Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
Add more filters

Publication year range
1.
Nature ; 506(7489): 445-50, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24553142

ABSTRACT

Ependymomas are common childhood brain tumours that occur throughout the nervous system, but are most common in the paediatric hindbrain. Current standard therapy comprises surgery and radiation, but not cytotoxic chemotherapy as it does not further increase survival. Whole-genome and whole-exome sequencing of 47 hindbrain ependymomas reveals an extremely low mutation rate, and zero significant recurrent somatic single nucleotide variants. Although devoid of recurrent single nucleotide variants and focal copy number aberrations, poor-prognosis hindbrain ependymomas exhibit a CpG island methylator phenotype. Transcriptional silencing driven by CpG methylation converges exclusively on targets of the Polycomb repressive complex 2 which represses expression of differentiation genes through trimethylation of H3K27. CpG island methylator phenotype-positive hindbrain ependymomas are responsive to clinical drugs that target either DNA or H3K27 methylation both in vitro and in vivo. We conclude that epigenetic modifiers are the first rational therapeutic candidates for this deadly malignancy, which is epigenetically deregulated but genetically bland.


Subject(s)
CpG Islands/genetics , Ependymoma/genetics , Epigenesis, Genetic/genetics , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , DNA Methylation/drug effects , Embryonic Stem Cells/metabolism , Ependymoma/drug therapy , Epigenomics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing/drug effects , Histones/drug effects , Histones/metabolism , Humans , Infant , Mice , Mice, Inbred NOD , Mice, SCID , Mutation/genetics , Phenotype , Polycomb Repressive Complex 2/metabolism , Prognosis , Rhombencephalon/pathology , Xenograft Model Antitumor Assays
2.
Nutr Metab Cardiovasc Dis ; 29(3): 306-315, 2019 03.
Article in English | MEDLINE | ID: mdl-30738642

ABSTRACT

BACKGROUND AND AIMS: Heparin-binding EGF-like growth factor (HB-EGF) is a representative EGF family member that interacts with EGFR under diverse stress environment. Previously, we reported that the HB-EGF-targeting using antisense oligonucleotide (ASO) effectively suppressed an aortic aneurysm in the vessel wall and circulatory lipid levels. In this study, we further examined the effects of the HB-EGF ASO administration on the development of hyperlipidemia-associated atherosclerosis using an atherogenic mouse model. METHODS AND RESULTS: The male and female LDLR deficient mice under Western diet containing 21% fat and 0.2% cholesterol content were cotreated with control and HB-EGF ASOs for 12 weeks. We observed that the HB-EGF ASO administration effectively downregulated circulatory VLDL- and LDL-associated lipid levels in circulation; concordantly, the HB-EGF targeting effectively suppressed the development of atherosclerosis in the aorta. An EGFR blocker BIBX1382 administration suppressed the hepatic TG secretion rate, suggesting a positive role of the HB-EGF signaling for the hepatic VLDL production. We newly observed that there was a significant improvement of the insulin sensitivity by the HB-EGF ASO administration in a mouse model under the Western diet as demonstrated by the improvement of the glucose and insulin tolerances. CONCLUSION: The HB-EGF ASO administration effectively downregulated circulatory lipid levels by suppressing hepatic VLDL production rate, which leads to effective protection against atherosclerosis in the vascular wall.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Heparin-binding EGF-like Growth Factor/metabolism , Hyperlipidemias/prevention & control , Lipoproteins, VLDL/blood , Oligonucleotides, Antisense/administration & dosage , Animals , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Biomarkers/blood , Cholesterol/blood , Disease Models, Animal , Down-Regulation , Female , Hep G2 Cells , Heparin-binding EGF-like Growth Factor/genetics , Humans , Hyperlipidemias/blood , Hyperlipidemias/genetics , Insulin Resistance , Liver/metabolism , Male , Mice, Knockout , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Triglycerides/blood
3.
Int J Obes (Lond) ; 40(9): 1424-34, 2016 09.
Article in English | MEDLINE | ID: mdl-27163748

ABSTRACT

BACKGROUND AND OBJECTIVES: Obesity is a global epidemic which increases the risk of the metabolic syndrome. Cathelicidin (LL-37 and mCRAMP) is an antimicrobial peptide with an unknown role in obesity. We hypothesize that cathelicidin expression correlates with obesity and modulates fat mass and hepatic steatosis. MATERIALS AND METHODS: Male C57BL/6 J mice were fed a high-fat diet. Streptozotocin was injected into mice to induce diabetes. Experimental groups were injected with cathelicidin and CD36 overexpressing lentiviruses. Human mesenteric fat adipocytes, mouse 3T3-L1 differentiated adipocytes and human HepG2 hepatocytes were used in the in vitro experiments. Cathelicidin levels in non-diabetic, prediabetic and type II diabetic patients were measured by enzyme-linked immunosorbent assay. RESULTS: Lentiviral cathelicidin overexpression reduced hepatic steatosis and decreased the fat mass of high-fat diet-treated diabetic mice. Cathelicidin overexpression reduced mesenteric fat and hepatic fatty acid translocase (CD36) expression that was reversed by lentiviral CD36 overexpression. Exposure of adipocytes and hepatocytes to cathelicidin significantly inhibited CD36 expression and reduced lipid accumulation. Serum cathelicidin protein levels were significantly increased in non-diabetic and prediabetic patients with obesity, compared with non-diabetic patients with normal body mass index (BMI) values. Prediabetic patients had lower serum cathelicidin protein levels than non-diabetic subjects. CONCLUSIONS: Cathelicidin inhibits the CD36 fat receptor and lipid accumulation in adipocytes and hepatocytes, leading to a reduction of fat mass and hepatic steatosis in vivo. Circulating cathelicidin levels are associated with increased BMI. Our results demonstrate that cathelicidin modulates the development of obesity.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Fatty Liver/drug therapy , Fatty Liver/prevention & control , Lipid Metabolism/drug effects , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , CD36 Antigens/biosynthesis , CD36 Antigens/genetics , Cell Differentiation/drug effects , Diabetes Mellitus, Experimental , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/complications , Fatty Liver/metabolism , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Humans , Immunohistochemistry , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/complications , Obesity/metabolism , Prediabetic State/complications , Prediabetic State/metabolism , Cathelicidins
4.
Nat Genet ; 31(1): 111-5, 2002 May.
Article in English | MEDLINE | ID: mdl-11923875

ABSTRACT

Regulation of glucose homeostasis by insulin depends on the maintenance of normal beta-cell mass and function. Insulin-like growth factor 1 (Igf1) has been implicated in islet development and differentiated function, but the factors controlling this process are poorly understood. Pancreatic islets produce Igf1 and Igf2, which bind to specific receptors on beta-cells. Igf1 has been shown to influence beta-cell apoptosis, and both Igf1 and Igf2 increase islet growth; Igf2 does so in a manner additive with fibroblast growth factor 2 (ref. 10). When mice deficient for the Igf1 receptor (Igf1r(+/-)) are bred with mice lacking insulin receptor substrate 2 (Irs2(-/-)), the resulting compound knockout mice show a reduction in mass of beta-cells similar to that observed in pancreas of Igf1r(-/-) mice (ref. 11), suggesting a role for Igf1r in growth of beta-cells. It is possible, however, that the effects in these mice occur secondary to changes in vascular endothelium or in the pancreatic ductal cells, or because of a decrease in the effects of other hormones implicated in islet growth. To directly define the role of Igf1, we have created a mouse with a beta-cell-specific knockout of Igf1r (betaIgf1r(-/-)). These mice show normal growth and development of beta-cells, but have reduced expression of Slc2a2 (also known as Glut2) and Gck (encoding glucokinase) in beta-cells, which results in defective glucose-stimulated insulin secretion and impaired glucose tolerance. Thus, Igf1r is not crucial for islet beta-cell development, but participates in control of differentiated function.


Subject(s)
Glucose Intolerance/etiology , Hyperinsulinism/etiology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Receptor, IGF Type 1/deficiency , Animals , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/genetics , Signal Transduction
5.
Nat Genet ; 27(4): 375-82, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11279518

ABSTRACT

Maturity-onset diabetes of the young type 3 (MODY3) is caused by haploinsufficiency of hepatocyte nuclear factor-1alpha (encoded by TCF1). Tcf1-/- mice have type 2 diabetes, dwarfism, renal Fanconi syndrome, hepatic dysfunction and hypercholestrolemia. Here we explore the molecular basis for the hypercholesterolemia using oligonucleotide microchip expression analysis. We demonstrate that Tcf1-/- mice have a defect in bile acid transport, increased bile acid and liver cholesterol synthesis, and impaired HDL metabolism. Tcf1-/- liver has decreased expression of the basolateral membrane bile acid transporters Slc10a1, Slc21a3 and Slc21a5, leading to impaired portal bile acid uptake and elevated plasma bile acid concentrations. In intestine and kidneys, Tcf1-/- mice lack expression of the ileal bile acid transporter (Slc10a2), resulting in increased fecal and urinary bile acid excretion. The Tcf1 protein (also known as HNF-1alpha) also regulates transcription of the gene (Nr1h4) encoding the farnesoid X receptor-1 (Fxr-1), thereby leading to reduced expression of small heterodimer partner-1 (Shp-1) and repression of Cyp7a1, the rate-limiting enzyme in the classic bile acid biosynthesis pathway. In addition, hepatocyte bile acid storage protein is absent from Tcf1-/- mice. Increased plasma cholesterol of Tcf1-/- mice resides predominantly in large, buoyant, high-density lipoprotein (HDL) particles. This is most likely due to reduced activity of the HDL-catabolic enzyme hepatic lipase (Lipc) and increased expression of HDL-cholesterol esterifying enzyme lecithin:cholesterol acyl transferase (Lcat). Our studies demonstrate that Tcf1, in addition to being an important regulator of insulin secretion, is an essential transcriptional regulator of bile acid and HDL-cholesterol metabolism.


Subject(s)
Bile Acids and Salts/metabolism , Cholesterol/blood , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Animals , Base Sequence , Bile Acids and Salts/biosynthesis , DNA Primers , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Ileum/metabolism , Kidney/metabolism , Mice , Mice, Knockout , Transcription Factors/genetics , Transcription Factors/physiology
6.
Diabetes Metab ; 47(2): 101183, 2021 03.
Article in English | MEDLINE | ID: mdl-32791310

ABSTRACT

AIMS: Trimethylamine N-oxide (TMAO), choline and betaine serum levels have been associated with metabolic diseases including type 2 diabetes (T2D) and non-alcoholic fatty liver disease (NAFLD). These associations could be mediated by insulin resistance. However, the relationships among these metabolites, insulin resistance and NAFLD have not been thoroughly investigated. Moreover, it has recently been suggested that TMAO could play a role in NAFLD by altering bile acid metabolism. We examined the association between circulating TMAO, choline and betaine levels and NAFLD in obese subjects. METHODS: Serum TMAO, choline, betaine and bile acid levels were measured in 357 Mexican obese patients with different grades of NAFLD as determined by liver histology. Associations of NAFLD with TMAO, choline and betaine levels were tested. Moreover, association of TMAO levels with non-alcoholic steatohepatitis (NASH) was tested separately in patients with and without T2D. RESULTS: TMAO and choline levels were significantly associated with NAFLD histologic features and NASH risk. While increased serum TMAO levels were significantly associated with NASH in patients with T2D, in non-T2D subjects this association lost significance after adjusting for sex, BMI and HOMA2-IR. Moreover, circulating secondary bile acids were associated both with increased TMAO levels and NASH. CONCLUSIONS: In obese patients, circulating TMAO levels were associated with NASH mainly in the presence of T2D. Functional studies are required to evaluate the role of insulin resistance and T2D in this association, both highly prevalent in NASH patients.


Subject(s)
Diabetes Mellitus, Type 2 , Methylamines/blood , Non-alcoholic Fatty Liver Disease , Adult , Betaine/blood , Bile Acids and Salts/blood , Biomarkers/blood , Biopsy , Choline/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Female , Humans , Insulin Resistance , Liver/pathology , Male , Mexican Americans , Middle Aged , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/epidemiology , Obesity/complications , Obesity/epidemiology
7.
J Cell Biol ; 122(6): 1361-71, 1993 Sep.
Article in English | MEDLINE | ID: mdl-7690763

ABSTRACT

Monoclonal antibodies (mAbs) have been produced against the chicken beta 1 subunit that affect integrin functions, including ligand binding, alpha subunit association, and regulation of ligand specificity. Epitope mapping of these antibodies was used to identify regions of the subunit involved in these functions. To accomplish this, we produced mouse/chicken chimeric beta 1 subunits and expressed them in mouse 3T3 cells. These chimeric subunits were fully functional with respect to heterodimer formation, cell surface expression, and cell adhesion. They differed in their ability to react with a panel anti-chicken beta 1 mAbs. Epitopes were identified by a loss of antibody binding upon substitution of regions of the chicken beta 1 subunit by homologous regions of the mouse beta 1 subunit. The identification of the epitope was confirmed by a reciprocal exchange of chicken and mouse beta 1 domains that resulted in the gain of the ability of the mouse subunit to interact with a particular anti-chicken beta 1 mAb. Using this approach, we found that the epitopes for one set of antibodies that block ligand binding mapped toward the amino terminal region of the beta 1 subunit. This region is homologous to a portion of the ligand-binding domain of the beta 3 subunit. In addition, a second set of antibodies that either block ligand binding, alter ligand specificity, or induce alpha/beta subunit dissociation mapped to the cysteine rich repeats near the transmembrane domain of the molecule. These data are consistent with a model in which a portion of beta 1 ligand binding domain rests within the amino terminal 200 amino acids and a regulatory domain, that affects ligand binding through secondary changes in the structure of the molecule resides in a region of the subunit, possibly including the cysteine-rich repeats, nearer the transmembrane domain. The data also suggest the possibility that the alpha subunit may exert an influence on ligand specificity by interacting with this regulatory domain of the beta 1 subunit.


Subject(s)
Epitopes/genetics , Integrins/genetics , Integrins/physiology , 3T3 Cells , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Base Sequence , Cell Adhesion , Chickens , Chimera , Cysteine/analysis , DNA/analysis , DNA/genetics , Epitopes/chemistry , Epitopes/immunology , Integrins/immunology , Ligands , Mice , Molecular Sequence Data , Peptide Mapping , Sequence Homology, Nucleic Acid
8.
Value Health ; 17(7): A539, 2014 Nov.
Article in English | MEDLINE | ID: mdl-27201728
9.
J Thromb Haemost ; 16(9): 1857-1872, 2018 09.
Article in English | MEDLINE | ID: mdl-29981269

ABSTRACT

Essentials Microbe-dependent production of trimethylamine N-oxide (TMAO) contributes to thrombosis risk. The impact of host flavin monooxygenase 3 (FMO3) modulation on platelet function is unknown. Genetic manipulation of FMO3 in mice alters systemic TMAO levels and thrombosis potential. Genetic manipulation of FMO3 is associated with alteration of gut microbial community structure. SUMMARY: Background Gut microbes play a critical role in the production of trimethylamine N-oxide (TMAO), an atherogenic metabolite that impacts platelet responsiveness and thrombosis potential. Involving both microbe and host enzymatic machinery, TMAO generation utilizes a metaorganismal pathway, beginning with ingestion of trimethylamine (TMA)-containing dietary nutrients such as choline, phosphatidylcholine and carnitine, which are abundant in a Western diet. Gut microbial TMA lyases use these nutrients as substrates to produce TMA, which upon delivery to the liver via the portal circulation, is converted into TMAO by host hepatic flavin monooxygenases (FMOs). Gut microbial production of TMA is rate limiting in the metaorganismal TMAO pathway because hepatic FMO activity is typically in excess. Objectives FMO3 is the major FMO responsible for host generation of TMAO; however, a role for FMO3 in altering platelet responsiveness and thrombosis potential in vivo has not yet been explored. Methods The impact of FMO3 suppression (antisense oligonucleotide-targeting) and overexpression (as transgene) on plasma TMAO levels, platelet responsiveness and thrombosis potential was examined using a murine FeCl3 -induced carotid artery injury model. Cecal microbial composition was examined using 16S analyses. Results Modulation of FMO3 directly impacts systemic TMAO levels, platelet responsiveness and rate of thrombus formation in vivo. Microbial composition analyses reveal taxa whose proportions are associated with both plasma TMAO levels and in vivo thrombosis potential. Conclusions The present studies demonstrate that host hepatic FMO3, the terminal step in the metaorganismal TMAO pathway, participates in diet-dependent and gut microbiota-dependent changes in both platelet responsiveness and thrombosis potential in vivo.


Subject(s)
Blood Platelets/physiology , Gastrointestinal Microbiome/physiology , Liver/enzymology , Methylamines/metabolism , Oxygenases/physiology , Thrombophilia/enzymology , Animals , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/chemically induced , Carotid Artery, Common , Chlorides/toxicity , Ferric Compounds/toxicity , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/pharmacology , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , Platelet-Rich Plasma , Ribotyping , Risk , Thrombophilia/microbiology , Transgenes
10.
J Clin Invest ; 97(7): 1630-9, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8601628

ABSTRACT

In an effort to identify genetic factors contributing to atherogenesis, we have studied inbred strains of mice that are susceptible (C57BL/6J) and resistant (C3H/HeJ) to diet-induced aortic fatty streak lesions. When maintained on a low-fat diet, HDL isolated from both strain C57BL/6J (B6) and C3H/HeJ (C3H) mice protect against LDL oxidation in a coculture model of the artery wall. However, when maintained on an atherogenic diet high in fat and cholesterol, the HDL isolated from B6 mice lose the capacity to protect, whereas HDL from C3H mice protect equally well. Associated with the loss in the ability of HDL to protect is a decrease in the activity of serum paraoxonase, a serum esterase carried on HDL that has previously been shown to protect against LDL oxidation in vitro. The levels of paraoxonase mRNA decreased in B6 mice upon challenge with the atherogenic diet but increased in C3H, indicating that paraoxonase production is under genetic control. In a set of recombinant inbred strains derived from the B6 and C3H parental strains, low paraoxonase mRNA levels segregated with aortic lesion development, supporting a role for paraoxonase in atherogenesis.


Subject(s)
Arteriosclerosis/etiology , Diet , Esterases/blood , Amino Acid Sequence , Animals , Arteriosclerosis/enzymology , Arteriosclerosis/genetics , Aryldialkylphosphatase , Base Sequence , Cloning, Molecular , Crosses, Genetic , DNA, Complementary/genetics , Diet, Atherogenic , Diet, Fat-Restricted , Disease Models, Animal , Esterases/genetics , Female , Gene Expression , Humans , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/pharmacology , Lipoproteins, LDL/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Data , Oxidation-Reduction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Species Specificity
11.
J Clin Invest ; 107(4): 419-30, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11181641

ABSTRACT

Myeloperoxidase (MPO), a heme enzyme secreted by activated phagocytes, generates an array of oxidants proposed to play critical roles in host defense and local tissue damage. Both MPO and its reaction products are present in human atherosclerotic plaque, and it has been proposed that MPO oxidatively modifies targets in the artery wall. We have now generated MPO-deficient mice, and show here that neutrophils from homozygous mutants lack peroxidase and chlorination activity in vitro and fail to generate chlorotyrosine or to kill Candida albicans in vivo. To examine the potential role of MPO in atherosclerosis, we subjected LDL receptor-deficient mice to lethal irradiation, repopulated their marrow with MPO-deficient or wild-type cells, and provided them a high-fat, high-cholesterol diet for 14 weeks. White cell counts and plasma lipoprotein profiles were similar between the two groups at sacrifice. Cross-sectional analysis of the aorta indicated that lesions in MPO-deficient mice were about 50% larger than controls. Similar results were obtained in a genetic cross with LDL receptor-deficient mice. In contrast to advanced human atherosclerotic lesions, the chlorotyrosine content of aortic lesions from wild-type as well as MPO-deficient mice was essentially undetectable. These data suggest an unexpected, protective role for MPO-generated reactive intermediates in murine atherosclerosis. They also identify an important distinction between murine and human atherosclerosis with regard to the potential involvement of MPO in protein oxidation.


Subject(s)
Arteriosclerosis/etiology , Peroxidase/physiology , Tyrosine/analogs & derivatives , Animals , Candida albicans/immunology , Humans , Hypochlorous Acid/metabolism , Mice , Mice, Inbred C57BL , Neutrophils/enzymology , Oxidation-Reduction , Peroxidase/deficiency , Peroxidase/genetics , Phagocytes/metabolism , Tyrosine/analysis
12.
Oncogene ; 36(37): 5231-5242, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28504719

ABSTRACT

Group3 medulloblastoma (MBG3) that predominantly occur in young children are usually associated with MYC amplification and/or overexpression, frequent metastasis and a dismal prognosis. Physiologically relevant MBG3 models are currently lacking, making inferences related to their cellular origin thus far limited. Using in utero electroporation, we here report that MBG3 mouse models can be developed in situ from different multipotent embryonic cerebellar progenitor cells via conditional expression of Myc and loss of Trp53 function in several Cre driver mouse lines. The Blbp-Cre driver that targets embryonic neural progenitors induced tumors exhibiting a large-cell/anaplastic histopathology adjacent to the fourth ventricle, recapitulating human MBG3. Enforced co-expression of luciferase together with Myc and a dominant-negative form of Trp53 revealed that GABAergic neuronal progenitors as well as cerebellar granule cells give rise to MBG3 with their distinct growth kinetics. Cross-species gene expression analysis revealed that these novel MBG3 models shared molecular characteristics with human MBG3, irrespective of their cellular origin. We here developed MBG3 mouse models in their physiological environment and we show that oncogenic insults drive this MB subgroup in different cerebellar lineages rather than in a specific cell of origin.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellum/embryology , Cerebellum/pathology , Medulloblastoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Cerebellum/cytology , Cerebellum/metabolism , Disease Models, Animal , Female , Humans , Male , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-myc/metabolism , Transfection
13.
Circ Res ; 86(10): 1078-84, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10827138

ABSTRACT

Lipids, monocytes, and arterial wall cells are primary components involved in atherogenesis. Using the inbred mouse strains C57BL/6J (B6) and C3H/HeJ (C3H), which have been extensively studied as models of the genetic control of diet-induced atherosclerosis, we examined which of these components determine genetic susceptibility. To test whether dietary responsiveness is involved, a congenic strain of C3H carrying an apoE-null allele (apoE(-/-)) was constructed. Although C3H.apoE(-/-) mice had higher plasma cholesterol levels, they developed much smaller lesions than their B6.apoE(-/-) counterpart on either chow or Western diets. Reciprocal bone marrow transplantation between the strains, with congenics carrying the same H-2 haplotype, was performed to examine the role of monocytes. The atherosclerosis susceptibility was not altered in the recipient mice, indicating that variations in monocyte function were not involved. Endothelial cells isolated from the aorta of B6 mice exhibited a dramatic induction of monocyte chemotactic protein-1, macrophage colony-stimulating factor, vascular cell adhesion molecule-1, and heme oxygenase-1 in response to minimally modified LDL, whereas endothelial cells from C3H mice showed little or no induction. In a set of recombinant inbred strains derived from the B6 and C3H parental strains, endothelial responses to minimally modified LDL cosegregated with aortic lesion size. These data provide strong evidence that endothelial cells, but not monocytes or plasma lipid levels, account for the difference in susceptibility to atherosclerosis between the 2 mouse strains.


Subject(s)
Arteriosclerosis/etiology , Animals , Aorta/pathology , Apolipoproteins E/deficiency , Blood Cells/physiology , Bone Marrow Transplantation , Cholesterol/metabolism , Disease Susceptibility , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Lipids/blood , Lipoproteins, LDL/chemistry , Lipoproteins, LDL/pharmacology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
15.
Circulation ; 102(1): 75-81, 2000 Jul 04.
Article in English | MEDLINE | ID: mdl-10880418

ABSTRACT

BACKGROUND: Oxidized LDL has been found within the subendothelial space, and it exhibits numerous atherogenic properties, including induction of inflammatory genes. We examined the possibility that variations in endothelial response to minimally modified LDL (MM-LDL) constitute one of the genetic components in atherosclerosis. METHODS AND RESULTS: By a novel explant technique, endothelial cells (ECs) were isolated from the aorta of inbred mouse strains with different susceptibilities to diet-induced atherosclerosis. Responses to MM-LDL were evaluated by examining the expression of inflammatory genes involved in atherosclerosis, including monocyte chemotactic protein-1 (MCP-1) and macrophage-colony-stimulating factor (M-CSF), an oxidative stress gene, heme oxygenase-1 (HO-1), and other, noninflammatory, genes. ECs from the susceptible mouse strain C57BL/6J exhibited dramatic induction of MCP-1, M-CSF, and HO-1, whereas ECs from the resistant strain C3H/HeJ showed little or no induction. In contrast, ECs from the 2 strains responded similarly to lipopolysaccharide. CONCLUSIONS: These data provide strong evidence that genetic factors in atherosclerosis act at the level of the vessel wall.


Subject(s)
Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Endothelium, Vascular/enzymology , Lipoproteins, LDL/metabolism , Animals , Aorta/cytology , Arteriosclerosis/immunology , Blotting, Northern , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemotaxis/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Membrane Proteins , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Vasculitis/enzymology
16.
Diabetes ; 48(7): 1459-65, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10389854

ABSTRACT

Genetic studies have shown that mutations in the gene encoding hepatocyte nuclear factor (HNF)-4alpha, a member of the steroid/thyroid hormone receptor superfamily, give rise to early-onset type 2 diabetes (MODY1). The functional properties of mutant HNF-4alpha proteins and the molecular mechanisms by which they impair insulin secretion are largely unknown. In the present study, we have investigated transcriptional activation, DNA binding properties, and protein dimerization activity of three HNF-4alpha missense mutations--HNF4(R127W), HNF4(V255M), and HNF4(E276Q)--that have been associated with type 2 diabetes. We demonstrate that HNF4(E276Q) has lost its ability to bind to HNF-4 consensus binding sites and activate transcription. HNF4(E276Q) had no effect on the functional activity of wild-type HNF-4alpha in the pancreatic beta-cell line HIT-T15, but it exhibited weak dominant-negative activity in other cell types. Analysis of HNF4(E276Q) protein showed that it exists in two forms: a full length 54-kDa protein and a 40-kDa COOH-terminal protein lacking the NH2-terminal transactivation domain and the DNA binding domain. Immunoprecipitation experiments indicate that this truncated protein can bind to wild-type HNF-4alpha and may be responsible for the weak dominant-negative effects seen in these cells. In addition, we show that the transcriptional transactivation of HNF4(R127W) and HNF4(V255M) is indistinguishable from that of wild-type HNF-4alpha, suggesting that they are sequence polymorphisms. Our results demonstrate that HNF4(E276Q) is a loss-of-function mutation and that it identifies glutamic acid 276 in alpha-helix 8 of the ligand-binding domain of HNF-4alpha protein as a critical residue for DNA binding, transcriptional activation, and protein stability in vivo.


Subject(s)
DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Mutation, Missense , Phosphoproteins/genetics , Receptors, Glucocorticoid/genetics , Transcription Factors/genetics , Animals , Cell Line , Hepatocyte Nuclear Factor 4 , Protein Biosynthesis , Rats , Transcriptional Activation
17.
Diabetes ; 49(5): 832-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10905494

ABSTRACT

Hepatocyte nuclear factor (HNF)-4alpha is a transcription factor that plays an important role in regulation of gene expression in pancreatic beta-cells and in the liver. Heterozygous mutations in the HNF-4alpha gene are responsible for maturity-onset diabetes of the young 1 (MODY1), which is characterized by pancreatic beta-cell-deficient insulin secretion. HNF-4alpha is a major transcriptional regulator of many genes expressed in the liver. However, no liver defect has been identified in individuals with HNF-4alpha mutations. In this study, we have identified HNF-4alpha target genes that are mainly expressed in the liver, including alpha1-antitrypsin, alpha1-antichymotrypsin, alpha-fetal protein, ceruloplasmin, IGF binding protein 1, transferrin, apolipoprotein(AI) [apo(AI)], apo(AII), apo(B), and apo(CIII). Serum levels of these proteins and Lp(a) and triglycerides were measured in 24 members of the HNF-4alpha/MODY1 RW pedigree (Q268X mutation), including 12 diabetic patients with HNF-4alpha mutations (D-HNF4+/-), 6 nondiabetic subjects with HNF-4alpha mutations (N-HNF4+/-), 6 normal relatives (N-HNF4+/+), 6 unrelated normal matched control subjects (N-HNF4+/+), and 12 matched diabetic (non-MODY1-5) patients (D-HNF4+/+). Serum levels of apo(AII), apo(CIII), lipoprotein(a) [Lp(a)], and triglyceride were significantly reduced in HNF4+/- subjects (26.9, 19.8, 12.1, and 72.1 mg/dl, respectively) compared with HNF4+/+ subjects (37.4, 26.5, 45.2, and 124.2 mg/dl, respectively) (P = 0.00001, P = 0.01, P = 0.00006, and P = 0.000003, respectively). This reduction was not found when apo(AII), apo(CIII), Lp(a), and triglyceride levels were compared in D-HNF4+/- versus N-HNF4+/- or in D-HNF4+/+ versus N-HNF4+/+ subjects, which indicates that HNF-4alpha haploinsufficiency rather than hyperglycemia is the primary cause of decreased serum protein and triglyceride concentrations. Furthermore, we determined that genetic or environmental modifiers other than HNF-4alpha do not appear to contribute to the observed decrease of HNF-4alpha-regulated serum proteins. This study demonstrates that a heterozygous HNF-4alpha mutation leads to an HNF-4alpha-dependent hepatocyte secretory defect of liver-specific proteins.


Subject(s)
DNA-Binding Proteins , Diabetes Mellitus, Type 2/genetics , Mutation , Phosphoproteins/genetics , Transcription Factors/genetics , Adult , Apolipoprotein A-II/blood , Apolipoprotein C-III , Apolipoproteins C/blood , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Diabetes Mellitus, Type 2/blood , Female , Genotype , Haplotypes , Hepatocyte Nuclear Factor 4 , Humans , Lipoprotein(a)/blood , Male , Phenotype , Triglycerides/blood
18.
Diabetes ; 50(11): 2472-80, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11679424

ABSTRACT

Mutations in hepatocyte nuclear factor 1alpha (HNF-1alpha) lead to maturity-onset diabetes of the young type 3 as a result of impaired insulin secretory response in pancreatic beta-cells. The expression of 50 genes essential for normal beta-cell function was studied to better define the molecular mechanism underlying the insulin secretion defect in Hnf-1alpha(-/-) mice. We found decreased steady-state mRNA levels of genes encoding glucose transporter 2 (Glut2), neutral and basic amino acid transporter, liver pyruvate kinase (L-Pk), and insulin in Hnf-1alpha(-/-) mice. In addition, we determined that the expression of several islet-enriched transcription factors, including Pdx-1, Hnf-4alpha, and Neuro-D1/Beta-2, was reduced in Hnf-1alpha(-/-) mice. These changes in pancreatic islet mRNA levels were already apparent in newborn animals, suggesting that loss of Hnf-1alpha function rather than chronic hyperglycemia is the primary cause of the altered gene expression. This expression profile was pancreatic islet-specific and distinct from hepatocytes, where we found normal expression of Glut2, L-Pk, and Hnf-4alpha in the liver of Hnf-1alpha(-/-) mice. The expression of small heterodimer partner (Shp-1), an orphan receptor that can heterodimerize with Hnf-4alpha and inhibit its transcriptional activity, was also reduced in Hnf-1alpha(-/-) islets. We characterized a 0.58-kb Shp-1 promoter and determined that the decreased expression of Shp-1 may be indirectly mediated by a downregulation of Hnf-4alpha. We further showed that Shp-1 can repress its own transcriptional activation by inhibiting Hnf-4alpha function, thereby establishing a feedback autoregulatory loop. Our results indicate that loss of Hnf-1alpha function leads to altered expression of genes involved in glucose-stimulated insulin secretion, insulin synthesis, and beta-cell differentiation.


Subject(s)
DNA-Binding Proteins , Gene Expression/physiology , Islets of Langerhans/embryology , Islets of Langerhans/metabolism , Nuclear Proteins , Transcription Factors/physiology , Animals , Cell Line , Fetus/physiology , Glucose/pharmacology , Hepatocyte Nuclear Factor 1 , Hepatocyte Nuclear Factor 1-alpha , Hepatocyte Nuclear Factor 1-beta , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Liver/physiology , Mice , Mice, Knockout/genetics , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Reference Values , Transcription Factors/deficiency , Transcription Factors/genetics , Transcriptional Activation/physiology
19.
J Mol Biol ; 195(2): 419-22, 1987 May 20.
Article in English | MEDLINE | ID: mdl-2821277

ABSTRACT

We have measured the contribution of the alkaline Bohr effect of the C-terminal histidine residues of the beta-chains of haemoglobin A by comparing haemoglobin A with haemoglobin Cowtown in which those histidine residues are replaced by leucine. Oxygenation of a stripped 2.5 mM (haem) solution of haemoglobin A yielded 0.19 H+/haem, while oxygenation of a similar solution of haemoglobin Cowtown produced no change of pH. Oxygen equilibria measured at 60 microM-haem in 0.1 M-Hepes buffer gave an alkaline Bohr effect of -0.21 H+/haem for haemoglobin A and only -0.01 H+/haem for haemoglobin Cowtown, even though its Hill's coefficient was greater than 2 throughout the pH range studied. These results prove that the chloride-independent part of the alkaline Bohr effect is due to the C-terminal histidine residues of the beta-chains. Oxygen equilibria measured in 0.095 M-bis-Tris buffers with minimal chloride or with 0.1 M-chloride showed the contribution of those histidine residues to the alkaline Bohr effect to be about 0.2 H+/haem, independent of chloride concentration. Determination of the individual Adair coefficients in the three different buffers indicated that pH and chloride tend to have their greatest effects at the second or third steps of oxygenation when the change of quaternary structure is most likely to occur; between pH 7 and 9, the fourth Adair coefficient is only very slightly affected by pH and not significantly by chloride.


Subject(s)
Anions , Hemoglobin A , Hemoglobins, Abnormal , Protons , Histidine , Humans , Hydrogen-Ion Concentration , Oxyhemoglobins/metabolism
20.
J Mol Biol ; 239(4): 555-60, 1994 Jun 17.
Article in English | MEDLINE | ID: mdl-8006967

ABSTRACT

Chloride reduces the oxygen affinity of mammalian haemoglobin by acting as an allosteric effector that stabilizes the quaternary deoxy (T) structure. Perutz and others showed evidence that it does so by neutralizing electrostatic repulsion by an excess of positive charges in the cavity that runs through the centre of the molecule, but without binding to any specific site. On the basis of this proposal, any amino acid substitutions in the central cavity that halve the number of excess positive charges should halve the chloride effect, neutralization of the excess positive charges should inhibit it and introduction of additional positive charges should enhance it. Charge changes on the surface of the molecule should leave it unaltered. We have tested this proposal by measuring the chloride effects in several abnormal human haemoglobins with replacements of polar residues in the central cavity or on the surface that we happened to come across. They all proved consistent with the proposal. It appears that diffusible electrolytes can modify allosteric equilibria without necessarily binding to any specific site. Our proposal also implies that amino acid substitutions that make the central cavity more electropositive should destabilize the T-structure and therefore increase the oxygen affinity, while substitutions that make it more electronegative should do the reverse. A survey of all substitutions reported in the literature shows that this is true, with a few exceptions due to special stereochemical effects.


Subject(s)
Chlorides/metabolism , Hemoglobins, Abnormal/chemistry , Hemoglobins, Abnormal/metabolism , Allosteric Regulation , Amino Acids/chemistry , Amino Acids/metabolism , Binding Sites , Humans , Hydrogen-Ion Concentration , Molecular Structure , Oxygen/metabolism , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL