Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 330
Filter
1.
Biol Reprod ; 78(6): 994-1001, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18322278

ABSTRACT

Amniotes, regardless of genetic sex, develop two sets of genital ducts: the Wolffian and Müllerian ducts. For normal sexual development to occur, one duct must differentiate into its corresponding organs, and the other must regress. In mammals, the Wolffian duct differentiates into the male reproductive tract, mainly the vasa deferentia, epididymides, and seminal vesicles, whereas the Müllerian duct develops into the four components of the female reproductive tract, the oviducts, uterus, cervix, and upper third of the vagina. In males, the fetal Leydig cells produce testosterone, which stimulates the differentiation of the Wolffian duct, whereas the Sertoli cells of the fetal testes express anti-Müllerian hormone, which activates the regression of the Müllerian duct. Anti-Müllerian hormone is a member of the transforming growth factor-beta (TGF-beta) family of secreted signaling molecules and has been shown to signal through the BMP pathway. It binds to its type II receptor, anti-Müllerian hormone receptor 2 (AMHR2), in the Müllerian duct mesenchyme and through an unknown mechanism(s); the mesenchyme induces the regression of the Müllerian duct mesoepithelium. Using tissue-specific gene inactivation with an Amhr2-Cre allele, we have determined that two TGF-beta type I receptors (Acvr1 and Bmpr1a) and all three BMP receptor-Smads (Smad1, Smad5, and Smad8) function redundantly in transducing the anti-Müllerian hormone signal required for Müllerian duct regression. Loss of these genes in the Müllerian duct mesenchyme results in male infertility due to retention of Müllerian duct derivatives in an otherwise virilized male.


Subject(s)
Activin Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Mullerian Ducts/embryology , Mullerian Ducts/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Activin Receptors, Type I/deficiency , Activin Receptors, Type I/genetics , Animals , Anti-Mullerian Hormone/pharmacology , Bone Morphogenetic Protein Receptors, Type I/deficiency , Bone Morphogenetic Protein Receptors, Type I/genetics , Female , Infertility, Male/embryology , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Models, Biological , Mullerian Ducts/drug effects , Pregnancy , Signal Transduction , Smad1 Protein/genetics , Smad5 Protein/genetics , Smad8 Protein/genetics
2.
Oncogene ; 27(17): 2507-12, 2008 Apr 10.
Article in English | MEDLINE | ID: mdl-17952112

ABSTRACT

The ability of transforming growth factor-beta (TGF-beta) to modulate various effects on distinct cell lineages has been a central feature of its multi-faceted nature. The purpose of this study was to access the effects of deletion of a key TGF-beta signal transducer, Smad3, on MAPK activation and v-Ras(Ha)-transformation of primary mouse embryonic fibroblasts (MEFs). We observe reduced TGF-beta1 and v-ras(Ha) mediated activation of the JNK and ERK MAPK pathway upon ablation of Smad3. Further, Smad3-deficient MEFs demonstrate resistance to v-ras(Ha)-induced transformation while the absence of Smad3 results in increased inhibition of farnesyl transferase activity. Taken together, these observations demonstrate that the absence of Smad3 protects fibroblasts from oncogenic transformation by (i) augmenting farnesyl transferase inhibition and (ii) suppressing the Ras-JNK MAPK pathway. These results provide new insights into the molecular mechanisms involved in v-Ras(Ha) oncogene-induced mesenchymal phenotypic transformation.


Subject(s)
Alkyl and Aryl Transferases/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Oncogene Protein p21(ras)/metabolism , Smad3 Protein/metabolism , Transgenes/genetics , Animals , Cell Line, Transformed , Cell Transformation, Neoplastic , Cells, Cultured , Chlorocebus aethiops , Enzyme Activation/drug effects , Gene Expression Regulation , MAP Kinase Signaling System/drug effects , Mice , Oncogene Protein p21(ras)/genetics , Phenotype , Smad3 Protein/deficiency , Smad3 Protein/genetics , Transforming Growth Factor beta/pharmacology
3.
Sci STKE ; 2001(113): pe43, 2001 Dec 18.
Article in English | MEDLINE | ID: mdl-11752631

ABSTRACT

The transforming growth factor-beta (TGF-beta) superfamily of signaling molecules regulates many developmental processes in a range of organisms from worms to humans. Understanding the mechanisms by which they exert their repertoire of effects has required identification of the components of signaling pathways that they control. Roberts and Derynck focus on this aspect of TGF-beta biology in their review of a recent Federation of American Societies for Experimental Biology (FASEB) meeting on TGF-beta signaling and development and summarize current signaling paradigms and future prospects in TGF-beta signaling from the cell surface to the nucleus.


Subject(s)
Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Animals , DNA-Binding Proteins/physiology , Drosophila melanogaster , Humans , Ligands , Mice , Trans-Activators/physiology , Transcriptional Activation/physiology , Xenopus laevis
4.
Adv Cancer Res ; 83: 1-54, 2001.
Article in English | MEDLINE | ID: mdl-11665716

ABSTRACT

Transforming growth factor-beta (TGF-beta) has been implicated in oncogenesis since the time of its discovery almost 20 years ago. The complex, multifunctional activities of TGF-beta endow it with both tumor suppressor and tumor promoting activities, depending on the stage of carcinogenesis and the responsivity of the tumor cell. Dysregulation or alteration of TGF-beta signaling in tumorigenesis can occur at many different levels, including activation of the ligand, mutation or transcriptional suppression of the receptors, or alteration of downstream signal transduction pathways resulting from mutation or changes in expression patterns of signaling intermediates or from changes in expression of other proteins which modulate signaling. New insights into signaling from the TGF-beta receptors, including the identification of Smad signaling pathways and their interaction with mitogen-activated protein (MAP) kinase pathways, are providing an understanding of the changes involved in the change from tumor suppressor to tumor promoting activities of TGF-beta. It is now appreciated that loss of sensitivity to inhibition of growth by TGF-beta by most tumor cells is not synonymous with complete loss of TGF-beta signaling but rather suggests that tumor cells gain advantage by selective inactivation of the tumor suppressor activities of TGF-beta with retention of its tumor promoting activities, especially those dependent on cross talk with MAP kinase pathways and AP-1.


Subject(s)
Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology , Animals , Cell Differentiation , Genes, Tumor Suppressor , Humans , Models, Biological , Mutation , Protein Isoforms , Signal Transduction , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/genetics
5.
J Biol Chem ; 276(49): 46297-304, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11567019

ABSTRACT

SNIP1 is a 396-amino acid nuclear protein shown to be an inhibitor of the TGF-beta signal transduction pathway and to be important in suppressing transcriptional activation dependent on the co-activators CBP and p300. In this report we show that SNIP1 potently inhibits the activity of NF-kappa B, which binds the C/H1 domain of CBP/p300, but does not interfere with the activity of transcription factors such as p53, which bind to other domains of p300, or factors such as VP16, which are independent of these co-activators. Inhibition of NF-kappa B activity is a function of the N-terminal domain of SNIP1 and involves competition of SNIP1 and the NF-kappa B subunit, RelA/p65, for binding to p300, similar to the mechanism of inhibition of Smad signaling by SNIP1. Immunohistochemical staining shows that expression of SNIP1 is strictly regulated in development and that it colocalizes, in certain tissues, with nuclear staining for RelA/p65 and for p300, suggesting that they may regulate NF-kappa B activity in vivo in a spatially and temporally controlled manner. These data led us to suggest that SNIP1 may be an inhibitor of multiple transcriptional pathways that require the C/H1 domain of CBP/p300.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , NF-kappa B/antagonists & inhibitors , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Animals , Binding, Competitive , Carrier Proteins/physiology , Cell Line , E1A-Associated p300 Protein , Embryonic and Fetal Development/physiology , Gene Expression Regulation/physiology , Humans , Mice , NF-kappa B/chemistry , NF-kappa B/metabolism , Nuclear Proteins/chemistry , RNA-Binding Proteins , Repressor Proteins/physiology , Trans-Activators/chemistry , Transcriptional Activation
6.
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
7.
Oncogene ; 20(26): 3332-40, 2001 Jun 07.
Article in English | MEDLINE | ID: mdl-11423983

ABSTRACT

Smad proteins transduce signals from TGF-beta receptors and regulate transcription of target genes either directly or in combination with other sequence-specific transcription factors. AP-1 sites and their cognate transcription factors also play important roles in the gene regulatory activities of TGF-beta. In this report, we have investigated the functional interactions of the Smad and AP-1 transcription factors. We demonstrate that Smad and AP-1 complexes specifically bind to their cognate cis-elements and do not interact with each other on-DNA, whereas off-DNA interactions occur between Smad3 and both c-Jun and JunB. Using both artificial constructs specific for either the Smad or AP-1 signaling pathways or natural promoters known to be TGF-beta-responsive, we have determined that Jun family members downregulate Smad3-mediated gene transactivation whereas AP-1-dependent promoters are synergistically activated by Smad3 and Jun proteins. We propose a model where the presence of Smad- and/or AP-1-specific cis-elements within TGF-beta-responsive genes allows dynamic modulation of gene expression, in contrast to the existing model where interactions between Smad and AP-1 proteins are merely an on/off mechanism to regulate TGF-beta/Smad targets.


Subject(s)
Activin Receptors, Type I , DNA-Binding Proteins/metabolism , Promoter Regions, Genetic/drug effects , Protein Serine-Threonine Kinases/drug effects , Receptors, Transforming Growth Factor beta/drug effects , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Base Sequence , COS Cells , Cells, Cultured , Chlorocebus aethiops , Consensus Sequence , DNA/metabolism , Fibroblasts/cytology , Humans , Macromolecular Substances , Molecular Sequence Data , Oligodeoxyribonucleotides/metabolism , Oligopeptides , Peptides/immunology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-jun/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/physiology , Regulatory Sequences, Nucleic Acid , Signal Transduction/drug effects , Smad3 Protein , Substrate Specificity , Transcriptional Activation , Transfection
8.
J Biol Chem ; 276(28): 26542-9, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11356828

ABSTRACT

X-linked inhibitor of apoptosis protein (XIAP) is a potent suppressor of apoptotic cell death, which functions by directly inhibiting caspases, the principal effectors of apoptosis. Here we report that XIAP can also function as a cofactor in the regulation of gene expression by transforming growth factor-beta (TGF-beta). XIAP, but not the related proteins c-IAP1 or c-IAP2, associated with several members of the type I class of the TGF-beta receptor superfamily and potentiated TGF-beta-induced signaling. Although XIAP-mediated activation of c-Jun N-terminal kinase and nuclear factor kappa B was found to require the TGF-beta signaling intermediate Smad4, the ability of XIAP to suppress apoptosis was found to be Smad4-independent. These data implicate a role for XIAP in TGF-beta-mediated signaling that is distinct from its anti-apoptotic functions.


Subject(s)
Proteins/physiology , Transforming Growth Factor beta/physiology , Apoptosis/physiology , Cell Line , Gene Expression Regulation/physiology , Humans , Signal Transduction , X-Linked Inhibitor of Apoptosis Protein
9.
Am J Obstet Gynecol ; 184(6): 1251-5, 2001 May.
Article in English | MEDLINE | ID: mdl-11349197

ABSTRACT

OBJECTIVE: This study was undertaken to audit ultrasonographic measurements of fetal liver length and middle cerebral artery peak velocity in cases of red blood cell alloimmunization between 1986 and 1999. STUDY DESIGN: A total of 200 fetuses at risk for anemia because of red blood cell alloimmunization underwent ultrasonographic measurement of the length of the right lobe of the liver, 45 underwent Doppler recording of middle cerebral artery peak velocity, and 119 underwent fetal blood sampling. RESULTS: The overall survival was 188 of 200 (94%). Among 69 fetuses found to have anemia, liver length values in 64 (93%) were at the 95th percentile or greater, and the other 5 were in the upper part of the normal range. The middle cerebral artery peak velocity was > or =95th percentile in 15 of the 19 cases of anemia in which this value was measured (79%). Among those measured within 1 week of birth, all liver lengths were at least in the upper part of the normal range, with most >95th percentile, including 1 case with a cord blood hemoglobin concentration <90 g/L. CONCLUSIONS: All fetuses with anemia identified at fetal blood sampling had enlarged livers with 93% at > or =95th percentile. The peak velocity in the middle cerebral artery was abnormal in most fetuses with anemia.


Subject(s)
Cerebral Arteries/diagnostic imaging , Cerebral Arteries/embryology , Liver/diagnostic imaging , Liver/embryology , Rh Isoimmunization , Ultrasonography, Prenatal , Blood Flow Velocity , Erythroblastosis, Fetal/blood , Erythroblastosis, Fetal/diagnosis , Erythroblastosis, Fetal/pathology , Erythroblastosis, Fetal/physiopathology , Female , Humans , Liver/pathology , Pregnancy
10.
J Biol Chem ; 276(22): 19495-502, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11278302

ABSTRACT

Members of the transforming growth factor-beta (TGF-beta) superfamily signal through unique cell membrane receptor serine-threonine kinases to activate downstream targets. TRAP1 is a previously described 96-kDa cytoplasmic protein shown to bind to TGF-beta receptors and suggested to play a role in TGF-beta signaling. We now fully characterize the binding properties of TRAP1, and show that it associates strongly with inactive heteromeric TGF-beta and activin receptor complexes and is released upon activation of signaling. Moreover, we demonstrate that TRAP1 plays a role in the Smad-mediated signal transduction pathway, interacting with the common mediator, Smad4, in a ligand-dependent fashion. While TRAP1 has only a small stimulatory effect on TGF-beta signaling in functional assays, deletion constructs of TRAP1 inhibit TGF-beta signaling and diminish the interaction of Smad4 with Smad2. These are the first data to identify a specific molecular chaperone for Smad4, suggesting a model in which TRAP1 brings Smad4 into the vicinity of the receptor complex and facilitates its transfer to the receptor-activated Smad proteins.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Trans-Activators/metabolism , Activins , Animals , COS Cells , Cell Line , Cytoplasm/metabolism , Epitopes/metabolism , Gene Deletion , Genes, Reporter , Green Fluorescent Proteins , Humans , Immunoblotting , Inhibins/metabolism , Ligands , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Models, Biological , Molecular Chaperones/metabolism , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , Signal Transduction , Smad2 Protein , Smad4 Protein , Transcription, Genetic , Transfection , Transforming Growth Factor beta/metabolism
11.
Genes Dev ; 15(4): 455-66, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11230153

ABSTRACT

Hepatitis B, one of the most common infectious diseases in the world, is closely associated with acute and chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Many clinical investigations have revealed that hepatic fibrosis is an important component of these liver diseases caused by chronic hepatitis B. TGF-beta signaling plays an important role in the pathogenesis of fibrosis in chronic hepatitis and cirrhosis. As these diseases are associated with hepatitis B virus (HBV) infection, we examined the possibility that the HBV-encoded pX oncoprotein regulates TGF-beta signaling. We show that pX enhances transcriptional activity in response to TGF-beta, BMP-2, and activin by stabilizing the complex of Smad4 with components of the basic transcriptional machinery. Additionally, confocal microscopic studies suggest that pX facilitates and potentiates the nuclear translocation of Smads, further enhancing TGF-beta signaling. Our studies suggest a new paradigm for amplification of Smad-mediated signaling by an oncoprotein and suggest that enhanced Smad-mediated signaling may contribute to HBV-associated liver fibrosis.


Subject(s)
DNA-Binding Proteins/metabolism , Hepatitis B virus/genetics , Liver Cirrhosis/virology , Oncogene Proteins, Viral/physiology , Signal Transduction/physiology , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , 3T3 Cells , Animals , Cell Nucleus/metabolism , Hepatitis B virus/pathogenicity , Mice , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Protein Binding , Smad4 Protein , Transcriptional Activation
12.
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
13.
J Biol Chem ; 276(22): 19332-9, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11279102

ABSTRACT

Sorting nexins (SNX) comprise a family of proteins with homology to several yeast proteins, including Vps5p and Mvp1p, that are required for the sorting of proteins to the yeast vacuole. Human SNX1, -2, and -4 have been proposed to play a role in receptor trafficking and have been shown to bind to several receptor tyrosine kinases, including receptors for epidermal growth factor, platelet-derived growth factor, and insulin as well as the long form of the leptin receptor, a glycoprotein 130-associated receptor. We now describe a novel member of this family, SNX6, which interacts with members of the transforming growth factor-beta family of receptor serine-threonine kinases. These receptors belong to two classes: type II receptors that bind ligand, and type I receptors that are subsequently recruited to transduce the signal. Of the type II receptors, SNX6 was found to interact strongly with ActRIIB and more moderately with wild type and kinase-defective mutants of TbetaRII. Of the type I receptors, SNX6 was found to interact only with inactivated TbetaRI. SNXs 1-4 also interacted with the transforming growth factor-beta receptor family, showing different receptor preferences. Conversely, SNX6 behaved similarly to the other SNX proteins in its interactions with receptor tyrosine kinases. Strong heteromeric interactions were also seen among SNX1, -2, -4, and -6, suggesting the formation in vivo of oligomeric complexes. These findings are the first evidence for the association of the SNX family of molecules with receptor serine-threonine kinases.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Transforming Growth Factor beta/chemistry , Amino Acid Sequence , Animals , Blotting, Western , COS Cells , Cell Line , Cloning, Molecular , Epitopes , Fluorescent Antibody Technique, Indirect , Humans , Ligands , Luciferases/metabolism , Molecular Sequence Data , Precipitin Tests , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Sorting Nexins , Tissue Distribution , Transfection , Two-Hybrid System Techniques , Vesicular Transport Proteins
14.
Dev Dyn ; 220(2): 141-54, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11169847

ABSTRACT

The eight mammalian Smad proteins mediate cellular signaling from members of the transforming growth factor-beta (TGF-beta), bone morphogenetic protein (BMP), and activin families. Smads 1, 5, and 8 transmit signals from BMPs, while Smads 2 and 3 transmit signals from TGF-betas and activin. Smad 4 is a common mediator of both pathways, while Smads 6 and 7 inhibit signaling. Signal transduction involves translocation of Smad complexes to the nucleus and subsequent gene activation. Little is known about the expression of endogenous Smad proteins during development. We identified commercially available Smad antibodies that specifically recognize a unique Smad protein and are suitable for immunohistochemistry. Here we compare the localization of Smads 1, 2, 3, 4, 5, and 6 in tissues of the 15-day gestation mouse embryo. Immunoreactive Smad proteins are seen in many tissues with differences in the localization being dependent upon the cell type. All tissues express Smad 4 and at least one each of the BMP-specific and TGF-beta-specific Smads, while expression of Smad 6 is more restricted. Differences are observed in the nuclear versus cytoplasmic localization among the Smads in different cell types or tissues, suggesting selective activation of Smads during this stage of development.


Subject(s)
Bone Morphogenetic Proteins/physiology , DNA-Binding Proteins/genetics , Embryo, Mammalian/physiology , Signal Transduction/physiology , Trans-Activators/genetics , Transforming Growth Factor beta/physiology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Gestational Age , Growth Plate/embryology , Humans , Immunohistochemistry , Mice , Organ Specificity , Recombinant Proteins/biosynthesis , Trans-Activators/analysis , Trans-Activators/metabolism , Transfection
15.
Dev Biol ; 240(1): 157-67, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11784053

ABSTRACT

The Smad family of intracellular signaling intermediates transduce signals downstream from the transforming growth factor beta (TGF-beta) family of receptor serine threonine kinases. The original member of this family, Smad1, has been shown to mediate signals from receptors for the bone morphogenetic proteins (BMPs), a large group of ligands in the TGF-beta superfamily that mediate important developmental events. We have targeted the Smad1 gene in mice and created mutants null at this locus. Smad1 mutant mice die at approximately 9.5 days postcoitum due to defects in allantois formation. In Smad1 mutant mice, the allantois fails to fuse to the chorion, resulting in a lack of placenta and failure to establish a definitive embryonic circulation. Although vasculogenesis is initiated in the mutant allantois, the vessels formed are disorganized, and VCAM-1 protein, a marker for distal allantois development, is not expressed. Smad1 null fibroblasts are still able to respond to BMP2, however, suggesting that the defect observed in the developing extraembryonic tissue is caused by a very specific loss of transcriptional activity regulated by Smad1. Our data further demonstrate that although highly similar structurally, Smad proteins are not functionally homologous.


Subject(s)
Allantoin/physiology , Chorion/physiology , DNA-Binding Proteins/genetics , Trans-Activators/genetics , Animals , Base Sequence , Blotting, Western , DNA Primers , Female , In Situ Hybridization , Male , Mice , Mice, Mutant Strains , Mutagenesis , Reverse Transcriptase Polymerase Chain Reaction , Smad Proteins , Smad1 Protein , Vascular Cell Adhesion Molecule-1/genetics
16.
Gene ; 258(1-2): 43-53, 2000 Nov 27.
Article in English | MEDLINE | ID: mdl-11111041

ABSTRACT

Smad1 belongs to a family of receptor-activated proteins which mediate signals from TGF-beta superfamily ligands, including TGF-beta and BMPs. Although much is known about the biochemistry of Smad1 signal transduction, the role of Smad1 in vivo is still unclear. Here we present the first description of the genomic structure of the mouse Smad1 gene and the characterization of its expression pattern in adult mouse tissues by immunohistochemistry. The Smad1 gene contains 7 exons and spans >42 kb of genomic DNA. Its coding region is contained within 6 exons and all introns, except intron 1, follow the GT/AG rule. Immunohistochemical analysis shows that Smad1 is widely expressed in adult mouse tissues, with a varying degree of nuclear localization in different cell types, suggesting a regulated function for this protein. This study assigns all of the exon-intron boundaries of the mouse Smad1 gene and provides the basis for assessing the functional significance of this gene using targeted gene manipulation in the mouse.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Profiling , Trans-Activators/genetics , 5' Untranslated Regions/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , DNA/chemistry , DNA/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA-Binding Proteins/metabolism , Exons , Female , Genes/genetics , Humans , Immunohistochemistry , Introns , Male , Mice , Molecular Sequence Data , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Smad Proteins , Smad1 Protein , Tissue Distribution , Trans-Activators/metabolism
17.
Arch Surg ; 135(11): 1265-70, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11074878

ABSTRACT

HYPOTHESIS: Anti-inflammatory corticosteroids significantly impair wound healing. Retinoids partially, but significantly, reverse this effect. Little is known about the mechanism of steroid retardation or retinoid reversal. We hypothesized that corticosteroids lower transforming growth factor-beta (TGF-beta) and insulin-like growth factor-I (IGF-I) levels and tissue deposition in wounds and that retinoids stimulate corticosteroid-impaired TGF-beta and IGF-I release and collagen production. DESIGN: Randomized controlled trial. SETTING: Wound healing research laboratory. PARTICIPANTS: Animal study. INTERVENTIONS: Four wire mesh wound cylinders were implanted subcutaneously into the backs of 72 male Sprague-Dawley rats. Wound healing was impaired by a single subcutaneous injection of 6 mg of methylprednisolone acetate (Depo-Medrol). Two preparations of retinoids were used in separate experiments: all-trans-retinoic acid and 9-cis-retinoic acid that were fed orally. MAIN OUTCOME MEASURES: Hydroxyproline content was measured in the healing tissue and TGF-beta and IGF-I levels were analyzed in the wound fluid. RESULTS: Methylprednisolone treatment significantly decreased TGF-beta and IGF-I levels in the wound fluid and hydroxyproline content in the tissue (P<.05). Oral all-trans- and 9-cis-retinoic acid partially reversed the TGF-beta and IGF-I decrease and significantly increased hydroxyproline content toward normal levels (P<.05). Oral all-trans-retinoic acid enhanced collagen deposition, TGF-beta and IGF-I levels over normal chow fed control animals (P<.05). CONCLUSIONS: Steroids and retinoids have antagonistic effects on growth factors and collagen deposition in wound healing. These effects can be relevant for treatment options in a clinical setting.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Methylprednisolone/analogs & derivatives , Tretinoin/pharmacology , Wound Healing/drug effects , Alitretinoin , Animals , Collagen/metabolism , Hydroxyproline/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Methylprednisolone/pharmacology , Methylprednisolone Acetate , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/metabolism
18.
Genesis ; 28(2): 47-57, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11064421

ABSTRACT

Bone morphogenetic proteins (BMP) are members of the TGFbeta superfamily of secreted factors with important regulatory functions during embryogenesis. We have isolated the zebrafish gene, nma, that encodes a protein with high sequence similarity to human NMA and Xenopus Bambi. It is also similar to TGFbeta type I serine/theronine kinase receptors in the extracellular ligand-binding domain but lacks a cytoplasmic kinase domain. During development, nma expression is similar to that of bmp2b and bmp4, and analysis in the dorsalized and ventralized zebrafish mutants swirl and chordino indicates that nma is regulated by BMP signaling. Overexpression of nma during zebrafish and Xenopus development resulted in phenotypes that appear to be based on inhibition of BMP signaling. Biochemically, NMA can associate with TGFbeta type II receptors and bind to TGFbeta ligand. We propose that nma is a BMP-regulated gene whose function is to attenuate BMP signaling during development through interactions with type II receptors and ligands.


Subject(s)
Activin Receptors, Type I/physiology , Gene Expression Regulation, Developmental/physiology , Membrane Proteins/physiology , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction/physiology , Xenopus Proteins , Zebrafish Proteins/physiology , Zebrafish/genetics , Amino Acid Sequence , Animals , Body Patterning , Bone Morphogenetic Proteins/physiology , Embryo, Nonmammalian/metabolism , Female , In Situ Hybridization , Membrane Proteins/genetics , Microinjections , Molecular Sequence Data , Mutation , Protein Serine-Threonine Kinases , RNA/metabolism , Radiation Hybrid Mapping , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Sequence Homology, Amino Acid , Xenopus/embryology , Xenopus/genetics , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics
19.
Dev Med Child Neurol ; 42(8): 518-24, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10981929

ABSTRACT

Little is known about the developmental course of striking action. This cross-sectional study explored the refinement of striking in 28 children aged between 4 and 12 years and investigated how well they could use kinaesthesis to gauge the length of an unseen bat. The kinematic data (including smoothness of movement) showed quantitative differences between the age groups. In contrast, no differences were found in the children's ability to judge the length of the unseen bat: within three strikes all of the children had made a clean hit, indicating that they had successfully judged bat length. The children then appeared to memorize the bat with which they had accurately hit the target and made: (1) minimal errors when using this bat in later trials and (2) predictable errors when using two other bats of different sizes. The results show that the striking action becomes optimized over childhood, with smoothness of movement providing an index of this refinement. The findings also suggest that young children have a higher level of kinaesthetic sensitivity than has been assumed previously on the basis of static limb positioning tasks. The results suggest that the striking task used in this study might be a useful tool for investigating the development of movement skills in children with developmental disorders.


Subject(s)
Child Development , Kinesthesis , Motor Skills/classification , Space Perception , Age Factors , Biomechanical Phenomena , Child , Child, Preschool , Cross-Sectional Studies , Female , Humans , Male , Memory
20.
J Natl Cancer Inst ; 92(17): 1388-402, 2000 Sep 06.
Article in English | MEDLINE | ID: mdl-10974075

ABSTRACT

Signaling from transforming growth factor-beta (TGF-beta) through its unique transmembrane receptor serine-threonine kinases plays a complex role in carcinogenesis, having both tumor suppressor and oncogenic activities. Tumor cells often escape from the antiproliferative effects of TGF-beta by mutational inactivation or dysregulated expression of components in its signaling pathway. Decreased receptor function and altered ratios of the TGF-beta type I and type II receptors found in many tumor cells compromise the tumor suppressor activities of TGF-beta and enable its oncogenic functions. Recent identification of a family of intracellular mediators, the Smads, has provided new paradigms for understanding mechanisms of subversion of TGF-beta signaling by tumor cells. In addition, several proteins recently have been identified that can modulate the Smad-signaling pathway and may also be targets for mutation in cancer. Other pathways such as various mitogen-activated protein kinase cascades also contribute substantially to TGF-beta signaling. Understanding the interplay between these signaling cascades as well as the complex patterns of cross-talk with other signaling pathways is an important area of investigation that will ultimately contribute to understanding of the bifunctional tumor suppressor/oncogene role of TGF-beta in carcinogenesis.


Subject(s)
Mutation , Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Animals , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/genetics , Humans , Neoplasms/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein , Smad3 Protein , Smad4 Protein , Trans-Activators/metabolism , Transforming Growth Factor beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...