Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Front Immunol ; 12: 631094, 2021.
Article in English | MEDLINE | ID: mdl-33841412

ABSTRACT

There is an urgent need to identify immunological markers of tuberculosis (TB) risk in HIV co-infected individuals. Previously we have shown that TB recurrence in HIV co-infected individuals on ART was associated with markers of systemic inflammation (IL-6, IL1ß and IL-1Rα). Here we examined the effect of additional acute inflammation and microbial translocation marker expression on risk of TB recurrence. Stored plasma samples were drawn from the TB Recurrence upon Treatment with HAART (TRuTH) study, in which individuals with previously treated pulmonary TB were screened for recurrence quarterly for up to 4 years. Recurrent TB cases (n = 37) were matched to controls (n = 102) by original trial study arm assignment and ART start date. Additional subsets of HIV infected (n = 41) and HIV uninfected (n = 37) individuals from Improving Recurrence Success (IMPRESS) study were sampled at active TB and post successful treatment completion. Plasma concentrations of soluble adhesion molecules (sMAdCAM, sICAM and sVCAM), lipopolysaccharide binding protein (LBP) and transforming growth factor-beta (TGF-ß1, TGF-ß2, TGF-ß3) were measured by multiplex immunoassays and ELISA. Cytokine data was square root transformed in order to reduce variability. Multivariable analysis adjusted for a number of potential confounders measured at sample time-point: age, BMI, CD4 count, viral load (VL) and measured at baseline: presence or absence of lung cavities, previous history of TB, and WHO disease stage (4 vs 3). The following analytes were associated with increased risk of TB recurrence in the multivariable model: sICAM (aOR 1.06, 95% CI: 1.02-1.12, p = 0.009), LBP (aOR 8.78, 95% CI: 1.23-62.66, p = 0.030) and TGF-ß3 (aOR 1.44, 95% CI 1.01-2.05, p = 0.044). Additionally, we observed a positive correlation between LBP and sICAM (r= 0.347, p<0.0001), and LBP and IL-6, identified to be one of the strongest predictors of TB risk in our previous study (r=0.623, p=0.03). These data show that increased risk of TB recurrence in HIV infected individuals on ART is likely associated with HIV mediated translocation of microbial products and the resulting chronic immune activation.


Subject(s)
HIV Infections/microbiology , Tuberculosis/blood , Tuberculosis/immunology , Acute-Phase Proteins , Adult , Antiretroviral Therapy, Highly Active , Bacterial Translocation/genetics , Biomarkers/blood , CD4 Lymphocyte Count , Carrier Proteins/blood , Cohort Studies , Cytokines/blood , Cytokines/immunology , Female , HIV Infections/drug therapy , HIV Infections/epidemiology , Humans , Male , Membrane Glycoproteins/blood , Recurrence , Risk Factors , South Africa/epidemiology , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/classification , Transforming Growth Factor beta/immunology , Tuberculosis/diagnosis , Tuberculosis/epidemiology , Viral Load
2.
Mol Med Rep ; 17(1): 699-704, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115550

ABSTRACT

Tumor progression can be affected by various cellular components of tumor cells and/or by tumor microenvironmental factors. The tumor microenvironment comprises a variety of nonmalignant stromal cells and inflammatory cytokines, which are pivotal in tumor promotion and progression. The transforming growth factor­ß (TGF­ß) ligands (TGF­ß1, 2 and 3) are secreted inflammatory cytokines, which are known to be involved in various aspects of tumor development through two transmembrane serine­threonine kinase receptors, TGFßR1 and TGFßR2. TGF­ß promotes or inhibits tumorigenesis depending on the concurrent gene mutations and tissue microenvironment present through the small mothers against decapentaplegic (Smad) and non­Smad pathways. This review aims to provide a comprehensive overview of the role of the TGF­ß pathway in tumor initiation and progression.


Subject(s)
Carcinogenesis/genetics , Neoplasms/genetics , Transforming Growth Factor beta/genetics , Humans , Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Transforming Growth Factor beta/classification , Tumor Microenvironment/genetics
3.
PLoS One ; 6(9): e24152, 2011.
Article in English | MEDLINE | ID: mdl-21931657

ABSTRACT

The TGF-ß signaling pathway is a metazoan-specific intercellular signaling pathway known to be important in many developmental and cellular processes in a wide variety of animals. We investigated the complexity and possible functions of this pathway in a member of one of the earliest branching metazoan phyla, the ctenophore Mnemiopsis leidyi. A search of the recently sequenced Mnemiopsis genome revealed an inventory of genes encoding ligands and the rest of the components of the TGF-ß superfamily signaling pathway. The Mnemiopsis genome contains nine TGF-ß ligands, two TGF-ß-like family members, two BMP-like family members, and five gene products that were unable to be classified with certainty. We also identified four TGF-ß receptors: three Type I and a single Type II receptor. There are five genes encoding Smad proteins (Smad2, Smad4, Smad6, and two Smad1s). While we have identified many of the other components of this pathway, including Tolloid, SMURF, and Nomo, notably absent are SARA and all of the known antagonists belonging to the Chordin, Follistatin, Noggin, and CAN families. This pathway likely evolved early in metazoan evolution as nearly all components of this pathway have yet to be identified in any non-metazoan. The complement of TGF-ß signaling pathway components of ctenophores is more similar to that of the sponge, Amphimedon, than to cnidarians, Trichoplax, or bilaterians. The mRNA expression patterns of key genes revealed by in situ hybridization suggests that TGF-ß signaling is not involved in ctenophore early axis specification. Four ligands are expressed during gastrulation in ectodermal micromeres along all three body axes, suggesting a role in transducing earlier maternal signals. Later expression patterns and experiments with the TGF-ß inhibitor SB432542 suggest roles in pharyngeal morphogenesis and comb row organization.


Subject(s)
Ctenophora/genetics , Evolution, Molecular , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Amino Acid Sequence , Animals , Bayes Theorem , Benzamides/pharmacology , Ctenophora/classification , Ctenophora/embryology , Dioxoles/pharmacology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , In Situ Hybridization , Molecular Sequence Data , Phylogeny , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Sequence Homology, Amino Acid , Signal Transduction/drug effects , Smad Proteins/genetics , Transforming Growth Factor beta/classification
4.
BMC Mol Biol ; 11: 37, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20459854

ABSTRACT

BACKGROUND: The transforming growth factor-beta (TGF-beta) family constitutes of dimeric proteins that regulate the growth, differentiation and metabolism of many cell types, including that of skeletal muscle in mammals. The potential role of TGF-betas in fish muscle growth is not known. RESULTS: Here we report the molecular characterization, developmental and tissue expression and regulation by nutritional state of a novel TGF-beta gene from a marine fish, the gilthead sea bream Sparus aurata. S. aurata TGF-beta6 is encoded by seven exons 361, 164, 133, 111, 181, 154, and 156 bp in length and is translated into a 420-amino acid peptide. The exons are separated by six introns: >643, 415, 93, 1250, 425 and >287 bp in length. Although the gene organization is most similar to mouse and chicken TGF-beta2, the deduced amino acid sequence represents a novel TGF-beta that is unique to fish that we have named TGF-beta6. The molecule has conserved putative functional residues, including a cleavage motif (RXXR) and nine cysteine residues that are characteristic of TGF-beta. Semi-quantitative analysis of TGF-beta6 expression revealed differential expression in various tissues of adult fish with high levels in skin and muscle, very low levels in liver, and moderate levels in other tissues including brain, eye and pituitary. TGF-beta6 is expressed in larvae on day of hatching and increases as development progresses. A fasting period of five days of juvenile fish resulted in increased levels of TGF-beta6 expression in white skeletal muscle compared to that in fed fish, which was slightly attenuated by one injection of growth hormone. CONCLUSION: Our findings provide valuable insights about genomic information and nutritional regulation of TGF-beta6 which will aid the further investigation of the S. aurata TGF-beta6 gene in association with muscle growth. The finding of a novel TGF-beta6 molecule, unique to fish, will contribute to the understanding of the evolution of the TGF-beta family of cytokines in vertebrates.


Subject(s)
Fish Proteins/genetics , Muscle, Skeletal/metabolism , Sea Bream/genetics , Transforming Growth Factor beta/genetics , Amino Acid Sequence , Animals , Fish Proteins/classification , Fish Proteins/metabolism , Gene Expression Regulation , Larva/metabolism , Molecular Sequence Data , Phylogeny , Sea Bream/growth & development , Sea Bream/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transforming Growth Factor beta/classification , Transforming Growth Factor beta/metabolism
5.
BMC Bioinformatics ; 10 Suppl 5: S3, 2009 May 06.
Article in English | MEDLINE | ID: mdl-19426460

ABSTRACT

BACKGROUND: The Protein Ontology (PRO) is designed as a formal and principled Open Biomedical Ontologies (OBO) Foundry ontology for proteins. The components of PRO extend from a classification of proteins on the basis of evolutionary relationships at the homeomorphic level to the representation of the multiple protein forms of a gene, including those resulting from alternative splicing, cleavage and/or post-translational modifications. Focusing specifically on the TGF-beta signaling proteins, we describe the building, curation, usage and dissemination of PRO. RESULTS: PRO is manually curated on the basis of PrePRO, an automatically generated file with content derived from standard protein data sources. Manual curation ensures that the treatment of the protein classes and the internal and external relationships conform to the PRO framework. The current release of PRO is based upon experimental data from mouse and human proteins wherein equivalent protein forms are represented by single terms. In addition to the PRO ontology, the annotation of PRO terms is released as a separate PRO association file, which contains, for each given PRO term, an annotation from the experimentally characterized sub-types as well as the corresponding database identifiers and sequence coordinates. The annotations are added in the form of relationship to other ontologies. Whenever possible, equivalent forms in other species are listed to facilitate cross-species comparison. Splice and allelic variants, gene fusion products and modified protein forms are all represented as entities in the ontology. Therefore, PRO provides for the representation of protein entities and a resource for describing the associated data. This makes PRO useful both for proteomics studies where isoforms and modified forms must be differentiated, and for studies of biological pathways, where representations need to take account of the different ways in which the cascade of events may depend on specific protein modifications. CONCLUSION: PRO provides a framework for the formal representation of protein classes and protein forms in the OBO Foundry. It is designed to enable data retrieval and integration and machine reasoning at the molecular level of proteins, thereby facilitating cross-species comparisons, pathway analysis, disease modeling and the generation of new hypotheses.


Subject(s)
Information Storage and Retrieval/methods , Intracellular Signaling Peptides and Proteins/classification , Transforming Growth Factor beta/chemistry , Computational Biology/methods , Databases, Genetic , Databases, Protein , Humans , Intracellular Signaling Peptides and Proteins/genetics , Transforming Growth Factor beta/classification , User-Computer Interface
6.
J Biol Chem ; 284(14): 9311-20, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19193648

ABSTRACT

The assembly and secretion of transforming growth factor beta superfamily ligands is dependent upon non-covalent interactions between their pro- and mature domains. Despite the importance of this interaction, little is known regarding the underlying regulatory mechanisms. In this study, the binding interface between the pro- and mature domains of the inhibin alpha-subunit was characterized using in vitro mutagenesis. Three hydrophobic residues near the N terminus of the prodomain (Leu(30), Phe(37), Leu(41)) were identified that, when mutated to alanine, disrupted heterodimer assembly and secretion. It is postulated that these residues mediate dimerization by interacting non-covalently with hydrophobic residues (Phe(271), Ile(280), Pro(283), Leu(338), and Val(340)) on the outer convex surface of the mature alpha-subunit. Homology modeling indicated that these mature residues are located at the interface between two beta-sheets of the alpha-subunit and that their side chains form a hydrophobic packing core. Mutation of these residues likely disturbs the conformation of this region, thereby disrupting non-covalent interactions with the prodomain. A similar hydrophobic interface was identified spanning the pro- and mature domains of the inhibin beta(A)-subunit. Mutation of key residues, including Ile(62), Leu(66), Phe(329), and Pro(341), across this interface was disruptive for the production of both inhibin A and activin A. In addition, mutation of Ile(62) and Leu(66) in the beta(A)-propeptide reduced its ability to bind, or inhibit the activity of, activin A. Conservation of the identified hydrophobic motifs in the pro- and mature domains of other transforming growth factor beta superfamily ligands suggests that we have identified a common biosynthetic pathway governing dimer assembly.


Subject(s)
Inhibins/metabolism , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Hydrophobic and Hydrophilic Interactions , Inhibins/chemistry , Inhibins/genetics , Ligands , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Sequence Alignment , Transforming Growth Factor beta/classification
7.
Front Oral Biol ; 12: 178-196, 2008.
Article in English | MEDLINE | ID: mdl-18391501

ABSTRACT

Premature suture obliteration results in an inability of cranial and facial bones to grow, with resulting craniofacial dysmorphology requiring surgical correction. Understanding the biological signaling associated with suture morphogenesis will enable less invasive treatment of patients with fused sutures, combined with therapy using biological molecules. While a number of advances have been made in identifying the genetic etiologies of various craniosynostotic syndromes, the pathogenesis of this condition is still not completely understood. Recently, it has been shown that differential expression of various transforming growth factor-beta (Tgf-beta) isoforms plays a crucial role in regulating suture patency once the sutures have formed. It has also been shown that differential expression of Tgf-beta isoforms may also play a role in craniosynostosis by altering proliferation, differentiation, and apoptosis within the suture. This chapter focuses on the role of Tgf-beta in suture morphogenesis and growth, exploring Tgf-beta biology, receptors, signaling pathways, animal models, and expression in both normal and pathological sutures.


Subject(s)
Cranial Sutures/growth & development , Morphogenesis/physiology , Transforming Growth Factor beta/physiology , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Proliferation , Cranial Sutures/physiology , Craniosynostoses/etiology , Craniosynostoses/physiopathology , Disease Models, Animal , Humans , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/classification
8.
BMC Genomics ; 9: 82, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18282302

ABSTRACT

BACKGROUND: Myostatin (MSTN) is a member of the transforming growth factor-beta superfamily that negatively regulates growth of skeletal muscle tissue. The gene encoding for the MSTN peptide is a consolidate candidate for the enhancement of productivity in terrestrial livestock. This gene potentially represents an important target for growth improvement of cultured finfish. RESULTS: Here we report molecular characterization, tissue expression and sequence variability of the barramundi (Lates calcarifer) MSTN-1 gene. The barramundi MSTN-1 was encoded by three exons 379, 371 and 381 bp in length and translated into a 376-amino acid peptide. Intron 1 and 2 were 412 and 819 bp in length and presented typical GT...AG splicing sites. The upstream region contained cis-regulatory elements such as TATA-box and E-boxes. A first assessment of sequence variability suggested that higher mutation rates are found in the 5' flanking region with several SNP's present in this species. A putative micro RNA target site has also been observed in the 3'UTR (untranslated region) and is highly conserved across teleost fish. The deduced amino acid sequence was conserved across vertebrates and exhibited characteristic conserved putative functional residues including a cleavage motif of proteolysis (RXXR), nine cysteines and two glycosilation sites. A qualitative analysis of the barramundi MSTN-1 expression pattern revealed that, in adult fish, transcripts are differentially expressed in various tissues other than skeletal muscles including gill, heart, kidney, intestine, liver, spleen, eye, gonad and brain. CONCLUSION: Our findings provide valuable insights such as sequence variation and genomic information which will aid the further investigation of the barramundi MSTN-1 gene in association with growth. The finding for the first time in finfish MSTN of a miRNA target site in the 3'UTR provides an opportunity for the identification of regulatory mutations on the expression of this gene.


Subject(s)
Fish Proteins/genetics , Gene Expression Profiling , Perciformes/genetics , Transforming Growth Factor beta/genetics , Amino Acid Sequence , Animals , Base Sequence , Fish Proteins/classification , Molecular Sequence Data , Myostatin , Phylogeny , Polymorphism, Single Nucleotide , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, DNA , Transforming Growth Factor beta/classification
9.
J Mammary Gland Biol Neoplasia ; 12(4): 249-57, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18027075

ABSTRACT

The molecular mechanisms controlling the onset of copious milk secretion are only now beginning to be elucidated. We have known for nearly four decades that progesterone suppresses milk secretion during pregnancy, and that the fall in progesterone near parturition is necessary for secretory activation. Similarly, we've known for 15 years that transforming growth factor beta (TGFbeta) also suppresses milk secretion. Yet no formal link between the two has ever been established. This work aims to review the evidence for and against a link between progesterone and TGFbeta, raise unanswered questions, and to propose further lines of research.


Subject(s)
Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Progesterone/metabolism , Transforming Growth Factor beta/metabolism , Animals , Female , Humans , Pregnancy , Protein Isoforms/classification , Protein Isoforms/metabolism , Signal Transduction , Transforming Growth Factor beta/classification
10.
Dev Biol ; 311(2): 500-11, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17936261

ABSTRACT

Vg1, a member of the TGF-beta superfamily of ligands, has been implicated in the induction of mesoderm, formation of primitive streak, and left-right patterning in Xenopus and chick embryos. In mice, GDF1 and GDF3 - two TGF-beta superfamily ligands that share high sequence identity with Vg1 - have been shown to independently mimic distinct aspects of Vg1's functions. However, the extent to which the developmental processes controlled by GDF1 and GDF3 and the underlying signaling mechanisms are evolutionarily conserved remains unclear. Here we show that phylogenetic and genomic analyses indicate that Gdf1 is the true Vg1 ortholog in mammals. In addition, and similar to GDF1, we find that GDF3 signaling can be mediated by the type I receptor ALK4, type II receptors ActRIIA and ActRIIB, and the co-receptor Cripto to activate Smad-dependent reporter genes. When expressed in heterologous cells, the native forms of either GDF1 or GDF3 were incapable of inducing downstream signaling. This could be circumvented by using chimeric constructs carrying heterologous prodomains, or by co-expression with the Furin pro-protein convertase, indicating poor processing of the native GDF1 and GDF3 precursors. Unexpectedly, co-expression with Nodal - another TGF-beta superfamily ligand involved in mesoderm formation - could also expose the activities of native GDF1 and GDF3, suggesting a potentially novel mode of cooperation between these ligands. Functional complementarity between GDF1 and GDF3 during embryonic development was investigated by analyzing genetic interactions between their corresponding genes. This analysis showed that Gdf1(-/-);Gdf3(-/-) compound mutants are more severely affected than either Gdf1(-/-) or Gdf3(-/-) single mutants, with defects in the formation of anterior visceral endoderm and mesoderm that recapitulate Vg1 loss of function, suggesting that GDF1 and GDF3 together represent the functional mammalian homologs of Vg1.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Transforming Growth Factor beta/metabolism , Xenopus Proteins/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Body Patterning , Cell Line , Chickens , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Growth Differentiation Factor 1 , Growth Differentiation Factor 3 , Humans , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/classification , Intercellular Signaling Peptides and Proteins/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mesoderm/cytology , Mesoderm/physiology , Mice , Mice, Knockout , Morphogenesis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nodal Protein , Phylogeny , Protein Precursors/genetics , Protein Precursors/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/classification , Transforming Growth Factor beta/genetics , Xenopus , Xenopus Proteins/classification , Xenopus Proteins/genetics
11.
Development ; 134(20): 3649-64, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17855430

ABSTRACT

The TGF-beta family member Nodal is essential for specification of the dorsal-ventral axis of the sea urchin embryo, but the molecular factors regulating its expression are not known. Analysis of the nodal promoter is an excellent entry point to identify these factors and to dissect the regulatory logic driving dorsal-ventral axis specification. Using phylogenetic footprinting, we delineated two regulatory regions located in the 5' region of the nodal promoter and in the intron that are required for correct spatial expression and for autoregulation. The 5' regulatory region contains essential binding sites for homeodomain, bZIP, Oct, Tcf/Lef, Sox and Smad transcription factors, and a binding site for an unidentified spatial repressor possibly related to Myb. Soon after its initiation, nodal expression critically requires autoregulation by Nodal and signaling by the maternal TGF-beta Univin. We show that Univin is related to Vg1, that both Nodal and Univin signal through Alk4/5/7, and that zygotic expression of univin, like that of nodal, is dependent on SoxB1 function and Tcf/beta-catenin signaling. This work shows that Tcf, SoxB1 and Univin play essential roles in the regulation of nodal expression in the sea urchin and suggests that some of the regulatory interactions controlling nodal expression predate the chordates. The data are consistent with a model of nodal regulation in which a maternal TGF-beta acts in synergy with maternal transcription factors and with spatial repressors to establish the dorsal-ventral axis of the sea urchin embryo.


Subject(s)
Body Patterning , Gene Expression Regulation, Developmental , Regulatory Sequences, Nucleic Acid , Sea Urchins , Transforming Growth Factor beta/metabolism , Animals , Base Sequence , Binding Sites , DNA Mutational Analysis , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/physiology , Genes, Reporter , Genetic Linkage , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , Introns , Molecular Sequence Data , Nodal Protein , Phylogeny , SOXB1 Transcription Factors , Sea Urchins/anatomy & histology , Sea Urchins/embryology , Sea Urchins/metabolism , Sequence Alignment , Signal Transduction/physiology , Smad Proteins/genetics , Smad Proteins/metabolism , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/classification , Transforming Growth Factor beta/genetics
12.
J Clin Invest ; 117(7): 1933-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17607365

ABSTRACT

Systemic iron balance is regulated by hepcidin, a peptide hormone secreted by the liver. By decreasing cell surface expression of the iron exporter ferroportin, hepcidin decreases iron absorption from the intestine and iron release from reticuloendothelial stores. Hepcidin excess has been implicated in the pathogenesis of anemia of chronic disease, while hepcidin deficiency has a key role in the pathogenesis of the iron overload disorder hemochromatosis. We have recently shown that hemojuvelin is a coreceptor for bone morphogenetic protein (BMP) signaling and that BMP signaling positively regulates hepcidin expression in liver cells in vitro. Here we show that BMP-2 administration increases hepcidin expression and decreases serum iron levels in vivo. We also show that soluble hemojuvelin (HJV.Fc) selectively inhibits BMP induction of hepcidin expression in vitro and that administration of HJV.Fc decreases hepcidin expression, increases ferroportin expression, mobilizes splenic iron stores, and increases serum iron levels in vivo. These data support a role for modulators of the BMP signaling pathway in treating diseases of iron overload and anemia of chronic disease.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Iron/metabolism , Signal Transduction , Animals , Antimicrobial Cationic Peptides/genetics , Bone Morphogenetic Proteins/classification , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/pharmacology , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Gene Expression Regulation , Hepcidins , Humans , Interleukin-6/pharmacology , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mononuclear Phagocyte System/metabolism , Signal Transduction/drug effects , Solubility , Transforming Growth Factor beta/classification , Transforming Growth Factor beta/pharmacology
13.
BMC Cell Biol ; 8: 25, 2007 Jun 25.
Article in English | MEDLINE | ID: mdl-17592637

ABSTRACT

BACKGROUND: Endocytosis is a key regulator of growth factor signaling pathways. Recent studies showed that the localization to endosomes of intracellular mediators of growth factor signaling may be required for their function. Although there is substantial evidence linking endocytosis and growth factor signaling in cultured cells, there has been little study of the endosomal localization of signaling components in intact tissues or organs. RESULTS: Proteins that are downstream of the transforming growth factor-beta superfamily signaling pathway were found on endosomes in chicken embryo and postnatal mouse lenses, which depend on signaling by members of the TGFbeta superfamily for their normal development. Phosphorylated Smad1 (pSmad1), pSmad2, Smad4, Smad7, the transcriptional repressors c-Ski and TGIF and the adapter molecules Smad anchor for receptor activation (SARA) and C184M, localized to EEA-1- and Rab5-positive vesicles in chicken embryo and/or postnatal mouse lenses. pSmad1 and pSmad2 also localized to Rab7-positive late endosomes. Smad7 was found associated with endosomes, but not caveolae. Bmpr1a conditional knock-out lenses showed decreased nuclear and endosomal localization of pSmad1. Many of the effectors in this pathway were distributed differently in vivo from their reported distribution in cultured cells. CONCLUSION: Based on the findings reported here and data from other signaling systems, we suggest that the localization of activated intracellular mediators of the transforming growth factor-beta superfamily to endosomes is important for the regulation of growth factor signaling.


Subject(s)
Endosomes/metabolism , Lens, Crystalline/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Biomarkers , Bone Morphogenetic Proteins/metabolism , Cell Nucleus/metabolism , Chick Embryo , Membrane Proteins/metabolism , Mice , Smad Proteins, Receptor-Regulated/metabolism , Time Factors , Transforming Growth Factor beta/classification , Vesicular Transport Proteins/metabolism
14.
Mini Rev Med Chem ; 6(11): 1255-61, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17100637

ABSTRACT

The transforming growth factor-beta (TGF-beta) superfamily includes TGF-betas, activin, myostatin and bone morphogenetic proteins. Misregulation of the activity of TGF-beta family members is involved in pathogenesis of cancer, muscular dystrophy, obesity and bone and tooth remodeling. Natural inhibitors for the TGF-beta superfamily regulate fine-tuning of activity of TGF-beta family in vivo. In addition to natural inhibitors for the TGF-beta family, soluble forms of receptors for the TGF-beta family, blocking monoclonal antibodies and small chemical TGF-beta inhibitors have been developed. In this review, we summarize recent advances in our understanding of inhibitors for the TGF-beta superfamily and their medical applications.


Subject(s)
Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , Humans , Muscular Diseases/drug therapy , Muscular Diseases/metabolism , Myostatin , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Binding , Transforming Growth Factor beta/classification
15.
PLoS Comput Biol ; 2(1): e3, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16446785

ABSTRACT

The TGF-beta pathway plays a central role in tissue homeostasis and morphogenesis. It transduces a variety of extracellular signals into intracellular transcriptional responses that control a plethora of cellular processes, including cell growth, apoptosis, and differentiation. We use computational modeling to show that coupling of signaling with receptor trafficking results in a highly versatile signal-processing unit, able to sense by itself absolute levels of ligand, temporal changes in ligand concentration, and ratios of multiple ligands. This coupling controls whether the response of the receptor module is transient or permanent and whether or not different signaling channels behave independently of each other. Our computational approach unifies seemingly disparate experimental observations and suggests specific changes in receptor trafficking patterns that can lead to phenotypes that favor tumor progression.


Subject(s)
Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/classification , Transforming Growth Factor beta/metabolism , Computer Simulation , Kinetics , Ligands , Models, Biological , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Protein Transport
16.
Biomaterials ; 27(6): 807-22, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16213014

ABSTRACT

The induction of bone formation starts by erecting scaffolds of smart biomimetic matrices acting as insoluble signals affecting the release of soluble osteogenic molecular signals. The cascade of bone differentiation by induction develops as a mosaic structure singly initiated by the osteogenic proteins of the transforming growth factor-beta (TGF-beta) supergene family. The osteogenic signals when combined with an insoluble signal or substratum initiate de novo bone formation by induction and are deployed singly, synergistically and synchronously to sculpt the architecture of the mineralized bone/bone marrow organ. The osteogenic proteins of the TGF-beta superfamily are the common molecular initiators deployed for embryonic development and the induction of bone in postnatal osteogenesis, whereby molecules exploited in embryonic development are re-deployed in postnatal tissue morphogenesis as a recapitulation of embryonic development. The pleiotropy of the osteogenic proteins of the TGF-beta superfamily is highlighted by the apparent redundancy of molecular signals initiating bone formation by induction including the TGF-beta isoforms per se, powerful inducers of endochondral bone but in the primate only. Bone induction by the TGF-beta isoforms in the primate is site and tissue specific with substantial endochondral bone induction in heterotopic sites but with absent osteoinductivity in orthotopic calvarial sites on day 30 and only limited osteogenesis pericranially on day 90. Ebaf/Lefty-A, a novel member of the TGF-beta superfamily, induces chondrogenesis in calvarial defects of Papio ursinus and bone regeneration across the defect on day 30 and 90, respectively. The strikingly pleiotropic effects of the bone morphogenetic and osteogenic proteins (BMPs/OPs) spring from amino acid sequence variations in the carboxy-terminal domain and in the transduction of distinct signalling pathways by individual Smad proteins after transmembrane serine/threonine kinase complexes of type I and II receptors. Predictable bone regeneration in clinical contexts requires information concerning the expression and cross regulation of gene products of the TGF-beta superfamily. OP-1, BMP-3, TGF-beta1 and type IV collagen mRNAs expression correlates to the morphological induction and maintenance of engineered ossicles by the hOP-1 osteogenic devices in the non-human primate P. ursinus. Amino-acid sequence variations amongst BMPs/OPs in the carboxy terminal domain confer the structure/activity profile responsible for the pleiotropic activity that controls tissue induction and morphogenesis of a variety of tissues and organs by different BMPs/OPs which are helping to engineer skeletal tissue regeneration in molecular terms.


Subject(s)
Osteogenesis , Signal Transduction , Animals , Biomimetic Materials/pharmacology , Extracellular Matrix/metabolism , Neovascularization, Physiologic/drug effects , Osteogenesis/drug effects , Papio , Protein Isoforms/metabolism , Signal Transduction/drug effects , Solubility , Transforming Growth Factor beta/classification , Transforming Growth Factor beta/metabolism
17.
Development ; 131(18): 4593-605, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15342483

ABSTRACT

The TGFbeta superfamily plays diverse and essential roles in kidney development. Gdf11 and Bmp4 are essential for outgrowth and positioning of the ureteric bud, the inducer of metanephric mesenchyme. During nephrogenesis, Bmp7 is required for renewal of the mesenchyme progenitor population. Additionally, in vitro studies demonstrate inhibitory effects of BMPs and TGFbetas on collecting duct branching and growth. Here, we explore the predicted models of TGFbeta superfamily function by cell-specific inactivation of Smad4, a key mediator of TGFbeta signaling. Using a HoxB7cre transgene expressed in ureteric bud and collecting duct, we find that development of the collecting duct is Smad4 independent. By contrast, removal of Smad4 in nephrogenic mesenchyme using the Bmp7(cre/+) allele leads to disorganization of the nephrogenic mesenchyme and impairment of mesenchyme induction. Smad4-deficient metanephric mesenchyme does not display defects in inducibility in LiCl or spinal cord induction assays. However, in situ hybridization and lineage analysis of Smad4 null mesenchyme cells at E11.5 show that the nephrogenic mesenchyme does not aggregate tightly around the ureteric bud tips, but remains loosely associated, embedded within a population of cells expressing markers of both nephrogenic mesenchyme and peripheral stroma. We conclude that the failure of recruitment of nephrogenic mesenchyme leaves a primitive population of mesenchyme at the periphery of the kidney. This population is gradually depleted, and by E16.5 the periphery is composed of cells of stromal phenotype. This study uncovers a novel role for TGFbeta superfamily signaling in the recruitment and/or organization of the nephrogenic mesenchyme at early time-points of kidney development. Additionally, we present conclusive genetic lineage mapping of the collecting duct and nephrogenic mesenchyme.


Subject(s)
Kidney/embryology , Kidney/metabolism , Mesoderm/metabolism , Morphogenesis , Signal Transduction , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Division , Cell Lineage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Deletion , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Kidney/pathology , Mesoderm/pathology , Mice , Mice, Transgenic , Multigene Family , Nephrosis/genetics , Nephrosis/metabolism , Nephrosis/pathology , Smad4 Protein , Stem Cells/pathology , Trans-Activators/deficiency , Trans-Activators/genetics , Trans-Activators/metabolism , Transforming Growth Factor beta/classification , Transforming Growth Factor beta/genetics
18.
Dev Comp Immunol ; 28(5): 461-85, 2004 May 03.
Article in English | MEDLINE | ID: mdl-15062644

ABSTRACT

Members of the transforming growth factor beta (TGF-beta) superfamily of cell signalling polypeptides have attracted much attention because of their ability, from nematodes to mammals, to control cellular functions that in turn, regulate embryo development and tissue homeostasis. On the basis of structure similarities, the TGF-beta members (ligands, receptors and Smads) are subdivided into TGF-beta sensu stricto, bone morphogenetic proteins (BMP) and activins. Although BMP is the best characterized pathway in metazoans, recent findings in molluscs and non-bilateria as well as the analysis of nematode and arthropod genomes, demonstrate the early origin of these distinct subfamilies of ligands, receptors and Smads. This report analyses the large diversity of ligands, receptors and Smads in metazoans from cnidarians and molluscs to mammals. The contribution of new data, mainly from the lophochotrozoan Crassostrea gigas and other organisms on the fringe of the 'branded model organisms', will help us to demonstrate that TGF-betas are probably the most ancestral active cytokines characterized at the molecular level in both Protostome and Deuterostome lineages.


Subject(s)
Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology , Amino Acid Sequence , Animals , Evolution, Molecular , Genome , Humans , Ligands , Molecular Sequence Data , Phylogeny , Protein Processing, Post-Translational , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/classification , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/physiology , Sequence Homology, Amino Acid , Signal Transduction , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/classification
SELECTION OF CITATIONS
SEARCH DETAIL