overexpression of Fgf10 in vivo and in vitro induces goblet cell differentiation while decreasing Paneth cells. Moreover, FGF10 decreases stem cell markers such as Lgr5, Lrig1, Hopx, Ascl2, and Sox9. FGF10 inhibited Hes1 expression in vitro, suggesting that FGF10 induces goblet cell differentiation likely through the inhibition of Notch signaling. Interestingly, Fgf10 overexpression for 3 days in vivo and in vitro increased the number of Mmp7/Muc2 double-positive cells, suggesting that goblet cells replace Paneth cells. Further studies are needed to determine the mechanism by which Fgf10 alters cell differentiation in the small intestine.


Assuntos
Fator 10 de Crescimento de Fibroblastos/metabolismo , Células Caliciformes/metabolismo , Intestino Delgado/metabolismo , Celulas de Paneth/metabolismo , Animais , Biomarcadores/metabolismo , Morte Celular , Diferenciação Celular , Proliferação de Células , Fator 10 de Crescimento de Fibroblastos/genética , Células Caliciformes/patologia , Humanos , Intestino Delgado/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Celulas de Paneth/patologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Fatores de Tempo , Técnicas de Cultura de Tecidos
8.
Gastroenterology ; 145(1): 138-148, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23562752

RESUMO

BACKGROUND & AIMS: Proprotein convertase 1/3 (PC1/3) deficiency, an autosomal-recessive disorder caused by rare mutations in the proprotein convertase subtilisin/kexin type 1 (PCSK1) gene, has been associated with obesity, severe malabsorptive diarrhea, and certain endocrine abnormalities. Common variants in PCSK1 also have been associated with obesity in heterozygotes in several population-based studies. PC1/3 is an endoprotease that processes many prohormones expressed in endocrine and neuronal cells. We investigated clinical and molecular features of PC1/3 deficiency. METHODS: We studied the clinical features of 13 children with PC1/3 deficiency and performed sequence analysis of PCSK1. We measured enzymatic activity of recombinant PC1/3 proteins. RESULTS: We identified a pattern of endocrinopathies that develop in an age-dependent manner. Eight of the mutations had severe biochemical consequences in vitro. Neonates had severe malabsorptive diarrhea and failure to thrive, required prolonged parenteral nutrition support, and had high mortality. Additional endocrine abnormalities developed as the disease progressed, including diabetes insipidus, growth hormone deficiency, primary hypogonadism, adrenal insufficiency, and hypothyroidism. We identified growth hormone deficiency, central diabetes insipidus, and male hypogonadism as new features of PCSK1 insufficiency. Interestingly, despite early growth abnormalities, moderate obesity, associated with severe polyphagia, generally appears. CONCLUSIONS: In a study of 13 children with PC1/3 deficiency caused by disruption of PCSK1, failure of enteroendocrine cells to produce functional hormones resulted in generalized malabsorption. These findings indicate that PC1/3 is involved in the processing of one or more enteric hormones that are required for nutrient absorption.


Assuntos
Diarreia/etiologia , Doenças do Sistema Endócrino/etiologia , Síndromes de Malabsorção/etiologia , Obesidade/complicações , Pró-Proteína Convertase 1/deficiência , Adolescente , Hormônio Adrenocorticotrópico/sangue , Criança , Pré-Escolar , Estudos de Coortes , Doenças do Sistema Endócrino/complicações , Doenças do Sistema Endócrino/congênito , Feminino , Humanos , Lactente , Masculino , Mutação , Obesidade/congênito , Pró-Proteína Convertase 1/genética
9.
Curr Opin Cell Biol ; 19(6): 634-45, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18061427

RESUMO

The elaboration of the pancreas from epithelial buds to the intricate organ requires complex patterning information that controls fundamental cellular processes such as differentiation and proliferation of pancreatic progenitor cells. During pancreatic organogenesis, endocrine cells are generated from a population of pancreatic progenitor cells. The progenitor cells during the early development simultaneously receive multiple signals, some mitogenic and some inducing differentiation. These extrinsic signals are interpreted through an intrinsic mechanism that either commits the progenitor cell to the mitotic cell cycle or leads to exit from the cell cycle in order to differentiate. The endocrine cells that differentiate from progenitor cells are postmitotic, and direct lineage tracing analyses indicate that a population of progenitor cells persists throughout embryogenesis to allow the differentiation of new endocrine cells. At the end of embryogenesis an early postnatal period is characterized by high rates of beta cell proliferation leading to massive increases in beta cell mass. The beta cell mass expansion considerably slows down in adult animals, though variations in insulin demand due to physiological and pathological states such as pregnancy and obesity can lead to adaptive changes in the beta cells that include hyperplasia, hypertrophy, and increased insulin synthesis and secretion. Deciphering the mechanisms that regulate the plasticity of beta cell mass can be an important step in developing effective strategies to treat diabetes.


Assuntos
Pâncreas/fisiologia , Regeneração , Animais , Diferenciação Celular , Humanos
10.
Cells ; 13(10)2024 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-38786091

RESUMO

The dysfunction of α and ß cells in pancreatic islets can lead to diabetes. Many questions remain on the subcellular organization of islet cells during the progression of disease. Existing three-dimensional cellular mapping approaches face challenges such as time-intensive sample sectioning and subjective cellular identification. To address these challenges, we have developed a subcellular feature-based classification approach, which allows us to identify α and ß cells and quantify their subcellular structural characteristics using soft X-ray tomography (SXT). We observed significant differences in whole-cell morphological and organelle statistics between the two cell types. Additionally, we characterize subtle biophysical differences between individual insulin and glucagon vesicles by analyzing vesicle size and molecular density distributions, which were not previously possible using other methods. These sub-vesicular parameters enable us to predict cell types systematically using supervised machine learning. We also visualize distinct vesicle and cell subtypes using Uniform Manifold Approximation and Projection (UMAP) embeddings, which provides us with an innovative approach to explore structural heterogeneity in islet cells. This methodology presents an innovative approach for tracking biologically meaningful heterogeneity in cells that can be applied to any cellular system.


Assuntos
Células Secretoras de Glucagon , Células Secretoras de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Glucagon/metabolismo , Animais , Tomografia por Raios X/métodos , Camundongos , Humanos , Insulina/metabolismo
11.
Diabetes ; 72(5): 575-589, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36607262

RESUMO

The molecular and functional heterogeneity of pancreatic ß-cells is well recognized, but the underlying mechanisms remain unclear. Pancreatic islets harbor a subset of ß-cells that co-express tyrosine hydroxylase (TH), an enzyme involved in synthesis of catecholamines that repress insulin secretion. Restriction of the TH+ ß-cells within islets is essential for appropriate function in mice, such that a higher proportion of these cells corresponds to reduced insulin secretion. Here, we use these cells as a model to dissect the developmental control of ß-cell heterogeneity. We define the specific molecular and metabolic characteristics of TH+ ß-cells and show differences in their developmental restriction in mice and humans. We show that TH expression in ß-cells is restricted by DNA methylation during ß-cell differentiation. Ablation of de novo DNA methyltransferase Dnmt3a in the embryonic progenitors results in a dramatic increase in the proportion of TH+ ß-cells, whereas ß-cell-specific ablation of Dnmt3a does not. We demonstrate that maintenance of Th promoter methylation is essential for its continued restriction in postnatal ß-cells. Loss of Th promoter methylation in response to chronic overnutrition increases the number of TH+ ß-cells, corresponding to impaired ß-cell function. These results reveal a regulatory role of DNA methylation in determining ß-cell heterogeneity.


Assuntos
Células Secretoras de Insulina , Ilhotas Pancreáticas , Tirosina 3-Mono-Oxigenase , Animais , Humanos , Camundongos , Metilação de DNA , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Regiões Promotoras Genéticas/genética , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
12.
Biomolecules ; 12(2)2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-35204716

RESUMO

As patients recently diagnosed with T1D and patients with T2D have residual beta cell mass, there is considerable effort in beta cell biology to understand the mechanisms that drive beta cell regeneration as a potential cellular therapy for expanding patients' residual beta cell population. Both mouse and human studies have established that beta cell mass expansion occurs rapidly during pregnancy. To investigate the mechanisms of beta cell mass expansion during pregnancy, we developed a novel in vivo and in vitro models of pseudopregnancy. Our models demonstrate that pseudopregnancy promotes beta cell mass expansion in parous mice, and this expansion is driven by beta cell proliferation rather than hypertrophy. Importantly, estrogen, progesterone, and placental lactogen induce STAT5A signaling in the pseudopregnancy model, demonstrating that this model successfully recapitulates pregnancy-induced beta cell replication. We then created an in vitro model of pseudopregnancy and found that the combination of estrogen and placental lactogen induced beta cell replication in human islets and rat insulinoma cells. Therefore, beta cells both in vitro and in vivo increase proliferation when subjected to the pseudopregnancy cocktail compared to groups treated with estradiol or placental lactogen alone. The pseudopregnancy models described here may help inform novel methods of inducing beta cell replication in patients with diabetes.


Assuntos
Células Secretoras de Insulina , Ilhotas Pancreáticas , Animais , Divisão Celular , Feminino , Humanos , Camundongos , Placenta , Lactogênio Placentário/farmacologia , Gravidez , Ratos
13.
Integr Biol (Camb) ; 14(2): 37-48, 2022 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-35368075

RESUMO

Patients with diabetes are unable to produce a sufficient amount of insulin to properly regulate their blood glucose levels. One potential method of treating diabetes is to increase the number of insulin-secreting beta cells in the pancreas to enhance insulin secretion. It is known that during pregnancy, pancreatic beta cells proliferate in response to the pregnancy hormone, prolactin (PRL). Leveraging this proliferative response to PRL may be a strategy to restore endogenous insulin production for patients with diabetes. To investigate this potential treatment, we previously developed a computational model to represent the PRL-mediated JAK-STAT signaling pathway in pancreatic beta cells. Here, we applied the model to identify the importance of particular signaling proteins in shaping the response of a population of beta cells. We simulated a population of 10 000 heterogeneous cells with varying initial protein concentrations responding to PRL stimulation. We used partial least squares regression to analyze the significance and role of each of the varied protein concentrations in producing the response of the cell. Our regression models predict that the concentrations of the cytosolic and nuclear phosphatases strongly influence the response of the cell. The model also predicts that increasing PRL receptor strengthens negative feedback mediated by the inhibitor suppressor of cytokine signaling. These findings reveal biological targets that can potentially be used to modulate the proliferation of pancreatic beta cells to enhance insulin secretion and beta cell regeneration in the context of diabetes.


Assuntos
Células Secretoras de Insulina , Prolactina , Feminino , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Gravidez , Prolactina/metabolismo , Prolactina/farmacologia , Transdução de Sinais/fisiologia
14.
Front Endocrinol (Lausanne) ; 12: 625415, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33868167

RESUMO

Childhood obesity is increasing at an alarming rate in the United States. This trend carries serious risk of children developing obesity-related diseases including Type 2 diabetes and cardiovascular disease. Non-nutritive sweeteners (NNS) are used as substitution for table sugar as a way to prevent weight gain. Their consumption is ubiquitous in adults and children; however the long-term health outcomes of chronic NNS consumption in children are unclear. Conflicting observational studies suggest that children consuming NNS are at risk of obesity and development of type 2 diabetes, while others concluded some benefits in weight reduction. Here, we review the physiological mechanisms that can contribute to the negative metabolic effects of NNS. We will focus on how NNS alters the sweet perception leading to increase caloric consumption, how NNs alters the gut microbiota, and how NNS may disrupt glucose homeostasis and initiate a vicious cycle of pancreatic endocrine dysfunction. Studies focused on the pediatric population are limited but necessary to determine whether the potential weight loss benefits outweigh the potential negative metabolic outcomes during this critical development period.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Dieta , Microbioma Gastrointestinal/fisiologia , Adoçantes não Calóricos , Obesidade Infantil/fisiopatologia , Criança , Humanos
15.
Diabetes Care ; 44(6): 1451-1453, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33905347

RESUMO

OBJECTIVE: The impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) on the incidence of new-onset type 2 diabetes and diabetic ketoacidosis (DKA) is unclear. It is unknown whether the coincidence of DKA noted in adult patients with type 2 diabetes is an issue for youth during the coronavirus disease 2019 pandemic. RESEARCH DESIGN AND METHODS: A retrospective single-center medical record review was conducted in a large, urban children's hospital of pediatric subjects presenting with new-onset type 2 diabetes between March and August of 2018 to 2020. RESULTS: The proportion of subjects presenting with new-onset type 2 diabetes in DKA dramatically increased in 2020 (9% in 2018, 3% in 2019, and 20% in 2020, P = 0.029). CONCLUSIONS: In 2020, youth with new-onset type 2 diabetes had a greater incidence of DKA at presentation than previously observed. Future studies should examine the impact of SARS-CoV2 exposure on the presentation of type 2 diabetes in all age-groups to inform better patient care.


Assuntos
COVID-19 , Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Cetoacidose Diabética , Adolescente , Adulto , Criança , Diabetes Mellitus Tipo 1/epidemiologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/epidemiologia , Cetoacidose Diabética/epidemiologia , Cetoacidose Diabética/etiologia , Humanos , Pandemias , RNA Viral , Estudos Retrospectivos , SARS-CoV-2
16.
Cell Mol Bioeng ; 14(1): 15-30, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33633812

RESUMO

INTRODUCTION: The expansion of insulin-producing beta cells during pregnancy is critical to maintain glucose homeostasis in the face of increasing insulin resistance. Prolactin receptor (PRLR) signaling is one of the primary mediators of beta cell expansion during pregnancy, and loss of PRLR signaling results in reduced beta cell mass and gestational diabetes. Harnessing the proliferative potential of prolactin signaling to expand beta cell mass outside of the context of pregnancy requires quantitative understanding of the signaling at the molecular level. METHODS: A mechanistic computational model was constructed to describe prolactin-mediated JAK-STAT signaling in pancreatic beta cells. The effect of different regulatory modules was explored through ensemble modeling. A Bayesian approach for likelihood estimation was used to fit the model to experimental data from the literature. RESULTS: Including receptor upregulation, with either inhibition by SOCS proteins, receptor internalization, or both, allowed the model to match experimental results for INS-1 cells treated with prolactin. The model predicts that faster dimerization and nuclear import rates of STAT5B compared to STAT5A can explain the higher STAT5B nuclear translocation. The model was used to predict the dose response of STAT5B translocation in rat primary beta cells treated with prolactin and reveal possible strategies to modulate STAT5 signaling. CONCLUSIONS: JAK-STAT signaling must be tightly controlled to obtain the biphasic response in STAT5 activation seen experimentally. Receptor up-regulation, combined with SOCS inhibition, receptor internalization, or both is required to match experimental data. Modulating reactions upstream in the signaling can enhance STAT5 activation to increase beta cell survival.

17.
Res Sq ; 2021 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-34312617

RESUMO

Increasing evidence of new-onset diabetes during the COVID19 pandemic indicates that the SARS-CoV2 virus may drive beta-cell dysfunction leading to diabetes, but it is unclear if it is a primary or secondary effect. Here, we present evidence of SARS-CoV-2 infection of pancreatic beta cells in vivo using a robust and reproducible non-human primates model of mild to moderate COVID19 pathogenesis. Pancreas from SARS-CoV-2 infected subjects were positive for the SARS-CoV2 spike protein by immunohistochemistry and structures indicative of viral replication were evident by electron microscopy. Total beta cell area was decreased in SARS-CoV-2-infected pancreas, attributable to beta cell atrophy. Beta cell granularity was decreased. These histologic phenotypes persisted beyond the duration of the clinical disease course. Detailed electron microscopy of SARS-CoV-2 infected beta-cells revealed ultrastructural hallmarks of beta cell stress that are seen in islets of patients with Type 2 diabetes, including disrupted mitochondria and dilated endoplasmic reticulum. To assess the metabolic status of beta cells from SARS-CoV-2-infected subjects, we used fluorescence life-time imaging to measure the ratio of free and bound NADH as a surrogate of glycolytic and oxidative metabolism. We report an increase in free NADH levels, suggesting that beta cells from SARS-CoV-2-infected subjects adopt a more glycolytic metabolic profile. Taken together, we conclude that SARS-CoV-2 infection induces beta cell stress that may compromise beta-cell function beyond the duration of the disease course. This raises the possibility that the beta cell stress and injury may have clinical implications of the long-term future health of patients that have recovered from COVID19.

18.
Am J Physiol Endocrinol Metab ; 299(5): E713-20, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20587750

RESUMO

Embryonic stem cell therapy has been proposed as a therapeutic strategy to restore ß-cell mass and function in T1DM. Recently, a group from Novocell (now ViaCyte) reported successful development of glucose-responsive islet-like structures after implantation of pancreatic endoderm (PE) derived from human embryonic stem cells (hESC) into immune-deficient mice. Our objective was to determine whether implantation of hESC-derived pancreatic endoderm from Novocell into athymic nude rats results in development of viable glucose-responsive pancreatic endocrine tissue. Athymic nude rats were implanted with PE derived from hESC either via implantation into the epididymal fat pads or by subcutaneous implantation into TheraCyte encapsulation devices for 20 wk. Blood glucose, weight, and human insulin/C-peptide secretion were monitored by weekly blood draws. Graft ß-cell function was assessed by a glucose tolerance test, and graft morphology was assessed by immunohistochemistry and immunofluorescence. At 20 wk postimplantation, epididymal fat-implanted PE progressed to develop islet-like structures in 50% of implants, with a mean ß-cell fractional area of 0.8 ± 0.3%. Human C-peptide and insulin were detectable, but at very low levels (C-peptide = 50 ± 26 pmol/l and insulin = 15 ± 7 pmol/l); however, there was no increase in human C-peptide/insulin levels after glucose challenge. There was no development of viable pancreatic tissue or meaningful secretory function when human PE was implanted in the TheraCyte encapsulation devices. These data confirm that islet-like structures develop from hESC differentiated to PE by the protocol developed by NovoCell. However, the extent of endocrine cell formation and secretory function is not yet sufficient to be clinically relevant.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Células-Tronco Embrionárias/transplante , Endoderma/transplante , Células Secretoras de Insulina/transplante , Animais , Glicemia/análise , Peso Corporal/fisiologia , Peptídeo C/sangue , Técnica Direta de Fluorescência para Anticorpo , Técnica Clamp de Glucose , Humanos , Imuno-Histoquímica , Insulina/sangue , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Estudos Longitudinais , Masculino , Ratos , Ratos Nus , Ratos Sprague-Dawley , Organismos Livres de Patógenos Específicos
19.
J Clin Invest ; 117(10): 2869-76, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17823659

RESUMO

Diabetes results from an inadequate mass of functional beta cells, due to either beta cell loss caused by immune assault or the lack of compensation to overcome insulin resistance. Elucidating the mechanisms that regulate beta cell mass has important ramifications for fostering beta cell regeneration and the treatment of diabetes. We report here that Skp2, a substrate recognition component of Skp1-Cul1-F-box (SCF) ubiquitin ligase, played an essential and specific role in regulating the cellular abundance of p27 and was a critical determinant of beta cell proliferation. In Skp2(-/-) mice, accumulation of p27 resulted in enlarged polyploid beta cells as a result of endoreduplication replacing proliferation. Despite beta cell hypertrophy, Skp2(-/-) mice exhibited diminished beta cell mass, hypoinsulinemia, and glucose intolerance. Increased insulin resistance resulting from diet-induced obesity caused Skp2(-/-) mice to become overtly diabetic, because beta cell growth in the absence of cell division was insufficient to compensate for increased metabolic demand. These results indicate that the Skp2-mediated degradation pathway regulating the cellular degradation of p27 is essential for establishing beta cell mass and to respond to increased metabolic demand associated with insulin resistance.


Assuntos
Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Resistência à Insulina/genética , Células Secretoras de Insulina/fisiologia , Proteínas Quinases Associadas a Fase S/fisiologia , Animais , Deleção de Genes , Glucose/metabolismo , Insulina/sangue , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/enzimologia , Camundongos , Camundongos Mutantes , Poliploidia , Proteínas Quinases Associadas a Fase S/genética
20.
JCI Insight ; 5(1)2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31805014

RESUMO

Biallelic mutations of the gene encoding the transcription factor NEUROG3 are associated with a rare disorder that presents in neonates as generalized malabsorption - due to a complete absence of enteroendocrine cells - followed, in early childhood or beyond, by insulin-dependent diabetes mellitus (IDDM). The commonly delayed onset of IDDM suggests a differential requirement for NEUROG3 in endocrine cell generation in the human pancreas versus the intestine. However, previously identified human mutations were hypomorphic and, hence, may have had residual function in pancreas. We report 2 patients with biallelic functionally null variants of the NEUROG3 gene who nonetheless did not present with IDDM during infancy but instead developed permanent IDDM during middle childhood ages. The variants showed no evidence of function in traditional promoter-based assays of NEUROG3 function and also failed to exhibit function in a variety of potentially novel in vitro and in vivo molecular assays designed to discern residual NEUROG3 function. These findings imply that, unlike in mice, pancreatic endocrine cell generation in humans is not entirely dependent on NEUROG3 expression and, hence, suggest the presence of unidentified redundant in vivo pathways in human pancreas capable of yielding ß cell mass sufficient to maintain euglycemia until early childhood.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diabetes Mellitus/genética , Predisposição Genética para Doença , Mutação com Perda de Função , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proliferação de Células , Criança , Diabetes Mellitus Tipo 1 , Células Enteroendócrinas/metabolismo , Feminino , Regulação da Expressão Gênica , Sequências Hélice-Alça-Hélice/genética , Humanos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas , Síndromes de Malabsorção , Masculino , Proteínas do Tecido Nervoso/metabolismo , Pâncreas , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA