Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
2.
Int J Obes Suppl ; 5(Suppl 1): S23-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-27152171

ABSTRACT

Brown adipose tissue (BAT) is capable of transforming chemically stored energy, in the form of triglycerides, into heat. Recent studies have shown that metabolically active BAT is present in a large proportion of adult humans, where its activity correlates with a favorable metabolic status. Hence, the tissue is now regarded as an interesting target for therapies against obesity and associated diseases such as type 2 diabetes, the hypothesis being that an induction of BAT would be beneficial for these disease states. Apart from the association between BAT activity and a healthier metabolic status, later studies have also shown a positive correlation between BAT volume and both bone cross-sectional area and bone mineral density, suggesting that BAT might stimulate bone anabolism. The aim of this review is to give the reader a brief overview of the BAT research field and to summarize and discuss recent findings regarding BAT being a potential player in bone metabolism.

3.
J Intern Med ; 276(4): 364-77, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24717051

ABSTRACT

Obesity and related diseases are a major cause of human morbidity and mortality and constitute a substantial economic burden for society. Effective treatment regimens are scarce, and new therapeutic targets are needed. Brown adipose tissue, an energy-expending tissue that produces heat, represents a potential therapeutic target. Its presence is associated with low body mass index, low total adipose tissue content and a lower risk of type 2 diabetes mellitus. Knowledge about the development and function of thermogenic adipocytes in brown adipose tissue has increased substantially in the last decade. Important transcriptional regulators have been identified, and hormones able to modulate the thermogenic capacity of the tissue have been recognized. Intriguingly, it is now clear that humans, like rodents, possess two types of thermogenic adipocytes: the classical brown adipocytes found in the interscapular brown adipose organ and the so-called beige adipocytes primarily found in subcutaneous white adipose tissue after adrenergic stimulation. The presence of two distinct types of energy-expending adipocytes in humans is conceptually important because these cells might be stimulated and recruited by different signals, raising the possibility that they might be separate potential targets for therapeutic intervention. In this review, we will discuss important features of the energy-expending brown adipose tissue and highlight those that may serve as potential targets for pharmacological intervention aimed at expanding the tissue and/or enhancing its function to counteract obesity.


Subject(s)
Adipose Tissue, Brown/physiology , Obesity/physiopathology , Obesity/therapy , Adipocytes/classification , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Energy Metabolism , Hormones/physiology , Humans , Subcutaneous Fat/cytology , Thermogenesis
4.
Int J Obes (Lond) ; 34 Suppl 1: S43-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20935666

ABSTRACT

Obesity is endemic in many regions of the world and a forerunner of several serious and sometimes fatal diseases such as ischemic heart disease, stroke, kidney failure and neoplasia. Although we know its origin--it results when energy intake exceeds energy expenditure--at present, the only proven therapy is bariatric surgery. This is a major abdominal procedure that, for reasons that are largely unknown (it cannot be explained solely by a reduction in ventricular volume), significantly reduces energy intake, but because of cost and limited availability, it will most likely be reserved for only a small fraction of those who stand to gain from effective antiobesity treatment. Clearly, alternative ways to treat obesity are needed. Another way to combat excessive accumulation of white adipose tissue would be to increase energy expenditure. Rodents, hibernators and human infants all have a specialized tissue--brown adipose tissue (BAT)--with the unique capacity to regulate energy expenditure by a process called adaptive thermogenesis. This process depends on the expression of uncoupling protein-1 (UCP1), which is a unique marker for BAT. UCP1 is an inner mitochondrial membrane protein that short circuits the mitochondrial proton gradient, so that oxygen consumption is no longer coupled to adenosine triphosphate synthesis. As a consequence, heat is generated. Mice lacking ucp-1 are severely compromised in their ability to maintain normal body temperature when acutely exposed to cold and they are also prone to become obese. We have shown that, in mice, BAT protects against diet-induced obesity, insulin resistance and type 2 diabetes. This is based on prevention of excessive accumulation of triglyceride in non-adipose tissues such as muscle and liver. Ectopic triglyceride storage at these locations is associated with initiation of insulin resistance and, ultimately, development of type 2 diabetes.


Subject(s)
Adipose Tissue, Brown/physiology , Insulin Resistance/physiology , Ion Channels/physiology , Mitochondrial Proteins/physiology , Obesity/physiopathology , Thermogenesis/physiology , Animals , Bariatric Surgery , Diabetes Mellitus, Type 2/etiology , Energy Intake/physiology , Humans , Insulin Resistance/genetics , Ion Channels/genetics , Mice , Mitochondrial Proteins/genetics , Obesity/genetics , Obesity/surgery , Uncoupling Protein 1
5.
Lymphology ; 39(2): 84-94, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16910099

ABSTRACT

FOXC2 mutations cause the lymphatic/ocular disorder Lymphedema-Distichiasis (LD), and Foxc2 haploinsufficient mice mimic this disorder. To determine if FOXC2 overexpression might also cause lymphatic and/or ocular abnormalities, we performed dynamic lymphatic imaging (Evans blue dye), ocular tissue examination, and metabolic profiles in mice: transgenic for FOXC2 with an adipocyte (aP2) promoter (aP2-FOXC2 Tg), heterozygous for targeted disruption of Foxc2 (Foxc2+/-), or compound heterozygous and transgenic (Foxc2+/-, Tg) compared to wild-type controls (WT). Foxc2+/-; aP2-FOXC2 Tg; and Foxc2+/-, Tg, exhibited LD's distinctive hyperplastic lymphatic phenotype characterized by increased number of lymphatic channels and lymph nodes as well as retrograde lymph reflux. Foxc2+/-, and Foxc2+/-, Tg but not aP2-FOXC2 Tg or WT showed an abnormal ocular phenotype. Previously described alterations in brown/ white fat distribution and lean phenotype in aP2-FOXC2 transgenics were confirmed. AP2-FOXC2 Tg immunohistochemistry disclosed aberrant FOXC2 expression in ectopic sites, especially embryonic heart. Lymphatic system links with fat metabolism are discussed.


Subject(s)
Disease Models, Animal , Eyelashes/abnormalities , Forkhead Transcription Factors/physiology , Lymphedema/genetics , Adipocytes/chemistry , Animals , Fatty Acid-Binding Proteins/genetics , Fatty Acids/metabolism , Female , Forkhead Transcription Factors/genetics , Glucose/metabolism , Heterozygote , Humans , Immunoenzyme Techniques , Insulin/metabolism , Lymphatic Abnormalities/genetics , Lymphatic Abnormalities/pathology , Lymphedema/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic
6.
Diabetologia ; 46(11): 1576-80, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14530861

ABSTRACT

AIMS/HYPOTHESIS: The transcription factor FOXC2 plays a key role in adipocyte differentiation and the FOXC2 gene is a candidate gene for Type 2 diabetes, obesity and dyslipidaemia. We investigated whether the FOXC2 -512C>T promoter variant is associated with Type 2 diabetes or its intermediary phenotypes in glucose tolerant subjects. METHODS: The variant was genotyped using PCR-RFLP in 705 unrelated Type 2 diabetic patients, 505 unrelated glucose-tolerant control subjects and 219 glucose-tolerant offspring of Type 2 diabetic probands. RESULTS: The frequency of the T-allele was 58% (95% CI 56-61%) and 59% (56-62%) among the Type 2 diabetic patients and the unrelated glucose-tolerant control subjects, respectively ( p=0.6). Among the glucose-tolerant subjects, the T-allele carriers had higher fasting serum triglyceride ( p=0.03), fasting serum C-peptide concentrations ( p=0.009) and insulinogenic index ( p=0.04). Furthermore, in glucose-tolerant women, the waist-to-hip ratio was significantly higher in carriers of the T-allele. CONCLUSION/INTERPRETATION: Our data suggest that the FOXC2 -512C>T variant is not associated with Type 2 diabetes. However, among glucose-tolerant subjects the variant is associated with hypertriglyceridaemia and increased fasting serum C-peptide.


Subject(s)
C-Peptide/blood , DNA-Binding Proteins/genetics , Diabetes Mellitus/genetics , Genetic Variation/genetics , Sequence Deletion , Transcription Factors/genetics , Base Sequence , Body Constitution , DNA Primers , Denmark , Female , Forkhead Transcription Factors , Glucose Tolerance Test , Humans , Male , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Reference Values , White People/genetics
7.
Cell ; 106(5): 563-73, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11551504

ABSTRACT

Obesity, hyperlipidemia, and insulin resistance are common forerunners of type 2 diabetes mellitus. We have identified the human winged helix/forkhead transcription factor gene FOXC2 as a key regulator of adipocyte metabolism. Increased FOXC2 expression, in adipocytes, has a pleiotropic effect on gene expression, which leads to a lean and insulin sensitive phenotype. FOXC2 affects adipocyte metabolism by increasing the sensitivity of the beta-adrenergic-cAMP-protein kinase A (PKA) signaling pathway through alteration of adipocyte PKA holoenzyme composition. Increased FOXC2 levels, induced by high fat diet, seem to counteract most of the symptoms associated with obesity, including hypertriglyceridemia and diet-induced insulin resistance--a likely consequence hereof would be protection against type 2 diabetes.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Hypertriglyceridemia/genetics , Insulin Resistance/genetics , Obesity/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/ultrastructure , Adrenergic beta-Agonists/pharmacology , Adult , Animals , Blood Glucose/metabolism , Blotting, Northern , Body Composition , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Diet , Enzyme Activation , Forkhead Transcription Factors , Gene Expression , Gene Expression Regulation , Genes, Reporter , Humans , Hypertriglyceridemia/metabolism , Insulin/blood , Isoenzymes/metabolism , Lipids/blood , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Obesity/metabolism , Organ Size , Oxygen Consumption
8.
Development ; 128(12): 2397-406, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11493558

ABSTRACT

The murine Foxf1 gene, encoding a forkhead - or winged helix - transcription factor, is expressed in splanchnic mesenchyme during organogenesis. The concentration of expression to subepithelial mesenchyme suggested that Foxf1 is activated by paracrine signals from endodermal epithelia. Homozygous Foxf1-null mice die before embryonic day 10, owing to defects in extra-embryonic mesoderm, and do not provide any information about the role of Foxf1 in morphogenesis of endodermally derived organs. We show that, on CD1 genetic background, Foxf1 heterozygote perinatal mortality is around 90%. The haploinsufficiency causes a variable phenotype that includes lung immaturity and hypoplasia, fusion of right lung lobes, narrowing of esophagus and trachea, esophageal atresia and tracheo-esophageal fistula. Similar malformations are observed in mutants that are defective in the sonic hedgehog (Shh) signaling pathway, and we show that exogenous Shh activates transcription of Foxf1 in developing lung. Foxf1 mRNA is absent in the lungs, foregut and sclerotomes of Shh(-/-) embryos, but persists in tissues where indian hedgehog (Ihh) is expressed. In lung organ cultures, activation of Foxf1 by Shh is counteracted by bone morphogenetic protein 4 (BMP4). Fibroblast growth factor (FGF) 10 and FGF7 both decrease Foxf1 expression and we speculate that this is mediated by transcriptional activation of epithelial Bmp4 (in the case of FGF10) and by inhibition of Shh expression for FGF7.


Subject(s)
Intestine, Small/embryology , Lung/embryology , Morphogenesis/physiology , Signal Transduction , Trans-Activators/metabolism , Transcription Factors/physiology , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Down-Regulation , Female , Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Hedgehog Proteins , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors/genetics , Transcriptional Activation
9.
Biochim Biophys Acta ; 1535(2): 174-85, 2001 Feb 14.
Article in English | MEDLINE | ID: mdl-11342006

ABSTRACT

UNLABELLED: It is well-established that high levels of cAMP or glucose can produce insulin resistance. The aim of this study was to characterize the interaction between these agents and insulin with respect to adipose tissue/muscle glucose transporter isoform (glucose transporter 4, GLUT4) gene regulation in cultured 3T3-F442A adipocytes and to further elucidate the GLUT4-related mechanisms in insulin resistance. Insulin (10(4) microU/ml) treatment for 16 h clearly increased GLUT4 mRNA level in cells cultured in medium containing 5.6 mM glucose but not in cells cultured in medium with high glucose (25 mM). 8-Bromo-cAMP (1 or 4 mM) or N(6)-monobutyryl cAMP, a hydrolyzable and a non-hydrolyzable cAMP analog, respectively, markedly decreased the GLUT4 mRNA level irrespective of glucose concentrations. In addition, these cAMP analogs also inhibited the upregulating effect of insulin on GLUT4 mRNA level. Interestingly, the tyrosine phosphatase inhibitor vanadate (1-50 microM) clearly increased GLUT4 mRNA level in a time- and concentration-dependent manner. Furthermore, cAMP-induced inhibition of the insulin effect was also prevented by vanadate. In parallel to the effects on GLUT4 gene expression, both insulin, vanadate and cAMP produced similar changes in cellular GLUT4 protein content and cAMP impaired the effect of insulin to stimulate (14)C-deoxyglucose uptake. In contrast, insulin, vanadate or cAMP did not alter insulin receptor (IR) mRNA or the cellular content of IR protein. IN CONCLUSION: (1) Both insulin and vanadate elicit a stimulating effect on GLUT4 gene expression in 3T3-F442A cells, but a prerequisite is that the surrounding glucose concentration is low. (2) Cyclic AMP impairs the insulin effect on GLUT4 gene expression, but this is prevented by vanadate, probably by enhancing the tyrosine phosphorylation of signalling peptides and/or transcription factors. (3) IR gene and protein expression is not altered by insulin, vanadate or cAMP in this cell type. (4) The changes in GLUT4 gene expression produced by cAMP or vanadate are accompanied by similar alterations in GLUT4 protein expression and glucose uptake, suggesting a role of GLUT4 gene expression for the long-term regulation of cellular insulin action on glucose transport.


Subject(s)
Adipocytes/drug effects , Cyclic AMP/pharmacology , Glucose/pharmacology , Insulin/pharmacology , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , 3T3 Cells , Adipocytes/metabolism , Animals , Blotting, Western , Culture Media , Deoxyglucose/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Glucose/analysis , Glucose Transporter Type 4 , Insulin Resistance/genetics , Mice , Monosaccharide Transport Proteins/analysis , Monosaccharide Transport Proteins/genetics , Protein Tyrosine Phosphatases/antagonists & inhibitors , RNA, Messenger/analysis , Vanadates/pharmacology
10.
Development ; 128(2): 155-66, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11124112

ABSTRACT

The murine Foxf1 gene encodes a forkhead transcription factor expressed in extra-embryonic and lateral plate mesoderm and later in splanchnic mesenchyme surrounding the gut and its derivatives. We have disrupted Foxf1 and show that mutant embryos die at midgestation due to defects in mesodermal differentiation and cell adhesion. The embryos do not turn and become deformed by the constraints of a small, inflexible amnion. Extra-embryonic structures exhibit a number of differentiation defects: no vasculogenesis occurs in yolk sac or allantois; chorioallantoic fusion fails; the amnion does not expand with the growth of the embryo, but misexpresses vascular and hematopoietic markers. Separation of the bulk of yolk sac mesoderm from the endodermal layer and adherence between mesoderm of yolk sac and amnion, indicate altered cell adhesion properties and enhanced intramesodermal cohesion. A possible cause of this is misexpression of the cell-adhesion protein VCAM1 in Foxf1-deficient extra-embryonic mesoderm, which leads to co-expression of VCAM with its receptor, alpha(4)-integrin. The expression level of Bmp4 is decreased in the posterior part of the embryo proper. Consistent with this, mesodermal proliferation in the primitive streak is reduced and somite formation is retarded. Expression of Foxf1 and the homeobox gene Irx3 defines the splanchnic and somatic mesodermal layers, respectively. In Foxf1-deficient embryos incomplete separation of splanchnic and somatic mesoderm is accompanied by misexpression of Irx3 in the splanchnopleure, which implicates Foxf1 as a repressor of Irx3 and as a factor involved in coelom formation.


Subject(s)
Mesoderm/cytology , Transcription Factors/physiology , Allantois/cytology , Amnion/cytology , Animals , Base Sequence , Blood Vessels/embryology , Cell Differentiation , Cell Division , DNA Primers/genetics , Female , Fetal Death/genetics , Gene Expression , Gene Targeting , In Situ Hybridization , Male , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Transcription Factors/deficiency , Transcription Factors/genetics , Vascular Cell Adhesion Molecule-1/genetics , Yolk Sac/blood supply
11.
Dev Dyn ; 218(1): 136-49, 2000 May.
Article in English | MEDLINE | ID: mdl-10822266

ABSTRACT

The growing family of forkhead transcription factors plays many important roles during embryonic development. In this study we have used in situ hybridization to explore the expression pattern of the forkhead transcription factor gene FoxF2 (FREAC-2, LUN) during mouse and rat embryogenesis, postnatal development, and in adult tissues. We demonstrate that FoxF2 is expressed in the mesenchyme adjacent to the epithelium in alimentary, respiratory, and urinary tracts, similar to FoxF1 (FREAC-1, HFH-8). FoxF2 mRNA was also observed in organs that do not express FoxF1 during embryogenesis, e.g., in the central nervous system, eye, ear, and limb buds. In organs that express both FoxF2 and FoxF1, these transcription factors may have similar functions in epithelio-mesenchymal cross-talk, but the fact that FoxF2 is more widely expressed than FoxF1 suggests that FoxF2 also has an independent role as a developmental regulator.


Subject(s)
DNA-Binding Proteins/genetics , Epithelial Cells/cytology , Gene Expression Regulation, Developmental/physiology , Trans-Activators/genetics , Age Factors , Animals , Cell Communication/genetics , Central Nervous System/embryology , Central Nervous System/physiology , DNA-Binding Proteins/metabolism , Embryonic and Fetal Development/genetics , Epithelial Cells/chemistry , Epithelial Cells/physiology , Extremities/embryology , Forkhead Transcription Factors , In Situ Hybridization , Lung/embryology , Lung/physiology , Mesoderm/chemistry , Mesoderm/cytology , Mesoderm/physiology , Mice , Mice, Inbred Strains , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Spine/embryology , Spine/physiology , Trans-Activators/metabolism , Urogenital System/embryology , Urogenital System/physiology
12.
Int J Cancer ; 86(3): 337-43, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10760820

ABSTRACT

Regulation of cell differentiation is most often impaired in malignant tumors and may represent a key mechanism for the progression of the disease. CCAAT-enhancer binding protein (C/EBP) is a family of transcription factors involved in the regulation of embryonic gut development in rodents, which has also been detected in various malignancies, e.g., liposarcomas and breast and ovarian epithelial tumors. We studied the relationship between C/EBP and tumor histology (Duke's invasive stage and pathological grade) in colorectal cancer. Immunoblotting techniques were used on microdissected fresh frozen tumor specimens, and expression of C/EBPalpha, C/EBPbeta and C/EBPzeta (CHOP) was analyzed in addition to that of the cell-cycle regulator p53 and the proliferation marker PCNA. Expression of C/EBPbeta (LAP isoforms) was markedly increased in all tumors compared with normal colon mucosa. Although the inter-patient variability was large, we found that LIP, the isoform of C/EBPbeta known to inhibit transcription, was expressed at higher levels in Duke's stage B tumors compared with Duke's stage A, whereas Duke's C tumors had the lowest LIP expression. A similar relationship was seen for CHOP. The cell-cycle regulator gene p53 was the only factor that clearly correlated with pathological grade: a decrease in p53 expression was demonstrated. Our data suggest that genetic and cellular events involving C/EBPbeta and CHOP are important for tumor invasion and that these events do not appear to be related to the pathological grade of the tumor.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Aged , Aged, 80 and over , CCAAT-Enhancer-Binding Proteins , Cell Division/genetics , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/biosynthesis , Female , Humans , Male , Middle Aged , Nuclear Proteins/biosynthesis , Transcription Factor CHOP , Transcription Factors/biosynthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
J Mol Biol ; 296(2): 351-9, 2000 Feb 18.
Article in English | MEDLINE | ID: mdl-10669593

ABSTRACT

Transcription factors of the forkhead type share a highly conserved DNA-binding domain of about 100 amino acid residues. FREAC-11, expressed in adipocytes, belongs to this class. Here, we report on NMR studies that established the three-dimensional structure of the FREAC-11, DNA-binding domain. Although apparent similarities to the structures of other members within the forkhead family are observed, the structure also reveals some remarkable differences. Along with the complementary dynamics, the data provide insight into the fundamentals of sequence specificity within a highly conserved motif.


Subject(s)
Adipose Tissue/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Amino Acid Sequence , Binding Sites , DNA/genetics , DNA/metabolism , Forkhead Transcription Factors , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Solutions , Substrate Specificity
14.
Genes Dev ; 14(2): 245-54, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10652278

ABSTRACT

In the mouse mutant dysgenetic lens (dyl) the lens vesicle fails to separate from the ectoderm, causing a fusion between the lens and the cornea. Lack of a proliferating anterior lens epithelium leads to absence of secondary lens fibers and a dysplastic, cataractic lens. We report the cloning of a gene, FoxE3, encoding a forkhead/winged helix transcription factor, which is expressed in the developing lens from the start of lens placode induction and becomes restricted to the anterior proliferating cells when lens fiber differentiation begins. We show that FoxE3 is colocalized with dyl in the mouse genome, that dyl mice have mutations in the part of FoxE3 encoding the DNA-binding domain, and that these mutations cosegregate with the dyl phenotype. During embryonic development, the primordial lens epithelium is formed in an apparently normal way in dyl mutants. However, instead of the proliferation characteristic of a normal lens epithelium, the posterior of these cells fail to divide and show signs of premature differentiation, whereas the most anterior cells are eliminated by apoptosis. This implies that FoxE3 is essential for closure of the lens vesicle and is a factor that promotes survival and proliferation, while preventing differentiation, in the lens epithelium.


Subject(s)
Epithelial Cells/cytology , Lens, Crystalline/physiology , Transcription Factors/genetics , Amino Acid Sequence , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Division/genetics , Chromosome Segregation/genetics , Chromosome Segregation/physiology , Cloning, Molecular , Epithelial Cells/pathology , Epithelial Cells/physiology , Forkhead Transcription Factors , Humans , Lens, Crystalline/cytology , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Molecular Sequence Data , Mutation/genetics , Mutation/physiology , Phenotype , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Transcription Factors/physiology
15.
Br J Cancer ; 79(7-8): 1240-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10098766

ABSTRACT

The CCAAT/enhancer binding protein (C/EBP) family of transcription factors is involved in metabolism and differentiation of cells, especially in rodent liver cells and adipocytes. Their roles in vivo and in particular during pathophysiological conditions in humans are largely unknown. We have investigated the presence of C/EBPalpha, -beta, -delta and -zeta in normal ovaries and in epithelial ovarian tumours of different stages. Immunohistochemical experiments demonstrated that C/EBPalpha and C/EBPbeta were preferentially expressed in epithelial/tumour cells irrespective of stage or grade of the tumour. C/EBPbeta was located in the nuclei of the cells, in contrast to C/EBPalpha, which was present only in the cytoplasm of these cells. The nuclear localization of C/EBPbeta indicates an active role of this transcription factor in tumour cells, whereas the cytoplasmic distribution suggests a more passive function of C/EBPalpha. C/EBPdelta and -zeta demonstrated a more diverse distribution with predominant localization to epithelial cells, but stromal distribution was also noted. The intracellular distribution was confined to both the nucleus and the cytoplasm for C/EBPdelta and -zeta. Western blotting demonstrated that C/EBPalpha, -beta, -delta and -zeta were present in a majority of the samples. The amount of C/EBPbeta increased markedly with malignancy, i.e. with degree of dedifferentiation, while the other members of the C/EBP family displayed a more constant expression level. These results demonstrate an association between the expression of members of the C/EBP family and the formation of epithelial ovarian tumours, with C/EBPbeta as a potential marker for these tumours. As C/EBPbeta is known to be expressed during proliferation of cells in vitro, it may participate in the proliferative process of ovarian epithelial tumour cells in vivo and play a central role in tumour progression.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovary/metabolism , Adenocarcinoma/metabolism , Adenofibroma/metabolism , Adenoma/metabolism , Blotting, Western , CCAAT-Enhancer-Binding Proteins , Cystadenocarcinoma, Serous/metabolism , Disease Progression , Female , Humans , Immunohistochemistry , Ovarian Neoplasms/pathology
16.
Gastroenterology ; 116(4): 842-54, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10092306

ABSTRACT

BACKGROUND & AIMS: One-kilobase sequence of the upstream fragment of the pig lactase-phlorizin hydrolase gene has been shown to control small intestinal-specific expression and postweaning decline of lactase-phlorizin hydrolase in transgenic mice. The aim of this study was to identify the regulatory DNA elements and transcription factors controlling lactase-phlorizin hydrolase expression. METHODS: The activity of different lactase-phlorizin hydrolase promoter fragments was investigated by transfection experiments using Caco-2 cells. Electrophoretic mobility shift assays and supershift analyses were used to characterize the interaction between intestinal transcription factors and the identified regulatory elements. RESULTS: Functional analysis revealed three previously undescribed regulatory regions in the lactase-phlorizin hydrolase promoter: a putative enhancer between -894 and -798 binding hepatocyte nuclear factor (HNF)-1 at position -894 to -880; a repressor-binding element between -278 to -264 to which an HNF-3-like factor is able to bind; and an element between -178 to -164 that binds an activating transcription factor. CONCLUSIONS: Identification of three new regulatory regions and HNF-1 and HNF-3-like transcription factor as players in the regulation of lactase-phlorizin hydrolase gene transcription has an impact on the understanding of the molecular mechanisms behind age-dependent, tissue-specific, differentiation-dependent, and regional regulation of expression in the intestine.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Enzymologic , Lactase-Phlorizin Hydrolase/genetics , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins , Caco-2 Cells , Cell Differentiation , Forkhead Transcription Factors , Hepatocyte Nuclear Factor 1 , Hepatocyte Nuclear Factor 1-alpha , Hepatocyte Nuclear Factor 1-beta , Humans , Nuclear Proteins/physiology , Promoter Regions, Genetic , Swine , Transcription, Genetic
17.
Endocrinology ; 140(2): 835-43, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9927313

ABSTRACT

The C/EBP (CCAAT/enhancer-binding protein) family of transcription factors is important for differentiation, lipid biosynthesis, and metabolism. Here, we demonstrate for the first time the presence of C/EBP alpha, beta, delta, and zeta messenger RNA (mRNA) and protein in Sertoli cell primary cultures. Treatment with FSH or 8-CPTcAMP strongly induced C/EBP beta mRNA above basal levels with rapid and transient kinetics in Sertoli cell primary cultures as well as in whole testes from hypophysectomized rats. Whereas C/EBP beta mRNA was induced approximately 50-fold, C/EBP delta mRNA was induced 5- to 8-fold by cAMP in Sertoli cells. Messenger RNA for C/EBP beta and delta were induced by inhibition of protein synthesis with cycloheximide and cycloheximide acted synergistically with cAMP. Immunoblots with C/EBP antibodies demonstrated a strong induction of C/EBP beta, delta, and zeta by cAMP. Electrophoretic mobility shift analysis of nuclear proteins from cAMP-treated Sertoli cells using a C/EBP consensus oligonucleotide and antibodies revealed specific binding of C/EBP/DNA complexes, the majority of which were supershifted by C/EBP beta antibody. Transfections of Sertoli cells with a C/EBP reporter construct showed approximately 3-fold induction of reporter gene activity by cAMP. In contrast, the reporter gene vector with a mutated form of the C/EBP binding site, was almost unresponsive to cAMP in transfections of Sertoli cells. Furthermore, C/EBP beta expression increased the activities of two promoters known to be cAMP-responsive in Sertoli cells. Thus, the early induction of C/EBP isoforms by cAMP may play a role in FSH-dependent regulation of late response genes in Sertoli cells.


Subject(s)
Cyclic AMP/physiology , Receptors, Invertebrate Peptide/metabolism , Sertoli Cells/physiology , Transcription Factors/metabolism , Animals , Cells, Cultured , Electrophoresis , Follicle Stimulating Hormone/pharmacology , Hypophysectomy , Isomerism , Male , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Invertebrate Peptide/genetics , Substrate Specificity , Testis/metabolism
18.
J Endocrinol ; 160(2): 247-55, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9924194

ABSTRACT

Changes in GH secretion are associated with changes in serum lipoproteins, utilisation of fuels and body composition. Since lipoprotein lipase (LPL) is a key enzyme in the regulation of lipid and lipoprotein metabolism, changes in LPL activity may contribute to these effects of GH. The present study was undertaken to investigate the role of GH and the GH-dependent growth factor, IGF-I, in the regulation of LPL in heart, skeletal muscle and adipose tissue. Female rats were hypophysectomised at 50 days of age. One week later, hormonal therapy was commenced. All hypophysectomised rats received l-thyroxine and cortisol. Adipose tissue, the heart, soleus and gastrocnemius muscles were excised after 1 week of hormonal therapy. The effect of insulin injections on adipose tissue and heart LPL activity was also studied. In separate experiments, LPL activity in post-heparin plasma was measured. Hypophysectomy had no effect on adipose tissue LPL activity, whereas activity was reduced in heart, soleus and gastrocnemius muscle tissues. GH treatment had no significant effect on LPL activity in adipose tissue or soleus muscle, but increased the LPL activity in heart and gastrocnemius muscle. GH treatment increased post-heparin plasma LPL activity. Recombinant human IGF-I treatment (1.25 mg/kg per day) markedly reduced LPL activity in adipose tissue, but had no effect in muscle tissues. The effect of IGF-I treatment on adipose tissue LPL was not reflected by a decrease in post-heparin plasma LPL activity. Daily injections of insulin for 7 days increased LPL activity in adipose tissue but had no effect on heart LPL activity. In adipose tissue, LPL mRNA levels tended to decrease as a result of IGF-I treatment. In the muscle tissues, no significant effects of hypophysectomy, GH or IGF-I treatment on LPL mRNA levels were observed.%It is concluded that GH increases heart and skeletal muscle tissue LPL activity, which probably contributes to an increased post-heparin plasma LPL activity. The effect of GH on muscle LPL activity is probably not mediated by IGF-I or insulin. Insulin and IGF-I have opposite effects on LPL activity in adipose tissue.


Subject(s)
Growth Hormone/pharmacology , Lipoprotein Lipase/metabolism , Muscle, Skeletal/enzymology , Adipose Tissue/drug effects , Adipose Tissue/enzymology , Animals , Enzyme Activation , Female , Humans , Hypophysectomy , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Muscle, Skeletal/drug effects , Myocardium/enzymology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology
19.
J Biol Chem ; 274(1): 165-9, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9867825

ABSTRACT

In this paper we show that the kidney-expressed winged helix transcription factor FREAC-4 is regulated by Ets-1, another kidney-expressed transcription factor. Through transfection experiments three Ets-1 cis-elements are identified within the first 152 nucleotides upstream of the transcription start in the freac-4 promoter. These sites are confirmed in a DNase I in vitro protection assay using recombinant Ets-1 protein. In cotransfection experiments using an Ets-1 expression vector, the induction of freac-4 reporter gene activity is attenuated approximately 6-fold when the three Ets-1 binding sites are mutated. Furthermore, we demonstrate that overexpression of Ets-1 in the human embryonic kidney cell line 293 is sufficient to increase freac-4 mRNA levels. These results are compatible with the hypothesis that Ets-1 acts as an upstream regulator of FREAC-4 expression during kidney development.


Subject(s)
DNA-Binding Proteins/metabolism , Kidney/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Cell Line , DNA Primers , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Genes, Reporter , Humans , Kidney/embryology , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins c-ets , Trans-Activators/genetics , Transfection
20.
Nat Genet ; 20(4): 374-6, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9843211

ABSTRACT

Fkhl0 is a member of the forkhead family of winged helix transcriptional regulators. Genes encoding forkhead proteins are instrumental during embryogenesis in mammals, in particular during development of the nervous system. Here we report that mice with a targeted disruption of the Fkh10 locus exhibit circling behaviour, poor swimming ability and abnormal reaching response-all common findings in mice with vestibular dysfunction. These animals also fail to elicit a Preyer reflex in response to a suprathreshold auditory stimulation, as seen in mice with profound hearing impairment. Histological examination of the inner ear reveals a gross structural malformation of the vestibulum as well as the cochlea. These structures have been replaced by a single irregular cavity in which neither proper semicircular ducts nor cochlea can be identified. We also show that at 9.5 days post coitum (dpc), Fkh10 is exclusively expressed in the otic vesicle. These findings implicate Fkh10 as an early regulator necessary for development of both cochlea and vestibulum and identify its human homologue FKHL10 as a previously unknown candidate deafness gene at 5q34.


Subject(s)
Ear, Inner/embryology , Nuclear Proteins/physiology , Transcription Factors/physiology , Acoustic Stimulation , Animals , Behavior, Animal , Ear, Inner/physiopathology , Forkhead Transcription Factors , Mice , Mice, Mutant Strains , Molecular Sequence Data , Vestibular Diseases/genetics , Vestibular Diseases/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...