Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Am Chem Soc ; 142(14): 6477-6482, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32175731

RESUMEN

The loss of insulin-producing ß-cells is the central pathological event in type 1 and 2 diabetes, which has led to efforts to identify molecules to promote ß-cell proliferation, protection, and imaging. However, the lack of ß-cell specificity of these molecules jeopardizes their therapeutic potential. A general platform for selective release of small-molecule cargoes in ß-cells over other islet cells ex vivo or other cell-types in an organismal context will be immensely valuable in advancing diabetes research and therapeutic development. Here, we leverage the unusually high Zn(II) concentration in ß-cells to develop a Zn(II)-based prodrug system to selectively and tracelessly deliver bioactive small molecules and fluorophores to ß-cells. The Zn(II)-targeting mechanism enriches the inactive cargo in ß-cells as compared to other pancreatic cells; importantly, Zn(II)-mediated hydrolysis triggers cargo activation. This prodrug system, with modular components that allow for fine-tuning selectivity, should enable the safer and more effective targeting of ß-cells.


Asunto(s)
Linfocitos B/metabolismo , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Zinc/uso terapéutico , Catálisis , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Humanos
2.
Curr Diab Rep ; 16(8): 70, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27313072

RESUMEN

Diabetes is a progressive disease affecting millions of people worldwide. There are several medications and treatment options to improve the life quality of people with diabetes. One of the strategies for the treatment of diabetes could be the use of human pluripotent stem cells or induced pluripotent stem cells. The recent advances in differentiation of stem cells into insulin-secreting beta-like cells in vitro make the transplantation of the stem cell-derived beta-like cells an attractive approach for treatment of type 1 and type 2 diabetes. While stem cell-derived beta-like cells provide an unlimited cell source for beta cell replacement therapies, these cells can also be used as a platform for drug screening or modeling diseases.


Asunto(s)
Diabetes Mellitus Tipo 2/terapia , Células Secretoras de Insulina/citología , Células Madre/citología , Ensayos Clínicos como Asunto , Diabetes Mellitus Tipo 2/etiología , Edición Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Trasplante de Células Madre
3.
J Proteome Res ; 14(8): 3111-3122, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26151086

RESUMEN

Compensatory islet response is a distinct feature of the prediabetic insulin-resistant state in humans and rodents. To identify alterations in the islet proteome that characterize the adaptive response, we analyzed islets from 5 month old male control, high-fat diet fed (HFD), or obese ob/ob mice by LC-MS/MS and quantified ~1100 islet proteins (at least two peptides) with a false discovery rate < 1%. Significant alterations in abundance were observed for ~350 proteins among groups. The majority of alterations were common to both models, and the changes of a subset of ~40 proteins and 12 proteins were verified by targeted quantification using selected reaction monitoring and western blots, respectively. The insulin-resistant islets in both groups exhibited reduced expression of proteins controlling energy metabolism, oxidative phosphorylation, hormone processing, and secretory pathways. Conversely, an increased expression of molecules involved in protein synthesis and folding suggested effects in endoplasmic reticulum stress response, cell survival, and proliferation in both insulin-resistant models. In summary, we report a unique comparison of the islet proteome that is focused on the compensatory response in two insulin-resistant rodent models that are not overtly diabetic. These data provide a valuable resource of candidate proteins to the scientific community to undertake further studies aimed at enhancing ß-cell mass in patients with diabetes. The data are available via the MassIVE repository, under accession no. MSV000079093.


Asunto(s)
Resistencia a la Insulina , Islotes Pancreáticos/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Secuencia de Aminoácidos , Animales , Western Blotting , Cromatografía Liquida , Dieta Alta en Grasa , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Datos de Secuencia Molecular , Espectrometría de Masas en Tándem
4.
Am J Physiol Endocrinol Metab ; 307(10): E906-18, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25249504

RESUMEN

Studies in both humans and rodents suggest that maternal diabetes leads to a higher risk of the fetus developing impaired glucose tolerance and obesity during adulthood. However, the impact of hyperinsulinemia in the mother on glucose homeostasis in the offspring has not been fully explored. We aimed to determine the consequences of maternal insulin resistance on offspring metabolism and endocrine pancreas development using the LIRKO mouse model, which exhibits sustained hyperinsulinemia and transient increase in blood glucose concentrations during pregnancy. We examined control offspring born to either LIRKO or control mothers on embryonic days 13.5, 15.5, and 17.5 and postpartum days 0, 4, and 10. Control offspring born to LIRKO mothers displayed low birth weights and subsequently rapidly gained weight, and their blood glucose and plasma insulin concentrations were higher than offspring born to control mothers in early postnatal life. In addition, concentrations of plasma leptin, glucagon, and active GLP-1 were higher in control pups from LIRKO mothers. Analyses of the endocrine pancreas revealed significantly reduced ß-cell area in control offspring of LIRKO mothers shortly after birth. ß-Cell proliferation and total islet number were also lower in control offspring of LIRKO mothers during early postnatal days. Together, these data indicate that maternal hyperinsulinemia and the transient hyperglycemia impair endocrine pancreas development in the control offspring and induce multiple metabolic alterations in early postnatal life. The relatively smaller ß-cell mass/area and ß-cell proliferation in these control offspring suggest cell-autonomous epigenetic mechanisms in the regulation of islet growth and development.


Asunto(s)
Glucemia/metabolismo , Diabetes Gestacional , Hiperglucemia/metabolismo , Hiperinsulinismo/metabolismo , Resistencia a la Insulina , Islotes Pancreáticos/patología , Complicaciones del Embarazo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Glucagón/sangre , Péptido 1 Similar al Glucagón/sangre , Hiperglucemia/sangre , Hiperglucemia/patología , Hiperinsulinismo/sangre , Hiperinsulinismo/patología , Insulina/sangre , Células Secretoras de Insulina/citología , Leptina/sangre , Ratones , Tamaño de los Órganos , Fenotipo , Embarazo , Complicaciones del Embarazo/sangre , Complicaciones del Embarazo/patología , Efectos Tardíos de la Exposición Prenatal/sangre , Efectos Tardíos de la Exposición Prenatal/patología , Aumento de Peso
5.
Nat Cell Biol ; 26(3): 421-437, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38409327

RESUMEN

Type 1 diabetes (T1D) is characterized by the destruction of pancreatic ß-cells. Several observations have renewed the interest in ß-cell RNA sensors and editors. Here, we report that N6-methyladenosine (m6A) is an adaptive ß-cell safeguard mechanism that controls the amplitude and duration of the antiviral innate immune response at T1D onset. m6A writer methyltransferase 3 (METTL3) levels increase drastically in ß-cells at T1D onset but rapidly decline with disease progression. m6A sequencing revealed the m6A hypermethylation of several key innate immune mediators, including OAS1, OAS2, OAS3 and ADAR1 in human islets and EndoC-ßH1 cells at T1D onset. METTL3 silencing enhanced 2'-5'-oligoadenylate synthetase levels by increasing its mRNA stability. Consistently, in vivo gene therapy to prolong Mettl3 overexpression specifically in ß-cells delayed diabetes progression in the non-obese diabetic mouse model of T1D. Mechanistically, the accumulation of reactive oxygen species blocked upregulation of METTL3 in response to cytokines, while physiological levels of nitric oxide enhanced METTL3 levels and activity. Furthermore, we report that the cysteines in position C276 and C326 in the zinc finger domains of the METTL3 protein are sensitive to S-nitrosylation and are important to the METTL3-mediated regulation of oligoadenylate synthase mRNA stability in human ß-cells. Collectively, we report that m6A regulates the innate immune response at the ß-cell level during the onset of T1D in humans.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Animales , Humanos , Ratones , Adenosina Desaminasa/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Inmunidad Innata , Células Secretoras de Insulina/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Oxidación-Reducción
6.
Elife ; 122023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37732504

RESUMEN

Pancreatic α-cells secrete glucagon, an insulin counter-regulatory peptide hormone critical for the maintenance of glucose homeostasis. Investigation of the function of human α-cells remains a challenge due to the lack of cost-effective purification methods to isolate high-quality α-cells from islets. Here, we use the reaction-based probe diacetylated Zinpyr1 (DA-ZP1) to introduce a novel and simple method for enriching live α-cells from dissociated human islet cells with ~95% purity. The α-cells, confirmed by sorting and immunostaining for glucagon, were cultured up to 10 days to form α-pseudoislets. The α-pseudoislets could be maintained in culture without significant loss of viability, and responded to glucose challenge by secreting appropriate levels of glucagon. RNA-sequencing analyses (RNA-seq) revealed that expression levels of key α-cell identity genes were sustained in culture while some of the genes such as DLK1, GSN, SMIM24 were altered in α-pseudoislets in a time-dependent manner. In conclusion, we report a method to sort human primary α-cells with high purity that can be used for downstream analyses such as functional and transcriptional studies.


Asunto(s)
Células Secretoras de Glucagón , Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Glucagón/metabolismo , Transcriptoma , Islotes Pancreáticos/metabolismo , Insulina/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucosa/metabolismo , Fluoresceínas/metabolismo , Células Secretoras de Insulina/metabolismo
7.
bioRxiv ; 2023 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-37577492

RESUMEN

N 6 -methyladenosine (m 6 A) is the most abundant chemical modification in mRNA, and plays important roles in human and mouse embryonic stem cell pluripotency, maintenance, and differentiation. We have recently reported, for the first time, the role of m 6 A in the postnatal control of ß-cell function in physiological states and in Type 1 and 2 Diabetes. However, the precise mechanisms by which m 6 A acts to regulate the development of human and mouse ß-cells are unexplored. Here, we show that the m 6 A landscape is dynamic during human pancreas development, and that METTL14, one of the m 6 A writer complex proteins, is essential for the early differentiation of both human and mouse ß-cells.

8.
bioRxiv ; 2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36824909

RESUMEN

Type 1 Diabetes (T1D) is characterized by autoimmune-mediated destruction of insulin-producing ß-cells. Several observations have renewed interest in the innate immune system as an initiator of the disease process against ß-cells. Here, we show that N 6 -Methyladenosine (m 6 A) is an adaptive ß-cell safeguard mechanism that accelerates mRNA decay of the 2'-5'-oligoadenylate synthetase (OAS) genes to control the antiviral innate immune response at T1D onset. m 6 A writer methyltransferase 3 (METTL3) levels increase drastically in human and mouse ß-cells at T1D onset but rapidly decline with disease progression. Treatment of human islets and EndoC-ßH1 cells with pro-inflammatory cytokines interleukin-1 ß and interferon α mimicked the METTL3 upregulation seen at T1D onset. Furthermore, m 6 A-sequencing revealed the m 6 A hypermethylation of several key innate immune mediators including OAS1, OAS2, and OAS3 in human islets and EndoC-ßH1 cells challenged with cytokines. METTL3 silencing in human pseudoislets or EndoC-ßH1 cells enhanced OAS levels by increasing its mRNA stability upon cytokine challenge. Consistently, in vivo gene therapy, to prolong Mettl3 overexpression specifically in ß-cells, delayed diabetes progression in the non-obese diabetic (NOD) mouse model of T1D by limiting the upregulation of Oas pointing to potential therapeutic relevance. Mechanistically, the accumulation of reactive oxygen species blocked METTL3 upregulation in response to cytokines, while physiological levels of nitric oxide promoted its expression in human islets. Furthermore, for the first time to our knowledge, we show that the cysteines in position C276 and C326 in the zinc finger domain of the METTL3 protein are sensitive to S-nitrosylation (SNO) and are significant for the METTL3 mediated regulation of OAS mRNA stability in human ß-cells in response to cytokines. Collectively, we report that m 6 A regulates human and mouse ß-cells to control the innate immune response during the onset of T1D and propose targeting METTL3 to prevent ß-cell death in T1D.

9.
JCI Insight ; 7(16)2022 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-35819843

RESUMEN

Adaptation to increased insulin demand is mediated by ß cell proliferation and neogenesis, among other mechanisms. Although it is known that pancreatic ß cells can arise from ductal progenitors, these observations have been limited mostly to the neonatal period. We have recently reported that the duct is a source of insulin-secreting cells in adult insulin-resistant states. To further explore the signaling pathways underlying the dynamic ß cell reserve during insulin resistance, we undertook human islet and duct transplantations under the kidney capsule of immunodeficient NOD/SCID-γ (NSG) mouse models that were pregnant, were insulin-resistant, or had insulin resistance superimposed upon pregnancy (insulin resistance + pregnancy), followed by single-nucleus RNA-Seq (snRNA-Seq) on snap-frozen graft samples. We observed an upregulation of proliferation markers (e.g., NEAT1) and expression of islet endocrine cell markers (e.g., GCG and PPY), as well as mature ß cell markers (e.g., INS), in transplanted human duct grafts in response to high insulin demand. We also noted downregulation of ductal cell identity genes (e.g., KRT19 and ONECUT2) coupled with upregulation of ß cell development and insulin signaling pathways. These results indicate that subsets of ductal cells are able to gain ß cell identity and reflect a form of compensation during the adaptation to insulin resistance in both physiological and pathological states.


Asunto(s)
Resistencia a la Insulina , Adulto , Animales , Femenino , Proteínas de Homeodominio , Humanos , Recién Nacido , Insulina/metabolismo , Resistencia a la Insulina/genética , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Embarazo , RNA-Seq , Factores de Transcripción
10.
Nat Metab ; 4(1): 76-89, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35058633

RESUMEN

MODY8 (maturity-onset diabetes of the young, type 8) is a dominantly inherited monogenic form of diabetes associated with mutations in the carboxyl ester lipase (CEL) gene expressed by pancreatic acinar cells. MODY8 patients develop childhood-onset exocrine pancreas dysfunction followed by diabetes during adulthood. However, it is unclear how CEL mutations cause diabetes. In the present study, we report the transfer of CEL proteins from acinar cells to ß-cells as a form of cross-talk between exocrine and endocrine cells. Human ß-cells show a relatively higher propensity for internalizing the mutant versus the wild-type CEL protein. After internalization, the mutant protein forms stable intracellular aggregates leading to ß-cell secretory dysfunction. Analysis of pancreas sections from a MODY8 patient reveals the presence of CEL protein in the few extant ß-cells. The present study provides compelling evidence for the mechanism by which a mutant gene expressed specifically in acinar cells promotes dysfunction and loss of ß-cells to cause diabetes.


Asunto(s)
Comunicación Celular , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Páncreas Exocrino/metabolismo , Células Acinares/metabolismo , Animales , Humanos , Inmunohistoquímica , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Lipasa/química , Lipasa/genética , Lipasa/metabolismo , Ratones , Mutación , Páncreas/metabolismo , Páncreas/patología , Transporte de Proteínas , Solubilidad
11.
Diabetes Metab Res Rev ; 27(6): 575-83, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21584921

RESUMEN

BACKGROUND: To increase the success rate in xenogeneic islet transplantation, proper assessment of graft mass is required following transplantation. For this reason, we aimed to develop a suitable fluorescence imaging system to monitor islet xenograft survival in diabetic mice. METHODS: Adenovirus vector encoding enhanced green fluorescent protein-transduced rat pancreatic islets were transplanted under the renal capsule of streptozotocin-induced diabetic mice and the fluorescence signal was quantified over time using a cooled charge-coupled device. Non-fasting blood glucose levels were recorded during the same period. Insulin release from transduced and control islets was detected via enzyme-linked immunosorbent assay. RESULTS: Adenovirus vector encoding enhanced green fluorescent protein infection did not alter the function or survival of pancreatic islets post transduction. A direct correlation was found between the number of islets (250-750) transplanted under the kidney capsule and the blood glucose recovery. CONCLUSIONS: Fluorescence imaging appears to be a useful tool for quantitative assessment of islet cell viability post transplantation and could permit earlier detection of graft rejection.


Asunto(s)
Supervivencia de Injerto/fisiología , Proteínas Fluorescentes Verdes , Trasplante de Islotes Pancreáticos/métodos , Trasplante Heterólogo , Adenoviridae/genética , Animales , Glucemia/metabolismo , Supervivencia Celular , Diabetes Mellitus Experimental , Femenino , Fluorescencia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratas , Ratas Wistar
12.
Genome Med ; 13(1): 128, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376240

RESUMEN

BACKGROUND: Human pancreatic islets are a central focus of research in metabolic studies. Transcriptomics is frequently used to interrogate alterations in cultured human islet cells using single-cell RNA-sequencing (scRNA-seq). We introduce single-nucleus RNA-sequencing (snRNA-seq) as an alternative approach for investigating transplanted human islets. METHODS: The Nuclei EZ protocol was used to obtain nuclear preparations from fresh and frozen human islet cells. Such preparations were first used to generate snRNA-seq datasets and compared to scRNA-seq output obtained from cells from the same donor. Finally, we employed snRNA-seq to obtain the transcriptomic profile of archived human islets engrafted in immunodeficient animals. RESULTS: We observed virtually complete concordance in identifying cell types and gene proportions as well as a strong association of global and islet cell type gene signatures between scRNA-seq and snRNA-seq applied to fresh and frozen cultured or transplanted human islet samples. CONCLUSIONS: We propose snRNA-seq as a reliable strategy to probe transcriptomic profiles of freshly harvested or frozen sources of transplanted human islet cells especially when scRNA-seq is not ideal.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Islotes Pancreáticos/metabolismo , Análisis de la Célula Individual , Transcriptoma , Animales , Biomarcadores , Humanos , Islotes Pancreáticos/citología , Trasplante de Islotes Pancreáticos , Ratones , Análisis de Secuencia de ADN , Análisis de la Célula Individual/métodos , Secuenciación del Exoma
13.
J Mol Endocrinol ; 66(4): 325-338, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33875613

RESUMEN

Strategies to increase functional pancreatic beta cell mass is of great interest in diabetes-related research. TNF-related apoptosis-inducing ligand (TRAIL) is well known to promote proliferation and survival in various cell types, including vascular smooth muscle and endothelial cells. Correlation between the protective nature of TRAIL on these cells and its proliferative effect is noteworthy. TRAIL's seemingly protective/therapeutic effect in diabetes prompted us to question whether it may act as an inducer of proliferation in pancreatic beta cells. We used rat primary islet cells and MIN6 mouse beta cell line to investigate TRAIL-induced proliferation. Cell viability and/or death was analyzed by MTT, WST-1, and Annexin-V/PI assays, while proliferation rates and pathways were assessed via immunocytochemical and Western blot analyses. Receptor neutralization antibodies identified the mediator receptors. Recombinant soluble TRAIL (sTRAIL) treatment led to 1.6-fold increased proliferation in insulin-positive cells in dispersed rat islets compared to the untreated group, while adenovirus-mediated overexpression of TRAIL increased the number of proliferating beta cells up to more than six-fold. sTRAIL or adenoviral vector-mediated TRAIL overexpression induced proliferation in MIN6 cells also. TRAIL's proliferative effect was mediated via AKT activation, which was suppressed upon specific inhibition. Neutralization of each TRAIL receptor reversed the proliferative effect to some degree, with the highest level of inhibition in death receptor 5 (DR5) blockage in MIN6 cells and in decoy receptor 1 (DcR1) blockage in primary rat beta cells. Thus, TRAIL induces proliferation in rodent pancreatic beta cells through activation of the AKT pathway.


Asunto(s)
Diabetes Mellitus/genética , Células Secretoras de Insulina/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Miembro 10c de Receptores del Factor de Necrosis Tumoral/genética , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Animales , Apoptosis/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Diabetes Mellitus/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación de la Expresión Génica/genética , Humanos , Células Secretoras de Insulina/patología , Ratones , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Proteínas Proto-Oncogénicas c-akt/genética , Ratas
14.
Life Sci Alliance ; 4(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33514654

RESUMEN

Highly sensitive approaches to target insulin-expressing cells would allow more effective imaging, sorting, and analysis of pancreatic ß-cells. Here, we introduce the use of a reaction-based probe, diacetylated Zinpyr1 (DA-ZP1), to image pancreatic ß-cells and ß-like cells derived from human pluripotent stem cells. We harness the high intracellular zinc concentration of ß-cells to induce a fluorescence signal in cells after administration of DA-ZP1. Given its specificity and rapid uptake by cells, we used DA-ZP1 to purify live stem cell-derived ß-like cells as confirmed by immunostaining analysis. We tested the ability of DA-ZP1 to image transplanted human islet grafts and endogenous mouse pancreatic islets in vivo after its systemic administration into mice. Thus, DA-ZP1 enables purification of insulin-secreting ß-like cells for downstream applications, such as functional studies, gene-expression, and cell-cell interaction analyses and can be used to label engrafted human islets and endogenous mouse islets in vivo.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Imagen Molecular/métodos , Sondas Moleculares , Animales , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Colorantes Fluorescentes/química , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Ratones , Sondas Moleculares/química , Estructura Molecular
15.
Mol Metab ; 47: 101164, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33453419

RESUMEN

The insulin and insulin-like growth factor-1 (IGF-1) receptors are important for the growth and development of embryonic tissues. To directly define their roles in the maintenance of pluripotency and differentiation of stem cells, we knocked out both receptors in induced pluripotent stem cells (iPSCs). iPSCs lacking both insulin and IGF-1 receptors (double knockout, DKO) exhibited preserved pluripotency potential despite decreased expression of transcription factors Lin28a and Tbx3 compared to control iPSCs. While embryoid body and teratoma assays revealed an intact ability of DKO iPSCs to form all three germ layers, the latter were composed of primitive neuroectodermal tumor-like cells in the DKO group. RNA-seq analyses of control vs DKO iPSCs revealed differential regulation of pluripotency, developmental, E2F1, and apoptosis pathways. Signaling analyses pointed to downregulation of the AKT/mTOR pathway and upregulation of the STAT3 pathway in DKO iPSCs in the basal state and following stimulation with insulin/IGF-1. Directed differentiation toward the three lineages was dysregulated in DKO iPSCs, with significant downregulation of key markers (Cebpα, Fas, Pparγ, and Fsp27) in adipocytes and transcription factors (Ngn3, Isl1, Pax6, and Neurod1) in pancreatic endocrine progenitors. Furthermore, differentiated pancreatic endocrine progenitor cells from DKO iPSCs showed increased apoptosis. We conclude that insulin and insulin-like growth factor-1 receptors are indispensable for normal lineage development and perturbations in the function and signaling of these receptors leads to upregulation of alternative compensatory pathways to maintain pluripotency.


Asunto(s)
Adipocitos/metabolismo , Desarrollo Embrionario , Células Madre Pluripotentes Inducidas/metabolismo , Insulina/metabolismo , Receptor IGF Tipo 1/metabolismo , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Proliferación Celular , Fibroblastos , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso , Receptor IGF Tipo 1/genética , Factor de Transcripción STAT3 , Transducción de Señal
16.
Stem Cell Reports ; 15(5): 1067-1079, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33125875

RESUMEN

The role of leptin receptor (OB-R) signaling in linking pluripotency with growth and development and the consequences of dysfunctional leptin signaling on progression of metabolic disease is poorly understood. Using a global unbiased proteomics approach we report that embryonic fibroblasts (MEFs) carrying the db/db mutation exhibit metabolic abnormalities, while their reprogrammed induced pluripotent stem cells (iPSCs) show altered expression of proteins involved in embryonic development. An upregulation in expression of eukaryotic translation initiation factor 4e (Eif4e) and Stat3 binding to the Eif4e promoter was supported by enhanced protein synthesis in mutant iPSCs. Directed differentiation of db/db iPSCs toward the neuronal lineage showed defects. Gene editing to correct the point mutation in db/db iPSCs using CRISPR-Cas9, restored expression of neuronal markers and protein synthesis while reversing the metabolic defects. These data imply a direct role for OB-R in regulating metabolism in embryonic fibroblasts and key developmental pathways in iPSCs.


Asunto(s)
Factor 4E Eucariótico de Iniciación/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Biosíntesis de Proteínas , Receptores de Leptina/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Sistemas CRISPR-Cas , Diferenciación Celular , Linaje de la Célula , Factor 4E Eucariótico de Iniciación/genética , Fibroblastos/metabolismo , Edición Génica , Regulación del Desarrollo de la Expresión Génica , Metaboloma , Ratones , Ratones Noqueados , Neurogénesis , Proteínas , Proteómica , Receptores de Leptina/genética
17.
Nat Metab ; 1(8): 765-774, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31867565

RESUMEN

The regulation of islet cell biology is critical for glucose homeostasis1.N6 -methyladenosine (m6A) is the most abundant internal messenger RNA (mRNA) modification in mammals2. Here we report that the m6A landscape segregates human type 2 diabetes (T2D) islets from controls significantly better than the transcriptome and that m6A is vital for ß-cell biology. m6A-sequencing in human T2D islets reveals several hypomethylated transcripts involved in cell-cycle progression, insulin secretion, and the Insulin/IGF1-AKT-PDX1 pathway. Depletion of m6A levels in EndoC-ßH1 induces cell-cycle arrest and impairs insulin secretion by decreasing AKT phosphorylation and PDX1 protein levels. ß-cell specific Mettl14 knock-out mice, which display reduced m6A levels, mimic the islet phenotype in human T2D with early diabetes onset and mortality due to decreased ß-cell proliferation and insulin degranulation. Our data underscore the significance of RNA methylation in regulating human ß-cell biology, and provide a rationale for potential therapeutic targeting of m6A modulators to preserve ß-cell survival and function in diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Células Secretoras de Insulina/fisiología , ARN Mensajero/metabolismo , Animales , Humanos , Secreción de Insulina , Metilación
18.
JCI Insight ; 4(8)2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30996131

RESUMEN

The identification of new sources of ß cells is an important endeavor with therapeutic implications for diabetes. Insulin resistance, in physiological states such as pregnancy or in pathological states such as type 2 diabetes (T2D), is characterized by a compensatory increase in ß cell mass. To explore the existence of a dynamic ß cell reserve, we superimposed pregnancy on the liver-specific insulin receptor-KO (LIRKO) model of insulin resistance that already exhibits ß cell hyperplasia and used lineage tracing to track the source of new ß cells. Although both control and LIRKO mice displayed increased ß cell mass in response to the relative insulin resistance of pregnancy, the further increase in mass in the latter supported a dynamic source that could be traced to pancreatic ducts. Two observations support the translational significance of these findings. First, NOD/SCID-γ LIRKO mice that became pregnant following cotransplantation of human islets and human ducts under the kidney capsule showed enhanced ß cell proliferation and an increase in ductal cells positive for transcription factors expressed during ß cell development. Second, we identified duct cells positive for immature ß cell markers in pancreas sections from pregnant humans and in individuals with T2D. Taken together, during increased insulin demand, ductal cells contribute to the compensatory ß cell pool by differentiation/neogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Diabetes Mellitus Tipo 2/terapia , Células Secretoras de Insulina/fisiología , Conductos Pancreáticos/citología , Embarazo/fisiología , Adulto , Animales , Proliferación Celular , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos , Ratones , Ratones Noqueados , Persona de Mediana Edad , Conductos Pancreáticos/trasplante , Receptor de Insulina/genética , Quimera por Trasplante , Adulto Joven
19.
Nat Metab ; 1(5): 509-518, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31423480

RESUMEN

Type 1 diabetes (T1D) is characterized by pancreatic islet infiltration by autoreactive immune cells and a near-total loss of ß-cells1. Restoration of insulin-producing ß-cells coupled with immunomodulation to suppress the autoimmune attack has emerged as a potential approach to counter T1D2-4. Here we report that enhancing ß-cell mass early in life, in two models of female NOD mice, results in immunomodulation of T-cells, reduced islet infiltration and lower ß-cell apoptosis, that together protect them from developing T1D. The animals displayed altered ß-cell antigens, and islet transplantation studies showed prolonged graft survival in the NOD-LIRKO model. Adoptive transfer of splenocytes from the NOD-LIRKOs prevented development of diabetes in pre-diabetic NOD mice. A significant increase in the splenic CD4+CD25+FoxP3+ regulatory T-cell (Treg) population was observed to underlie the protected phenotype since Treg depletion rendered NOD-LIRKO mice diabetic. The increase in Tregs coupled with activation of TGF-ß/SMAD3 signaling pathway in pathogenic T-cells favored reduced ability to kill ß-cells. These data support a previously unidentified observation that initiating ß-cell proliferation, alone, prior to islet infiltration by immune cells alters the identity of ß-cells, decreases pathologic self-reactivity of effector cells and increases Tregs to prevent progression of T1D.


Asunto(s)
Proliferación Celular , Diabetes Mellitus Tipo 1/patología , Sistema Inmunológico/inmunología , Células Secretoras de Insulina/patología , Animales , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Progresión de la Enfermedad , Humanos , Ratones
20.
Mol Metab ; 18: 153-163, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30316806

RESUMEN

OBJECTIVES: Insulin receptor (IR)-mediated signaling is involved in the regulation of pluripotent stem cells; however, its direct effects on regulating the maintenance of pluripotency and lineage development are not fully understood. The main objective of this study is to understand the role of IR signaling in pluripotency and lineage development. METHODS: To explore the role of IR signaling, we generated IR knock-out (IRKO) mouse induced pluripotent stem cells (miPSCs) from E14.5 mouse embryonic fibroblasts (MEFs) of global IRKO mice using a cocktail of four reprogramming factors: Oct4, Sox2, Klf4, cMyc. We performed pluripotency characterization and directed the differentiation of control and IRKO iPSCs into neural progenitors (ectoderm), adipocyte progenitors (mesoderm), and pancreatic beta-like cells (endoderm). We mechanistically confirmed these findings via phosphoproteomics analyses of control and IRKO iPSCs. RESULTS: Interestingly, expression of pluripotency markers including Klf4, Lin28a, Tbx3, and cMyc were upregulated, while abundance of Oct4 and Nanog were enhanced by 4-fold and 3-fold, respectively, in IRKO iPSCs. Analyses of signaling pathways demonstrated downregulation of phospho-STAT3, p-mTor and p-Erk and an increase in the total mTor and Erk proteins in IRKO iPSCs in the basal unstimulated state. Stimulation with leukemia inhibitory factor (LIF) showed a ∼33% decrease of phospho-ERK in IRKO iPSCs. On the contrary, Erk phosphorylation was increased during in vitro spontaneous differentiation of iPSCs lacking IRs. Lineage-specific directed differentiation of the iPSCs revealed that cells lacking IR showed enhanced expression of neuronal lineage markers (Pax6, Tubb3, Ascl1 and Oligo2) while exhibiting a decrease in adipocyte (Fas, Acc, Pparγ, Fabp4, C/ebpα, and Fsp27) and pancreatic beta cell markers (Ngn3, Isl1, and Sox9). Further molecular characterization by phosphoproteomics confirmed the novel IR-mediated regulation of the global pluripotency network including several key proteins involved in diverse aspects of growth and embryonic development. CONCLUSION: We report, for the first time to our knowledge, the phosphoproteome of insulin, IGF1, and LIF stimulation in mouse iPSCs to reveal the importance of insulin receptor signaling for the maintenance of pluripotency and lineage determination.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal , Adipocitos/citología , Adipocitos/metabolismo , Animales , Linaje de la Célula , Células Cultivadas , Células Madre Pluripotentes Inducidas/citología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Factor 4 Similar a Kruppel , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteoma/genética , Proteoma/metabolismo , Receptor de Insulina/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA