Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Matern Child Nutr ; 19(1): e13417, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36111423

RESUMO

Inadequate diet and frequent symptomatic infections are considered major causes of growth stunting in low-income countries, but interventions targeting these risk factors have achieved limited success. Asymptomatic infections can restrict growth, but little is known about their role in global stunting prevalence. We investigated factors related to length-for-age Z-score (LAZ) at 24 months by constructing an interconnected network of various infections, biomarkers of inflammation (as assessed by alpha-1-acid glycoprotein [AGP]), and growth (insulin-like growth factor 1 [IGF-1] and collagen X biomarker [CXM]) at 18 months, as well as other children, maternal, and household level factors. Among 604 children, there was a continuous decline in mean LAZ and increased mean length deficit from birth to 24 months. At 18 months of age, the percentage of asymptomatic children who carried each pathogen was: 84.5% enterovirus, 15.5% parechovirus, 7.7% norovirus, 4.6% rhinovirus, 0.6% rotavirus, 69.6% Campylobacter, 53.8% Giardia lamblia, 11.9% malaria parasites, 10.2% Shigella, and 2.7% Cryptosporidium. The mean plasma IGF-1 concentration was 12.5 ng/ml and 68% of the children had systemic inflammation (plasma AGP concentration >1 g/L). Shigella infection was associated with lower LAZ at 24 months through both direct and indirect pathways, whereas enterovirus, norovirus, Campylobacter, Cryptosporidium, and malaria infections were associated with lower LAZ at 24 months indirectly, predominantly through increased systemic inflammation and reduced plasma IGF-1 and CXM concentration at 18 months.


Assuntos
Criptosporidiose , Cryptosporidium , Malária , Pré-Escolar , Humanos , Lactente , Infecções Assintomáticas/epidemiologia , Biomarcadores , Cryptosporidium/metabolismo , Transtornos do Crescimento/epidemiologia , Inflamação , Fator de Crescimento Insulin-Like I
2.
Calcif Tissue Int ; 111(1): 66-72, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35275235

RESUMO

Collagen X marker (CXM) is a degradation fragment of collagen type X. It is a real-time biomarker of height velocity with established norms. Plasma C-type natriuretic peptide (CNP) and NTproCNP levels have also been found to correlate with growth velocity in the general population and are elevated in individuals with achondroplasia compared with age- and sex-matched controls. Collagen X marker levels in people with fibroblast growth factor receptor 3 (FGFR3)-opathies have never been systematically measured. The objective of this study was to measure CXM in a population of dwarfism caused by FGFR3-opathies. Using the same cohort in which CNP and NTproCNP levels were previously measured, archived serum aliquots from 63 children with achondroplasia, six with hypochondroplasia, and two with thanatophoric dysplasia had CXM concentrations measured. Results were plotted against age- and sex-specific norms, and standard deviation scores were plotted for comparison between clinical diagnoses. CXM levels were significantly decreased (p < 0.0001) in children with achondroplasia compared with age- and sex-matched controls. Temporal patterns of change in CXM levels were sex-dependent. As the FGFR3 pathway was more constitutively active, CXM levels decreased. New tools are emerging to study impact of skeletal dysplasia on growth plate regulation and function.


Assuntos
Acondroplasia , Deformidades Congênitas dos Membros , Displasia Tanatofórica , Biomarcadores , Criança , Colágeno Tipo X , Feminino , Humanos , Masculino
3.
Clin Pharmacokinet ; 61(2): 263-280, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34431071

RESUMO

BACKGROUND AND OBJECTIVE: Vosoritide, an analog of C-type natriuretic peptide, has been developed for the treatment of children with achondroplasia. The pharmacokinetics of vosoritide and relationships between plasma exposure and efficacy, biomarkers, and safety endpoints were evaluated in a phase II, open-label, dose-escalation study (N = 35 patients aged 5-14 years who received daily subcutaneous injections for 24 months) and a phase III, double-blind, placebo-controlled study (N = 60 patients aged 5-18 years randomized to receive daily subcutaneous injections for 52 weeks). METHODS: Pharmacokinetic parameters for both studies were obtained from non-compartmental analysis. Potential correlations between vosoritide exposure and changes in annualized growth velocity, collagen type X marker (CXM; a biomarker of endochondral ossification), cyclic guanosine monophosphate (cGMP; a biomarker of pharmacological activity), heart rate, and systolic and diastolic blood pressures were then evaluated. RESULTS: The exposure-response relationships for changes in both annualized growth velocity and the CXM biomarker saturated at 15 µg/kg, while systemic pharmacological activity, as measured by urinary cGMP, was near maximal or saturated at exposures obtained at the highest dose studied (i.e. 30 µg/kg). This suggested that the additional bioactivity was likely in tissues not related to endochondral bone formation. In the phase III study, following subcutaneous administration at the recommended dose of 15 µg/kg to patients with achondroplasia aged 5-18 years, vosoritide was rapidly absorbed with a median time to maximal plasma concentration (Cmax) of 15 minutes, and cleared with a mean half-life of 27.9 minutes after 52 weeks of treatment. Vosoritide exposure (Cmax and area under the concentration-time curve [AUC]) was consistent across visits. No evidence of accumulation with once-daily dosing was observed. Total anti-vosoritide antibody (TAb) responses were detected in the serum of 25 of 60 (42%) treated patients in the phase III study, with no apparent impact of TAb development noted on annualized growth velocity or vosoritide exposure. Across the exposure range obtained with 15 µg/kg in the phase III study, no meaningful correlations between vosoritide plasma exposure and changes in annualized growth velocity or CXM, or changes from predose heart rate, and systolic or diastolic blood pressures were observed. CONCLUSIONS: The results support the recommended dose of vosoritide 15 µg/kg for once-daily subcutaneous administration in patients with achondroplasia aged ≥ 5 years whose epiphyses are not closed. CLINICAL TRIALS REGISTRATION: NCT02055157, NCT03197766, and NCT01603095.


Assuntos
Acondroplasia , Peptídeo Natriurético Tipo C , Acondroplasia/induzido quimicamente , Acondroplasia/tratamento farmacológico , Adolescente , Área Sob a Curva , Biomarcadores , Criança , Pré-Escolar , Método Duplo-Cego , Humanos , Injeções Subcutâneas , Peptídeo Natriurético Tipo C/análogos & derivados , Peptídeo Natriurético Tipo C/farmacocinética , Peptídeo Natriurético Tipo C/uso terapêutico
4.
J Clin Endocrinol Metab ; 106(1): e255-e264, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33034649

RESUMO

CONTEXT: Height velocity (HV) is difficult to assess because growth is very slow. The current practice of calculating it from measurements taken at several-month intervals is insufficient for managing children with growth disorders. We identified a bone growth by-product (collagen X biomarker, CXM) in blood that in preliminary analysis in healthy children correlated strongly with conventionally determined HV and displayed a pattern resembling published norms for HV vs age. OBJECTIVE: The goal was to confirm our initial observations supporting the utility of CXM as an HV biomarker in a larger number of individuals and establish working reference ranges for future studies. DESIGN, SETTINGS, AND PARTICIPANTS: CXM was assessed in archived blood samples from 302 healthy children and 10 healthy adults yielding 961 CXM measurements. A total of 432 measurements were plotted by age, and sex-specific reference ranges were calculated. Serial values from 116 participants were plotted against observed HV. Matched plasma, serum, and dried blood spot readings were compared. RESULTS: A correlation of blood CXM with conventional HV was confirmed. Scatter plots of CXM vs age showed a similar pattern to current HV norms, and CXM levels demarcated the pubertal growth spurt both in girls and boys. CXM levels differed little in matched serum, plasma, and dried blood spot samples. CONCLUSIONS: Blood CXM offers a potential means to estimate HV in real time. Our results establish sex-specific, working reference ranges for assessing skeletal growth, especially over time. CXM stability in stored samples makes it well suited for retrospective studies.


Assuntos
Estatura/fisiologia , Desenvolvimento Infantil/fisiologia , Colágeno Tipo X/sangue , Adolescente , Biomarcadores/análise , Biomarcadores/sangue , Desenvolvimento Ósseo/fisiologia , Criança , Pré-Escolar , Colágeno Tipo X/análise , Endocrinologia/métodos , Endocrinologia/normas , Feminino , Gráficos de Crescimento , Humanos , Lactente , Masculino , Padrões de Prática Médica/normas , Padrões de Referência , Valores de Referência , Estados Unidos , Adulto Jovem
5.
J Orthop Res ; 39(1): 53-62, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32533783

RESUMO

Currently, there are no standardized methods for quantitatively measuring fracture repair. Physicians rely on subjective physical examinations and qualitative evaluation of radiographs to detect mineralized tissue. Since most fractures heal indirectly through a cartilage intermediate, these tools are limited in their diagnostic utility of early repair. Prior to converting to the bone, cartilage undergoes hypertrophic maturation, characterized by the deposition of a provisional collagen X matrix. The objective of this study was to characterize the kinetics of a novel collagen X biomarker relative to other biological measurements of fracture healing using a murine model of endochondral fracture repair in which a closed, mid-shaft tibia fracture was created using the classic drop-weight technique. Serum was collected 5 to 42 days post-fracture in male and female mice and compared to uninjured controls (n = 8-12). Collagen X in the serum was quantified using a recently validated ELISA-based bioassay ("Cxm")1 and compared to genetic and histological markers of fracture healing and inflammation. We found the Cxm biomarker reliably increased from baseline to a statistically unique peak 14 days post-fracture that then resolved to pre-fracture levels by 3 weeks following injury. The shape and timing of the Cxm curve followed the genetic and histological expression of collagen X but did not show a strong correlation with local inflammatory states. Assessment of fracture healing progress is crucial to making correct and timely clinical decisions for patients. This Cxm bioassay represents a minimally invasive, inexpensive technique that could provide reliable information on the biology of the fracture to significantly improve clinical care.


Assuntos
Colágeno Tipo X/sangue , Consolidação da Fratura , Fraturas da Tíbia/sangue , Animais , Biomarcadores/sangue , Feminino , Masculino , Camundongos Endogâmicos C57BL , Caracteres Sexuais
6.
Sci Transl Med ; 9(419)2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-29212713

RESUMO

Despite its importance as a key parameter of child health and development, growth velocity is difficult to determine in real time because skeletal growth is slow and clinical tools to accurately detect very small increments of growth do not exist. We report discovery of a marker for skeletal growth in infants and children. The intact trimeric noncollagenous 1 (NC1) domain of type X collagen, the marker we designated as CXM for Collagen X Marker, is a degradation by-product of endochondral ossification that is released into the circulation in proportion to overall growth plate activity. This marker corresponds to the rate of linear bone growth at time of measurement. Serum concentrations of CXM plotted against age show a pattern similar to well-established height growth velocity curves and correlate with height growth velocity calculated from incremental height measurements in this study. The CXM marker is stable once collected and can be accurately assayed in serum, plasma, and dried blood spots. CXM testing may be useful for monitoring growth in the pediatric population, especially responses of infants and children with genetic and acquired growth disorders to interventions that target the underlying growth disturbances. The utility of CXM may potentially extend to managing other conditions such as fracture healing, scoliosis, arthritis, or cancer.


Assuntos
Desenvolvimento Ósseo/fisiologia , Colágeno Tipo X/metabolismo , Consolidação da Fratura/fisiologia , Adulto , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Camundongos , Adulto Jovem
7.
Hum Mol Genet ; 25(R1): R2-8, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26443596

RESUMO

Achondroplasia (ACH) is the prototype and most common of the human chondrodysplasias. It results from gain-of-function mutations that exaggerate the signal output of the fibroblast growth factor receptor 3 (FGFR3), a receptor tyrosine kinase that negatively regulates growth plate activity and linear bone growth. Several approaches to reduce FGFR3 signaling by blocking receptor activation or inhibiting downstream signals have been proposed. Five show promise in preclinical mouse studies. Two candidate therapies target the extracellular domain of FGFR3. The first is a decoy receptor that competes for activating ligands. The second is a synthetic blocking peptide that prevents ligands from binding and activating FGFR3. Two established drugs, statins and meclozine, improve growth of ACH mice. The strongest candidate therapy employs an analog of C-type natriuretic peptide (CNP), which antagonizes the mitogen-activated-protein (MAP) kinase pathway downstream of the FGFR3 receptor and may also act independently in the growth plate. Only the CNP analog has reached clinical trials. Preliminary results of Phase 2 studies show a substantial increase in growth rate of ACH children after six months of therapy with no serious adverse effects. A challenge for drug therapy in ACH is targeting agents to the avascular growth plate. The application of gene therapy in osteoarthritis offers insights because it faces similar technical obstacles. Major advances in gene therapy include the emergence of recombinant adeno-associated virus as the vector of choice, capsid engineering to target vectors to specific tissues, and development of methods to direct vectors to articular chondrocytes.


Assuntos
Acondroplasia/terapia , Osteoartrite/terapia , Animais , Terapia Genética , Humanos , Camundongos
8.
Connect Tissue Res ; 56(6): 427-33, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26075305

RESUMO

Fibroblast Growth Factor Receptor 3 (FGFR3) is one of four high-affinity receptors for canonical FGF ligands. It acts in many tissues and plays a special role in skeletal development, especially post-embryonic bone growth, where it inhibits chondrocyte proliferation and differentiation. Gain of function mutations cause the most common forms of dwarfism in humans, and they are also detected in cancer. Triggered by ligand binding or in some cases mutation, FGFR3 activation involves dimerization of receptor monomers, phosphorylation of specific tyrosine residues in the receptor's kinase domain and in the tightly linked scaffold protein Fibroblast Receptor Factor Substrate 2 (FRS2). Signaling molecules recruited to these phosphorylation sites propagate signals through cascades that are subject to modulation. Signal output is also regulated by the fate of the receptor and the interval between its activation and degradation. Trafficking pathways have been identified for both lysosomal and proteasomal degradation, as well as, an alternative fate that involves intramembrane cleavage that produces an intracellular domain fragment capable of nuclear transport and potential function.


Assuntos
Neoplasias Ósseas , Nanismo , Mutação , Proteínas de Neoplasias , Proteólise , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Nanismo/genética , Nanismo/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo
9.
Methods Cell Biol ; 124: 391-417, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25287851

RESUMO

Correlated imaging is the process of imaging a specimen with two complementary modalities and then registering and overlaying the fields obtained in each modality to create a composite view. One of the images is made somewhat transparent, allowing detail in the underlying image to be visible and assisting in the registration of the two images. As an example, an image localizing a specific tissue component by fluorescence may be overlaid atop a TEM image of the same field. The resulting composite image would demonstrate specific ultrastructural features in the high-resolution TEM field, which are colorized in the overlay. Other examples include composites from MicroCT or soft X-ray images overlaid atop light microscopy or TEM images. Automated image registration may be facilitated by a variety of sophisticated computer programs utilized by high-throughput laboratories. This chapter is meant for the more occasional user wishing to align images manually. ImageJ is a public domain, image processing program developed at the National Institutes of Health and is available to anyone as a free download. ImageJ performs marvelously well for the purpose of image registration; therefore, step-by-step instructions are included here. Specimen handling, including fixation and choice of embedding media, is not straightforward for correlative imaging. A step-by-step description of the protocols which work in our laboratory is included for simultaneous localization in LM, EM and micro-CT, as well as maintaining GFP emission in tissue embedded for TEM.


Assuntos
Imageamento Tridimensional , Animais , Células Cultivadas , Marcadores Fiduciais , Humanos , Microscopia Confocal/métodos , Microscopia Confocal/normas , Microscopia Eletrônica de Transmissão/métodos , Microscopia Eletrônica de Transmissão/normas , Microtomia , Coloração e Rotulagem , Inclusão do Tecido , Interface Usuário-Computador , Microtomografia por Raio-X/métodos , Microtomografia por Raio-X/normas
10.
J Cell Physiol ; 229(12): 1999-2014, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24777663

RESUMO

The endocrine feedback loop between vitamin D3(1,25(OH)2D3) and parathyroid hormone (PTH) plays a central role in skeletal development. PTH-related protein (PTHrP) shares homology and its receptor (PTHR1) with PTH. The aim of this study was to investigate whether there is a functional paracrine feedback loop between 1,25(OH)2D3 and PTHrP in the growth plate, in parallel with the endocrine feedback loop between 1,25(OH)2D3 and PTH. This was investigated in ATDC5 cells treated with 10(-8) M 1,25(OH)2D3 or PTHrP, Col2-pd2EGFP transgenic mice, and primary Col2-pd2EGFP growth plate chondrocytes isolated by FACS, using RT-qPCR, Western blot, PTHrP ELISA, chromatin immunoprecipitation (ChIP) assay, silencing of the 1,25(OH)2D3 receptor (VDR), immunofluorescent staining, immunohistochemistry, and histomorphometric analysis of the growth plate. The ChIP assay confirmed functional binding of the VDR to the PTHrP promoter, but not to the PTHR1 promoter. Treatment with 1,25(OH)2D3 decreased PTHrP protein production, an effect which was prevented by silencing of the VDR. Treatment with PTHrP significantly induced VDR production, but did not affect 1α- and 24-hydroxylase expression. Hypertrophic differentiation was inhibited by PTHrP and 1,25(OH)2D3 treatment. Taken together, these findings indicate that there is a functional paracrine feedback loop between 1,25(OH)2D3 and PTHrP in the growth plate. 1,25(OH)2D3 decreases PTHrP production, while PTHrP increases chondrocyte sensitivity to 1,25(OH)2D3 by increasing VDR production. In light of the role of 1,25(OH)2D3 and PTHrP in modulating chondrocyte differentiation, 1,25(OH)2D3 in addition to PTHrP could potentially be used to prevent undesirable hypertrophic chondrocyte differentiation during cartilage repair or regeneration.


Assuntos
Colecalciferol/metabolismo , Condrócitos/metabolismo , Comunicação Parácrina/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Animais , Diferenciação Celular/genética , Colecalciferol/administração & dosagem , Condrócitos/patologia , Retroalimentação Fisiológica , Regulação da Expressão Gênica no Desenvolvimento , Lâmina de Crescimento/metabolismo , Humanos , Camundongos , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo
11.
Expert Rev Mol Med ; 14: e11, 2012 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-22559284

RESUMO

Mutations that exaggerate signalling of the receptor tyrosine kinase fibroblast growth factor receptor 3 (FGFR3) give rise to achondroplasia, the most common form of dwarfism in humans. Here we review the clinical features, genetic aspects and molecular pathogenesis of achondroplasia and examine several therapeutic strategies designed to target the mutant receptor or its signalling pathways, including the use of kinase inhibitors, blocking antibodies, physiologic antagonists, RNAi and chaperone inhibitors. We conclude by discussing the challenges of treating growth plate disorders in children.


Assuntos
Acondroplasia/tratamento farmacológico , Acondroplasia/genética , Terapia de Alvo Molecular , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Acondroplasia/metabolismo , Anticorpos/farmacologia , Humanos , Mutação , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Relação Estrutura-Atividade
12.
Mol Biol Cell ; 22(20): 3861-73, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21865593

RESUMO

Fibroblast growth factor receptor 3 (FGFR3) is a major negative regulator of bone growth that inhibits the proliferation and differentiation of growth plate chondrocytes. Activating mutations of its c isoform cause dwarfism in humans; somatic mutations can drive oncogenic transformation in multiple myeloma and bladder cancer. How these distinct activities arise is not clear. FGFR3 was previously shown to undergo proteolytic cleavage in the bovine rib growth plate, but this was not explored further. Here, we show that FGF1 induces regulated intramembrane proteolysis (RIP) of FGFR3. The ectodomain is proteolytically cleaved (S1) in response to ligand-induced receptor activation, but unlike most RIP target proteins, it requires endocytosis and does not involve a metalloproteinase. S1 cleavage generates a C-terminal domain fragment that initially remains anchored in the membrane, is phosphorylated, and is spatially distinct from the intact receptor. Ectodomain cleavage is followed by intramembrane cleavage (S2) to generate a soluble intracellular domain that is released into the cytosol and can translocate to the nucleus. We identify the S1 cleavage site and show that γ-secretase mediates the S2 cleavage event. In this way we demonstrate a mechanism for the nuclear localization of FGFR3 in response to ligand activation, which may occur in both development and disease.


Assuntos
Diferenciação Celular/fisiologia , Membrana Celular/enzimologia , Fator 1 de Crescimento de Fibroblastos/metabolismo , Lâmina de Crescimento/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Células COS , Bovinos , Membrana Celular/genética , Chlorocebus aethiops , Condrócitos/citologia , Condrócitos/metabolismo , Endocitose , Fator 1 de Crescimento de Fibroblastos/genética , Lâmina de Crescimento/citologia , Imunoprecipitação , Fosforilação , Plasmídeos , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteólise , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Transfecção
13.
Biochim Biophys Acta ; 1808(8): 2045-50, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21536014

RESUMO

The A391E mutation in the transmembrane domain of fibroblast growth factor receptor 3 leads to aberrant development of the cranium. It has been hypothesized that the mutant glutamic acid stabilizes the dimeric receptor due to hydrogen bonding and enhances its ligand-independent activation. We previously tested this hypothesis in lipid bilayers and showed that the mutation stabilizes the isolated transmembrane domain dimer by -1.3°kcal/mol. Here we further test the hypothesis, by investigating the effect of the A391E mutation on the activation of full-length fibroblast growth factor receptor 3 in human embryonic kidney 293T cells in the absence of ligand. We find that the mutation enhances the ligand-independent activation propensity of the receptor by -1.7°kcal/mol. This value is consistent with the observed strength of hydrogen bonds in membranes, and supports the above hypothesis.


Assuntos
Membrana Celular/metabolismo , Mutação , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Membrana Celular/química , Fator 1 de Crescimento de Fibroblastos/metabolismo , Células HEK293 , Humanos , Ligação de Hidrogênio , Ligantes , Modelos Biológicos , Modelos Químicos , Fosforilação , Multimerização Proteica , Estabilidade Proteica , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Relação Estrutura-Atividade , Transfecção
14.
J Biol Chem ; 286(22): 19597-604, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21487019

RESUMO

Fibroblast growth factor receptor 3 (FGFR3) is a key regulator of growth and differentiation, whose aberrant activation causes a number of genetic diseases including achondroplasia and cancer. Hsp90 is a specialized molecular chaperone involved in stabilizing a select set of proteins termed clients. Here, we delineate the relationship of Hsp90 and co-chaperone Cdc37 with FGFR3 and the FGFR family. FGFR3 strongly associates with these chaperone complexes and depends on them for stability and function. Inhibition of Hsp90 function using the geldanamycin analog 17-AAG induces the ubiquitination and degradation of FGFR3 and reduces the signaling capacity of FGFR3. Other FGFRs weakly interact with these chaperones and are differentially influenced by Hsp90 inhibition. The Hsp90-related ubiquitin ligase CHIP is able to interact and destabilize FGFR3. Our results establish FGFR3 as a strong Hsp90 client and suggest that modulating Hsp90 chaperone complexes may beneficially influence the stability and function of FGFR3 in disease.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Ubiquitinação , Acondroplasia/genética , Acondroplasia/metabolismo , Animais , Benzoquinonas/farmacologia , Células COS , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Chaperoninas/genética , Chaperoninas/metabolismo , Chlorocebus aethiops , Estabilidade Enzimática/efeitos dos fármacos , Estabilidade Enzimática/genética , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/genética , Humanos , Lactamas Macrocíclicas/farmacologia , Camundongos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética
15.
Dev Dyn ; 240(3): 663-73, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21337464

RESUMO

We report the generation of a new mouse strain harboring a Col2-pd2EGFP reporter transgene; pd2EGFP has a much shorter half-life than EGFP, making it a near real-time reporter for Col2α1 expression in vivo and in vitro. In the post-natal growth plate, pd2EGFP fluorescence was expressed in almost all proliferative chondrocytes and in some hypertrophic chondrocytes based on localization with type X collagen. In articular cartilage, pd2EGFP fluorescence diminished over time, nicely illustrating the decrease of type II collagen synthesis in articular chondrocytes during growth. Monolayers of FACS-sorted chondrocytes from P1-2 mice showed faster loss of pd2EGFP compared to EGFP, reflecting rapid chondrocyte de-differentiation. High-density culture of FACS-pd2EGFP- growth plate chondrocytes revealed the typical temporal expression pattern in which type II collagen preceded type X collagen matrix deposition. The Col2-pd2EGFP reporter mouse will be a valuable tool for studies of growth plate chondrocyte biology.


Assuntos
Colágeno Tipo II/metabolismo , Animais , Condrócitos/citologia , Condrócitos/metabolismo , Citometria de Fluxo , Lâmina de Crescimento/citologia , Lâmina de Crescimento/metabolismo , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Microscopia Confocal
16.
Curr Opin Pediatr ; 22(4): 516-23, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20601886

RESUMO

PURPOSE OF REVIEW: Although the genetic defect underlying achondroplasia has been known for over a decade, no effective therapies to stimulate bone growth have emerged. Here we review the recent literature and summarize the molecular mechanisms underlying disease pathology and examine their potential as therapeutic targets. Currently used preclinical models are discussed in the context of recent advances with a special focus on C-type natriuretic peptide. RECENT FINDINGS: Research on the mutation in Fibroblast Growth Factor Receptor 3 (FGFR3) that causes achondroplasia suggests that disease results from increased signal transduction from the mutant receptor. Thus, current therapeutic strategies have focused on reducing signals emanating from FGFR3. First-generation therapies directly targeting FGFR3, such as kinase inhibitors and neutralizing antibodies, designed for targeting FGFR3 in cancer, are still in the preclinical phase and have yet to translate into the management of achondroplasia. Counteracting signal transduction pathways downstream of FGFR3 holds promise with the discovery that administration of C-type natriuretic peptide to achondroplastic mice ameliorates their clinical phenotype. However, more research into long-term effectiveness and safety of this strategy is needed. Direct targeting of therapeutic agents to growth plate cartilage may enhance efficacy and minimize side effects of these and future therapies. SUMMARY: Current research into the pathogenesis of achondroplasia has expanded our understanding of the mechanisms of FGFR3-induced disease and has increased the number of approaches that we may use to potentially correct it. Further research is needed to validate these approaches in preclinical models of achondroplasia.


Assuntos
Acondroplasia , Predisposição Genética para Doença , Natriuréticos/uso terapêutico , Peptídeo Natriurético Tipo C/uso terapêutico , Acondroplasia/diagnóstico , Acondroplasia/tratamento farmacológico , Acondroplasia/genética , Animais , Humanos , Mutação , Prognóstico , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética
17.
J Cell Biochem ; 110(5): 1046-57, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20564212

RESUMO

The mammalian skeleton developments and grows through two complementary pathways: membranous ossification, which gives rise to the calvarial bones and distal clavicle, and endochondral ossification, which is responsible for the bones of the limbs, girdles, vertebrae, face and base of the skull and the medial clavicle. Fibroblast growth factors (FGFs) and their cognate FGF receptors (FGFRs) play important roles in regulating both pathways. However, the details of how FGF signals are initiated, propagated and modulated within the developing skeleton are only slowly emerging. This prospect will focus on the current understanding of these events during endochondral skeletal development with special attention given to concepts that have emerged in the past few years.


Assuntos
Desenvolvimento Ósseo/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Transdução de Sinais/fisiologia , Animais , Osso e Ossos/fisiologia , Lâmina de Crescimento/crescimento & desenvolvimento , Lâmina de Crescimento/fisiologia , Humanos , Modelos Biológicos , Osteogênese/fisiologia
18.
Mol Cell Biochem ; 338(1-2): 215-24, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20033473

RESUMO

Cartilage oligomeric matrix protein (COMP) is a pentameric approximately 524 kDa multidomain extracellular matrix protein and is the fifth member of the thrombospondin family. COMP is abundantly expressed in proliferating and hypertrophic chondrocytes of the growth plate, articular cartilage, synovium, tendon, and ligament. The spatial localization of COMP highlights its importance in the phenotypes of pseudoachondroplasia (PSACH) and multiple epiphyseal dysplasia (MED), COMP disorders that are characterized by disproportionate short stature, brachydactyly, scoliosis, early-onset osteoarthritis, and joint hypermobility. In this study, the role of COMP in ligament was investigated with a series of cell attachment assays using ligament cells binding to COMP. A dose-dependent cell attachment activity was found, which was inhibited by a peptide containing the SFYVVMWK amino acid sequence derived from the globular C-terminal domain of COMP. This activity was independent of the recently described RGD-dependent attachment activity. Function-blocking antibodies to CD47 and alphaVbeta3 integrin reduced cell attachment to COMP, implicating the participation of these cell surface molecules in COMP cell binding. Immunofluorescence studies showed that cell attachment to COMP induced the formation of lamellae containing F-actin microspikes associated with fascin. We propose that COMP promotes cell attachment via two independent mechanisms involving cell surface CD47 and alphaVbeta3 integrin and that a consequence of cell attachment to COMP is the specific induction of fascin-stabilized actin microspikes.


Assuntos
Antígeno CD47/metabolismo , Adesão Celular/fisiologia , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Integrina alfaVbeta3/metabolismo , Actinas/metabolismo , Animais , Antígeno CD47/genética , Proteínas de Transporte/metabolismo , Proteína de Matriz Oligomérica de Cartilagem , Extensões da Superfície Celular/metabolismo , Extensões da Superfície Celular/ultraestrutura , Condrócitos/citologia , Condrócitos/fisiologia , Proteínas da Matriz Extracelular/genética , Glicoproteínas/genética , Humanos , Integrina alfaVbeta3/genética , Ligamentos/citologia , Ligamentos/metabolismo , Proteínas Matrilinas , Camundongos , Proteínas dos Microfilamentos/metabolismo , Peptídeos/genética , Peptídeos/metabolismo
19.
BMC Res Notes ; 2: 243, 2009 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20003239

RESUMO

BACKGROUND: The expression and study of recombinant proteins in mammalian culture systems can be complicated during the cell lysis procedure by contaminating proteins from cellular compartments distinct from those within which the protein of interest resides and also by solubility issues that may arise from the use of a single lysis buffer. Partial subcellular fractionation using buffers of increasing stringency, rather than whole cell lysis is one way in which to avoid or reduce this contamination and ensure complete recovery of the target protein. Currently published protocols involve time consuming centrifugation steps which may require expensive equipment and commercially available kits can be prohibitively expensive when handling large or multiple samples. FINDINGS: We have established a protocol to sequentially extract proteins from cultured mammalian cells in fractions enriched for cytosolic, membrane bound organellar, nuclear and insoluble proteins. All of the buffers used can be made inexpensively and easily and the protocol requires no costly equipment. While the method was optimized for a specific cell type, we demonstrate that the protocol can be applied to a variety of commonly used cell lines and anticipate that it can be applied to any cell line via simple optimization of the primary extraction step. CONCLUSION: We describe a protocol for the crude subcellular fractionation of cultured mammalian cells that is both straightforward and cost effective and may facilitate the more accurate study of recombinant proteins and the generation of purer preparations of said proteins from cell extracts.

20.
Trends Endocrinol Metab ; 20(7): 341-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19716710

RESUMO

The mammalian skeleton forms and grows through two developmental pathways: membranous ossification, which gives rise to calvarial bones and the distal clavicle, and endochondral ossification, which is responsible for the bones of the limbs, girdles, vertebrae, face, base of the skull and the medial clavicle. The regulation of both pathways is extremely complex, and the rules that govern it are still emerging. However, it has become clear that fibroblast growth factors (FGFs) and their cognate receptors (FGFRs) play essential roles. This review focuses on the roles of FGFs and FGFRs in endochondral skeletal development, with special attention given to concepts that have emerged in the past few years.


Assuntos
Desenvolvimento Ósseo/fisiologia , Osso e Ossos/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Osteogênese/fisiologia , Animais , Lâmina de Crescimento/fisiologia , Humanos , Mamíferos/crescimento & desenvolvimento , Mamíferos/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Esqueleto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...