Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 297(3): 101055, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34411563

RESUMO

Fibrillin-1 (FBN1) is the major component of extracellular matrix microfibrils, which are required for proper development of elastic tissues, including the heart and lungs. Through protein-protein interactions with latent transforming growth factor (TGF) ß-binding protein 1 (LTBP1), microfibrils regulate TGF-ß signaling. Mutations within the 47 epidermal growth factor-like (EGF) repeats of FBN1 cause autosomal dominant disorders including Marfan Syndrome, which is characterized by disrupted TGF-ß signaling. We recently identified two novel protein O-glucosyltransferases, Protein O-glucosyltransferase 2 (POGLUT2) and 3 (POGLUT3), that modify a small fraction of EGF repeats on Notch. Here, using mass spectral analysis, we show that POGLUT2 and POGLUT3 also modify over half of the EGF repeats on FBN1, fibrillin-2 (FBN2), and LTBP1. While most sites are modified by both enzymes, some sites show a preference for either POGLUT2 or POGLUT3. POGLUT2 and POGLUT3 are homologs of POGLUT1, which stabilizes Notch proteins by addition of O-glucose to Notch EGF repeats. Like POGLUT1, POGLUT2 and 3 can discern a folded versus unfolded EGF repeat, suggesting POGLUT2 and 3 are involved in a protein folding pathway. In vitro secretion assays using the N-terminal portion of recombinant FBN1 revealed reduced FBN1 secretion in POGLUT2 knockout, POGLUT3 knockout, and POGLUT2 and 3 double-knockout HEK293T cells compared with wild type. These results illustrate that POGLUT2 and 3 function together to O-glucosylate protein substrates and that these modifications play a role in the secretion of substrate proteins. It will be interesting to see how disease variants in these proteins affect their O-glucosylation.


Assuntos
Fibrilina-1/metabolismo , Fibrilina-2/metabolismo , Proteínas de Ligação a TGF-beta Latente/metabolismo , Síndrome de Marfan/metabolismo , Motivos de Aminoácidos , Fibrilina-1/química , Fibrilina-1/genética , Fibrilina-2/química , Fibrilina-2/genética , Glicosilação , Humanos , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/genética , Síndrome de Marfan/enzimologia , Síndrome de Marfan/genética , Sistemas de Translocação de Proteínas , Transdução de Sinais
2.
BMC Med Genomics ; 13(1): 183, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33302946

RESUMO

BACKGROUND: Autosomal recessive cutis laxa type IC (ARCL IC, MIM: #613177) results from a mutation in the LTBP4 gene (MIM: #604710) on chromosome 19q13. CASE PRESENTATION: A 28-day-old Chinese infant with generalized cutis laxa accompanied by impaired pulmonary, gastrointestinal, genitourinary, retinal hemorrhage, abnormality of coagulation and hyperbilirubinemia was admitted to our hospital. To find out the possible causes of these symptoms, whole-exome sequencing was performed on the infant. Two novel pathogenic frame-shift variants [c.605_606delGT (p.Ser204fs * 8) and c.1719delC (p.Arg574fs * 199)] of the LTBP4 gene associated with ARCL IC were found which was later verified by Sanger sequencing. The pathogenicity of mutations was subsequently assessed by several software programs and databases. In addition, an analytical review on the clinical phenotypes of the disease previously reported in literature was performed. CONCLUSIONS: This is the first report of a Chinese infant with ARCL IC in China due to novel pathogenic variations of LTBP4. Our study extends the cutis laxa type IC mutation spectrum as well as the phenotypes associated with the disease in different populations.


Assuntos
Anormalidades Múltiplas/genética , Cútis Laxa/genética , Mutação da Fase de Leitura , Proteínas de Ligação a TGF-beta Latente/genética , Povo Asiático/genética , Cromossomos Humanos Par 19/genética , Códon sem Sentido , Cútis Laxa/etnologia , Feminino , Heterozigoto , Humanos , Lactente , Recém-Nascido , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/fisiologia , Modelos Moleculares , Fases de Leitura Aberta/genética , Linhagem , Conformação Proteica , Enfisema Pulmonar/diagnóstico por imagem , Enfisema Pulmonar/genética , Sequenciamento do Exoma
3.
Cell ; 180(3): 490-501.e16, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-31955848

RESUMO

Integrin αvß8 binds with exquisite specificity to latent transforming growth factor-ß (L-TGF-ß). This binding is essential for activating L-TGF-ß presented by a variety of cell types. Inhibiting αvß8-mediated TGF-ß activation blocks immunosuppressive regulatory T cell differentiation, which is a potential therapeutic strategy in cancer. Using cryo-electron microscopy, structure-guided mutagenesis, and cell-based assays, we reveal the binding interactions between the entire αvß8 ectodomain and its intact natural ligand, L-TGF-ß, as well as two different inhibitory antibody fragments to understand the structural underpinnings of αvß8 binding specificity and TGF-ß activation. Our studies reveal a mechanism of TGF-ß activation where mature TGF-ß signals within the confines of L-TGF-ß and the release and diffusion of TGF-ß are not required. The structural details of this mechanism provide a rational basis for therapeutic strategies to inhibit αvß8-mediated L-TGF-ß activation.


Assuntos
Microscopia Crioeletrônica/métodos , Integrinas/química , Integrinas/metabolismo , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/metabolismo , Fator de Crescimento Transformador beta1/química , Fator de Crescimento Transformador beta1/metabolismo , Animais , Anticorpos/imunologia , Sítios de Ligação , Brônquios/citologia , Células CHO , Cricetulus , Feminino , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Integrinas/imunologia , Ativação Linfocitária , Masculino , Vison , Ligação Proteica , Conformação Proteica em alfa-Hélice , Domínios e Motivos de Interação entre Proteínas , Linfócitos T Reguladores/imunologia
4.
Proc Natl Acad Sci U S A ; 116(41): 20428-20437, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31548410

RESUMO

Elastogenesis is a hierarchical process by which cells form functional elastic fibers, providing elasticity and the ability to regulate growth factor bioavailability in tissues, including blood vessels, lung, and skin. This process requires accessory proteins, including fibulin-4 and -5, and latent TGF binding protein (LTBP)-4. Our data demonstrate mechanisms in elastogenesis, focusing on the interaction and functional interdependence between fibulin-4 and LTBP-4L and its impact on matrix deposition and function. We show that LTBP-4L is not secreted in the expected extended structure based on its domain composition, but instead adopts a compact conformation. Interaction with fibulin-4 surprisingly induced a conformational switch from the compact to an elongated LTBP-4L structure. This conversion was only induced by fibulin-4 multimers associated with increased avidity for LTBP-4L; fibulin-4 monomers were inactive. The fibulin-4-induced conformational change caused functional consequences in LTBP-4L in terms of binding to other elastogenic proteins, including fibronectin and fibrillin-1, and of LTBP-4L assembly. A transient exposure of LTBP-4L with fibulin-4 was sufficient to stably induce conformational and functional changes; a stable complex was not required. These data define fibulin-4 as a molecular extracellular chaperone for LTBP-4L. The altered LTBP-4L conformation also promoted elastogenesis, but only in the presence of fibulin-4, which is required to escort tropoelastin onto the extended LTBP-4L molecule. Altogether, this study provides a dual mechanism for fibulin-4 in 1) inducing a stable conformational and functional change in LTBP-4L, and 2) promoting deposition of tropoelastin onto the elongated LTBP-4L.


Assuntos
Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/fisiologia , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/metabolismo , Animais , Células Cultivadas , Elastina , Fibronectinas/metabolismo , Humanos , Camundongos , Ligação Proteica , Conformação Proteica , Tropoelastina/metabolismo
5.
J Biol Chem ; 294(20): 8037-8045, 2019 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-30926607

RESUMO

The protease ADAMTS7 functions in the extracellular matrix (ECM) of the cardiovascular system. However, its physiological substrate specificity and mechanism of regulation remain to be explored. To address this, we conducted an unbiased substrate analysis using terminal amine isotopic labeling of substrates (TAILS). The analysis identified candidate substrates of ADAMTS7 in the human fibroblast secretome, including proteins with a wide range of functions, such as collagenous and noncollagenous extracellular matrix proteins, growth factors, proteases, and cell-surface receptors. It also suggested that autolysis occurs at Glu-729-Val-730 and Glu-732-Ala-733 in the ADAMTS7 Spacer domain, which was corroborated by N-terminal sequencing and Western blotting. Importantly, TAILS also identified proteolysis of the latent TGF-ß-binding proteins 3 and 4 (LTBP3/4) at a Glu-Val and Glu-Ala site, respectively. Using purified enzyme and substrate, we confirmed ADAMTS7-catalyzed proteolysis of recombinant LTBP4. Moreover, we identified multiple additional scissile bonds in an N-terminal linker region of LTBP4 that connects fibulin-5/tropoelastin and fibrillin-1-binding regions, which have an important role in elastogenesis. ADAMTS7-mediated cleavage of LTBP4 was efficiently inhibited by the metalloprotease inhibitor TIMP-4, but not by TIMP-1 and less efficiently by TIMP-2 and TIMP-3. As TIMP-4 expression is prevalent in cardiovascular tissues, we propose that TIMP-4 represents the primary endogenous ADAMTS7 inhibitor. In summary, our findings reveal LTBP4 as an ADAMTS7 substrate, whose cleavage may potentially impact elastogenesis in the cardiovascular system. We also identify TIMP-4 as a likely physiological ADAMTS7 inhibitor.


Assuntos
Proteínas ADAMTS , Fibroblastos/enzimologia , Proteínas de Ligação a TGF-beta Latente , Proteólise , Inibidores Teciduais de Metaloproteinases , Proteínas ADAMTS/química , Proteínas ADAMTS/genética , Proteínas ADAMTS/metabolismo , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Células HEK293 , Humanos , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/genética , Proteínas de Ligação a TGF-beta Latente/metabolismo , Domínios Proteicos , Proteômica , Inibidor Tecidual de Metaloproteinase-1/química , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Inibidores Teciduais de Metaloproteinases/química , Inibidores Teciduais de Metaloproteinases/genética , Inibidores Teciduais de Metaloproteinases/metabolismo , Tropoelastina/química , Tropoelastina/genética , Tropoelastina/metabolismo
6.
J Cell Sci ; 131(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29361522

RESUMO

Dysregulated secretion and extracellular activation of TGF-ß1 stimulates myofibroblasts to accumulate disordered and stiff extracellular matrix (ECM) leading to fibrosis. Fibronectin immobilizes latent TGF-ß-binding protein-1 (LTBP-1) and thus stores TGF-ß1 in the ECM. Because the ED-A fibronectin splice variant is prominently expressed during fibrosis and supports myofibroblast activation, we investigated whether ED-A promotes LTBP-1-fibronectin interactions. Using stiffness-tuneable substrates for human dermal fibroblast cultures, we showed that high ECM stiffness promotes expression and colocalization of LTBP-1 and ED-A-containing fibronectin. When rescuing fibronectin-depleted fibroblasts with specific fibronectin splice variants, LTBP-1 bound more efficiently to ED-A-containing fibronectin than to ED-B-containing fibronectin and fibronectin lacking splice domains. Function blocking of the ED-A domain using antibodies and competitive peptides resulted in reduced LTBP-1 binding to ED-A-containing fibronectin, reduced LTBP-1 incorporation into the fibroblast ECM and reduced TGF-ß1 activation. Similar results were obtained by blocking the heparin-binding stretch FNIII12-13-14 (HepII), adjacent to the ED-A domain in fibronectin. Collectively, our results suggest that the ED-A domain enhances association of the latent TGF-ß1 by promoting weak direct binding to LTBP-1 and by enhancing heparin-mediated protein interactions through HepII in fibronectin.


Assuntos
Fibronectinas/genética , Fibrose/genética , Proteínas de Ligação a TGF-beta Latente/genética , Fator de Crescimento Transformador beta1/genética , Animais , Proteínas de Transporte , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibronectinas/química , Fibrose/patologia , Células HEK293 , Humanos , Proteínas de Ligação a TGF-beta Latente/química , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Ligação Proteica/genética , Domínios Proteicos/genética , Isoformas de Proteínas/genética , Ratos
7.
Structure ; 25(8): 1208-1221.e5, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28669633

RESUMO

Fibrillin-1 (FBN1) mutations associated with Marfan syndrome lead to an increase in transforming growth factor ß (TGF-ß) activation in connective tissues resulting in pathogenic changes including aortic dilatation and dissection. Since FBN1 binds latent TGF-ß binding proteins (LTBPs), the major reservoir of TGF-ß in the extracellular matrix (ECM), we investigated the structural basis for the FBN1/LTBP1 interaction. We present the structure of a four-domain FBN1 fragment, EGF2-EGF3-Hyb1-cbEGF1 (FBN1E2cbEGF1), which reveals a near-linear domain organization. Binding studies demonstrate a bipartite interaction between a C-terminal LTBP1 fragment and FBN1E2cbEGF1, which lies adjacent to the latency-associated propeptide (LAP)/TGF-ß binding site of LTBP1. Modeling of the binding interface suggests that, rather than interacting along the longitudinal axis, LTBP1 anchors itself to FBN1 using two independent epitopes. As part of this mechanism, a flexible pivot adjacent to the FBN1/LTBP1 binding site allows LTBP1 to make contacts with different ECM networks while presumably facilitating a force-induced/traction-based TGF-ß activation mechanism.


Assuntos
Fibrilina-1/química , Proteínas de Ligação a TGF-beta Latente/química , Sítios de Ligação , Fibrilina-1/metabolismo , Humanos , Proteínas de Ligação a TGF-beta Latente/metabolismo , Simulação de Acoplamento Molecular , Ligação Proteica
8.
PLoS One ; 11(2): e0150358, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26918958

RESUMO

Latent TGFß binding proteins are extracellular matrix proteins that bind latent TGFß to form the large latent complex. Nonsynonymous polymorphisms in LTBP4, a member of the latent TGFß binding protein gene family, have been linked to several human diseases, underscoring the importance of TGFß regulation for a range of phenotypes. Because of strong linkage disequilibrium across the LTBP4 gene, humans have two main LTBP4 alleles that differ at four amino acid positions, referred to as IAAM and VTTT for the encoded residues. VTTT is considered the "risk" allele and associates with increased intracellular TGFß signaling and more deleterious phenotypes in muscular dystrophy and other diseases. We now evaluated LTBP4 nsSNPs in dilated cardiomyopathy, a distinct disorder associated with TGFß signaling. We stratified based on self-identified ethnicity and found that the LTBP4 VTTT allele is associated with increased risk of dilated cardiomyopathy in European Americans extending the diseases that associate with LTBP4 genotype. However, the association of LTBP4 SNPs with dilated cardiomyopathy was not observed in African Americans. To elucidate the mechanism by which LTBP4 genotype exerts this differential effect, TGFß's association with LTBP4 protein was examined. LTBP4 protein with the IAAM residues bound more latent TGFß compared to the LTBP4 VTTT protein. Together these data provide support that LTBP4 genotype exerts its effect through differential avidity for TGFß accounting for the differences in TGFß signaling attributed to these two alleles.


Assuntos
Cardiomiopatia Dilatada/genética , Etnicidade/genética , Haplótipos/genética , Proteínas de Ligação a TGF-beta Latente/genética , Fator de Crescimento Transformador beta1/metabolismo , Adulto , Negro ou Afro-Americano/genética , Alelos , Sequência de Aminoácidos , Cardiomiopatia Dilatada/metabolismo , Chicago/epidemiologia , Europa (Continente)/etnologia , Feminino , Sangue Fetal , Frequência do Gene , Genótipo , Células HEK293 , Humanos , Recém-Nascido , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/metabolismo , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Recombinantes de Fusão/metabolismo , Adulto Jovem
9.
PLoS One ; 10(8): e0135577, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26263555

RESUMO

Latent transforming growth factor-beta-1 binding protein-2 (LTBP-2) belongs to the fibrillin-LTBP superfamily of extracellular matrix proteins. LTBPs and fibrillins are involved in the sequestration and storage of latent growth factors, particularly transforming growth factor ß (TGF-ß), in tissues. Unlike other LTBPs, LTBP-2 does not covalently bind TGF-ß, and its molecular functions remain unclear. We are screening LTBP-2 for binding to other growth factors and have found very strong saturable binding to fibroblast growth factor-2 (FGF-2) (Kd = 1.1 nM). Using a series of recombinant LTBP-2 fragments a single binding site for FGF-2 was identified in a central region of LTBP-2 consisting of six tandem epidermal growth factor-like (EGF-like) motifs (EGFs 9-14). This region was also shown to contain a heparin/heparan sulphate-binding site. FGF-2 stimulation of fibroblast proliferation was completely negated by the addition of 5-fold molar excess of LTBP-2 to the assay. Confocal microscopy showed strong co-localisation of LTBP-2 and FGF-2 in fibrotic keloid tissue suggesting that the two proteins may interact in vivo. Overall the study indicates that LTBP-2 is a potent inhibitor of FGF-2 that may influence FGF-2 bioactivity during wound repair particularly in fibrotic tissues.


Assuntos
Sítios de Ligação , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/química , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Fibrilinas , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/metabolismo , Heparina/metabolismo , Humanos , Queloide/metabolismo , Proteínas de Ligação a TGF-beta Latente/farmacologia , Proteínas dos Microfilamentos/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/agonistas , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Proteínas Recombinantes , Pele/metabolismo
10.
Dis Model Mech ; 8(4): 403-15, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25713297

RESUMO

Recent studies have revealed an important role for LTBP-4 in elastogenesis. Its mutational inactivation in humans causes autosomal recessive cutis laxa type 1C (ARCL1C), which is a severe disorder caused by defects of the elastic fiber network. Although the human gene involved in ARCL1C has been discovered based on similar elastic fiber abnormalities exhibited by mice lacking the short Ltbp-4 isoform (Ltbp4S(-/-)), the murine phenotype does not replicate ARCL1C. We therefore inactivated both Ltbp-4 isoforms in the mouse germline to model ARCL1C. Comparative analysis of Ltbp4S(-/-) and Ltbp4-null (Ltbp4(-/-)) mice identified Ltbp-4L as an important factor for elastogenesis and postnatal survival, and showed that it has distinct tissue expression patterns and specific molecular functions. We identified fibulin-4 as a previously unknown interaction partner of both Ltbp-4 isoforms and demonstrated that at least Ltbp-4L expression is essential for incorporation of fibulin-4 into the extracellular matrix (ECM). Overall, our results contribute to the current understanding of elastogenesis and provide an animal model of ARCL1C.


Assuntos
Cútis Laxa/genética , Cútis Laxa/patologia , Genes Recessivos , Proteínas de Ligação a TGF-beta Latente/genética , Animais , Animais Recém-Nascidos , Aorta/anormalidades , Aorta/patologia , Cardiomegalia/complicações , Cardiomegalia/patologia , Tecido Elástico/metabolismo , Elastina/metabolismo , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Feminino , Inativação Gênica , Glicosilação , Ventrículos do Coração/patologia , Humanos , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/deficiência , Proteínas de Ligação a TGF-beta Latente/metabolismo , Pulmão/anormalidades , Pulmão/patologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Pele/patologia , Redução de Peso
11.
Sci Transl Med ; 6(259): 259ra144, 2014 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-25338755

RESUMO

Latent transforming growth factor-ß (TGFß) binding proteins (LTBPs) bind to inactive TGFß in the extracellular matrix. In mice, muscular dystrophy symptoms are intensified by a genetic polymorphism that changes the hinge region of LTBP, leading to increased proteolytic susceptibility and TGFß release. We have found that the hinge region of human LTBP4 was also readily proteolysed and that proteolysis could be blocked by an antibody to the hinge region. Transgenic mice were generated to carry a bacterial artificial chromosome encoding the human LTBP4 gene. These transgenic mice displayed larger myofibers, increased damage after muscle injury, and enhanced TGFß signaling. In the mdx mouse model of Duchenne muscular dystrophy, the human LTBP4 transgene exacerbated muscular dystrophy symptoms and resulted in weaker muscles with an increased inflammatory infiltrate and greater LTBP4 cleavage in vivo. Blocking LTBP4 cleavage may be a therapeutic strategy to reduce TGFß release and activity and decrease inflammation and muscle damage in muscular dystrophy.


Assuntos
Proteínas de Ligação a TGF-beta Latente/metabolismo , Distrofia Muscular Animal/metabolismo , Sequência de Aminoácidos , Animais , Cromossomos Artificiais Bacterianos/metabolismo , Fibrose , Células HEK293 , Humanos , Hipertrofia , Proteínas de Ligação a TGF-beta Latente/antagonistas & inibidores , Proteínas de Ligação a TGF-beta Latente/química , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular Animal/patologia , Serina Proteases/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Transgenes
12.
Semin Cell Dev Biol ; 32: 85-97, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24508081

RESUMO

Secreted cytokines of the TGFß family are found in all multicellular organisms and implicated in regulating fundamental cell behaviors such as proliferation, differentiation, migration and survival. Signal transduction involves complexes of specific type I and II receptor kinases that induce the nuclear translocation of Smad transcription factors to regulate target genes. Ligands of the BMP and Nodal subgroups act at a distance to specify distinct cell fates in a concentration-dependent manner. These signaling gradients are shaped by multiple factors, including proteases of the proprotein convertase (PC) family that hydrolyze one or several peptide bonds between an N-terminal prodomain and the C-terminal domain that forms the mature ligand. This review summarizes information on the proteolytic processing of TGFß and related precursors, and its spatiotemporal regulation by PCs during development and various diseases, including cancer. Available evidence suggests that the unmasking of receptor binding epitopes of TGFß is only one (and in some cases a non-essential) function of precursor processing. Future studies should consider the impact of proteolytic maturation on protein localization, trafficking and turnover in cells and in the extracellular space.


Assuntos
Precursores de Proteínas/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Humanos , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/metabolismo , Modelos Moleculares , Pró-Proteína Convertases/química , Pró-Proteína Convertases/metabolismo , Ligação Proteica , Precursores de Proteínas/química , Estrutura Terciária de Proteína , Fator de Crescimento Transformador beta/química
13.
PLoS One ; 9(1): e87125, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489852

RESUMO

Proteins from the LTBP/fibrillin family perform key structural and functional roles in connective tissues. LTBP1 forms the large latent complex with TGFß and its propeptide LAP, and sequesters the latent growth factor to the extracellular matrix. Bioinformatics studies suggest the main structural features of the LTBP1 C-terminus are conserved through evolution. NMR studies were carried out on three overlapping C-terminal fragments of LTBP1, comprising four domains with characterised homologues, cbEGF14, TB3, EGF3 and cbEGF15, and three regions with no homology to known structures. The NMR data reveal that the four domains adopt canonical folds, but largely lack the interdomain interactions observed with homologous fibrillin domains; the exception is the EGF3-cbEGF15 domain pair which has a well-defined interdomain interface. (15)N relaxation studies further demonstrate that the three interdomain regions act as flexible linkers, allowing a wide range of motion between the well-structured domains. This work is consistent with the LTBP1 C-terminus adopting a flexible "knotted rope" structure, which may facilitate cell matrix interactions, and the accessibility to proteases or other factors that could contribute to TGFß activation.


Assuntos
Proteínas de Ligação a TGF-beta Latente/química , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia Estrutural de Proteína
14.
Biomol NMR Assign ; 8(1): 159-63, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23494870

RESUMO

Latent TGFß binding protein 1 (LTBP1) is a large extracellular protein that has been shown to bind covalently to the propeptide of TGFß cytokines and form a large latent complex, which is then incapable of binding TGFß receptors. LTBP1 has also been demonstrated to interact with a number of insoluble extracellular matrix components, such as fibrillin, which may play a role in TGFß regulation. Here we present the backbone (1)H, (13)C and (15)N assignments for two EGF domains of human LTBP1, and flanking regions, together forming a 12 kDa protein fragment at the C-terminus of LTBP1. This region is of particular interest as it is postulated to be involved in interactions with fibrillin microfibrils.


Assuntos
Aminoácidos/química , Cálcio/metabolismo , Fator de Crescimento Epidérmico/química , Proteínas de Ligação a TGF-beta Latente/química , Ressonância Magnética Nuclear Biomolecular , Sequência de Aminoácidos , Isótopos de Carbono , Humanos , Hidrogênio , Dados de Sequência Molecular , Isótopos de Nitrogênio , Estrutura Terciária de Proteína
15.
Mol Vis ; 19: 333-47, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23401661

RESUMO

PURPOSE: To assess for the first time the possible contribution of latent transforming growth factor (TGF)-beta binding protein 2 (LTBP2), an extracellular matrix (ECM) protein that associates with fibrillin-1-containing microfibrils, to the etiology of primary open angle glaucoma (POAG) and pseudoexfoliation (PEX) syndrome. Mutations in LTBP2 have previously been shown to be the cause of primary congenital glaucoma (PCG) and other disorders that often manifest as secondary glaucoma. METHODS: All exons of LTBP2 were sequenced in the DNA of 42 unrelated patients with POAG and 48 unrelated patients with PEX syndrome. Contribution of candidate variations to disease was assessed by screening in control individuals and use of biochemical, bioinformatics, and evolutionary criteria, and in one case by segregation analysis within the family of a proband with POAG. Microscopy was performed on the skin of a patient with PEX syndrome whose condition developed into PEX glaucoma during the course of the study and on the skin of her son previously identified with PCG who harbored the same LTBP2 mutation. RESULTS: Among the 30 sequence variations observed in LTBP2, five found in five patients with POAG and two found in two patients with PEX glaucoma syndrome may contribute to their diseases. One of the mutations was observed in a patient with POAG and in a patient with PEX glaucoma syndrome. Light, fluorescent, and electron microscopy showed that a mutation present in one of the individuals affected with PEX glaucoma syndrome and in her son affected with PCG causes disruptions in the ECM. CONCLUSIONS: Some LTBP2 sequence variations can contribute to the etiology of POAG and PEX glaucoma syndrome. It is not expected that in these diseases LTBP2 mutations behave in a strictly Mendelian fashion with complete penetrance. In conjunction with recent findings, the results suggest that anomalies in the ECM are among the factors that can contribute to POAG and PEX glaucoma syndrome. LTBP2 and other related ECM protein coding genes should be screened in larger cohorts with these diseases, which are common disorders and important to the public health.


Assuntos
Síndrome de Exfoliação/genética , Glaucoma de Ângulo Aberto/genética , Proteínas de Ligação a TGF-beta Latente/genética , Mutação , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Estudos de Coortes , Análise Mutacional de DNA , Síndrome de Exfoliação/etiologia , Síndrome de Exfoliação/patologia , Matriz Extracelular/genética , Matriz Extracelular/patologia , Feminino , Variação Genética , Glaucoma de Ângulo Aberto/etiologia , Glaucoma de Ângulo Aberto/patologia , Humanos , Proteínas de Ligação a TGF-beta Latente/química , Masculino , Pessoa de Meia-Idade , Linhagem , Penetrância , Pele/patologia , Síndrome , Adulto Jovem
16.
Biochem Biophys Res Commun ; 432(1): 163-8, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23348228

RESUMO

Fucoidan, a sulfated, fucose-rich polysaccharide isolated from marine brown algae, has antifibrotic effects. We investigated the biologic effects of interactions of fucoidan with transforming growth factor-ß1 (TGF-ß1) and latent TGF-ß1 (LTGF-ß1). TGF-ß1 bound to fucoidan was unable to interact with its receptor. In agreement with this, fucoidan attenuated the cellular effect of TGF-ß1 as measured by phosphorylation of Smad2. Binding of fucoidan rendered LTGF-ß1 resistant to activation as follows. Fucoidan inhibited furin-like proprotein convertase-mediated activation of platelet LTGF-ß1 without suppression of the enzyme. In addition, acid- or heat-activation of small recombinant LTGF-ß1 and acid-activation of large LTGF-ß1 in cultured cell supernatant were also inhibited by fucoidan. Fucoidan is a mixture of polysaccharides of different sizes. As molecular weight of fucoidan increases, it had more inhibitory effects on TGF-ß1 and LTGF-ß1. In conclusion, inhibitions of LTGF-ß1 activation and TGF-ß1 receptor binding by fucoidan may in part account for its antifibrotic effects.


Assuntos
Proteínas de Ligação a TGF-beta Latente/antagonistas & inibidores , Proteínas de Ligação a TGF-beta Latente/metabolismo , Polissacarídeos/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Ácidos , Plaquetas/metabolismo , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Temperatura Alta , Humanos , Proteínas de Ligação a TGF-beta Latente/química , Peso Molecular , Polissacarídeos/metabolismo , Pró-Proteína Convertases/metabolismo , Proteínas Recombinantes/metabolismo
17.
J Cell Biochem ; 113(2): 410-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22223425

RESUMO

Latent transforming growth factor beta (TGF-ß) binding proteins (LTBPs) are large extracellular glycoproteins structurally similar to fibrillins. They perform intricate and important roles in the extracellular matrix (ECM) and perturbations of their function manifest as a wide range of diseases. LTBPs are major regulators of TGF-ß bioavailability and action. In addition, LTBPs interact with other ECM proteins-from cytokines to large multi-factorial aggregates like microfibrils and elastic fibers, affecting their genesis, structure, and performance. In the present article, we review recent advancements in the field and relate the complex roles of LTBP in development and homeostasis.


Assuntos
Proteínas de Ligação a TGF-beta Latente/metabolismo , Animais , Matriz Extracelular/metabolismo , Doenças Genéticas Inatas/genética , Humanos , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/genética , Mutação , Ligação Proteica , Estrutura Terciária de Proteína
18.
Nature ; 474(7351): 343-9, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21677751

RESUMO

Transforming growth factor (TGF)-ß is stored in the extracellular matrix as a latent complex with its prodomain. Activation of TGF-ß1 requires the binding of α(v) integrin to an RGD sequence in the prodomain and exertion of force on this domain, which is held in the extracellular matrix by latent TGF-ß binding proteins. Crystals of dimeric porcine proTGF-ß1 reveal a ring-shaped complex, a novel fold for the prodomain, and show how the prodomain shields the growth factor from recognition by receptors and alters its conformation. Complex formation between α(v)ß(6) integrin and the prodomain is insufficient for TGF-ß1 release. Force-dependent activation requires unfastening of a 'straitjacket' that encircles each growth-factor monomer at a position that can be locked by a disulphide bond. Sequences of all 33 TGF-ß family members indicate a similar prodomain fold. The structure provides insights into the regulation of a family of growth and differentiation factors of fundamental importance in morphogenesis and homeostasis.


Assuntos
Fator de Crescimento Transformador beta1/química , Fator de Crescimento Transformador beta1/metabolismo , Ativinas/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Síndrome de Camurati-Engelmann/genética , Linhagem Celular , Cristalografia por Raios X , Células HEK293 , Humanos , Integrinas/química , Integrinas/metabolismo , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Família Multigênica , Mutação/genética , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Estrutura Terciária de Proteína , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Suínos , Fator de Crescimento Transformador beta1/biossíntese , Fator de Crescimento Transformador beta1/genética
19.
Biochem J ; 433(2): 263-76, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21175431

RESUMO

Fibrillins and LTBPs [latent TGFß (transforming growth factor ß)-binding proteins] perform vital and complex roles in the extracellular matrix and are relevant to a wide range of human diseases. These proteins share a signature 'eight cysteine' or 'TB (TGFß-binding protein-like)' domain that is found nowhere else in the human proteome, and which has been shown to mediate a variety of protein-protein interactions. These include covalent binding of the TGFß propeptide, and RGD-directed interactions with a repertoire of integrins. TB domains are found interspersed with long arrays of EGF (epidermal growth factor)-like domains, which occur more widely in extracellular proteins, and also mediate binding to a large number of proteins and proteoglycans. In the present paper, newly available protein sequence information from a variety of sources is reviewed and related to published findings on the structure and function of fibrillins and LTBPs. These sequences give valuable insight into the evolution of TB domain proteins and suggest that the fibrillin domain organization emerged first, over 600 million years ago, prior to the divergence of Cnidaria and Bilateria, after which it has remained remarkably unchanged. Comparison of sequence features and domain organization in such a diverse group of organisms also provides important insights into how fibrillins and LTBPs might perform their roles in the extracellular matrix.


Assuntos
Evolução Molecular , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/metabolismo , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/metabolismo , Animais , Fibrilinas , Humanos , Proteínas de Ligação a TGF-beta Latente/genética , Proteínas dos Microfilamentos/genética , Ligação Proteica , Estrutura Terciária de Proteína
20.
Dev Dyn ; 240(1): 176-87, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21181942

RESUMO

Transforming Growth Factor ß (TGF-ß) is crucial for valve development and homeostasis. The long form of Latent TGF-ß binding protein 1 (LTBP1L) covalently binds all TGF-ß isoforms and regulates their bioavailability. Ltbp1L expression analysis during valvulogenesis revealed two patterns of Ltbp1L production: an early one (E9.5-11.5) associated with endothelial-to-mesenchymal transformation (EMT); and a late one (E12.5 to birth) contemporaneous with valve remodeling. Similarly, histological analysis of Ltbp1L(-/-) developing valves identified two different pathologies: generation of hypoplastic endocardial cushions in early valvulogenesis, followed by development of hyperplastic valves in late valvulogenesis. Ltbp1L promotes valve EMT, as Ltbp1L absence yields hypoplastic endocardial cushions in vivo and attenuated EMT in vitro. Ltbp1L(-/-) valve hyperplasia in late valvuogenesis represents a consequence of prolonged EMT. We demonstrate that Ltbp1L is a major regulator of Tgf-ß activity during valvulogenesis since its absence results in a perturbed Tgf-ß pathway that causes all Ltbp1L(-/-) valvular defects.


Assuntos
Valvas Cardíacas/embriologia , Proteínas de Ligação a TGF-beta Latente/fisiologia , Animais , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Cardiopatias Congênitas/genética , Valvas Cardíacas/anormalidades , Valvas Cardíacas/metabolismo , Hiperplasia , Proteínas de Ligação a TGF-beta Latente/química , Proteínas de Ligação a TGF-beta Latente/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Valva Mitral/anormalidades , Valva Mitral/embriologia , Valva Mitral/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Crescimento Transformadores/genética , Fatores de Crescimento Transformadores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...