Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Int J Obes (Lond) ; 42(3): 384-390, 2018 03.
Article in English | MEDLINE | ID: mdl-29381148

ABSTRACT

OBJECTIVE: Body mass index (BMI) is commonly used to assess obesity, which is associated with numerous diseases and negative health outcomes. BMI has been shown to be a heritable, polygenic trait, with close to 100 loci previously identified and replicated in multiple populations. We aim to replicate known BMI loci and identify novel associations in a trans-ethnic study population. SUBJECTS: Using eligible participants from the Population Architecture using Genomics and Epidemiology consortium, we conducted a trans-ethnic meta-analysis of 102 514 African Americans, Hispanics, Asian/Native Hawaiian, Native Americans and European Americans. Participants were genotyped on over 200 000 SNPs on the Illumina Metabochip custom array, or imputed into the 1000 Genomes Project (Phase I). Linear regression of the natural log of BMI, adjusting for age, sex, study site (if applicable), and ancestry principal components, was conducted for each race/ethnicity within each study cohort. Race/ethnicity-specific, and combined meta-analyses used fixed-effects models. RESULTS: We replicated 15 of 21 BMI loci included on the Metabochip, and identified two novel BMI loci at 1q41 (rs2820436) and 2q31.1 (rs10930502) at the Metabochip-wide significance threshold (P<2.5 × 10-7). Bioinformatic functional investigation of SNPs at these loci suggests a possible impact on pathways that regulate metabolism and adipose tissue. CONCLUSION: Conducting studies in genetically diverse populations continues to be a valuable strategy for replicating known loci and uncovering novel BMI associations.


Subject(s)
Body Mass Index , Racial Groups/genetics , Racial Groups/statistics & numerical data , Genome-Wide Association Study , Genomics , Humans , Polymorphism, Single Nucleotide/genetics
2.
Clin Pharmacol Ther ; 100(5): 427-430, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27393744

ABSTRACT

Implementation of pharmacogenetic-guided warfarin dosing has been hindered by inconsistent results from reported clinical trials and a lack of available algorithms that include alleles prevalent in non-white populations. However, current evidence indicates that algorithm-guided dosing is more accurate than empirical dosing. To facilitate multiethnic algorithm-guided warfarin dosing using preemptive genetic testing, we developed a strategy that accounts for the complexity of race and leverages electronic health records for algorithm variables and deploying point-of-care dose recommendations.


Subject(s)
Algorithms , Cytochrome P-450 CYP2C9/genetics , Electronic Health Records , Genetic Testing , Vitamin K Epoxide Reductases/genetics , Warfarin/administration & dosage , Anticoagulants/administration & dosage , Dose-Response Relationship, Drug , Drug Dosage Calculations , Ethnicity , Humans , Pharmacogenetics/methods , Polymorphism, Genetic
3.
Contemp Clin Trials ; 47: 101-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26747051

ABSTRACT

People of African ancestry (Blacks) have increased risk of kidney failure due to numerous socioeconomic, environmental, and clinical factors. Two variants in the APOL1 gene are now thought to account for much of the racial disparity associated with hypertensive kidney failure in Blacks. However, this knowledge has not been translated into clinical care to help improve patient outcomes and address disparities. GUARDD is a randomized trial to evaluate the effects and challenges of incorporating genetic risk information into primary care. Hypertensive, non-diabetic, adults with self-reported African ancestry, without kidney dysfunction, are recruited from diverse clinical settings and randomized to undergo APOL1 genetic testing at baseline (intervention) or at one year (waitlist control). Providers are educated about genomics and APOL1. Guided by a genetic counselor, trained staff return APOL1 results to patients and provide low-literacy educational materials. Real-time clinical decision support tools alert clinicians of their patients' APOL1 results and associated risk status at the point of care. Our academic-community-clinical partnership designed a study to generate information about the impact of genetic risk information on patient care (blood pressure and renal surveillance) and on patient and provider knowledge, attitudes, beliefs, and behaviors. GUARDD will help establish the effective implementation of APOL1 risk-informed management of hypertensive patients at high risk of CKD, and will provide a robust framework for future endeavors to implement genomic medicine in diverse clinical practices. It will also add to the important dialog about factors that contribute to and may help eliminate racial disparities in kidney disease.


Subject(s)
Apolipoproteins/genetics , Black or African American/genetics , Genetic Testing/methods , Hypertension/genetics , Lipoproteins, HDL/genetics , Primary Health Care/methods , Renal Insufficiency, Chronic/genetics , Adolescent , Adult , Aged , Apolipoprotein L1 , Decision Support Techniques , Genetic Counseling/methods , Genetic Predisposition to Disease , Humans , Middle Aged , Polymorphism, Genetic , Risk Assessment , Young Adult
4.
Pharmacogenomics J ; 16(3): 231-7, 2016 06.
Article in English | MEDLINE | ID: mdl-26169577

ABSTRACT

The most common side effect of angiotensin-converting enzyme inhibitor (ACEi) drugs is cough. We conducted a genome-wide association study (GWAS) of ACEi-induced cough among 7080 subjects of diverse ancestries in the Electronic Medical Records and Genomics (eMERGE) network. Cases were subjects diagnosed with ACEi-induced cough. Controls were subjects with at least 6 months of ACEi use and no cough. A GWAS (1595 cases and 5485 controls) identified associations on chromosome 4 in an intron of KCNIP4. The strongest association was at rs145489027 (minor allele frequency=0.33, odds ratio (OR)=1.3 (95% confidence interval (CI): 1.2-1.4), P=1.0 × 10(-8)). Replication for six single-nucleotide polymorphisms (SNPs) in KCNIP4 was tested in a second eMERGE population (n=926) and in the Genetics of Diabetes Audit and Research in Tayside, Scotland (GoDARTS) cohort (n=4309). Replication was observed at rs7675300 (OR=1.32 (1.01-1.70), P=0.04) in eMERGE and at rs16870989 and rs1495509 (OR=1.15 (1.01-1.30), P=0.03 for both) in GoDARTS. The combined association at rs1495509 was significant (OR=1.23 (1.15-1.32), P=1.9 × 10(-9)). These results indicate that SNPs in KCNIP4 may modulate ACEi-induced cough risk.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/adverse effects , Cough/chemically induced , Cough/genetics , Kv Channel-Interacting Proteins/genetics , Polymorphism, Single Nucleotide , Case-Control Studies , Computational Biology , Cough/ethnology , Databases, Genetic , Electronic Health Records , Female , Gene Frequency , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Logistic Models , Male , Multivariate Analysis , Odds Ratio , Phenotype , Risk Assessment , Risk Factors , Scotland , United States
6.
J Clin Invest ; 108(6): 807-16, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11560950

ABSTRACT

Primary and secondary forms of focal segmental glomerulosclerosis (FSGS) are characterized by depletion of podocytes and constitute a central manifestation of chronic progressive glomerular diseases. Here we report that podocytes undergo apoptosis at early stages in the course of progressive glomerulosclerosis in TGF-beta1 transgenic mice. Apoptosis is associated with progressive depletion of podocytes and precedes mesangial expansion. Smad7 protein expression is strongly induced specifically in damaged podocytes of transgenic mice and in cultured murine podocytes treated with TGF-beta. TGF-beta1 and Smad7 each induce apoptosis in podocytes, and their coexpression has an additive effect. Activation of p38 MAP kinase and caspase-3 is required for TGF-beta-mediated apoptosis, but not for apoptosis induced by Smad7. Unlike TGF-beta, Smad7 inhibits nuclear translocation and transcriptional activity of the cell survival factor NF-kappaB. Our results suggest a novel functional role for Smad7 as amplifier of TGF-beta-induced apoptosis in podocytes and a new pathomechanism for podocyte depletion in progressive glomerulosclerosis.


Subject(s)
Apoptosis/drug effects , DNA-Binding Proteins/pharmacology , Kidney Glomerulus/cytology , Kidney Glomerulus/drug effects , Trans-Activators/pharmacology , Transforming Growth Factor beta/pharmacology , Animals , Caspase 3 , Caspases/metabolism , Cells, Cultured , Disease Models, Animal , Glomerular Mesangium/pathology , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Smad7 Protein , Transforming Growth Factor beta/genetics , p38 Mitogen-Activated Protein Kinases
7.
Chest ; 120(1 Suppl): 43S-47S, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11451911

ABSTRACT

Transforming growth factor (TGF)-beta plays a central role in fibrosis, contributing both to the influx and activation of inflammatory cells, as well as to activation of fibroblasts to elaborate extracellular matrix. In the past few years, new insight has been gained into signal transduction pathways downstream of the TGF-beta receptor serine-threonine kinases with the identification of a family of evolutionarily conserved Smad proteins. Two receptor-activated Smad proteins, Smad2 and Smad3, are phosphorylated by the activated TGF-beta type I receptor kinase, after which they partner with the common mediator, Smad4, and are translocated to the nucleus to where they participate in transcriptional complexes to control expression of target genes. We have shown in wound healing studies of mice null for Smad3, that loss of this key signaling intermediate interferes with the chemotaxis of inflammatory cells to TGF-beta as well as with their ability to autoinduce TGF-beta. Moreover, studies with mouse embryo fibroblasts null for Smad3 show that TGF-beta-dependent induction of c-Jun and c-Fos, important in induction of collagen as well as in autoinduction of TGF-beta, is mediated by Smad3. Based on these observations, we hypothesize that loss of Smad3 will confer resistance to fibrosis and result in reduced inflammatory cell infiltrates, reduced autoinduction of TGF-beta, important to sustain the process, and reduced elaboration of collagen. Preliminary observations in a model of radiation-induced fibrosis confirm this hypothesis and suggest that inhibitors of Smad3 might have clinical application both to improve wound healing and to reduce fibrosis.


Subject(s)
DNA-Binding Proteins/physiology , Pulmonary Fibrosis/physiopathology , Signal Transduction/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/physiology , Wound Healing/physiology , Humans , MAP Kinase Signaling System/physiology , Phosphorylation , Smad3 Protein
8.
J Mammary Gland Biol Neoplasia ; 6(1): 67-82, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11467453

ABSTRACT

Ligands of the TGF-beta superfamily are unique in that they signal through transmembrane receptor serine-threonine kinases, rather than tyrosine kinases. The receptor complex couples to a signal transduction pathway involving a novel family of proteins, the Smads. On phosphorylation, Smads translocate to the nucleus where they modulate transcriptional responses. However, TGF-betas can also activate the mitogen-activated protein kinase (MAPK)4 pathway, and the different biological responses to TGF-beta depend to varying degrees on activation of either or both of these two pathways. The Smad pathway is a nexus for cross-talk with other signal transduction pathways and for modulation by many different interacting proteins. Despite compelling evidence that TGF-beta has tumor suppressor activity in the mammary gland, neither TGF-beta receptors nor Smads are genetically inactivated in human breast cancer, though receptor expression is reduced. Possible reasons are discussed in relation to the dual role of TGF-beta as tumor suppressor and oncogene.


Subject(s)
Breast/growth & development , Mammary Neoplasms, Animal/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Female , Humans , Transforming Growth Factor beta/genetics
9.
Proc Natl Acad Sci U S A ; 98(12): 6686-91, 2001 Jun 05.
Article in English | MEDLINE | ID: mdl-11390996

ABSTRACT

Epithelial-mesenchymal transitions (EMTs) are an essential manifestation of epithelial cell plasticity during morphogenesis, wound healing, and tumor progression. Transforming growth factor-beta (TGF-beta) modulates epithelial plasticity in these physiological contexts by inducing EMT. Here we report a transcriptome screen of genetic programs of TGF-beta-induced EMT in human keratinocytes and propose functional roles for extracellular response kinase (ERK) mitogen-activated protein kinase signaling in cell motility and disruption of adherens junctions. We used DNA arrays of 16,580 human cDNAs to identify 728 known genes regulated by TGF-beta within 4 hours after treatment. TGF-beta-stimulated ERK signaling mediated regulation of 80 target genes not previously associated with this pathway. This subset is enriched for genes with defined roles in cell-matrix interactions, cell motility, and endocytosis. ERK-independent genetic programs underlying the onset of EMT involve key pathways and regulators of epithelial dedifferentiation, undifferentiated transitional and mesenchymal progenitor phenotypes, and mediators of cytoskeletal reorganization. The gene expression profiling approach delineates complex context-dependent signaling pathways and transcriptional events that determine epithelial cell plasticity controlled by TGF-beta. Investigation of the identified pathways and genes will advance the understanding of molecular mechanisms that underlie tumor invasiveness and metastasis.


Subject(s)
Transcription, Genetic/drug effects , Transforming Growth Factor beta/pharmacology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Cytoskeleton/physiology , Epithelial Cells/physiology , Gene Expression Regulation/drug effects , Humans , Integrins/physiology , Keratinocytes/physiology , Mitogen-Activated Protein Kinases/physiology
10.
J Biol Chem ; 276(23): 19945-53, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11262418

ABSTRACT

A prominent pathway of transforming growth factor (TGF)-beta signaling involves receptor-dependent phosphorylation of Smad2 and Smad3, which then translocate to the nucleus to activate transcription of target genes. To investigate the relative importance of these two Smad proteins in TGF-beta1 signal transduction, we have utilized a loss of function approach, based on analysis of the effects of TGF-beta1 on fibroblasts derived from mouse embryos deficient in Smad2 (S2KO) or Smad3 (S3KO). TGF-beta1 caused 50% inhibition of cellular proliferation in wild-type fibroblasts as assessed by [(3)H]thymidine incorporation, whereas the growth of S2KO or S3KO cells was only weakly inhibited by TGF-beta1. Lack of Smad2 or Smad3 expression did not affect TGF-beta1-induced fibronectin synthesis but resulted in markedly suppressed induction of plasminogen activator inhibitor-1 by TGF-beta1. Moreover, TGF-beta1-mediated induction of matrix metalloproteinase-2 was selectively dependent on Smad2, whereas induction of c-fos, Smad7, and TGF-beta1 autoinduction relied on expression of Smad3. Investigation of transcriptional activation of TGF-beta-sensitive reporter genes in the different fibroblasts showed that activation of the (Smad binding element)(4)-Lux reporter by TGF-beta1 was dependent on expression of Smad3, but not Smad2, whereas activation of the activin response element-Lux reporter was strongly suppressed in S2KO fibroblasts but, on the contrary, enhanced in S3KO cells. Our findings indicate specific roles for Smad2 and Smad3 in TGF-beta1 signaling.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins , Animals , Cell Division , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA-Binding Proteins/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Genes, Immediate-Early , Genes, Reporter , Genes, fos , Mice , Mice, Knockout , Smad2 Protein , Smad3 Protein , Trans-Activators/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transforming Growth Factor beta/biosynthesis
11.
J Biol Chem ; 276(9): 6727-38, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11102446

ABSTRACT

Transforming growth factor-beta (TGF-beta) signaling proceeds from the cell membrane to the nucleus through the cooperation of the type I and II serine/threonine kinase receptors and their downstream SMAD effectors. Although various regulatory proteins affecting TGF-beta-mediated events have been described, relatively little is known about receptor interactions at the level of the plasma membrane. Caveolae are cholesterol-rich membrane microdomains that, along with their marker protein caveolin-1 (Cav-1), have been implicated in the compartmentalization and regulation of certain signaling events. Here, we demonstrate that specific components of the TGF-beta cascade are associated with caveolin-1 in caveolae and that Cav-1 interacts with the Type I TGF-beta receptor. Additionally, Cav-1 is able to suppress TGF-beta-mediated phosphorylation of Smad-2 and subsequent downstream events. We localize the Type I TGF-beta receptor interaction to the scaffolding domain of Cav-1 and show that it occurs in a physiologically relevant time frame, acting to rapidly dampen signaling initiated by the TGF-beta receptor complex.


Subject(s)
Caveolins/physiology , DNA-Binding Proteins/physiology , Receptors, Transforming Growth Factor beta/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/physiology , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites , Caveolin 1 , Cell Differentiation , DNA-Binding Proteins/chemistry , Mice , Molecular Sequence Data , Phosphorylation , Receptors, Transforming Growth Factor beta/analysis , Smad2 Protein , Tacrolimus Binding Protein 1A/physiology , Trans-Activators/chemistry
12.
Kidney Int Suppl ; 77: S45-52, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10997690

ABSTRACT

It is now generally accepted that transforming growth factor-beta (TGF-beta) has an important role in the pathogenesis of both acute and chronic forms of renal disease. Although TGF-beta's potent fibrogenic activity is considered a major factor in chronic progression of renal disease, this cytokine participates in the control of several fundamental cellular responses in the kidney including inflammation, programmed cell death, cell growth, cell differentiation, and cellular hypertrophy. Recent identification of Smad proteins as intracellular mediators of TGF-beta signaling has provided important insights into mechanisms that may determine the specificity of TGF-beta action in different renal and inflammatory cells. Thus, Smads are characterized by an astonishingly complex array of molecular and functional interactions with other signaling pathways. These emerging patterns of signaling cross talk involving Smad proteins suggest a dynamic profile of positive or negative transmodulation of TGF-beta signaling, depending on the cellular context. Understanding the interplay between these signaling cascades is an important field of investigation that will ultimately reveal new targets for precise and selective modulation of TGF-beta's diverse actions in renal diseases.


Subject(s)
DNA-Binding Proteins/physiology , Signal Transduction , Trans-Activators/physiology , Transforming Growth Factor beta/physiology , Animals , Cytoskeleton/physiology , Humans , Kidney/physiology , Smad2 Protein , Smad3 Protein , Smad4 Protein , Smad7 Protein , Transcription, Genetic
13.
J Cell Sci ; 113 ( Pt 13): 2355-61, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10852815

ABSTRACT

Transforming growth factor-(beta) (TGF(beta)) has a wide range of activities on vascular cells and inflammatory cells, suggesting it may have different functions during various stages of atherogenesis. We report that mice heterozygous for the deletion of the tgfb1 gene (tgfb1(+/-) mice) have reduced levels of TGF(beta)1 in the artery wall until at least 8 weeks of age. On a normal mouse chow diet, the vascular endothelium of tgfb1(+/-) mice is indistinguishable from wild-type littermates, assessed by morphology and intercellular cell adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) expression. In contrast, levels of the smooth muscle isoforms of actin and myosin in medial smooth muscle cells of tgfb1(+/-) mice are significantly reduced. Following feeding a cholesterol-enriched diet for 12 weeks, high levels of ICAM-1 and VCAM-1 were detected in the vascular endothelial cells of tgfb1(+/-) mice, but not wild-type mice. Furthermore, marked deposition of lipid into the artery wall was only observed in the tgfb1(+/-) mice on the cholesterol-enriched diet. These vascular lipid lesions were accompanied by local invasion of macrophages. We conclude that deletion of a single allele of the tgfb1 gene results in a reduced level of TGFbeta1 antigen in the aorta together with reduced smooth muscle cell differentiation, whereas the addition of a high fat dietary challenge is required to activate the vascular endothelium and to promote the formation of fatty streaks resembling early atherosclerosis in humans.


Subject(s)
Arteriosclerosis/etiology , Arteriosclerosis/physiopathology , Dietary Fats/adverse effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Lipid Metabolism , Muscle, Smooth, Vascular/metabolism , Transforming Growth Factor beta/deficiency , Animals , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Arteriosclerosis/pathology , Endothelium, Vascular/pathology , Inflammation/pathology , Inflammation/physiopathology , Lipoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology
14.
Genes Dev ; 14(2): 187-97, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10652273

ABSTRACT

A number of pathogenic and proinflammatory stimuli, and the transforming growth factor-beta (TGF-beta) exert opposing activities in cellular and immune responses. Here we show that the RelA subunit of nuclear factor kappaB (NF-kappaB/RelA) is necessary for the inhibition of TGF-beta-induced phosphorylation, nuclear translocation, and DNA binding of SMAD signaling complexes by tumor necrosis factor-alpha (TNF-alpha). The antagonism is mediated through up-regulation of Smad7 synthesis and induction of stable associations between ligand-activated TGF-beta receptors and inhibitory Smad7. Down-regulation of endogenous Smad7 by expression of antisense mRNA releases TGF-beta/SMAD-induced transcriptional responses from suppression by cytokine-activated NF-kappaB/RelA. Following stimulation with bacterial lipopolysaccharide (LPS), or the proinflammatory cytokines TNF-alpha and interleukin-1beta (IL-1beta, NF-kappaB/RelA induces Smad7 synthesis through activation of Smad7 gene transcription. These results suggest a mechanism of suppression of TGF-beta/SMAD signaling by opposing stimuli mediated through the activation of inhibitory Smad7 by NF-kappaB/RelA.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/physiology , Ligases/physiology , NF-kappa B/physiology , Signal Transduction/physiology , Trans-Activators/antagonists & inhibitors , Trans-Activators/physiology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/physiology , 3T3 Cells , Animals , COS Cells , DNA-Binding Proteins/genetics , Ligases/metabolism , Mice , Mice, Mutant Strains , NF-kappa B/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Smad7 Protein , Trans-Activators/genetics , Transcription, Genetic , Tumor Necrosis Factor-alpha/physiology
15.
J Biol Chem ; 275(15): 11320-6, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10753944

ABSTRACT

Smad7 is an inducible intracellular inhibitor of transforming growth factor-beta (TGF-beta) signaling that is regulated by diverse stimuli including members of the TGF-beta superfamily. To define the molecular mechanisms of negative control of TGF-beta signaling, we have isolated the human SMAD7 gene and characterized its promoter region. A -303 to +672 SMAD7 region contained a palindromic GTCTAGAC Smad binding element (SBE) between nucleotides -179 and -172 that was necessary for the induction of a Smad7 promoter luciferase reporter gene by TGF-beta. Electrophoretic mobility shift assays using oligonucleotide probes demonstrated that TGF-beta rapidly induced the binding of an endogenous SBE-binding complex (SBC) containing Smad2, Smad3, and Smad4. Transfection assays in mouse embryonic fibroblasts (MEFs), with targeted deletions of either Smad2 or Smad3, and the Smad4-deficient cell line MD-MBA-468 revealed that both Smad3 and Smad4, but not Smad2, were absolutely required for induction of the Smad7 promoter reporter gene by TGF-beta. Furthermore, the TGF-beta-inducible SBE-binding complex was diminished in Smad2-deficient MEFs when compared with wild type MEFs and not detectable in Smad3-deficient MEFs and MD-MBA-468 cells. Taken together, our data demonstrate that TGF-beta induces transcription of the human SMAD7 gene through activation of Smad3 and Smad4 transcription factor binding to its proximal promoter.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Promoter Regions, Genetic , Trans-Activators/genetics , Trans-Activators/physiology , Transforming Growth Factor beta/pharmacology , 3T3 Cells , Animals , Base Sequence , Cell Line , DNA/metabolism , Humans , Mice , Molecular Sequence Data , Smad3 Protein , Smad4 Protein , Smad7 Protein
16.
Cancer Res ; 59(19): 4834-42, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10519393

ABSTRACT

Transforming growth factor (TGF)-betas are multifunctional growth factors, the properties of which include the potent inhibition of epithelial cell growth. Expression patterns of TGF-betas and TGF-beta receptors in the normal prostate indicate that these growth regulators play key roles in prostatic development and proliferative homeostasis. Importantly, TGF-beta receptor levels are frequently diminished in malignant human prostate tissue. To test the hypothesis that loss of TGF-beta responsiveness is causally involved in the tumorigenic process, we have used retroviral transduction to introduce a dominant-negative mutant type II TGF-beta receptor (DNR) into the premalignant rat prostatic epithelial cell line, NRP-152. High-level expression of the DNR abolished the ability of TGF-beta to inhibit cell growth, to promote cell differentiation, and to induce apoptosis, and it partially blocked the induction of extracellular matrix gene expression. When injected into nude mice, NRP-152-DNR cells formed carcinomas at 13 of 34 sites, compared with 0 of 30 sites for parental and control cells (P = 0.0001). We conclude that the type II TGF-beta receptor is an important tumor suppressor in the prostate, and furthermore, that loss of TGF-beta responsiveness can contribute early in the tumorigenic process by causing the malignant transformation of preneoplastic cells.


Subject(s)
Cell Transformation, Neoplastic , Extracellular Matrix Proteins/genetics , Gene Expression Regulation/drug effects , Prostatic Neoplasms/pathology , Receptors, Transforming Growth Factor beta/physiology , Transforming Growth Factor beta/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Epithelial Cells , Humans , Male , Mice , Mice, Nude , Prostate , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases , Rats , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Recombinant Proteins/metabolism , Transfection , Transplantation, Heterologous
17.
J Am Soc Nephrol ; 10(2): 271-80, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10215326

ABSTRACT

Renal pathology in mice that are transgenic for the murine albumin enhancer/promoter linked to a full-length porcine transforming growth factor-beta1 (TGF-beta1) gene has been described previously. In these mice, transgene expression is limited to the liver and the plasma level of TGF-beta is increased. The earliest renal pathologic change is glomerulosclerosis, at 3 wk of age, and this is followed by tubulointerstitial fibrosis. In this study, it was hypothesized that circulating TGF-beta1 increases renal extracellular matrix accumulation and activates local TGF-beta gene expression. Immunostaining at 5 wk revealed increased amounts of collagen I and III within the mesangium, glomerular capillary loops, and interstitium, while the amount of collagen IV was normal. Similarly, Northern analysis showed increased expression of mRNA encoding collagen I and III, as well as biglycan and decorin, while the expression of collagen IV was unchanged. These changes began as early as 1 wk of age, a time before the appearance of glomerulosclerosis. To evaluate matrix degradation, collagenase IV activity was evaluated by gelatin zymography and an increase in matrix metalloproteinase-2 was found. Finally, the production of tissue inhibitors of metalloproteinase was evaluated. Tissue inhibitor of metalloproteinase-1 (TIMP-1) mRNA was increased 18-fold, while TIMP-2 and TIMP-3 were unchanged. In 2-wk-old transgenic kidney, local expression of TGF-beta1, beta2, and beta3 protein was similar to wild-type mice. In 5-wk-old transgenic mice, TGF-beta1 and beta2 protein was present in increased amounts within glomeruli, and renal TGF-beta1 mRNA was increased threefold. It is concluded that elevated levels of circulating TGF-beta1 may act on the kidney to increase matrix protein production and decrease matrix remodeling. Only after glomerulosclerosis is established does local glomerular overproduction of TGF-beta become manifest.


Subject(s)
Extracellular Matrix Proteins/metabolism , Kidney/metabolism , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Animals , Collagen/genetics , Collagen/metabolism , Fibrosis , Gene Expression/physiology , Isomerism , Kidney/pathology , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Transgenic/anatomy & histology , Proteoglycans/genetics , RNA, Messenger/metabolism , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism
19.
Kidney Blood Press Res ; 21(1): 1-12, 1998.
Article in English | MEDLINE | ID: mdl-9661131

ABSTRACT

An extensive number of animal and clinical studies indicate that transforming growth factor-beta (TGF-beta s) play an important role in inflammatory and fibrotic diseases, including renal fibrosis. Recent mouse models harboring genetically engineered alterations in TGF-beta pathways reveal complicated mechanisms of regulation of TGF-beta activity in vivo. The purpose of this review is to present recent advances relevant to our understanding of the TGF-beta-signaling system in renal physiology and pathophysiology.


Subject(s)
Kidney Diseases/metabolism , Transforming Growth Factor beta/metabolism , Animals , Disease Models, Animal , Fibrosis/metabolism , Humans , Mice , Mice, Transgenic , Receptors, Transforming Growth Factor beta/metabolism
20.
Nat Med ; 4(7): 802-7, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9662371

ABSTRACT

Components of the transforming growth factor-beta (TGF-beta) signal pathway function as classic tumor suppressors, but the role of the TGF-betas themselves is less clear. Here we show that mice heterozygous for deletion of the TGF-beta1 gene express only 10-30% of wild-type TGF-beta1 protein levels. Although grossly normal, these mice have a subtly altered proliferative phenotype, with increased cell turnover in the liver and lung. Treatment of these mice with chemical carcinogens resulted in enhanced tumorigenesis when compared with wild-type littermates. However, tumors in the heterozygous mice did not lose the remaining wild-type TGF-beta1 allele, indicating that the TGF-beta1 ligand is a new form of tumor suppressor that shows true haploid insufficiency in its ability to protect against tumorigenesis.


Subject(s)
Genes, Tumor Suppressor , Transforming Growth Factor beta/genetics , Animals , Apoptosis , Carcinogenicity Tests , Cell Cycle Proteins/genetics , Cell Division , Gene Targeting , Liver/cytology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...