Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Proc Natl Acad Sci U S A ; 113(12): 3293-8, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26951663

ABSTRACT

This study provides an assessment of the Fluidigm C1 platform for RNA sequencing of single mouse pancreatic islet cells. The system combines microfluidic technology and nanoliter-scale reactions. We sequenced 622 cells, allowing identification of 341 islet cells with high-quality gene expression profiles. The cells clustered into populations of α-cells (5%), ß-cells (92%), δ-cells (1%), and pancreatic polypeptide cells (2%). We identified cell-type-specific transcription factors and pathways primarily involved in nutrient sensing and oxidation and cell signaling. Unexpectedly, 281 cells had to be removed from the analysis due to low viability, low sequencing quality, or contamination resulting in the detection of more than one islet hormone. Collectively, we provide a resource for identification of high-quality gene expression datasets to help expand insights into genes and pathways characterizing islet cell types. We reveal limitations in the C1 Fluidigm cell capture process resulting in contaminated cells with altered gene expression patterns. This calls for caution when interpreting single-cell transcriptomics data using the C1 Fluidigm system.


Subject(s)
Islets of Langerhans/metabolism , Sequence Analysis, RNA/methods , Animals , Islets of Langerhans/cytology , Mice , Mice, Inbred C57BL , Transcription Factors/metabolism
2.
Cells ; 12(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36980244

ABSTRACT

The rare SLC30A8 mutation encoding a truncating p.Arg138* variant (R138X) in zinc transporter 8 (ZnT8) is associated with a 65% reduced risk for type 2 diabetes. To determine whether ZnT8 is required for beta cell development and function, we derived human pluripotent stem cells carrying the R138X mutation and differentiated them into insulin-producing cells. We found that human pluripotent stem cells with homozygous or heterozygous R138X mutation and the null (KO) mutation have normal efficiency of differentiation towards insulin-producing cells, but these cells show diffuse granules that lack crystalline zinc-containing insulin granules. Insulin secretion is not compromised in vitro by KO or R138X mutations in human embryonic stem cell-derived beta cells (sc-beta cells). Likewise, the ability of sc-beta cells to secrete insulin and maintain glucose homeostasis after transplantation into mice was comparable across different genotypes. Interestingly, sc-beta cells with the SLC30A8 KO mutation showed increased cytoplasmic zinc, and cells with either KO or R138X mutation were resistant to apoptosis when extracellular zinc was limiting. These findings are consistent with a protective role of zinc in cell death and with the protective role of zinc in T2D.


Subject(s)
Cation Transport Proteins , Diabetes Mellitus, Type 2 , Human Embryonic Stem Cells , Zinc Transporter 8 , Zinc , Animals , Humans , Mice , Apoptosis/genetics , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/physiology , Insulin/metabolism , Loss of Function Mutation , Mutation/genetics , Zinc/metabolism , Zinc Transporter 8/genetics , Zinc Transporter 8/metabolism
3.
Endocrinology ; 162(1)2021 01 01.
Article in English | MEDLINE | ID: mdl-33206168

ABSTRACT

The liver plays a critical role in maintaining ammonia homeostasis. Urea cycle defects, liver injury, or failure and glutamine synthetase (GS) deficiency result in hyperammonemia, serious clinical conditions, and lethality. In this study we used a mouse model with a defect in the urea cycle enzyme ornithine transcarbamylase (Otcspf-ash) to test the hypothesis that glucagon receptor inhibition using a monoclonal blocking antibody will reduce the hyperammonemia and associated lethality induced by a high-protein diet, which exacerbates disease. We found reduced expression of glutaminase, which degrades glutamine and increased expression of GS in livers of Otcspf-ash mice treated with the glucagon receptor blocking antibody. The gene expression changes favor ammonia consumption and were accompanied by increased circulating glutamine levels and diminished hyperammonemia. Otcspf-ash mice treated with the glucagon receptor-blocking antibody gained lean and body mass and had increased survival. These data suggest that glucagon receptor inhibition using a monoclonal antibody could reduce the risk for hyperammonemia and other clinical manifestations of patients suffering from defects in the urea cycle, liver injury, or failure and GS deficiency.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hyperammonemia/therapy , Ornithine Carbamoyltransferase Deficiency Disease/therapy , Receptors, Glucagon/antagonists & inhibitors , Amino Acids/blood , Ammonia/blood , Animals , Body Weight , Gene Expression Regulation/drug effects , Glutamate-Ammonia Ligase/genetics , Glutamate-Ammonia Ligase/metabolism , Glutaminase/genetics , Glutaminase/metabolism , Male , Mice , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/mortality
4.
Commun Biol ; 4(1): 1218, 2021 10 22.
Article in English | MEDLINE | ID: mdl-34686758

ABSTRACT

Bulk RNA sequencing provides the opportunity to understand biology at the whole transcriptome level without the prohibitive cost of single cell profiling. Advances in spatial transcriptomics enable to dissect tissue organization and function by genome-wide gene expressions. However, the readout of both technologies is the overall gene expression across potentially many cell types without directly providing the information of cell type constitution. Although several in-silico approaches have been proposed to deconvolute RNA-Seq data composed of multiple cell types, many suffer a deterioration of performance in complex tissues. Here we present AdRoit, an accurate and robust method to infer the cell composition from transcriptome data of mixed cell types. AdRoit uses gene expression profiles obtained from single cell RNA sequencing as a reference. It employs an adaptive learning approach to alleviate the sequencing technique difference between the single cell and the bulk (or spatial) transcriptome data, enhancing cross-platform readout comparability. Our systematic benchmarking and applications, which include deconvoluting complex mixtures that encompass 30 cell types, demonstrate its preferable sensitivity and specificity compared to many existing methods as well as its utilities. In addition, AdRoit is computationally efficient and runs orders of magnitude faster than most methods.


Subject(s)
Gene Expression Profiling/methods , Genome , Transcriptome , Sensitivity and Specificity
5.
JCI Insight ; 6(5)2021 03 08.
Article in English | MEDLINE | ID: mdl-33529174

ABSTRACT

Limitations in cell proliferation are important for normal function of differentiated tissues and essential for the safety of cell replacement products made from pluripotent stem cells, which have unlimited proliferative potential. To evaluate whether these limitations can be established pharmacologically, we exposed pancreatic progenitors differentiating from human pluripotent stem cells to small molecules that interfere with cell cycle progression either by inducing G1 arrest or by impairing S phase entry or S phase completion and determined growth potential, differentiation, and function of insulin-producing endocrine cells. We found that the combination of G1 arrest with a compromised ability to complete DNA replication promoted the differentiation of pancreatic progenitor cells toward insulin-producing cells and could substitute for endocrine differentiation factors. Reduced replication fork speed during differentiation improved the stability of insulin expression, and the resulting cells protected mice from diabetes without the formation of cystic growths. The proliferative potential of grafts was proportional to the reduction of replication fork speed during pancreatic differentiation. Therefore, a compromised ability to enter and complete S phase is a functionally important property of pancreatic endocrine differentiation, can be achieved by reducing replication fork speed, and is an important determinant of cell-intrinsic limitations of growth.


Subject(s)
Cell Cycle , Cell Differentiation , DNA Replication , Diabetes Mellitus , Induced Pluripotent Stem Cells , Insulin-Secreting Cells , Stem Cell Transplantation , Animals , Aphidicolin , Cell Proliferation , Diabetes Mellitus/therapy , Humans , Insulin/metabolism , Islets of Langerhans , Mice , Pancreas , Pluripotent Stem Cells , Transplants
6.
J Vis Exp ; (149)2019 07 18.
Article in English | MEDLINE | ID: mdl-31380847

ABSTRACT

Pancreatic islets comprise of endocrine cells with distinctive hormone expression patterns. The endocrine cells show functional differences in response to normal and pathological conditions. The goal of this protocol is to generate high-quality, large-scale transcriptome data of each endocrine cell type with the use of a droplet-based microfluidic single-cell RNA sequencing technology. Such data can be utilized to build the gene expression profile of each endocrine cell type in normal or specific conditions. The process requires careful handling, accurate measurement, and rigorous quality control. In this protocol, we describe detailed steps for human pancreatic islets dissociation, sequencing, and data analysis. The representative results of about 20,000 human single islet cells demonstrate the successful application of the protocol.


Subject(s)
Islets of Langerhans/cytology , Sequence Analysis, RNA , Single-Cell Analysis , Animals , Base Sequence , Humans , Exome Sequencing
7.
Endocrinology ; 160(5): 979-988, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30938753

ABSTRACT

Plasma amino acids and their transporters constitute an important part of the feedback loop between the liver and pancreatic α-cell function, and glucagon regulates hepatic amino acid turnover. Disruption of hepatic glucagon receptor action activates the loop and results in high plasma amino acids and hypersecretion of glucagon associated with α-cell hyperplasia. In the present study, we report a technique to rescue implanted human pancreatic islets from the mouse kidney capsule. Using this model, we have demonstrated that expression of the amino acid transporter SLC38A4 increases in α-cells after administration of a glucagon receptor blocking antibody. The increase in SLC38A4 expression and associated α-cell proliferation was dependent on mechanistic target of rapamycin pathway. We confirmed increased α-cell proliferation and expression of SLC38A4 in pancreas sections from patients with glucagon cell hyperplasia and neoplasia (GCHN) with loss-of-function mutations in the glucagon receptor. Collectively, using a technique to rescue implanted human islets from the kidney capsule in mice and pancreas sections from patients with GCHN, we found that expression of SLC38A4 was increased under conditions of disrupted glucagon receptor signaling. These data provide support for the existence of a liver-human α-cell endocrine feedback loop.


Subject(s)
Amino Acid Transport System A/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Islets of Langerhans Transplantation/methods , Receptors, Glucagon/metabolism , Adult , Amino Acid Transport System A/genetics , Animals , Cell Proliferation/genetics , Female , Glucagon-Secreting Cells/cytology , Humans , Hyperplasia/blood , Hyperplasia/metabolism , Male , Mice , Middle Aged , Receptors, Glucagon/genetics , Signal Transduction , Transplantation, Heterologous
8.
Diabetes ; 67(9): 1783-1794, 2018 09.
Article in English | MEDLINE | ID: mdl-29950394

ABSTRACT

Proinsulin is a misfolding-prone protein, making its biosynthesis in the endoplasmic reticulum (ER) a stressful event. Pancreatic ß-cells overcome ER stress by activating the unfolded protein response (UPR) and reducing insulin production. This suggests that ß-cells transition between periods of high insulin biosynthesis and UPR-mediated recovery from cellular stress. We now report the pseudotime ordering of single ß-cells from humans without diabetes detected by large-scale RNA sequencing. We identified major states with 1) low UPR and low insulin gene expression, 2) low UPR and high insulin gene expression, or 3) high UPR and low insulin gene expression. The latter state was enriched for proliferating cells. Stressed human ß-cells do not dedifferentiate and show little propensity for apoptosis. These data suggest that human ß-cells transition between states with high rates of biosynthesis to fulfill the body's insulin requirements to maintain normal blood glucose levels and UPR-mediated recovery from ER stress due to high insulin production.


Subject(s)
Endoplasmic Reticulum Stress , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Proinsulin/metabolism , Transcription Factors/metabolism , Unfolded Protein Response , Biomarkers/metabolism , Cell Proliferation , Cells, Cultured , Databases, Genetic , Gene Expression Profiling , Humans , In Situ Hybridization, Fluorescence , Insulin/chemistry , Insulin/genetics , Insulin Secretion , Insulin-Secreting Cells/cytology , Kinetics , Multigene Family , Nucleotide Mapping , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , Proinsulin/chemistry , Proinsulin/genetics , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Transcription Factors/genetics
9.
Endocrinology ; 159(9): 3177-3186, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30010845

ABSTRACT

Pancreatic α cells proliferate at a low rate, and little is known about the control of this process. Here we report the characterization of human α cells by large-scale, single-cell RNA sequencing coupled with pseudotime ordering. We identified two large subpopulations and a smaller cluster of proliferating α cells with increased expression of genes involved in cell-cycle regulation. The proliferating α cells were differentiated, had normal levels of GCG expression, and showed no signs of cellular stress. Proliferating α cells were detected in both the G1S and G2M phases of the cell cycle. Human α cells proliferate at a fivefold higher rate than human ß cells and express lower levels of the cell-cycle inhibitors CDKN1A and CDKN1C. Collectively, this study provides the gene signatures of human α cells and the genes involved in their cell division. The lower expression of two cell-cycle inhibitors in human α cells could account for their higher rate of proliferation compared with their insulin-producing counterparts.


Subject(s)
Cell Proliferation/genetics , Glucagon-Secreting Cells/metabolism , RNA, Messenger/metabolism , Transcriptome , Adult , Cell Cycle , Female , Humans , Male , Middle Aged , Sequence Analysis, RNA , Single-Cell Analysis , Young Adult
10.
Endocrinology ; 159(12): 4023-4032, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30380031

ABSTRACT

The ghrelin-producing ε cell represents the fifth endocrine cell type in human pancreatic islets. The abundance of ε cells in adult pancreas is extremely low, which has hampered the investigation on the molecular pathways regulating the development and the function of this cell type. In this study, we explored the molecular features defining the function of pancreatic ε cells isolated from adult nondiabetic donors using single-cell RNA sequencing technology. We focus on transcription factors, cell surface receptors, and genes involved in metabolic pathways that contribute to regulation of cellular function. Furthermore, the genes that separate ε cells from the other islet endocrine cell types are presented. This study expands prior knowledge about the genes important for ε cell functioning during development and provides a resource to interrogate the transcriptome of this rare human islet cell type.


Subject(s)
Ghrelin/metabolism , Pancreas/cytology , Pancreas/metabolism , Transcriptome , Adult , Cell Count , Cell Separation , Cells, Cultured , Gene Expression Profiling , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Microarray Analysis , Signal Transduction/genetics
11.
Biochim Biophys Acta ; 1757(8): 942-68, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16905113

ABSTRACT

A protein structure should provide the information needed to understand its observed properties. Significant progress has been made in developing accurate calculations of acid/base and oxidation/reduction reactions in proteins. Current methods and their strengths and weaknesses are discussed. The distribution and calculated ionization states in a survey of proteins is described, showing that a significant minority of acidic and basic residues are buried in the protein and that most of these remain ionized. The electrochemistry of heme and quinones are considered. Proton transfers in bacteriorhodopsin and coupled electron and proton transfers in photosynthetic reaction centers, 5-coordinate heme binding proteins and cytochrome c oxidase are highlighted as systems where calculations have provided insight into the reaction mechanism.


Subject(s)
Bacteriorhodopsins/chemistry , Electrons , Proteins/chemistry , Protons , Water/chemistry , Models, Theoretical , Solutions , Thermodynamics
12.
Stem Cell Reports ; 8(2): 264-277, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28132887

ABSTRACT

We recently developed a technique for generating hypothalamic neurons from human pluripotent stem cells. Here, as proof of principle, we examine the use of these cells in modeling of a monogenic form of severe obesity: PCSK1 deficiency. The cognate enzyme, PC1/3, processes many prohormones in neuroendocrine and other tissues. We generated PCSK1 (PC1/3)-deficient human embryonic stem cell (hESC) lines using both short hairpin RNA and CRISPR-Cas9, and investigated pro-opiomelanocortin (POMC) processing using hESC-differentiated hypothalamic neurons. The increased levels of unprocessed POMC and the decreased ratios (relative to POMC) of processed POMC-derived peptides in both PCSK1 knockdown and knockout hESC-derived neurons phenocopied POMC processing reported in PC1/3-null mice and PC1/3-deficient patients. PC1/3 deficiency was associated with increased expression of melanocortin receptors and PRCP (prolylcarboxypeptidase, a catabolic enzyme for α-melanocyte stimulating hormone (αMSH)), and reduced adrenocorticotropic hormone secretion. We conclude that the obesity accompanying PCSK1 deficiency may not be primarily due to αMSH deficiency.


Subject(s)
Human Embryonic Stem Cells/cytology , Neurons/cytology , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Proprotein Convertase 1/deficiency , Adrenocorticotropic Hormone/metabolism , Animals , Apoptosis , CRISPR-Cas Systems , Cell Differentiation/genetics , Cells, Cultured , Endoplasmic Reticulum Stress , Gene Expression , Gene Knockdown Techniques , Gene Targeting , Humans , Immunohistochemistry , Mice , Mutation , Proprotein Convertase 1/genetics , Proteolysis , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , alpha-MSH/metabolism
13.
Cell Metab ; 25(6): 1348-1361.e8, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28591637

ABSTRACT

Glucagon supports glucose homeostasis by stimulating hepatic gluconeogenesis, in part by promoting the uptake and conversion of amino acids into gluconeogenic precursors. Genetic disruption or pharmacologic inhibition of glucagon signaling results in elevated plasma amino acids and compensatory glucagon hypersecretion involving expansion of pancreatic α cell mass. Recent findings indicate that hyperaminoacidemia triggers pancreatic α cell proliferation via an mTOR-dependent pathway. We confirm and extend these findings by demonstrating that glucagon pathway blockade selectively increases expression of the sodium-coupled neutral amino acid transporter Slc38a5 in a subset of highly proliferative α cells and that Slc38a5 controls the pancreatic response to glucagon pathway blockade; most notably, mice deficient in Slc38a5 exhibit markedly decreased α cell hyperplasia to glucagon pathway blockade-induced hyperaminoacidemia. These results show that Slc38a5 is a key component of the feedback circuit between glucagon receptor signaling in the liver and amino-acid-dependent regulation of pancreatic α cell mass in mice.


Subject(s)
Amino Acid Transport Systems, Neutral/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Receptors, Glucagon/metabolism , Signal Transduction , Amino Acid Transport Systems, Neutral/genetics , Animals , Glucagon/genetics , Glucagon-Secreting Cells/pathology , Hyperplasia , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Receptors, Glucagon/genetics
14.
J Mol Biol ; 348(5): 1283-98, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15854661

ABSTRACT

Ionizable residues play essential roles in proteins, modulating protein stability, fold and function. Asp, Glu, Arg, and Lys make up about a quarter of the residues in an average protein. Multi-conformation continuum electrostatic (MCCE) calculations were used to predict the ionization states of all acidic and basic residues in 490 proteins. Of all 36,192 ionizable residues, 93.5% were predicted to be ionized. Thirty-five percent have lost 4.08 kcal/mol solvation energy (DeltaDeltaG(rxn)) sufficient to shift a pK(a) by three pH units in the absence of other interactions and 17% have DeltaDeltaG(rxn) sufficient to shift pK(a) by five pH units. Overall 85% of these buried residues (DeltaDeltaG(rxn)>5DeltapK units) are ionized, including 92% of the Arg, 86% of the Asp, 77% of the Glu, and 75% of the Lys. Ion-pair interactions stabilize the ionization of both acids and bases. The backbone dipoles stabilize anions more than cations. The interactions with polar side-chains are also different for acids and bases. Asn and Gln stabilize all charges, Ser and Thr stabilize only acids while Tyr rarely stabilize Lys. Thus, hydroxyls are better hydrogen bond donors than acceptors. Buried ionized residues are more likely to be conserved than those on the surface. There are 3.95 residues buried per 100 residues in an average protein.


Subject(s)
Amino Acids, Acidic/chemistry , Amino Acids, Basic/chemistry , Proteins/chemistry , Arginine/chemistry , Aspartic Acid/chemistry , Crystallography , Glutamic Acid/chemistry , Ions/chemistry , Lysine/chemistry , Static Electricity
15.
Cell Metab ; 24(4): 608-615, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27667665

ABSTRACT

Pancreatic islet cells are critical for maintaining normal blood glucose levels, and their malfunction underlies diabetes development and progression. We used single-cell RNA sequencing to determine the transcriptomes of 1,492 human pancreatic α, ß, δ, and PP cells from non-diabetic and type 2 diabetes organ donors. We identified cell-type-specific genes and pathways as well as 245 genes with disturbed expression in type 2 diabetes. Importantly, 92% of the genes have not previously been associated with islet cell function or growth. Comparison of gene profiles in mouse and human α and ß cells revealed species-specific expression. All data are available for online browsing and download and will hopefully serve as a resource for the islet research community.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Gene Expression Profiling , Humans , Mice , Signal Transduction/genetics , Transcriptome/genetics
16.
Endocrinology ; 157(9): 3431-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27466694

ABSTRACT

Aging improves pancreatic ß-cell function in mice. This is a surprising finding because aging is typically associated with functional decline. We performed single-cell RNA sequencing of ß-cells from 3- and 26-month-old mice to explore how changes in gene expression contribute to improved function with age. The old mice were healthy and had reduced blood glucose levels and increased ß-cell mass, which correlated to their body weight. ß-Cells from young and old mice had similar transcriptome profiles. In fact, only 193 genes (0.89% of all detected genes) were significantly regulated (≥2-fold; false discovery rate < 0.01; normalized counts > 5). Of these, 183 were down-regulated and mainly associated with pathways regulating gene expression, cell cycle, cell death, and survival as well as cellular movement, function, and maintenance. Collectively our data show that ß-cells from very old mice have transcriptome profiles similar to those of young mice. These data support previous findings that aging is not associated with reduced ß-cell mass or functional ß-cell decline in mice.


Subject(s)
Aging/metabolism , Insulin-Secreting Cells/metabolism , Animals , Cellular Senescence , Glucose/metabolism , Homeostasis , Male , Mice, Inbred C57BL , Sequence Analysis, RNA , Single-Cell Analysis , Transcription Factors/metabolism , Transcriptome
17.
Endocrinology ; 156(8): 2781-94, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26020795

ABSTRACT

Antagonizing glucagon action represents an attractive therapeutic option for reducing hepatic glucose production in settings of hyperglycemia where glucagon excess plays a key pathophysiological role. We therefore generated REGN1193, a fully human monoclonal antibody that binds and inhibits glucagon receptor (GCGR) signaling in vitro. REGN1193 administration to diabetic ob/ob and diet-induced obese mice lowered blood glucose to levels observed in GCGR-deficient mice. In diet-induced obese mice, REGN1193 reduced food intake, adipose tissue mass, and body weight. REGN1193 increased circulating levels of glucagon and glucagon-like peptide 1 and was associated with reversible expansion of pancreatic α-cell area. Hyperglucagonemia and α-cell hyperplasia was observed in fibroblast growth factor 21-deficient mice treated with REGN1193. Single administration of REGN1193 to diabetic cynomolgus monkeys normalized fasting blood glucose and glucose tolerance and increased circulating levels of glucagon and amino acids. Finally, administration of REGN1193 for 8 weeks to normoglycemic cynomolgus monkeys did not cause hypoglycemia or increase pancreatic α-cell area. In summary, the GCGR-blocking antibody REGN1193 normalizes blood glucose in diabetic mice and monkeys but does not produce hypoglycemia in normoglycemic monkeys. Thus, REGN1193 provides a potential therapeutic modality for diabetes mellitus and acute hyperglycemic conditions.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Blood Glucose/drug effects , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/therapeutic use , Receptors, Glucagon/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/complications , Female , Fibroblast Growth Factors/genetics , Humans , Hypoglycemic Agents/pharmacology , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Obesity/complications , Obesity/drug therapy , Obesity/pathology , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/genetics
SELECTION OF CITATIONS
SEARCH DETAIL