Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(18): e2318619121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38657050

ABSTRACT

Nonalcoholic fatty liver disease, recently renamed metabolic dysfunction-associated steatotic liver disease (MASLD), is a progressive metabolic disorder that begins with aberrant triglyceride accumulation in the liver and can lead to cirrhosis and cancer. A common variant in the gene PNPLA3, encoding the protein PNPLA3-I148M, is the strongest known genetic risk factor for MASLD. Despite its discovery 20 y ago, the function of PNPLA3, and now the role of PNPLA3-I148M, remain unclear. In this study, we sought to dissect the biogenesis of PNPLA3 and PNPLA3-I148M and characterize changes induced by endogenous expression of the disease-causing variant. Contrary to bioinformatic predictions and prior studies with overexpressed proteins, we demonstrate here that PNPLA3 and PNPLA3-I148M are not endoplasmic reticulum-resident transmembrane proteins. To identify their intracellular associations, we generated a paired set of isogenic human hepatoma cells expressing PNPLA3 and PNPLA3-I148M at endogenous levels. Both proteins were enriched in lipid droplet, Golgi, and endosomal fractions. Purified PNPLA3 and PNPLA3-I148M proteins associated with phosphoinositides commonly found in these compartments. Despite a similar fractionation pattern as the wild-type variant, PNPLA3-I148M induced morphological changes in the Golgi apparatus, including increased lipid droplet-Golgi contact sites, which were also observed in I148M-expressing primary human patient hepatocytes. In addition to lipid droplet accumulation, PNPLA3-I148M expression caused significant proteomic and transcriptomic changes that resembled all stages of liver disease. Cumulatively, we validate an endogenous human cellular system for investigating PNPLA3-I148M biology and identify the Golgi apparatus as a central hub of PNPLA3-I148M-driven cellular change.


Subject(s)
Acyltransferases , Golgi Apparatus , Lipid Droplets , Phospholipases A2, Calcium-Independent , Humans , Acyltransferases/metabolism , Golgi Apparatus/metabolism , Lipase/metabolism , Lipase/genetics , Lipid Droplets/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Phospholipases A2, Calcium-Independent/metabolism
2.
bioRxiv ; 2023 Oct 14.
Article in English | MEDLINE | ID: mdl-37873239

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD), recently renamed metabolic dysfunction-associated steatotic liver disease (MASLD), is a progressive metabolic disorder that begins with aberrant triglyceride accumulation in the liver and can lead to cirrhosis and cancer. A common variant in the gene PNPLA3, encoding the protein PNPLA3-I148M, is the strongest known genetic risk factor for MASLD to date. Despite its discovery twenty years ago, the function of PNPLA3, and now the role of PNPLA3-I148M, remain unclear. In this study, we sought to dissect the biogenesis of PNPLA3 and PNPLA3-I148M and characterize changes induced by endogenous expression of the disease-causing variant. Contrary to bioinformatic predictions and prior studies with overexpressed proteins, we demonstrate here that PNPLA3 and PNPLA3-I148M are not endoplasmic reticulum-resident transmembrane proteins. To identify their intracellular associations, we generated a paired set of isogenic human hepatoma cells expressing PNPLA3 and PNPLA3-I148M at endogenous levels. Both proteins were enriched in lipid droplet, Golgi, and endosomal fractions. Purified PNPLA3 and PNPLA3-I148M proteins associated with phosphoinositides commonly found in these compartments. Despite a similar fractionation pattern as the wild-type variant, PNPLA3-I148M induced morphological changes in the Golgi apparatus, including increased lipid droplet-Golgi contact sites, which were also observed in I148M-expressing primary human patient hepatocytes. In addition to lipid droplet accumulation, PNPLA3-I148M expression caused significant proteomic and transcriptomic changes that resembled all stages of liver disease. Cumulatively, we validate an endogenous human cellular system for investigating PNPLA3-I148M biology and identify the Golgi apparatus as a central hub of PNPLA3-I148M-driven cellular change.

3.
Nat Genet ; 54(11): 1652-1663, 2022 11.
Article in English | MEDLINE | ID: mdl-36280732

ABSTRACT

Nonalcoholic fatty liver (NAFL) and its sequelae are growing health problems. We performed a genome-wide association study of NAFL, cirrhosis and hepatocellular carcinoma, and integrated the findings with expression and proteomic data. For NAFL, we utilized 9,491 clinical cases and proton density fat fraction extracted from 36,116 liver magnetic resonance images. We identified 18 sequence variants associated with NAFL and 4 with cirrhosis, and found rare, protective, predicted loss-of-function variants in MTARC1 and GPAM, underscoring them as potential drug targets. We leveraged messenger RNA expression, splicing and predicted coding effects to identify 16 putative causal genes, of which many are implicated in lipid metabolism. We analyzed levels of 4,907 plasma proteins in 35,559 Icelanders and 1,459 proteins in 47,151 UK Biobank participants, identifying multiple proteins involved in disease pathogenesis. We show that proteomics can discriminate between NAFL and cirrhosis. The present study provides insights into the development of noninvasive evaluation of NAFL and new therapeutic options.


Subject(s)
Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/genetics , Proteomics , Genome-Wide Association Study , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism
4.
Sci Adv ; 8(10): eabj2917, 2022 Mar 11.
Article in English | MEDLINE | ID: mdl-35263131

ABSTRACT

Circulating corticosteroids orchestrate stress adaptation, including inhibition of inflammation. While pathways governing corticosteroid biosynthesis and intracellular signaling are well understood, less is known about mechanisms controlling plasma corticosteroid transport. Here, we show that hepatocyte KLF15 (Kruppel-like factor 15) controls plasma corticosteroid transport and inflammatory responses through direct transcriptional activation of Serpina6, which encodes corticosteroid-binding globulin (CBG). Klf15-deficient mice have profoundly low CBG, reduced plasma corticosteroid binding capacity, and heightened mortality during inflammatory stress. These defects are completely rescued by reconstituting CBG, supporting that KLF15 works primarily through CBG to control plasma corticosterone homeostasis. To understand transcriptional mechanisms, we generated the first KLF15 cistromes using newly engineered Klf153xFLAG mice. Unexpectedly, liver KLF15 is predominantly promoter enriched, including Serpina6, where it binds a palindromic GC-rich motif, opens chromatin, and transactivates genes with minimal associated direct gene repression. Overall, we provide critical mechanistic insight into KLF15 function and identify a hepatocyte-intrinsic transcriptional module that potently regulates systemic corticosteroid transport and inflammation.

5.
Nucleic Acid Ther ; 31(5): 324-340, 2021 10.
Article in English | MEDLINE | ID: mdl-34297902

ABSTRACT

Human genome wide association studies confirm the association of the rs738409 single nucleotide polymorphism (SNP) in the gene encoding protein patatin like phospholipase domain containing 3 (PNPLA3) with nonalcoholic fatty liver disease (NAFLD); the presence of the resulting mutant PNPLA3 I148M protein is a driver of nonalcoholic steatohepatitis (NASH). While Pnpla3-deficient mice do not display an adverse phenotype, the safety of knocking down endogenous wild type PNPLA3 in humans remains unknown. To expand the scope of a potential targeted NAFLD therapeutic to both homozygous and heterozygous PNPLA3 rs738409 populations, we sought to identify a minor allele-specific small interfering RNA (siRNA). Limiting our search to SNP-spanning triggers, a series of chemically modified siRNA were tested in vitro for activity and selectivity toward PNPLA3 rs738409 mRNA. Conjugation of the siRNA to a triantennary N-acetylgalactosamine (GalNAc) ligand enabled in vivo screening using adeno-associated virus to overexpress human PNPLA3I148M versus human PNPLA3I148I in mouse livers. Structure-activity relationship optimization yielded potent and minor allele-specific compounds that achieved high levels of mRNA and protein knockdown of human PNPLA3I148M but not PNPLA3I148I. Testing of the minor allele-specific siRNA in PNPLA3I148M-expressing mice fed a NASH-inducing diet prevented PNPLA3I148M-driven disease phenotypes, thus demonstrating the potential of a precision medicine approach to treating NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Alleles , Animals , Genome-Wide Association Study , Lipase/genetics , Liver , Membrane Proteins/genetics , Mice , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/therapy , Phospholipases A2, Calcium-Independent , RNA, Small Interfering/genetics
6.
J Biol Chem ; 292(5): 1951-1969, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28011645

ABSTRACT

The discovery of brown adipose tissue (BAT) as a key regulator of energy expenditure has sparked interest in identifying novel soluble factors capable of activating inducible BAT (iBAT) to combat obesity. Using a high content cell-based screen, we identified fibroblast growth factor 16 (FGF16) as a potent inducer of several physical and transcriptional characteristics analogous to those of both "classical" BAT and iBAT. Overexpression of Fgf16 in vivo recapitulated several of our in vitro findings, specifically the significant induction of the Ucp1 gene and UCP1 protein expression in inguinal white adipose tissue (iWAT), a common site for emergent active iBAT. Despite significant UCP1 up-regulation in iWAT and dramatic weight loss, the metabolic improvements observed due to Fgf16 overexpression in vivo were not the result of increased energy expenditure, as measured by indirect calorimetric assessment. Instead, a pattern of reduced food and water intake, combined with feces replete with lipid and bile acid, indicated a phenotype more akin to that of starvation and intestinal malabsorption. Gene expression analysis of the liver and ileum indicated alterations in several steps of bile acid metabolism, including hepatic synthesis and reabsorption. Histological analysis of intestinal tissue revealed profound abnormalities in support of this conclusion. The in vivo data, together with FGF receptor binding analysis, indicate that the in vivo outcome observed is the likely result of both direct and indirect mechanisms and probably involves multiple receptors. These results highlight the complexity of FGF signaling in the regulation of various metabolic processes.


Subject(s)
Adipose Tissue, White/metabolism , Fibroblast Growth Factors/metabolism , Signal Transduction , Thermogenesis , Ubiquitin-Specific Proteases/biosynthesis , Adipose Tissue, White/pathology , Animals , Cell Line , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Fibroblast Growth Factors/genetics , Humans , Mice , Obesity/chemically induced , Obesity/genetics , Obesity/metabolism , Ubiquitin-Specific Proteases/genetics
7.
PLoS One ; 11(2): e0147254, 2016.
Article in English | MEDLINE | ID: mdl-26840340

ABSTRACT

Pancreatic amyloid formation by islet amyloid polypeptide (IAPP) is a hallmark pathological feature of type 2 diabetes. IAPP is stored in the secretory granules of pancreatic beta-cells and co-secreted with insulin to maintain glucose homeostasis. IAPP is innocuous under homeostatic conditions but imbalances in production or processing of IAPP may result in homodimer formation leading to the rapid production of cytotoxic oligomers and amyloid fibrils. The consequence is beta-cell dysfunction and the accumulation of proteinaceous plaques in and around pancreatic islets. Beta-site APP-cleaving enzyme 2, BACE2, is an aspartyl protease commonly associated with BACE1, a related homolog responsible for amyloid processing in the brain and strongly implicated in Alzheimer's disease. Herein, we identify two distinct sites of the mature human IAPP sequence that are susceptible to BACE2-mediated proteolytic activity. The result of proteolysis is modulation of human IAPP fibrillation and human IAPP protein degradation. These results suggest a potential therapeutic role for BACE2 in type 2 diabetes-associated hyperamylinaemia.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Islet Amyloid Polypeptide/metabolism , Amino Acid Sequence , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cell Line , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/genetics , Mass Spectrometry , Mice , Molecular Sequence Data , Mutation , Plaque, Amyloid/metabolism , Proteolysis/drug effects , Recombinant Proteins , Substrate Specificity
8.
EBioMedicine ; 2(3): 214-24, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26151067

ABSTRACT

The polarization of tissue resident macrophages toward the alternatively activated, anti-inflammatory M2 phenotype is believed to positively impact obesity and insulin resistance. Here we show that the soluble form of the extracellular domain (ECD) of C-type lectin-like receptor 2, CLEC2, regulates Kupffer cell polarization in the liver and improves glucose and lipid parameters in diabetic animal models. Over-expression of Fc-CLEC2(ECD) in mice via in vivo gene delivery, or injection of recombinant Fc-CLEC2(ECD) protein, results in a reduction of blood glucose and liver triglyceride levels and improves glucose tolerance. Furthermore, Fc-CLEC2(ECD) treatment improves cytokine profiles and increases both the M2 macrophage population and the genes involved in the oxidation of lipid metabolism in the liver. These data reveal a previously unidentified role for CLEC2 as a regulator of macrophage polarity, and establish CLEC2 as a promising therapeutic target for treatment of diabetes and liver disease.


Subject(s)
Glucose/metabolism , Kupffer Cells/metabolism , Lectins, C-Type/metabolism , Lipid Metabolism/physiology , Animals , Cell Polarity , Fatty Liver/genetics , Fatty Liver/metabolism , Homeostasis/drug effects , Humans , Kupffer Cells/cytology , Kupffer Cells/drug effects , Lectins, C-Type/genetics , Lipid Metabolism/drug effects , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred Strains , Protein Structure, Tertiary , Receptors, Fc/genetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Solubility
9.
Am J Physiol Endocrinol Metab ; 307(12): E1144-52, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25370851

ABSTRACT

Elucidating the role of secreted frizzled-related protein 5 (SFRP5) in metabolism and obesity has been complicated by contradictory findings when knockout mice were used to determine metabolic phenotypes. By overexpressing SFRP5 in obese, prediabetic mice we consistently observed elevated hyperglycemia and glucose intolerance, supporting SFRP5 as a negative regulator of glucose metabolism. Accordingly, Sfrp5 mRNA expression analysis of both epididymal and subcutaneous adipose depots of mice indicated a correlation with obesity. Thus, we generated a monoclonal antibody (mAb) against SFRP5 to ascertain the effect of SFRP5 inhibition in vivo. Congruent with SFRP5 overexpression worsening blood glucose levels and glucose intolerance, anti-SFRP5 mAb therapy improved these phenotypes in vivo. The results from both the overexpression and mAb inhibition studies suggest a role for SFRP5 in glucose metabolism and pancreatic ß-cell function and thus establish the use of an anti-SFRP5 mAb as a potential approach to treat type 2 diabetes.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Monoclonal/immunology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Immunoglobulin G/immunology , Insulin-Secreting Cells/drug effects , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/complications , Obesity/genetics , Obesity/metabolism
10.
Metabolism ; 62(1): 90-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22982177

ABSTRACT

OBJECTIVE: To develop a rapid, easy and clinically relevant in vivo model to evaluate novel insulin secretagogues on human islets, we investigated the effect of insulin secretagogues on functional human islets in a humanized mouse model. MATERIALS/METHODS: Human islets were transplanted under the kidney capsule of streptozotocin (STZ)-induced diabetic mice with immunodeficiency. Human islet graft function was monitored by measuring non-fasting blood glucose levels. After diabetes was reversed, human islet transplanted mice were characterized physiologically by oral glucose tolerance and pharmacologically with clinically proven insulin secretagogues, glucagon-like peptide-1 (GLP-1), exenatide, glyburide, nateglinide and sitagliptin. Additionally, G protein-coupled receptor 40 (GPR40) agonists were evaluated in this model. RESULTS: Long-term human islet graft survival could be achieved in immunodeficient mice. Oral glucose challenge in human islet transplanted mice resulted in an immediate incremental increase of plasma human C-peptide, while the plasma mouse C-peptide was undetectable. Treatments with GLP-1, exenatide, glyburide, nateglinide and sitagliptin effectively increased plasma human C-peptide levels and improved postprandial glucose concentrations. GPR40 agonists also stimulated human C-peptide secretion and significantly improved postprandial glucose in the human islet transplanted mice. CONCLUSIONS: Our studies indicate that a humanized mouse model with human islet grafts could mimic the in vivo characteristics of human islets and could be a powerful tool for the evaluation of novel insulin secretagogues or other therapeutic agents that directly and/or indirectly target human ß cells.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Incretins/pharmacology , Insulin/metabolism , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Animals , Blood Glucose/analysis , Cyclohexanes/pharmacology , Diabetes Mellitus, Experimental/metabolism , Exenatide , Glucagon-Like Peptide 1/pharmacology , Glucose Tolerance Test , Glyburide/pharmacology , Humans , Insulin Secretion , Male , Mice , Mice, Nude , Nateglinide , Peptides/pharmacology , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Pyrazines/pharmacology , Sitagliptin Phosphate , Specific Pathogen-Free Organisms , Triazoles/pharmacology , Venoms/pharmacology
11.
Am J Physiol Endocrinol Metab ; 299(4): E624-32, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20647556

ABSTRACT

Antagonism of the glucagon receptor (GCGR) is associated with increased circulating levels of glucagon-like peptide-1 (GLP-1). To investigate the contribution of GLP-1 to the antidiabetic actions of GCGR antagonism, we administered an anti-GCGR monoclonal antibody (mAb B) to wild-type mice and GLP-1 receptor knockout (GLP-1R KO) mice. Treatment of wild-type mice with mAb B lowered fasting blood glucose, improved glucose tolerance, and enhanced glucose-stimulated insulin secretion during an intraperitoneal glucose tolerance test (ipGTT). In contrast, treatment of GLP-1R KO mice with mAb B had little efficacy during an ipGTT. Furthermore, pretreatment with the GLP-1R antagonist exendin-(9-39) diminished the antihyperglycemic effects of mAb B in wild-type mice. To determine the mechanism whereby mAb B improves glucose tolerance, we generated a monoclonal antibody that specifically antagonizes the human GLP-1R. Using a human islet transplanted mouse model, we demonstrated that pancreatic islet GLP-1R signaling is required for the full efficacy of the GCGR antagonist. To identify the source of the elevated GLP-1 observed in GCGR mAb-treated mice, we measured active GLP-1 content in pancreas and intestine from db/db mice treated with anti-GCGR mAb for 8 wk. Elevated GLP-1 in GCGR mAb-treated mice was predominantly derived from increased pancreatic GLP-1 synthesis and processing. All together, these data show that pancreatic GLP-1 is a significant contributor to the glucose-lowering effects observed in response to GCGR antagonist treatment.


Subject(s)
Glucagon-Like Peptide 1/physiology , Glucagon/physiology , Islets of Langerhans/physiology , Receptors, Glucagon/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Female , Glucagon/blood , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Islets of Langerhans/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Peptide Fragments/pharmacology , Receptors, Glucagon/blood , Receptors, Glucagon/physiology , Signal Transduction/drug effects
12.
Diabetes ; 57(4): 958-66, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18083786

ABSTRACT

Several studies have shown that the adult pancreas possesses a limited potential for beta-cell regeneration upon tissue injury. One of the difficulties in studying beta-cell regeneration has been the lack of a robust, synchronized animal model system that would allow controlled regulation of beta-cell loss and subsequent proliferation in adult pancreas. Here we present a transgenic mouse regeneration model in which the c-Myc transcription factor/mutant estrogen receptor (cMycER(TAM)) fusion protein can be specifically activated in mature beta-cells. We have studied these transgenic mice by immunohistochemical and biochemical methods to assess the ablation and posterior regeneration of beta-cells. Activation of the cMycER(TAM) fusion protein results in synchronous and selective beta-cell apoptosis followed by the onset of acute diabetes. Inactivation of c-Myc leads to gradual regeneration of insulin-expressing cells and reversal of diabetes. Our results demonstrate that the mature pancreas has the ability to fully recover from almost complete ablation of all existing beta-cells. Our results also suggest the regeneration of beta-cells is mediated by replication of beta-cells rather than neogenesis from pancreatic ducts.


Subject(s)
Insulin-Secreting Cells/physiology , Pancreas/physiology , Animals , Cell Division , Crosses, Genetic , Glucagon/analysis , Immunohistochemistry , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Proto-Oncogene Proteins c-myc/genetics , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/physiology , Regeneration
13.
Proc Natl Acad Sci U S A ; 104(15): 6247-52, 2007 Apr 10.
Article in English | MEDLINE | ID: mdl-17404238

ABSTRACT

There is widespread interest in defining factors and mechanisms that stimulate proliferation of pancreatic islet cells. Wnt signaling is an important regulator of organ growth and cell fates, and genes encoding Wnt-signaling factors are expressed in the pancreas. However, it is unclear whether Wnt signaling regulates pancreatic islet proliferation and differentiation. Here we provide evidence that Wnt signaling stimulates islet beta cell proliferation. The addition of purified Wnt3a protein to cultured beta cells or islets promoted expression of Pitx2, a direct target of Wnt signaling, and Cyclin D2, an essential regulator of beta cell cycle progression, and led to increased beta cell proliferation in vitro. Conditional pancreatic beta cell expression of activated beta-catenin, a crucial Wnt signal transduction protein, produced similar phenotypes in vivo, leading to beta cell expansion, increased insulin production and serum levels, and enhanced glucose handling. Conditional beta cell expression of Axin, a potent negative regulator of Wnt signaling, led to reduced Pitx2 and Cyclin D2 expression by beta cells, resulting in reduced neonatal beta cell expansion and mass and impaired glucose tolerance. Thus, Wnt signaling is both necessary and sufficient for islet beta cell proliferation, and our study provides previously unrecognized evidence of a mechanism governing endocrine pancreas growth and function.


Subject(s)
Cell Proliferation , Insulin-Secreting Cells/physiology , Signal Transduction/physiology , Wnt Proteins/metabolism , Animals , Cyclin D2 , Cyclins/metabolism , Homeodomain Proteins/metabolism , Immunohistochemistry , Mice , Mice, Mutant Strains , Transcription Factors/metabolism , Wnt3 Protein , Wnt3A Protein , Homeobox Protein PITX2
14.
Proc Natl Acad Sci U S A ; 99(25): 16105-10, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12441403

ABSTRACT

The use of embryonic stem cells for cell-replacement therapy in diseases like diabetes mellitus requires methods to control the development of multipotent cells. We report that treatment of mouse embryonic stem cells with inhibitors of phosphoinositide 3-kinase, an essential intracellular signaling regulator, produced cells that resembled pancreatic beta cells in several ways. These cells aggregated in structures similar, but not identical, to pancreatic islets of Langerhans, produced insulin at levels far greater than previously reported, and displayed glucose-dependent insulin release in vitro. Transplantation of these cell aggregates increased circulating insulin levels, reduced weight loss, improved glycemic control, and completely rescued survival in mice with diabetes mellitus. Graft removal resulted in rapid relapse and death. Graft analysis revealed that transplanted insulin-producing cells remained differentiated, enlarged, and did not form detectable tumors. These results provide evidence that embryonic stem cells can serve as the source of insulin-producing replacement tissue in an experimental model of diabetes mellitus. Strategies for producing cells that can replace islet functions described here can be adapted for similar uses with human cells.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Enzyme Inhibitors/pharmacology , Growth Inhibitors/pharmacology , Insulin/biosynthesis , Phosphoinositide-3 Kinase Inhibitors , Stem Cell Transplantation , Stem Cells/drug effects , Androstadienes/pharmacology , Animals , Biomarkers , Cell Aggregation , Cell Differentiation/drug effects , Cell Line/cytology , Cell Line/drug effects , Cell Line/metabolism , Cell Line/transplantation , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 1/blood , Embryo, Mammalian/cytology , Glucagon/biosynthesis , Homeostasis , Insulin/blood , Insulin/metabolism , Insulin Secretion , Islets of Langerhans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/etiology , Niacinamide/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Stem Cell Transplantation/adverse effects , Stem Cells/cytology , Stem Cells/metabolism , Streptozocin , Weight Loss , Wortmannin
15.
Am J Transplant ; 2(6): 510-9, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12118894

ABSTRACT

Both the direct and indirect antigen presentation pathways are important mechanisms for T cell-mediated allograft rejection. Studies using knockout mice and monoclonal antibodies have demonstrated that CD4+ T cells are both necessary and sufficient for the rejection of allogeneic tissues, including skin, heart, and islet. Furthermore, combined blockade of the CD28/B7 and CD154/CD40 costimulatory pathways induces tolerance in multiple CD4+ T-cell dependent allograft models. In this study, we addressed the T-cell requirement for costimulation in direct antigen presentation. We demonstrated that class II-specific alloreactive T-cell receptor transgenic T cells were sufficient to mediate allograft rejection independent of costimulatory blockade. Analysis of the costimulatory capacity of different antigen presenting cell (APC) populations demonstrated that APCs resident within the donor skin, Langerhans cells, are potent stimulators not requiring CD28- or CD154-dependent costimulation for direct major histocompatibility complex (MHC) antigen presentation. These results complement previous work examining the role of costimulation on CD8+ T cells, supporting a model in which the effectiveness of costimulatory blockade in the setting of transplantation may be selective for the indirect pathway of MHC alloantigen presentation.


Subject(s)
Antigen Presentation , Graft Rejection/immunology , Immune Tolerance , Animals , CD28 Antigens/immunology , CD40 Antigens/immunology , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...