Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Bone Miner Res ; 39(2): 177-189, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38477760

RESUMO

Bone histomorphometry is a well-established approach to assessing skeletal pathology, providing a standard evaluation of the cellular components, architecture, mineralization, and growth of bone tissue. However, it depends in part on the subjective interpretation of cellular morphology by an expert, which introduces bias. In addition, diseases like osteogenesis imperfecta (OI) and fibrous dysplasia are accompanied by changes in the morphology and function of skeletal tissue and cells, hindering consistent evaluation of some morphometric parameters and interpretation of the results. For instance, traditional histomorphometry combined with collagen turnover markers suggested that reduced bone formation in classical OI is accompanied by increased bone resorption. In contrast, the well-documented postpubertal reduction in fractures would be easier to explain by reduced bone resorption after puberty, highlighting the need for less ambiguous measurements. Here we propose an approach to histomorphometry based on in situ mRNA hybridization, which uses Col1a1 as osteoblast and Ctsk as osteoclast markers. This approach can be fully automated and eliminates subjective identification of bone surface cells. We validate these markers based on the expression of Bglap, Ibsp, and Acp5. Comparison with traditional histological and tartrate-resistant acid phosphatase staining of the same sections suggests that mRNA-based analysis is more reliable. Unlike inconclusive traditional histomorphometry of mice with α2(I)-Gly610 to Cys substitution in the collagen triple helix, mRNA-based measurements reveal reduced osteoclastogenesis in 11-wk-old animals consistent with the postpubertal catch-up osteogenesis observed by microCT. We optimize the technique for cryosections of mineralized bone and sections of paraffin-embedded decalcified tissue, simplifying and broadening its applications. We illustrate the application of the mRNA-based approach to human samples using the example of a McCune-Albright syndrome patient. By eliminating confounding effects of altered cellular morphology and the need for subjective morphological evaluation, this approach may provide a more reproducible and accessible evaluation of bone pathology.


Assuntos
Osso e Ossos , Colágeno Tipo I , Modelos Animais de Doenças , Osteogênese Imperfeita , Osteogênese Imperfeita/patologia , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/genética , Animais , Camundongos , Osso e Ossos/patologia , Osso e Ossos/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Osteoclastos/metabolismo , Osteoclastos/patologia , Puberdade , Osteoblastos/metabolismo , Osteoblastos/patologia , Biomarcadores/metabolismo , Osteogênese
2.
Matrix Biol ; 93: 79-94, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32562852

RESUMO

Efficient quality control and export of procollagen from the cell is crucial for extracellular matrix homeostasis, yet it is still incompletely understood. One of the debated questions is the role of a collagen-specific ER chaperone HSP47 in these processes. Most ER chaperones preferentially bind to unfolded polypeptide chains, enabling selective export of natively folded proteins from the ER after chaperone release. In contrast, HSP47 preferentially binds to the natively folded procollagen and is believed to be released only in the ER-Golgi intermediate compartment (ERGIC) or cis-Golgi. HSP47 colocalization with procollagen in punctate structures observed by immunofluorescence imaging of fixed cells has thus been interpreted as evidence for HSP47 export from the ER together with procollagen in transport vesicles destined for ERGIC or Golgi. To understand the mechanism of this co-trafficking and its physiological significance, we imaged the dynamics of fluorescently tagged type I procollagen and HSP47 punctate structures in live MC3T3 murine osteoblasts with up to 120 nm spatial and 500 ms time resolution. Contrary to the prevailing model, we discovered that most bona fide carriers delivering procollagen from ER exit sites (ERESs) to Golgi contained no HSP47, unless the RDEL signal for ER retention in HSP47 was deleted or mutated. These transport intermediates exhibited characteristic rapid, directional motion along microtubules, while puncta with colocalized HSP47 and procollagen similar to the ones described before had only limited, stochastic motion. Live cell imaging and fluorescence recovery after photobleaching revealed that the latter puncta (including the ones induced by ARF1 inhibition) were dilated regions of ER lumen, ERESs, or autophagic structures surrounded by lysosomal membranes. Procollagen was colocalized with HSP47 and ERGIC53 at ERESs. It was colocalized with ERGIC53 but not HSP47 in Golgi-bound transport intermediates. Our results suggest that procollagen and HSP47 sorting occurs at ERES before procollagen is exported from the ER in Golgi-bound transport intermediates, providing new insights into mechanisms of procollagen trafficking.


Assuntos
Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Choque Térmico HSP47/metabolismo , Osteoblastos/citologia , Pró-Colágeno/metabolismo , Células 3T3 , Animais , Lisossomos/metabolismo , Lectinas de Ligação a Manose/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Microscopia de Fluorescência , Osteoblastos/metabolismo , Transporte Proteico
3.
Matrix Biol ; 90: 20-39, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32112888

RESUMO

Null mutations in CRTAP or P3H1, encoding cartilage-associated protein and prolyl 3-hydroxylase 1, cause the severe bone dysplasias, types VII and VIII osteogenesis imperfecta. Lack of either protein prevents formation of the ER prolyl 3-hydroxylation complex, which catalyzes 3Hyp modification of types I and II collagen and also acts as a collagen chaperone. To clarify the role of the A1 3Hyp substrate site in recessive bone dysplasia, we generated knock-in mice with an α1(I)P986A substitution that cannot be 3-hydroxylated. Mutant mice have normal survival, growth, femoral breaking strength and mean bone mineralization. However, the bone collagen HP/LP crosslink ratio is nearly doubled in mutant mice, while collagen fibril diameter and bone yield energy are decreased. Thus, 3-hydroxylation of the A1 site α1(I)P986 affects collagen crosslinking and structural organization, but its absence does not directly cause recessive bone dysplasia. Our study suggests that the functions of the modification complex as a collagen chaperone are thus distinct from its role as prolyl 3-hydroxylase.


Assuntos
Substituição de Aminoácidos , Colágeno Tipo I/genética , Osteoblastos/citologia , Osteogênese Imperfeita/genética , Animais , Células Cultivadas , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Introdução de Genes , Humanos , Hidroxilação , Masculino , Camundongos , Osteoblastos/metabolismo , Osteogênese Imperfeita/metabolismo , Fenótipo
4.
PLoS One ; 13(7): e0200264, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29990383

RESUMO

OI is a clinically and genetically heterogeneous disorder characterized by bone fragility. More than 90% of patients are heterozygous for mutations in type I collagen genes, COL1A1 and COL1A2, and a common mutation is substitution for an obligatory glycine in the triple helical Gly-X-Y repeats. Few non-glycine substitutions in the triple helical domain have been reported; most result in Y-position substitutions of arginine by cysteine. Here, we investigated leucine and cysteine substitutions for one Y-position arginine, p.Arg958 (Arg780 in the triple helical domain) of proα1(I) chains that cause mild OI. We compared their effects with two substitutions for glycine located in close proximity. Like substitutions for glycine, those for arginine reduced the denaturation temperature of the whole molecule and caused asymmetric posttranslational overmodification of the chains. Circular dichroism and increased susceptibility to cleavage by MMP1, MMP2 and catalytic domain of MMP1 revealed significant destabilization of the triple helix near the collagenase cleavage site. On a cellular level, we observed slower triple helix folding and intracellular collagen retention, which disturbed the Endoplasmic Reticulum function and affected matrix deposition. Molecular dynamic modeling suggested that Arg780 substitutions disrupt the triple helix structure and folding by eliminating hydrogen bonds of arginine side chains, in addition to preventing HSP47 binding. The pathogenic effects of these non-glycine substitutions in bone are probably caused mostly by procollagen misfolding and its downstream effects.


Assuntos
Arginina/metabolismo , Colágeno Tipo I/metabolismo , Osteogênese Imperfeita/metabolismo , Pró-Colágeno/metabolismo , Dicroísmo Circular , Colágeno Tipo I/genética , Humanos , Mutação , Osteogênese Imperfeita/genética , Pró-Colágeno/genética , Dobramento de Proteína
5.
Hum Mol Genet ; 26(12): 2207-2217, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28419360

RESUMO

Collagen prolyl 4-hydroxylases (C-P4Hs) play a central role in the formation and stabilization of the triple helical domain of collagens. P4HA1 encodes the catalytic α(I) subunit of the main C-P4H isoenzyme (C-P4H-I). We now report human bi-allelic P4HA1 mutations in a family with a congenital-onset disorder of connective tissue, manifesting as early-onset joint hypermobility, joint contractures, muscle weakness and bone dysplasia as well as high myopia, with evidence of clinical improvement of motor function over time in the surviving patient. Similar to P4ha1 null mice, which die prenatally, the muscle tissue from P1 and P2 was found to have reduced collagen IV immunoreactivity at the muscle basement membrane. Patients were compound heterozygous for frameshift and splice site mutations leading to reduced, but not absent, P4HA1 protein level and C-P4H activity in dermal fibroblasts compared to age-matched control samples. Differential scanning calorimetry revealed reduced thermal stability of collagen in patient-derived dermal fibroblasts versus age-matched control samples. Mutations affecting the family of C-P4Hs, and in particular C-P4H-I, should be considered in patients presenting with congenital connective tissue/myopathy overlap disorders with joint hypermobility, contractures, mild skeletal dysplasia and high myopia.


Assuntos
Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Prolil Hidroxilases/genética , Animais , Membrana Basal/metabolismo , Osso e Ossos/metabolismo , Criança , Colágeno Tipo IV/genética , Tecido Conjuntivo , Humanos , Masculino , Camundongos , Camundongos Knockout , Músculos/metabolismo , Mutação , Osteocondrodisplasias/genética , Prolil Hidroxilases/metabolismo , Tendões/metabolismo
6.
Nat Commun ; 7: 11920, 2016 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-27380894

RESUMO

Osteogenesis imperfecta (OI) is a collagen-related bone dysplasia. We identified an X-linked recessive form of OI caused by defects in MBTPS2, which encodes site-2 metalloprotease (S2P). MBTPS2 missense mutations in two independent kindreds with moderate/severe OI cause substitutions at highly conserved S2P residues. Mutant S2P has normal stability, but impaired functioning in regulated intramembrane proteolysis (RIP) of OASIS, ATF6 and SREBP transcription factors, consistent with decreased proband secretion of type I collagen. Further, hydroxylation of the collagen lysine residue (K87) critical for crosslinking is reduced in proband bone tissue, consistent with decreased lysyl hydroxylase 1 in proband osteoblasts. Reduced collagen crosslinks presumptively undermine bone strength. Also, proband osteoblasts have broadly defective differentiation. These mutations provide evidence that RIP plays a fundamental role in normal bone development.


Assuntos
Membrana Celular/patologia , Colágeno Tipo I/genética , Metaloendopeptidases/genética , Mutação de Sentido Incorreto , Osteoblastos/metabolismo , Osteogênese Imperfeita/genética , Fator 6 Ativador da Transcrição/genética , Fator 6 Ativador da Transcrição/metabolismo , Adulto , Idoso , Diferenciação Celular , Membrana Celular/metabolismo , Colágeno Tipo I/deficiência , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Genes Recessivos , Humanos , Hidroxilação , Masculino , Metaloendopeptidases/metabolismo , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Osteoblastos/patologia , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/patologia , Linhagem , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Proteólise , Índice de Gravidade de Doença , Proteínas de Ligação a Elemento Regulador de Esterol/genética , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo
7.
J Bone Miner Res ; 31(8): 1608-1616, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26925839

RESUMO

Glycine (Gly) substitutions in collagen Gly-X-Y repeats disrupt folding of type I procollagen triple helix and cause severe bone fragility and malformations (osteogenesis imperfecta [OI]). However, these mutations do not elicit the expected endoplasmic reticulum (ER) stress response, in contrast to other protein-folding diseases. Thus, it has remained unclear whether cell stress and osteoblast malfunction contribute to the bone pathology caused by Gly substitutions. Here we used a mouse with a Gly610 to cysteine (Cys) substitution in the procollagen α2(I) chain to show that misfolded procollagen accumulation in the ER leads to an unusual form of cell stress, which is neither a conventional unfolded protein response (UPR) nor ER overload. Despite pronounced ER dilation, there is no upregulation of binding immunoglobulin protein (BIP) expected in the UPR and no activation of NF-κB signaling expected in the ER overload. Altered expression of ER chaperones αB crystalline and HSP47, phosphorylation of EIF2α, activation of autophagy, upregulation of general stress response protein CHOP, and osteoblast malfunction reveal some other adaptive response to the ER disruption. We show how this response alters differentiation and function of osteoblasts in culture and in vivo. We demonstrate that bone matrix deposition by cultured osteoblasts is rescued by activation of misfolded procollagen autophagy, suggesting a new therapeutic strategy for OI. © 2016 American Society for Bone and Mineral Research.


Assuntos
Colágeno Tipo I/genética , Mutação/genética , Osteoblastos/metabolismo , Osteogênese Imperfeita/patologia , Pró-Colágeno/química , Pró-Colágeno/metabolismo , Dobramento de Proteína , Estresse Fisiológico , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Embrião de Mamíferos/patologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Matriz Extracelular/metabolismo , Camundongos Endogâmicos C57BL , Osteoblastos/patologia , Osteoblastos/ultraestrutura , Osteogênese Imperfeita/metabolismo , Processamento de Proteína Pós-Traducional , Proteólise
8.
Am J Hum Genet ; 97(4): 521-34, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26365339

RESUMO

The evolutionarily conserved transmembrane anterior posterior transformation 1 protein, encoded by TAPT1, is involved in murine axial skeletal patterning, but its cellular function remains unknown. Our study demonstrates that TAPT1 mutations underlie a complex congenital syndrome, showing clinical overlap between lethal skeletal dysplasias and ciliopathies. This syndrome is characterized by fetal lethality, severe hypomineralization of the entire skeleton and intra-uterine fractures, and multiple congenital developmental anomalies affecting the brain, lungs, and kidneys. We establish that wild-type TAPT1 localizes to the centrosome and/or ciliary basal body, whereas defective TAPT1 mislocalizes to the cytoplasm and disrupts Golgi morphology and trafficking and normal primary cilium formation. Knockdown of tapt1b in zebrafish induces severe craniofacial cartilage malformations and delayed ossification, which is shown to be associated with aberrant differentiation of cranial neural crest cells.


Assuntos
Cílios/genética , Transtornos da Motilidade Ciliar/genética , Anormalidades Craniofaciais/genética , Proteínas de Membrana/genética , Mutação/genética , Ossificação Heterotópica/genética , Osteocondrodisplasias/genética , Sequência de Aminoácidos , Animais , Padronização Corporal , Diferenciação Celular , Movimento Celular , Cílios/metabolismo , Cílios/patologia , Embrião não Mamífero/anormalidades , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ , Masculino , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Crista Neural/citologia , Crista Neural/metabolismo , Linhagem , Transporte Proteico , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Peixe-Zebra/embriologia , Peixe-Zebra/genética
9.
PLoS Genet ; 10(6): e1004465, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24968150

RESUMO

Cyclophilin B (CyPB), encoded by PPIB, is an ER-resident peptidyl-prolyl cis-trans isomerase (PPIase) that functions independently and as a component of the collagen prolyl 3-hydroxylation complex. CyPB is proposed to be the major PPIase catalyzing the rate-limiting step in collagen folding. Mutations in PPIB cause recessively inherited osteogenesis imperfecta type IX, a moderately severe to lethal bone dysplasia. To investigate the role of CyPB in collagen folding and post-translational modifications, we generated Ppib-/- mice that recapitulate the OI phenotype. Knock-out (KO) mice are small, with reduced femoral areal bone mineral density (aBMD), bone volume per total volume (BV/TV) and mechanical properties, as well as increased femoral brittleness. Ppib transcripts are absent in skin, fibroblasts, femora and calvarial osteoblasts, and CyPB is absent from KO osteoblasts and fibroblasts on western blots. Only residual (2-11%) collagen prolyl 3-hydroxylation is detectable in KO cells and tissues. Collagen folds more slowly in the absence of CyPB, supporting its rate-limiting role in folding. However, treatment of KO cells with cyclosporine A causes further delay in folding, indicating the potential existence of another collagen PPIase. We confirmed and extended the reported role of CyPB in supporting collagen lysyl hydroxylase (LH1) activity. Ppib-/- fibroblast and osteoblast collagen has normal total lysyl hydroxylation, while increased collagen diglycosylation is observed. Liquid chromatography/mass spectrometry (LC/MS) analysis of bone and osteoblast type I collagen revealed site-specific alterations of helical lysine hydroxylation, in particular, significantly reduced hydroxylation of helical crosslinking residue K87. Consequently, underhydroxylated forms of di- and trivalent crosslinks are strikingly increased in KO bone, leading to increased total crosslinks and decreased helical hydroxylysine- to lysine-derived crosslink ratios. The altered crosslink pattern was associated with decreased collagen deposition into matrix in culture, altered fibril structure in tissue, and reduced bone strength. These studies demonstrate novel consequences of the indirect regulatory effect of CyPB on collagen hydroxylation, impacting collagen glycosylation, crosslinking and fibrillogenesis, which contribute to maintaining bone mechanical properties.


Assuntos
Colágeno Tipo I/genética , Ciclofilinas/genética , Osteogênese Imperfeita/genética , Processamento de Proteína Pós-Traducional/genética , Animais , Colágeno/química , Colágeno/genética , Colágeno/metabolismo , Colágeno Tipo I/química , Colágeno Tipo I/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/patologia , Genes Recessivos , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mutação , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/patologia , Dobramento de Proteína
10.
Arch Biochem Biophys ; 535(2): 111-4, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23541862

RESUMO

In normal soft tissues, collagen is degraded primarily by collagenases from the matrix metalloproteinase family. Yet, collagenase-like activity of tumor-associated isoforms of other enzymes might be involved in cancer invasion as well. In the present study, we systematically examined collagen degradation by non-sulfated isoforms of trypsins, which were proposed to possess such an activity. We found that non-sulfated trypsin-1, -2, and -3 were able to cleave non-helical and unfolded regions of collagen chains but not the intact triple helix, similar to sulfated trypsins produced by the pancreas. Trypsin-2 sulfation did not affect the cleavage rate either. An apparent triple helix cleavage by tumor-associated trypsin-2 reported earlier likely occurred after triple helix unfolding during sample denaturation for gel electrophoresis. Nevertheless, tumor-associated trypsins might be important for releasing collagen from fibers through telopeptide cleavage as well as for degrading unfolded collagen chains, e.g. after initial cleavage and destabilization of triple helices by collagenases.


Assuntos
Colágeno Tipo I/química , Neoplasias/química , Tripsina/química , Humanos , Isoenzimas/química , Desnaturação Proteica , Estrutura Secundária de Proteína , Desdobramento de Proteína
11.
Am J Hum Genet ; 92(4): 565-74, 2013 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-23499309

RESUMO

We report that hypofunctional alleles of WNT1 cause autosomal-recessive osteogenesis imperfecta, a congenital disorder characterized by reduced bone mass and recurrent fractures. In consanguineous families, we identified five homozygous mutations in WNT1: one frameshift mutation, two missense mutations, one splice-site mutation, and one nonsense mutation. In addition, in a family affected by dominantly inherited early-onset osteoporosis, a heterozygous WNT1 missense mutation was identified in affected individuals. Initial functional analysis revealed that altered WNT1 proteins fail to activate canonical LRP5-mediated WNT-regulated ß-catenin signaling. Furthermore, osteoblasts cultured in vitro showed enhanced Wnt1 expression with advancing differentiation, indicating a role of WNT1 in osteoblast function and bone development. Our finding that homozygous and heterozygous variants in WNT1 predispose to low-bone-mass phenotypes might advance the development of more effective therapeutic strategies for congenital forms of bone fragility, as well as for common forms of age-related osteoporosis.


Assuntos
Densidade Óssea/genética , Osso e Ossos/patologia , Mutação/genética , Osteogênese Imperfeita/genética , Osteoporose/genética , Proteína Wnt1/genética , Animais , Sequência de Bases , Células Cultivadas , Criança , Pré-Escolar , Feminino , Heterozigoto , Humanos , Recém-Nascido , Proteínas Relacionadas a Receptor de LDL/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteogênese Imperfeita/patologia , Osteoporose/patologia , Linhagem , Fenótipo , Gravidez
12.
J Biol Chem ; 285(29): 22276-81, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20463013

RESUMO

Type I collagen cleavage is crucial for tissue remodeling, but its homotrimeric isoform is resistant to all collagenases. The homotrimers occur in fetal tissues, fibrosis, and cancer, where their collagenase resistance may play an important physiological role. To understand the mechanism of this resistance, we studied interactions of alpha1(I)(3) homotrimers and normal alpha1(I)(2)alpha2(I) heterotrimers with fibroblast collagenase (MMP-1). Similar MMP-1 binding to the two isoforms and similar cleavage efficiency of unwound alpha1(I) and alpha2(I) chains suggested increased stability and less efficient unwinding of the homotrimer triple helix at the collagenase cleavage site. The unwinding, necessary for placing individual chains inside the catalytic cleft of the enzyme, was the rate-limiting cleavage step for both collagen isoforms. Comparative analysis of the homo- and heterotrimer cleavage kinetics revealed that MMP-1 binding promotes stochastic helix unwinding, resolving the controversy between different models of collagenase action.


Assuntos
Colágeno Tipo I/química , Colágeno Tipo I/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Multimerização Proteica , Animais , Colágenos Fibrilares/metabolismo , Humanos , Cinética , Camundongos , Microscopia Confocal , Modelos Biológicos , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Temperatura
13.
Cancer Res ; 70(11): 4366-74, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20460529

RESUMO

Collagen fibers affect metastasis in two opposing ways, by supporting invasive cells but also by generating a barrier to invasion. We hypothesized that these functions might be performed by different isoforms of type I collagen. Carcinomas are reported to contain alpha1(I)(3) homotrimers, a type I collagen isoform normally not present in healthy tissues, but the role of the homotrimers in cancer pathophysiology is unclear. In this study, we found that these homotrimers were resistant to all collagenolytic matrix metalloproteinases (MMP). MMPs are massively produced and used by cancer cells and cancer-associated fibroblasts for degrading stromal collagen at the leading edge of tumor invasion. The MMP-resistant homotrimers were produced by all invasive cancer cell lines tested, both in culture and in tumor xenografts, but they were not produced by cancer-associated fibroblasts, thereby comprising a specialized fraction of tumor collagen. We observed the homotrimer fibers to be resistant to pericellular degradation, even upon stimulation of the cells with proinflammatory cytokines. Furthermore, we confirmed an enhanced proliferation and migration of invasive cancer cells on the surface of homotrimeric versus normal (heterotrimeric) type I collagen fibers. In summary, our findings suggest that invasive cancer cells may use homotrimers for building MMP-resistant invasion paths, supporting local proliferation and directed migration of the cells whereas surrounding normal stromal collagens are cleaved. Because the homotrimers are universally secreted by cancer cells and deposited as insoluble, MMP-resistant fibers, they offer an appealing target for cancer diagnostics and therapy.


Assuntos
Colágeno Tipo I/metabolismo , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Metaloproteinases da Matriz/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Movimento Celular/fisiologia , Fibroblastos/metabolismo , Fibrossarcoma/enzimologia , Humanos , Isoenzimas , Camundongos , Camundongos Nus , Invasividade Neoplásica
14.
PLoS One ; 2(10): e1029, 2007 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-17925877

RESUMO

Fibers composed of type I collagen triple helices form the organic scaffold of bone and many other tissues, yet the energetically preferred conformation of type I collagen at body temperature is a random coil. In fibers, the triple helix is stabilized by neighbors, but how does it fold? The observations reported here reveal surprising features that may represent a new paradigm for folding of marginally stable proteins. We find that human procollagen triple helix spontaneously folds into its native conformation at 30-34 degrees C but not at higher temperatures, even in an environment emulating Endoplasmic Reticulum (ER). ER-like molecular crowding by nonspecific proteins does not affect triple helix folding or aggregation of unfolded chains. Common ER chaperones may prevent aggregation and misfolding of procollagen C-propeptide in their traditional role of binding unfolded polypeptide chains. However, such binding only further destabilizes the triple helix. We argue that folding of the triple helix requires stabilization by preferential binding of chaperones to its folded, native conformation. Based on the triple helix folding temperature measured here and published binding constants, we deduce that HSP47 is likely to do just that. It takes over 20 HSP47 molecules to stabilize a single triple helix at body temperature. The required 50-200 microM concentration of free HSP47 is not unusual for heat-shock chaperones in ER, but it is 100 times higher than used in reported in vitro experiments, which did not reveal such stabilization.


Assuntos
Chaperonas Moleculares/química , Pró-Colágeno/química , Bioquímica/métodos , Varredura Diferencial de Calorimetria , Dicroísmo Circular , Proteínas de Choque Térmico HSP47/química , Humanos , Íons , Pepsina A/química , Conformação Proteica , Desnaturação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Temperatura , Termodinâmica
15.
Hum Mutat ; 28(4): 396-405, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17206620

RESUMO

The most common mutations in type I collagen causing types II-IV osteogenesis imperfecta (OI) result in substitution for glycine in a Gly-Xaa-Yaa triplet by another amino acid. We delineated a Y-position substitution in a small pedigree with a combined OI/Ehlers-Danlos Syndrome (EDS) phenotype, characterized by moderately decreased DEXA z-score (-1.3 to -2.6), long bone fractures, and large-joint hyperextensibility. Affected individuals have an alpha1(I)R888C (p.R1066C) substitution in one COL1A1 allele. Polyacrylamide gel electrophoresis (PAGE) of [(3)H]-proline labeled steady-state collagen reveals slight overmodification of the alpha1(I) monomer band, much less than expected for a substitution of a neighboring glycine residue, and a faint alpha1(I) dimer. Dimers form in about 10% of proband type I collagen. Dimer formation is inefficient compared to a possible 25%, probably because the SH-side chains have less proximity in this Y-position than when substituting for a glycine. Theoretical stability calculations, differential scanning calorimetry (DSC) thermograms, and thermal denaturation curves showed only weak local destabilization from the Y-position substitution in one or two chains of a collagen helix, but greater destabilization is seen in collagen containing dimers. Y-position collagen dimers cause kinking of the helix, resulting in a register shift that is propagated the full length of the helix and causes resistance to procollagen processing by N-proteinase. Collagen containing the Y-position substitution is incorporated into matrix deposited in culture, including immaturely and maturely cross-linked fractions. In vivo, proband dermal fibrils have decreased density and increased diameter compared to controls, with occasional aggregate formation. This report on Y-position substitutions in type I collagen extends the range of phenotypes caused by nonglycine substitutions and shows that, similar to X- and Y-position substitutions in types II and III collagen, the phenotypes resulting from nonglycine substitutions in type I collagen are distinct from those caused by glycine substitutions.


Assuntos
Colágeno Tipo I/genética , Síndrome de Ehlers-Danlos/genética , Osteogênese Imperfeita/genética , Adulto , Sequência de Aminoácidos , Substituição de Aminoácidos , Células Cultivadas , Criança , Colágeno Tipo I/química , Colágeno Tipo I/ultraestrutura , Cisteína/genética , Dimerização , Síndrome de Ehlers-Danlos/diagnóstico , Humanos , Lactente , Masculino , Microscopia Eletrônica de Transmissão , Mutação de Sentido Incorreto , Osteogênese Imperfeita/diagnóstico , Linhagem , Fenótipo , Estrutura Terciária de Proteína , Análise de Sequência de Proteína
16.
J Biol Chem ; 280(19): 19259-69, 2005 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-15728585

RESUMO

Patients with OI/EDS form a distinct subset of osteogenesis imperfecta (OI) patients. In addition to skeletal fragility, they have characteristics of Ehlers-Danlos syndrome (EDS). We identified 7 children with types III or IV OI, plus severe large and small joint laxity and early progressive scoliosis. In each child with OI/EDS, we identified a mutation in the first 90 residues of the helical region of alpha1(I) collagen. These mutations prevent or delay removal of the procollagen N-propeptide by purified N-proteinase (ADAMTS-2) in vitro and in pericellular assays. The mutant pN-collagen which results is efficiently incorporated into matrix by cultured fibroblasts and osteoblasts and is prominently present in newly incorporated and immaturely cross-linked collagen. Dermal collagen fibrils have significantly reduced cross-sectional diameters, corroborating incorporation of pN-collagen into fibrils in vivo. Differential scanning calorimetry revealed that these mutant collagens are less stable than the corresponding procollagens, which is not seen with other type I collagen helical mutations. These mutations disrupt a distinct folding region of high thermal stability in the first 90 residues at the amino end of type I collagen and alter the secondary structure of the adjacent N-proteinase cleavage site. Thus, these OI/EDS collagen mutations are directly responsible for the bone fragility of OI and indirectly responsible for EDS symptoms, by interference with N-propeptide removal.


Assuntos
Colágeno Tipo I/genética , Síndrome de Ehlers-Danlos/genética , Mutação , Osteogênese Imperfeita/genética , Proteínas ADAM , Proteínas ADAMTS , Proteína ADAMTS4 , Adolescente , Adulto , Sequência de Aminoácidos , Varredura Diferencial de Calorimetria , Células Cultivadas , Pré-Escolar , Colágeno/metabolismo , Reagentes de Ligações Cruzadas/farmacologia , Análise Mutacional de DNA , Eletroforese em Gel de Poliacrilamida , Matriz Extracelular/metabolismo , Feminino , Fibroblastos/citologia , Temperatura Alta , Humanos , Lactente , Masculino , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Osteoblastos/metabolismo , Peptídeos/química , Fenótipo , Pró-Colágeno N-Endopeptidase/metabolismo , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Pele/citologia , Pele/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA