Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Autoimmun Rev ; 21(5): 103058, 2022 May.
Article in English | MEDLINE | ID: mdl-35108619

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease that causes a deficit of pancreatic islet ß cells. Millions of individuals worldwide have T1D, and its incidence increases annually. Recent clinical trials have highlighted the limits of conventional immunotherapy in T1D and underscore the need for novel treatments that not only overcome multiple immune dysfunctions, but also help restore islet ß-cell function. To address these two key issues, we have developed a unique and novel procedure designated the Stem Cell Educator therapy, based on the immune education by cord-blood-derived multipotent stem cells (CB-SC). Over the last 10 years, this technology has been evaluated through international multi-center clinical studies, which have demonstrated its clinical safety and efficacy in T1D and other autoimmune diseases. Mechanistic studies revealed that Educator therapy could fundamentally correct the autoimmunity and induce immune tolerance through multiple molecular and cellular mechanisms such as the expression of a master transcription factor autoimmune regulator (AIRE) in CB-SC for T-cell modulation, an expression of Galectin-9 on CB-SC to suppress activated B cells, and secretion of CB-SC-derived exosomes to polarize human blood monocytes/macrophages into type 2 macrophages. Educator therapy is the leading immunotherapy to date to safely and efficiently correct autoimmunity and restore ß cell function in T1D patients.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Autoimmunity , Diabetes Mellitus, Type 1/therapy , Fetal Blood/metabolism , Humans , Insulin-Secreting Cells/metabolism , Stem Cells
3.
Cell Rep ; 11(11): 1797-808, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26074075

ABSTRACT

Obesity is associated with higher incidence of cancer, but the predisposing mechanisms remain poorly understood. The NAD(+)-dependent deacetylase SirT1 orchestrates metabolism, cellular survival, and growth. However, there is no unifying mechanism to explain the metabolic and tumor-related effects of SirT1. In this work, we demonstrate that genetic ablation of the endogenous inhibitor of SirT1, Deleted-in-Breast-Cancer-1 (Dbc1), unexpectedly results in obesity and insulin resistance. Dbc1 deficiency promoted SirT1-dependent gain of function of stearoyl-coenzyme A desaturase 1 (Scd1), increasing plasma and tissue levels of unsaturated fatty acids. The metabolic abnormalities in Dbc1(-/-) mice were reversed by ablation of hepatic SirT1 or by inhibition of Scd1 activity. Furthermore, loss of Dbc1 impaired activation of the master tumor suppressor p53 and treatment with an Scd1 inhibitor extended survival of tumor-prone TP53(-/-) mice by decreasing tumor-related death. Together, our findings illustrate a shared mechanism of obesity and tumor progression mediated by hepatic SirT1 and resulting in the activation of a key lipid synthetic enzyme, with potential therapeutic implications.


Subject(s)
Liver Neoplasms/genetics , Nerve Tissue Proteins/metabolism , Obesity/genetics , Sirtuin 1/metabolism , Stearoyl-CoA Desaturase/metabolism , Animals , Cell Cycle Proteins , HEK293 Cells , Humans , Insulin Resistance/genetics , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Obesity/metabolism , Sirtuin 1/genetics , Stearoyl-CoA Desaturase/antagonists & inhibitors , Stearoyl-CoA Desaturase/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
J Alzheimers Dis ; 46(4): 843-8, 2015.
Article in English | MEDLINE | ID: mdl-25869787

ABSTRACT

Patients with Alzheimer's disease (AD) have a higher risk for developing insulin resistance and diabetes. Amyloid plaques, a hallmark of AD, are composed of amyloid-ß (Aß). Because the mediobasal hypothalamus controls hepatic glucose production, we examined the hypothesis that its exposure to Aß perturbs the regulation of glucose metabolism. The infusion of Aß25-35, but not its scrambled counterpart, into the mediobasal hypothalamus of young rats, increased circulating glucose as a consequence of enhanced hepatic glucose production during pancreatic clamp studies. These findings suggest a link between AD and alterations of glucose metabolism.


Subject(s)
Amyloid beta-Peptides/pharmacology , Blood Glucose/metabolism , Hypothalamus, Middle/drug effects , Liver/metabolism , Peptide Fragments/pharmacology , Animals , Glucose Clamp Technique , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver/drug effects , Male , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
5.
Nat Commun ; 5: 5190, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25307742

ABSTRACT

Insulin integrates hepatic glucose and lipid metabolism, directing nutrients to storage as glycogen and triglyceride. In type 2 diabetes, levels of the former are low and the latter are exaggerated, posing a pathophysiologic and therapeutic conundrum. A branching model of insulin signalling, with FoxO1 presiding over glucose production and Srebp-1c regulating lipogenesis, provides a potential explanation. Here we illustrate an alternative mechanism that integrates glucose production and lipogenesis under the unifying control of FoxO. Liver-specific ablation of three FoxOs (L-FoxO1,3,4) prevents the induction of glucose-6-phosphatase and the repression of glucokinase during fasting, thus increasing lipogenesis at the expense of glucose production. We document a similar pattern in the early phases of diet-induced insulin resistance, and propose that FoxOs are required to enable the liver to direct nutritionally derived carbons to glucose versus lipid metabolism. Our data underscore the heterogeneity of hepatic insulin resistance during progression from the metabolic syndrome to overt diabetes, and the conceptual challenge of designing therapies that curtail glucose production without promoting hepatic lipid accumulation.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Forkhead Transcription Factors/metabolism , Glucose/metabolism , Lipogenesis , Liver/metabolism , Animals , Cell Cycle Proteins , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Fasting/metabolism , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Glucokinase/genetics , Glucokinase/metabolism , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Insulin/metabolism , Lipid Metabolism , Liver/enzymology , Male , Mice , Mice, Inbred C57BL
6.
Diabetes ; 62(4): 1152-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23274895

ABSTRACT

The metabolism of lactate to pyruvate in the mediobasal hypothalamus (MBH) regulates hepatic glucose production. Because astrocytes and neurons are functionally linked by metabolic coupling through lactate transfer via the astrocyte-neuron lactate shuttle (ANLS), we reasoned that astrocytes might be involved in the hypothalamic regulation of glucose metabolism. To examine this possibility, we used the gluconeogenic amino acid proline, which is metabolized to pyruvate in astrocytes. Our results showed that increasing the availability of proline in rats either centrally (MBH) or systemically acutely lowered blood glucose. Pancreatic clamp studies revealed that this hypoglycemic effect was due to a decrease of hepatic glucose production secondary to an inhibition of glycogenolysis, gluconeogenesis, and glucose-6-phosphatase flux. The effect of proline was mimicked by glutamate, an intermediary of proline metabolism. Interestingly, proline's action was markedly blunted by pharmacological inhibition of hypothalamic lactate dehydrogenase (LDH) suggesting that metabolic flux through LDH was required. Furthermore, short hairpin RNA-mediated knockdown of hypothalamic LDH-A, an astrocytic component of the ANLS, also blunted the glucoregulatory action of proline. Thus our studies suggest not only a new role for proline in the regulation of hepatic glucose production but also indicate that hypothalamic astrocytes are involved in the regulatory mechanism as well.


Subject(s)
Astrocytes/metabolism , Glucose/metabolism , Hypothalamus/cytology , Proline/metabolism , Animals , Blood Glucose , Gene Expression Regulation, Enzymologic/drug effects , Glucose/administration & dosage , Glucose/pharmacology , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Insulin/pharmacology , Liver/drug effects , Liver/enzymology , Male , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Rats , Rats, Sprague-Dawley , Somatostatin/pharmacology
7.
Diabetes ; 60(11): 2691-700, 2011 11.
Article in English | MEDLINE | ID: mdl-21896928

ABSTRACT

OBJECTIVE: Sirtuin 1 (SIRT1) and its activator resveratrol are emerging as major regulators of metabolic processes. We investigate the site of resveratrol action on glucose metabolism and the contribution of SIRT1 to these effects. Because the arcuate nucleus in the mediobasal hypothalamus (MBH) plays a pivotal role in integrating peripheral metabolic responses to nutrients and hormones, we examined whether the actions of resveratrol are mediated at the MBH. RESEARCH DESIGN AND METHODS: Sprague Dawley (SD) male rats received acute central (MBH) or systemic injections of vehicle, resveratrol, or SIRT1 inhibitor during basal pancreatic insulin clamp studies. To delineate the pathway(s) by which MBH resveratrol modulates hepatic glucose production, we silenced hypothalamic SIRT1 expression using a short hairpin RNA (shRNA) inhibited the hypothalamic ATP-sensitive potassium (K(ATP)) channel with glibenclamide, or selectively transected the hepatic branch of the vagus nerve while infusing resveratrol centrally. RESULTS: Our studies show that marked improvement in insulin sensitivity can be elicited by acute administration of resveratrol to the MBH or during acute systemic administration. Selective inhibition of hypothalamic SIRT1 using a cell-permeable SIRT1 inhibitor or SIRT1-shRNA negated the effect of central and peripheral resveratrol on glucose production. Blockade of the K(ATP) channel and hepatic vagotomy significantly attenuated the effect of central resveratrol on hepatic glucose production. In addition, we found no evidence for hypothalamic AMPK activation after MBH resveratrol administration. CONCLUSIONS: Taken together, these studies demonstrate that resveratrol improves glucose homeostasis mainly through a central SIRT1-dependent pathway and that the MBH is a major site of resveratrol action.


Subject(s)
Enzyme Activators/pharmacology , Glucose/metabolism , Hypothalamus, Middle/drug effects , Insulin/pharmacology , Liver/drug effects , Sirtuin 1/metabolism , Stilbenes/pharmacology , Animals , Enzyme Activators/administration & dosage , Enzyme Activators/chemistry , Enzyme Inhibitors/administration & dosage , Gene Silencing , Hypoglycemic Agents/pharmacology , Hypothalamus, Middle/metabolism , Insulin Antagonists/pharmacology , Insulin Resistance , KATP Channels/antagonists & inhibitors , Liver/innervation , Liver/metabolism , Male , Organ Specificity , Potassium Channel Blockers/pharmacology , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Resveratrol , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Stilbenes/administration & dosage , Stilbenes/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...