Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Circulation ; 123(20): 2219-25, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555707

ABSTRACT

BACKGROUND: In-hospital pulmonary embolism (PE) has been extensively studied in large populations; however, out-of-hospital fatal PE studies are rare. Here, we systematically evaluated a large number of decedents who suffered fatal PE outside of hospitals and were subsequently investigated by the New York City Office of Chief Medical Examiner. METHODS AND RESULTS: A total of 578 consecutive out-of-hospital fatal PE cases were analyzed. All underwent autopsy, toxicology, microbiology, and genetic testing. Incidence rates and baseline characteristics were analyzed. Race-adjusted incidence rates of out-of-hospital fatal PE (per 100 000 people per year) were as follows: blacks, 3.73 (95% confidence interval, 3.31 to 4.11); whites, 1.15 (95% confidence interval, 0.96 to 1.33); and Hispanics, 0.93 (95% confidence interval, 0.72 to 1.10). Overall, obesity (body mass index ≥30 kg/m(2)) was 2.5- to 3-fold higher in fatal PE cases than in the New York City population as a whole. Carrier frequencies for prothrombin G20210A in fatal PE were 2- to 10-fold higher than reported frequencies in ethnically matched controls. Cumulative distribution curves showed that compared with whites, blacks and Hispanics died at significantly younger ages (P<0.001). Univariate and multiple linear regression analyses showed that in addition to nonwhite ethnicity, heterozygous carriers for factor V Leiden (P=0.001) and obesity (P=0.002) are significantly associated with younger age at death. CONCLUSION: There are unique epidemiological differences in out-of-hospital fatal PE between ethnic groups in New York City.


Subject(s)
Emergency Medical Services/statistics & numerical data , Ethnicity/statistics & numerical data , Pulmonary Embolism/ethnology , Pulmonary Embolism/mortality , Adult , Black or African American/statistics & numerical data , Age Distribution , Aged , Aged, 80 and over , Female , Hispanic or Latino/statistics & numerical data , Humans , Incidence , Male , Middle Aged , New York City/epidemiology , Sex Distribution , Venous Thromboembolism/ethnology , Venous Thromboembolism/mortality , White People/statistics & numerical data , Young Adult
2.
J Mol Endocrinol ; 46(3): 175-92, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21321096

ABSTRACT

Adipose-specific inactivation of both AP-1 and CCAAT-enhancer-binding protein (C/EBP) families of B-ZIP transcription factors in transgenic mice causes severe lipoatrophy. To evaluate whether inactivation of only C/EBP members was critical for lipoatrophy, A-C/EBP, a dominant-negative protein that specifically inhibits the DNA binding of the C/EBP members, was expressed in adipose tissue. For the first 2 weeks after birth, aP2-A-C/EBP mice had no white adipose tissue (WAT), drastically reduced brown adipose tissue (BAT), and exhibited marked hepatic steatosis, hyperinsulinemia, and hyperlipidemia. However, WAT appeared during the third week, coinciding with significantly improved metabolic functioning. In adults, BAT remained reduced, causing cold intolerance. At 30 weeks, the aP2-A-C/EBP mice had only 35% reduced WAT, with clear morphological signs of lipodystrophy in subcutaneous fat. Circulating leptin and adiponectin levels were less than the wild-type levels, and these mice exhibited impaired triglyceride clearance. Insulin resistance, glucose intolerance, and reduced free fatty acid release in response to ß3-adrenergic agonist suggest improper functioning of the residual WAT. Gene expression analysis of inguinal WAT identified reduced mRNA levels of several enzymes involved in fatty acid synthesis and glucose metabolism that are known C/EBPα transcriptional targets. There were increased levels for genes involved in inflammation and muscle differentiation. However, when dermal fibroblasts from aP2-A-C/EBP mice were differentiated into adipocytes in tissue culture, muscle markers were elevated more than the inflammatory markers. These results demonstrate that the C/EBP family is essential for adipose tissue development during the early postnatal period, the regulation of glucose and lipid homeostasis in adults, and the suppression of the muscle lineage.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Lipodystrophy/etiology , Lipodystrophy/metabolism , Proteins/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Blotting, Western , Body Composition/genetics , Body Composition/physiology , CCAAT-Enhancer-Binding Proteins/genetics , Calorimetry, Indirect , Cells, Cultured , Eating/genetics , Eating/physiology , Fatty Acid-Binding Proteins/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Lipodystrophy/genetics , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Proteins/genetics , Triglycerides/metabolism
3.
Virol J ; 6: 61, 2009 May 18.
Article in English | MEDLINE | ID: mdl-19450275

ABSTRACT

BACKGROUND: Pancreatic islet transplantation is a promising treatment for type I diabetes mellitus, but current immunosuppressive strategies do not consistently provide long-term survival of transplanted islets. We are therefore investigating the use of adeno-associated viruses (AAVs) as gene therapy vectors to transduce rat islets with immunosuppressive genes prior to transplantation into diabetic mice. RESULTS: We compared the transduction efficiency of AAV2 vectors with an AAV2 capsid (AAV2/2) to AAV2 vectors pseudotyped with AAV5 (AAV2/5), AAV8 (AAV2/8) or bovine adeno-associated virus (BAAV) capsids, or an AAV2 capsid with an insertion of the low density lipoprotein receptor ligand from apolipoprotein E (AAV2apoE), on cultured islets, in the presence of helper adenovirus infection to speed expression of a GFP transgene. Confocal microscopy and flow cytometry were used. The AAV2/5 vector was superior to AAV2/2 and AAV2/8 in rat islets. Flow cytometry indicated AAV2/5-mediated gene expression in approximately 9% of rat islet cells and almost 12% of insulin-positive cells. The AAV2/8 vector had a higher dependence on the helper virus multiplicity of infection than the AAV 2/5 vector. In addition, the BAAV and AAV2apoE vectors were superior to AAV2/2 for transducing rat islets. Rat islets (300 per mouse) transduced with an AAV2/5 vector harboring the immunosuppressive transgene, tgf beta 1, retain the ability to correct hyperglycemia when transplanted into immune-deficient diabetic mice. CONCLUSION: AAV2/5 vectors may therefore be useful for pre-treating donor islets prior to transplantation.


Subject(s)
Dependovirus/genetics , Diabetes Mellitus, Type 1/therapy , Genetic Therapy/methods , Genetic Vectors/genetics , Islets of Langerhans/virology , Transduction, Genetic/methods , Animals , Cell Line , Dependovirus/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/surgery , Female , Genetic Vectors/metabolism , Humans , In Vitro Techniques , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation , Mice , Mice, Inbred NOD , Mice, SCID , Rats , Rats, Wistar
4.
PLoS Comput Biol ; 5(3): e1000324, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19325873

ABSTRACT

Adipose tissue grows by two mechanisms: hyperplasia (cell number increase) and hypertrophy (cell size increase). Genetics and diet affect the relative contributions of these two mechanisms to the growth of adipose tissue in obesity. In this study, the size distributions of epididymal adipose cells from two mouse strains, obesity-resistant FVB/N and obesity-prone C57BL/6, were measured after 2, 4, and 12 weeks under regular and high-fat feeding conditions. The total cell number in the epididymal fat pad was estimated from the fat pad mass and the normalized cell-size distribution. The cell number and volume-weighted mean cell size increase as a function of fat pad mass. To address adipose tissue growth precisely, we developed a mathematical model describing the evolution of the adipose cell-size distributions as a function of the increasing fat pad mass, instead of the increasing chronological time. Our model describes the recruitment of new adipose cells and their subsequent development in different strains, and with different diet regimens, with common mechanisms, but with diet- and genetics-dependent model parameters. Compared to the FVB/N strain, the C57BL/6 strain has greater recruitment of small adipose cells. Hyperplasia is enhanced by high-fat diet in a strain-dependent way, suggesting a synergistic interaction between genetics and diet. Moreover, high-fat feeding increases the rate of adipose cell size growth, independent of strain, reflecting the increase in calories requiring storage. Additionally, high-fat diet leads to a dramatic spreading of the size distribution of adipose cells in both strains; this implies an increase in size fluctuations of adipose cells through lipid turnover.


Subject(s)
Adipocytes/pathology , Adipose Tissue/growth & development , Adipose Tissue/pathology , Dietary Fats/metabolism , Models, Biological , Obesity/pathology , Obesity/physiopathology , Animals , Cell Enlargement , Cell Proliferation , Cell Size , Computer Simulation , Hyperplasia/pathology , Hyperplasia/physiopathology , Hypertrophy/pathology , Hypertrophy/physiopathology , Mice , Mice, Inbred C57BL
5.
Cell Metab ; 4(5): 363-75, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17084710

ABSTRACT

Most animal models of obesity and hyperinsulinemia are associated with increased vagal cholinergic activity. The M3 muscarinic acetylcholine receptor subtype is widely expressed in the brain and peripheral tissues and plays a key role in mediating the physiological effects of vagal activation. Here, we tested the hypothesis that the absence of M3 receptors in mice might protect against various forms of experimentally or genetically induced obesity and obesity-associated metabolic deficits. In all cases, the lack of M3 receptors greatly ameliorated impairments in glucose homeostasis and insulin sensitivity but had less robust effects on overall adiposity. Under all experimental conditions tested, M3 receptor-deficient mice showed a significant elevation in basal and total energy expenditure, most likely due to enhanced central sympathetic outflow and increased rate of fatty-acid oxidation. These findings suggest that the M3 receptor may represent a potential pharmacologic target for the treatment of obesity and associated metabolic disorders.


Subject(s)
Aortic Bodies/metabolism , Metabolic Diseases/etiology , Obesity , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/physiology , Animals , Epinephrine/urine , Ion Channels/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/metabolism , Norepinephrine/urine , Receptor, Muscarinic M3/deficiency , Uncoupling Protein 3
6.
Am J Physiol Endocrinol Metab ; 290(6): E1227-36, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16682489

ABSTRACT

The antiobesity and antidiabetic effects of the beta3-adrenergic agonists were investigated on nonobese type 2 diabetic MKR mice after injection with a beta3-adrenergic agonist, CL-316243. An intact response to acute CL-316243 treatment was observed in MKR mice. Chronic intraperitoneal CL-316243 treatment of MKR mice reduced blood glucose and serum insulin levels. Hyperinsulinemic euglycemic clamps exhibited improvement of the whole body insulin sensitivity and glucose homeostasis concurrently with enhanced insulin action in liver and adipose tissue. Treating MKR mice with CL-316243 significantly lowered serum and hepatic lipid levels, in part due to increased whole body triglyceride clearance and fatty acid oxidation in adipocytes. A significant reduction in total body fat content and epididymal fat weight was observed along with enhanced metabolic rate in both wild-type and MKR mice after treatment. These data demonstrate that beta3-adrenergic activation improves the diabetic state of nonobese diabetic MKR mice by potentiation of free fatty acid oxidation by adipose tissue, suggesting a potential therapeutic role for beta3-adrenergic agonists in nonobese diabetic subjects.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Dioxoles/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Adrenergic beta-3 Receptor Antagonists , Adrenergic beta-Agonists/pharmacology , Animals , Blood Glucose/metabolism , Body Composition , Glucose/administration & dosage , Glucose/pharmacokinetics , Insulin/metabolism , Lipid Metabolism/drug effects , Liver/metabolism , Mice , Mice, Inbred NOD , Oxidation-Reduction
7.
J Biol Chem ; 281(28): 18989-99, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16672216

ABSTRACT

The complex imprinted Gnas locus encodes several gene products including G(s)alpha, the ubiquitously expressed G protein alpha-subunit required for receptor-stimulated cAMP generation, and the neuroendocrine-specific G(s)alpha isoform XLalphas. XLalphas is only expressed from the paternal allele, whereas G(s)alpha is biallelically expressed in most tissues. XLalphas knock-out mice (Gnasxl(m+/p-)) have poor suckling and perinatal lethality, implicating XLalphas as critical for postnatal feeding. We have now examined the metabolic phenotype of adult Gnasxl(m+/p-) mice. Gnasxl(m+/p-) mice had reduced fat mass and lipid accumulation in adipose tissue, with increased food intake and metabolic rates. Gene expression profiling was consistent with increased lipid metabolism in adipose tissue. These changes likely result from increased sympathetic nervous system activity rather than adipose cell-autonomous effects, as we found that XLalphas is not normally expressed in adult adipose tissue, and Gnasxl(m+/p-) mice had increased urinary norepinephrine levels but not increased metabolic responsiveness to a beta3-adrenergic agonist. Gnasxl(m+/p-) mice were hypolipidemic and had increased glucose tolerance and insulin sensitivity. The similar metabolic profile observed in some prior paternal Gnas knock-out models results from XLalphas deficiency (or deficiency of the related alternative truncated protein XLN1). XLalphas (or XLN1) is a negative regulator of sympathetic nervous system activity in mice.


Subject(s)
Energy Metabolism , GTP-Binding Protein alpha Subunits, Gs/chemistry , Glucose/chemistry , Glucose/metabolism , Adipose Tissue/metabolism , Animals , Body Composition , Catecholamines/metabolism , Chromogranins , Cyclic AMP/metabolism , Gene Expression Regulation , Lipids/chemistry , Mice , Mice, Knockout , Models, Genetic , Sympathetic Nervous System/pathology , Triglycerides/metabolism
8.
J Clin Invest ; 115(11): 3217-27, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16239968

ABSTRACT

The G protein G(s)alpha is essential for hormone-stimulated cAMP generation and is an important metabolic regulator. We investigated the role of liver G(s)-signaling pathways by developing mice with liver-specific G(s)alpha deficiency (LGsKO mice). LGsKO mice had increased liver weight and glycogen content and reduced adiposity, whereas survival, body weight, food intake, and metabolic rates at ambient temperature were unaffected. LGsKO mice had increased glucose tolerance with both increased glucose-stimulated insulin secretion and increased insulin sensitivity in liver and muscle. Fed LGsKO mice were hypoglycemic and hypoinsulinemic, with low expression of hepatic gluconeogenic enzymes and PPARgamma coactivator-1. However, LGsKO mice maintained normal fasting glucose and insulin levels, probably due to prolonged breakdown of glycogen stores and possibly increased extrahepatic gluconeogenesis. Lipid metabolism was unaffected in fed LGsKO mice, but fasted LGsKO mice had increased lipogenic and reduced lipid oxidation gene expression in liver and increased serum triglyceride and FFA levels. LGsKO mice had very high serum glucagon and glucagon-like peptide-1 levels and pancreatic alpha cell hyperplasia, probably secondary to hepatic glucagon resistance and/or chronic hypoglycemia. Our results define novel roles for hepatic G(s)-signaling pathways in glucose and lipid regulation, which may prove useful in designing new therapeutic targets for diabetes and obesity.


Subject(s)
Adiposity/genetics , Fasting/physiology , GTP-Binding Protein alpha Subunits, Gs/deficiency , GTP-Binding Protein alpha Subunits, Gs/genetics , Glucose/metabolism , Liver/metabolism , Animals , Eating/genetics , Fasting/metabolism , GTP-Binding Protein alpha Subunits, Gs/physiology , Glucagon/blood , Glucagon-Like Peptide 1/blood , Glucose Tolerance Test , Hypoglycemia/genetics , Hypoglycemia/metabolism , Hypoglycemia/physiopathology , Insulin/metabolism , Insulin Resistance/genetics , Insulin Secretion , Islets of Langerhans/metabolism , Lipid Metabolism/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Oxygen Consumption/genetics , Signal Transduction/genetics
9.
Endocrinology ; 146(9): 4024-35, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15947005

ABSTRACT

Leptin has metabolic effects on peripheral tissues including muscle, liver, and pancreas, and it has been successfully used to treat lipodystrophic diabetes, a leptin-deficient state. To study whether leptin therapy can be used for treatment of more common cases of type 2 diabetes, we used a mouse model of type 2 diabetes (MKR mice) that show normal leptin levels and are diabetic due to a primary defect in both IGF-I and insulin receptors signaling in skeletal muscle. Here we show that leptin administration to the MKR mice resulted in improvement of diabetes, an effect that was independent of the reduced food intake. The main effect of leptin therapy was enhanced hepatic insulin responsiveness possibly through decreasing gluconeogenesis. In addition, the reduction of lipid stores in liver and muscle induced by enhancing fatty acid oxidation and inhibiting lipogenesis led to an improvement of the lipotoxic condition. Our data suggest that leptin could be a potent antidiabetic drug in cases of type 2 diabetes that are not leptin resistant.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hyperglycemia/drug therapy , Insulin Resistance , Leptin/pharmacology , AMP-Activated Protein Kinases , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Eating , Energy Metabolism/drug effects , Hyperglycemia/metabolism , Hyperinsulinism/drug therapy , Hyperinsulinism/metabolism , Insulin/metabolism , Insulin Secretion , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Multienzyme Complexes/metabolism , Muscle, Skeletal/enzymology , Protein Serine-Threonine Kinases/metabolism , Triglycerides/metabolism
10.
Endocrinology ; 145(10): 4667-76, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15231693

ABSTRACT

Insulin resistance is one of the primary characteristics of type 2 diabetes. Mice overexpressing a dominant-negative IGF-I receptor specifically in muscle (MKR mice) demonstrate severe insulin resistance with high levels of serum and tissue lipids and eventually develop type 2 diabetes at 5-6 wk of age. To determine whether lipotoxicity plays a role in the progression of the disease, we crossed MKR mice with mice overexpressing a fatty acid translocase, CD36, in skeletal muscle. The double-transgenic MKR/CD36 mice showed normalization of the hyperglycemia and the hyperinsulinemia as well as a marked improvement in liver insulin sensitivity. The MKR/CD36 mice also exhibited normal rates of fatty acid oxidation in skeletal muscle when compared with the decreased rate of fatty acid oxidation in MKR. With the reduction in insulin resistance, beta-cell function returned to normal. These and other results suggest that the insulin resistance in the MKR mice is associated with increased muscle triglycerides levels and that whole-body insulin resistance can be, at least partially, reversed in association with a reduction in muscle triglycerides levels, although the mechanisms are yet to be determined.


Subject(s)
CD36 Antigens/metabolism , Diabetes Mellitus, Type 2/physiopathology , Insulin Resistance , Muscle, Skeletal/metabolism , Animals , Diabetes Mellitus, Type 2/pathology , Fatty Acids/blood , Glucose/pharmacokinetics , Glucose Clamp Technique , Glycogen/metabolism , Hyperglycemia/blood , Hyperinsulinism/blood , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/pathology , Liver/metabolism , Male , Mice , Mice, Transgenic , Oxidation-Reduction , Triglycerides/blood , Triglycerides/metabolism
11.
Biochem Biophys Res Commun ; 310(2): 641-5, 2003 Oct 17.
Article in English | MEDLINE | ID: mdl-14521959

ABSTRACT

Resistin, a recently described adipocyte factor, is regulated by peroxisome proliferator-activated receptor gamma (PPARgamma) agonists. While resistin has been proposed to mediate insulin resistance in rodents, little is known about human resistin and its expression in pancreatic islets has not been tested. The goal of the present study was therefore to analyze whether resistin, like PPARgamma, is expressed in islets. Human islets from seven donors were analyzed by quantitative RT-PCR revealing resistin expression in all samples. Immunohistochemistry using a resistin-specific antibody on human pancreatic sections localized resistin protein to the islets. Mouse resistin was also detected in the Min6 beta cell line. Interestingly, we found a 4-fold increase in islet resistin expression in insulin resistant A-ZIP transgenic compared to wild-type mice. Our results demonstrate that resistin is expressed in islets and up-regulated in insulin resistance and thereby shed new light on the role of resistin in mice and humans.


Subject(s)
Hormones, Ectopic/metabolism , Intercellular Signaling Peptides and Proteins , Islets of Langerhans/metabolism , Proteins , Animals , Cell Line, Tumor , Cells, Cultured , Hormones, Ectopic/genetics , Hormones, Ectopic/immunology , Humans , Immunohistochemistry , Insulin Resistance , Islets of Langerhans/anatomy & histology , Mice , Nerve Growth Factor , RNA, Messenger/metabolism , Resistin , Transcription, Genetic , Up-Regulation
12.
Proc Natl Acad Sci U S A ; 100(10): 5920-5, 2003 May 13.
Article in English | MEDLINE | ID: mdl-12719535

ABSTRACT

Posttranslational modifications and remodeling of nucleosomes are critical factors in the regulation of transcription. Higher-order folding of chromatin also is likely to contribute to the control of gene expression, but the absence of a detailed description of the structure of the chromatin fiber has impaired progress in this area. Mammalian somatic cells contain a set of H1 linker-histone subtypes, H1 (0) and H1a to H1e, that bind to nucleosome core particles and to the linker DNA between nucleosomes. To determine whether the H1 histone subtypes play differential roles in the regulation of gene expression, we combined mice lacking specific H1 histone subtypes with mice carrying transgenes subject to position effects. Because position effects result from the unique chromatin structure created by the juxtaposition of regulatory elements in the transgene and at the site of integration, transgenes can serve as exquisitely sensitive indicators of chromatin structure. We report that some, but not all, linker histones can attenuate or accentuate position effects. The results suggest that the linker-histone subtypes play differential roles in the control of gene expression and that the sequential arrangement of the linker histones on the chromatin fiber might regulate higher-order chromatin structure and fine-tune expression levels.


Subject(s)
Anemia, Hemolytic/blood , Globins/genetics , Globins/metabolism , Histones/genetics , Anemia, Hemolytic/chemically induced , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/physiology , Gene Deletion , Gene Expression Regulation , Histones/metabolism , In Situ Hybridization, Fluorescence , Mice , Mice, Transgenic , Phenylhydrazines , Protein Processing, Post-Translational , Spleen/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...