Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Elife ; 122024 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-38277211

RESUMEN

Adolescent idiopathic scoliosis (AIS) is a common and progressive spinal deformity in children that exhibits striking sexual dimorphism, with girls at more than fivefold greater risk of severe disease compared to boys. Despite its medical impact, the molecular mechanisms that drive AIS are largely unknown. We previously defined a female-specific AIS genetic risk locus in an enhancer near the PAX1 gene. Here, we sought to define the roles of PAX1 and newly identified AIS-associated genes in the developmental mechanism of AIS. In a genetic study of 10,519 individuals with AIS and 93,238 unaffected controls, significant association was identified with a variant in COL11A1 encoding collagen (α1) XI (rs3753841; NM_080629.2_c.4004C>T; p.(Pro1335Leu); p=7.07E-11, OR = 1.118). Using CRISPR mutagenesis we generated Pax1 knockout mice (Pax1-/-). In postnatal spines we found that PAX1 and collagen (α1) XI protein both localize within the intervertebral disc-vertebral junction region encompassing the growth plate, with less collagen (α1) XI detected in Pax1-/- spines compared to wild-type. By genetic targeting we found that wild-type Col11a1 expression in costal chondrocytes suppresses expression of Pax1 and of Mmp3, encoding the matrix metalloproteinase 3 enzyme implicated in matrix remodeling. However, the latter suppression was abrogated in the presence of the AIS-associated COL11A1P1335L mutant. Further, we found that either knockdown of the estrogen receptor gene Esr2 or tamoxifen treatment significantly altered Col11a1 and Mmp3 expression in chondrocytes. We propose a new molecular model of AIS pathogenesis wherein genetic variation and estrogen signaling increase disease susceptibility by altering a PAX1-COL11a1-MMP3 signaling axis in spinal chondrocytes.


Adolescent idiopathic scoliosis (AIS) is a twisting deformity of the spine that occurs during periods of rapid growth in children worldwide. Children with severe cases of AIS require surgery to stop it from getting worse, presenting a significant financial burden to health systems and families. Although AIS is known to cluster in families, its genetic causes and its inheritance pattern have remained elusive. Additionally, AIS is known to be more prevalent in females, a bias that has not been explained. Advances in techniques to study the genetics underlying diseases have revealed that certain variations that increase the risk of AIS affect cartilage and connective tissue. In humans, one such variation is near a gene called Pax1, and it is female-specific. The extracellular matrix is a network of proteins and other molecules in the space between cells that help connect tissues together, and it is particularly important in cartilage and other connective tissues. One of the main components of the extracellular matrix is collagen. Yu, Kanshour, Ushiki et al. hypothesized that changes in the extracellular matrix could affect the cartilage and connective tissues of the spine, leading to AIS. To show this, the scientists screened over 100,000 individuals and found that AIS is associated with variants in two genes coding for extracellular matrix proteins. One of these variants was found in a gene called Col11a1, which codes for one of the proteins that makes up collagen. To understand the relationship between Pax1 and Col11a1, Yu, Kanshour, Ushiki et al. genetically modified mice so that they would lack the Pax1 gene. In these mice, the activation of Col11a1 was reduced in the mouse spine. They also found that the form of Col11a1 associated with AIS could not suppress the activation of a gene called Mmp3 in mouse cartilage cells as effectively as unmutated Col11a1. Going one step further, the researchers found that lowering the levels of an estrogen receptor altered the activation patterns of Pax1, Col11a1, and Mmp3 in mouse cartilage cells. These findings suggest a possible mechanism for AIS, particularly in females. The findings of Yu, Kanshour, Ushiki et al. highlight that cartilage cells in the spine are particularly relevant in AIS. The results also point to specific molecules within the extracellular matrix as important for maintaining proper alignment in the spine when children are growing rapidly. This information may guide future therapies aimed at maintaining healthy spinal cells in adolescent children, particularly girls.


Asunto(s)
Escoliosis , Masculino , Animales , Niño , Ratones , Humanos , Femenino , Adolescente , Escoliosis/genética , Metaloproteinasa 3 de la Matriz/genética , Columna Vertebral , Factores de Transcripción/genética , Colágeno/genética , Variación Genética , Colágeno Tipo XI/genética
2.
bioRxiv ; 2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37292598

RESUMEN

Adolescent idiopathic scoliosis (AIS) is a common and progressive spinal deformity in children that exhibits striking sexual dimorphism, with girls at more than five-fold greater risk of severe disease compared to boys. Despite its medical impact, the molecular mechanisms that drive AIS are largely unknown. We previously defined a female-specific AIS genetic risk locus in an enhancer near the PAX1 gene. Here we sought to define the roles of PAX1 and newly-identified AIS-associated genes in the developmental mechanism of AIS. In a genetic study of 10,519 individuals with AIS and 93,238 unaffected controls, significant association was identified with a variant in COL11A1 encoding collagen (α1) XI (rs3753841; NM_080629.2_c.4004C>T; p.(Pro1335Leu); P=7.07e-11, OR=1.118). Using CRISPR mutagenesis we generated Pax1 knockout mice (Pax1-/-). In postnatal spines we found that PAX1 and collagen (α1) XI protein both localize within the intervertebral disc (IVD)-vertebral junction region encompassing the growth plate, with less collagen (α1) XI detected in Pax1-/- spines compared to wildtype. By genetic targeting we found that wildtype Col11a1 expression in costal chondrocytes suppresses expression of Pax1 and of Mmp3, encoding the matrix metalloproteinase 3 enzyme implicated in matrix remodeling. However, this suppression was abrogated in the presence of the AIS-associated COL11A1P1335L mutant. Further, we found that either knockdown of the estrogen receptor gene Esr2, or tamoxifen treatment, significantly altered Col11a1 and Mmp3 expression in chondrocytes. We propose a new molecular model of AIS pathogenesis wherein genetic variation and estrogen signaling increase disease susceptibility by altering a Pax1-Col11a1-Mmp3 signaling axis in spinal chondrocytes.

3.
J Bone Miner Res ; 38(2): 288-299, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36459048

RESUMEN

Neurofibromatosis type 1 (NF1) is a tumor predisposition syndrome caused by heterozygous NF1 gene mutations. Patients with NF1 present with pleiotropic somatic secondary manifestations, including development of bone pseudarthrosis after fracture. Somatic NF1 gene mutations were reproducibly identified in patient-derived pseudarthrosis specimens, suggesting a local mosaic cell population including somatic pathologic cells. The somatic cellular pathogenesis of NF1 pseudarthroses remains unclear, though defects in osteogenesis have been posited. Here, we applied time-series single-cell RNA-sequencing (scRNA-seq) to patient-matched control and pseudarthrosis-derived primary bone stromal cells (BSCs). We show that osteogenic specification to an osteoblast progenitor cell population was evident for control bone-derived cells and haploinsufficient pseudarthrosis-derived cells. Similar results were observed for somatic patient fracture-derived NF1-/- cells; however, expression of genetic pathways associated with skeletal mineralization were significantly reduced in NF1-/- cells compared with fracture-derived NF1+/- cells. In mice, we show that Nf1 expressed in bone marrow osteoprogenitors is required for the maintenance of the adult skeleton. Results from our study implicate impaired Clec11a-Itga11-Wnt signaling in the pathogenesis of NF1-associated skeletal disease. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Fracturas Óseas , Neurofibromatosis 1 , Seudoartrosis , Ratones , Animales , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Seudoartrosis/genética , Seudoartrosis/metabolismo , Seudoartrosis/patología , Fracturas Óseas/patología , Osteoblastos/metabolismo , Osteogénesis/genética
4.
Hum Mol Genet ; 29(22): 3717-3728, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33105483

RESUMEN

Talipes equinovarus (clubfoot, TEV) is a congenital rotational foot deformity occurring in 1 per 1000 births with increased prevalence in males compared with females. The genetic etiology of isolated clubfoot (iTEV) remains unclear. Using a genome-wide association study, we identified a locus within FSTL5, encoding follistatin-like 5, significantly associated with iTEV. FSTL5 is an uncharacterized gene whose potential role in embryonic and postnatal development was previously unstudied. Utilizing multiple model systems, we found that Fstl5 was expressed during later stages of embryonic hindlimb development, and, in mice, expression was restricted to the condensing cartilage anlage destined to form the limb skeleton. In the postnatal growth plate, Fstl5 was specifically expressed in prehypertrophic chondrocytes. As Fstl5 knockout rats displayed no gross malformations, we engineered a conditional transgenic mouse line (Fstl5LSL) to overexpress Fstl5 in skeletal osteochondroprogenitors. We observed that hindlimbs were slightly shorter and that bone mineral density was reduced in adult male, but not female, Prrx1-cre;Fstl5LSL mice compared with control. No overt clubfoot-like deformity was observed in Prrx1-cre;Fstl5LSL mice, suggesting FSTL5 may function in other cell types to contribute to iTEV pathogenesis. Interrogating published mouse embryonic single-cell expression data showed that Fstl5 was expressed in cell lineage subclusters whose transcriptomes were associated with neural system development. Moreover, our results suggest that lineage-specific expression of the Fstl genes correlates with their divergent roles as modulators of transforming growth factor beta and bone morphogenetic protein signaling. Results from this study associate FSTL5 with iTEV and suggest a potential sexually dimorphic role for Fstl5 in vivo.


Asunto(s)
Pie Equinovaro/genética , Proteínas Relacionadas con la Folistatina/genética , Predisposición Genética a la Enfermedad , Proteínas de Homeodominio/genética , Animales , Pie Equinovaro/patología , Modelos Animales de Enfermedad , Extremidades/patología , Regulación de la Expresión Génica/genética , Técnicas de Inactivación de Genes , Estudios de Asociación Genética , Humanos , Ratones , Ratas
5.
Nat Commun ; 6: 6452, 2015 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-25784220

RESUMEN

Idiopathic scoliosis (IS) is a common paediatric musculoskeletal disease that displays a strong female bias. By performing a genome-wide association study (GWAS) of 3,102 individuals, we identify significant associations with 20p11.22 SNPs for females (P=6.89 × 10(-9)) but not males (P=0.71). This association with IS is also found in independent female cohorts from the United States of America and Japan (overall P=2.15 × 10(-10), OR=1.30 (rs6137473)). Unexpectedly, the 20p11.22 IS risk alleles were previously associated with protection from early-onset alopecia, another sexually dimorphic condition. The 174-kb associated locus is distal to PAX1, which encodes paired box 1, a transcription factor involved in spine development. We identify a sequence in the associated locus with enhancer activity in zebrafish somitic muscle and spinal cord, an activity that is abolished by IS-associated SNPs. We thus identify a sexually dimorphic IS susceptibility locus, and propose the first functionally defined candidate mutations in an enhancer that may regulate expression in specific spinal cells.


Asunto(s)
Elementos de Facilitación Genéticos , Predisposición Genética a la Enfermedad , Factores de Transcripción Paired Box/genética , Escoliosis/genética , Alelos , Animales , Mapeo Cromosómico , Estudios de Cohortes , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Japón , Desequilibrio de Ligamiento , Masculino , Mutación , Factores de Transcripción Paired Box/fisiología , Fenotipo , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Análisis de Secuencia de ADN , Factores Sexuales , Estados Unidos , Pez Cebra
6.
Hum Mol Genet ; 22(3): 444-51, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23100325

RESUMEN

Macrodactyly is a discrete congenital anomaly consisting of enlargement of all tissues localized to the terminal portions of a limb, typically within a 'nerve territory'. The classic terminology for this condition is 'lipofibromatous hamartoma of nerve' or Type I macrodactyly. The peripheral nerve, itself, is enlarged both in circumference and in length. It is not related to neurofibromatosis (NF1), nor is it associated with vascular malformations, such as in the recently reported CLOVES syndrome. The specific nerve pathophysiology in this form of macrodactyly has not been well described and a genetic etiology for this specific form of enlargement is unknown. To identify the genetic cause of macrodactyly, we used whole-exome sequencing to identify somatic mutations present in the affected nerve of a single patient. We confirmed a novel mutation in PIK3CA (R115P) present in the patient's affected nerve tissue but not in blood DNA. Sequencing PIK3CA exons identified gain-of-function mutations (E542K, H1047L or H1047R) in the affected tissue of five additional unrelated patients; mutations were absent in blood DNA available from three patients. Immunocytochemistry confirmed AKT activation in cultured cells from the nerve of a macrodactyly patient. Additionally, we found that the most abnormal structure within the involved nerve in a macrodactylous digit is the perineurium, with additional secondary effects on the axon number and size. Thus, isolated congenital macrodactyly is caused by somatic activation of the PI3K/AKT cell-signaling pathway and is genetically and biochemically related to other overgrowth syndromes.


Asunto(s)
Deformidades Congénitas de las Extremidades/genética , Mutación , Fosfatidilinositol 3-Quinasas/genética , Células Cultivadas , Preescolar , Fosfatidilinositol 3-Quinasa Clase I , Femenino , Dedos/anomalías , Estudio de Asociación del Genoma Completo/métodos , Humanos , Inmunohistoquímica , Lactante , Microscopía Electrónica , Tejido Nervioso/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Análisis de Secuencia de ADN , Transducción de Señal , Síndrome
7.
Sci Rep ; 2: 650, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22970342

RESUMEN

Juvenile femoral head osteonecrosis is due to disruption of blood supply which results in ischemic injury. Angiogenesis is an essential component for the healing of damaged head. Hypoxia-inducible factor-1α (HIF-1α) is a master regulator of cellular response to hypoxia. Our histological studies showed increased vessel formation in cartilage in the ischemic group compared to the control group in a pig model of femoral head osteonecrosis. Microarray and RT-PCR indicated that VEGF expression was upregulated along with HIF-1α in the ischemic side. Immunohistochemistry assay demonstrated that HIF-1α and VEGF were upregulated in chondrocytes in ischemic femoral heads. Both HIF-1α and VEGF expression increased in primary chondrocytes under hypoxia station. Interestingly, an HIF-1α activator DFO further enhanced VEGF expression. Moreover, transfection of siRNA directed against HIF-1α led to inhibition of VEGF expression. Taken together, our data indicated that upregulation of VEGF during hypoxia in chondrocyte is mediated partially through HIF-1α.


Asunto(s)
Cartílago Articular/metabolismo , Necrosis de la Cabeza Femoral/mortalidad , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Cartílago Articular/irrigación sanguínea , Cartílago Articular/patología , Hipoxia de la Célula , Células Cultivadas , Condrocitos/metabolismo , Cabeza Femoral/irrigación sanguínea , Cabeza Femoral/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isquemia/metabolismo , Neovascularización Fisiológica , Sus scrofa , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética
8.
Bone ; 48(3): 507-13, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20950722

RESUMEN

Legg-Calve-Perthes disease (LCPD) is a juvenile form of ischemic osteonecrosis of the femoral head leading to femoral head deformity and premature osteoarthritis. Femoral head osteonecrosis occurs due to blood supply disruption which results in hypoxic injury to the femoral head. Hypoxia-inducible factor-1 α (HIF-1α) is a master regulator of cellular response to hypoxia. A pig model of ischemic osteonecrosis of femoral head has been shown to have radiographic and histopathologic changes resembling LCPD. Our preliminary studies showed that the cartilage layer was thicker in the hypoxia group compared to the control group. The mechanism underlying this cartilage response is not known. To explore the hypoxia-induced downstream gene activity following the femoral head ischemia, porcine microarray analysis of gene profiles of chondrocytes from normal and ischemic femoral heads was performed. In the ischemic side, the expression of Sox9, a transcription factor required for chondrocyte differentiation, was upregulated along with HIF-1α. Expressions of Sox9 target genes, such as type II collagen and aggrecan, were also increased. Microarray results were confirmed by quantitative real-time RT-PCR. In addition, immunohistochemistry assay demonstrated that both HIF-1α and Sox9 were upregulated in chondrocytes in ischemic femoral heads compared with normal controls. To investigate the possible molecular mechanisms of hypoxia on Sox9 activity, we tested the effect of HIF-1α on Sox9 expression in vitro. We made a luciferase reporter construct driven by 2kb Sox9 promoter. Transient transfection assay showed that HIF-1α activated Sox9 promoter activity in a dose-dependent manner. Sox9 is known to activate type II collagen target gene expression. To test the effect of HIF-1α on Sox9-mediated transcription, HIF-1α was cotransfected with Sox9 in type II collagen reporter assay. Our results demonstrated that HIF-1α enhanced Sox9-mediated transcriptional activity. Moreover, coimmunoprecipitation assay showed that HIF-1α associated with Sox9 directly. Taken together, these findings indicate that HIF-1α activates Sox9 expression and enhances Sox9-mediated transcriptional activity and that HIF-1α physically interacts with Sox9. We speculate that HIF-1α upregulation of Sox9 activity may have a chondroprotective role following femoral head ischemia.


Asunto(s)
Necrosis de la Cabeza Femoral/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor de Transcripción SOX9/metabolismo , Animales , Células COS , Cartílago/metabolismo , Cartílago/patología , Diferenciación Celular , Chlorocebus aethiops , Condrocitos/metabolismo , Condrocitos/patología , Cabeza Femoral/metabolismo , Cabeza Femoral/patología , Necrosis de la Cabeza Femoral/complicaciones , Necrosis de la Cabeza Femoral/genética , Necrosis de la Cabeza Femoral/patología , Regulación de la Expresión Génica , Hipoxia/complicaciones , Hipoxia/genética , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Factor de Transcripción SOX9/genética , Sus scrofa , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...