Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
1.
Hum Mol Genet ; 31(22): 3777-3788, 2022 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-35660865

RESUMO

Mutations in the fibrillin-1 (FBN1) gene are responsible for the autosomal dominant form of geleophysic dysplasia (GD), which is characterized by short stature and extremities, thick skin and cardiovascular disease. All known FBN1 mutations in patients with GD are localized within the region encoding the transforming growth factor-ß binding protein-like 5 (TB5) domain of this protein. Herein, we generated a knock-in mouse model, Fbn1Y1698C by introducing the p.Tyr1696Cys mutation from a patient with GD into the TB5 domain of murine Fbn1 to elucidate the specific role of this domain in endochondral ossification. We found that both Fbn1Y1698C/+ and Fbn1Y1698C/Y1698C mice exhibited a reduced stature reminiscent of the human GD phenotype. The Fbn1 point mutation introduced in these mice affected the growth plate formation owing to abnormal chondrocyte differentiation such that mutant chondrocytes failed to establish a dense microfibrillar network composed of FBN1. This original Fbn1 mutant mouse model offers new insight into the pathogenic events underlying GD. Our findings suggest that the etiology of GD involves the dysregulation of the extracellular matrix composed of an abnormal FBN1 microfibril network impacting the differentiation of the chondrocytes.


Assuntos
Doenças do Desenvolvimento Ósseo , Fibrilina-1 , Deformidades Congênitas dos Membros , Síndrome de Marfan , Animais , Humanos , Camundongos , Doenças do Desenvolvimento Ósseo/metabolismo , Fibrilina-1/genética , Deformidades Congênitas dos Membros/genética , Síndrome de Marfan/genética , Mutação , Osteogênese/genética
2.
Nat Commun ; 12(1): 5013, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34408147

RESUMO

Human families with chromosomal rearrangements at 2q31, where the human HOXD locus maps, display mesomelic dysplasia, a severe shortening and bending of the limb. In mice, the dominant Ulnaless inversion of the HoxD cluster produces a similar phenotype suggesting the same origin for these malformations in humans and mice. Here we engineer 1 Mb inversion including the HoxD gene cluster, which positioned Hoxd13 close to proximal limb enhancers. Using this model, we show that these enhancers contact and activate Hoxd13 in proximal cells, inducing the formation of mesomelic dysplasia. We show that a secondary Hoxd13 null mutation in-cis with the inversion completely rescues the alterations, demonstrating that ectopic HOXD13 is directly responsible for this bone anomaly. Single-cell expression analysis and evaluation of HOXD13 binding sites suggests that the phenotype arises primarily by acting through genes normally controlled by HOXD13 in distal limb cells. Altogether, these results provide a conceptual and mechanistic framework to understand and unify the molecular origins of human mesomelic dysplasia associated with 2q31.


Assuntos
Anormalidades Múltiplas/genética , Doenças do Desenvolvimento Ósseo/genética , Proteínas de Homeodomínio/genética , Deformidades Congênitas dos Membros/genética , Fatores de Transcrição/genética , Anormalidades Múltiplas/embriologia , Anormalidades Múltiplas/metabolismo , Animais , Doenças do Desenvolvimento Ósseo/embriologia , Doenças do Desenvolvimento Ósseo/metabolismo , Modelos Animais de Doenças , Feminino , Deleção de Genes , Proteínas de Homeodomínio/metabolismo , Humanos , Deformidades Congênitas dos Membros/embriologia , Deformidades Congênitas dos Membros/metabolismo , Mutação com Perda de Função , Masculino , Camundongos Endogâmicos C57BL , Família Multigênica , Fatores de Transcrição/metabolismo
3.
J Cell Physiol ; 236(12): 8208-8225, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34170011

RESUMO

Cohesin loader nipped-B-like protein (Nipbl) is increasingly recognized for its important role in development and cancer. Cornelia de Lange Syndrome (CdLS), mostly caused by heterozygous mutations of Nipbl, is an autosomal dominant disease characterized by multiorgan malformations. However, the regulatory role and underlying mechanism of Nipbl in skeletal development remain largely elusive. In this study, we constructed a Nipbl-a Cas9-knockout (KO) zebrafish, which displayed severe retardation of global growth and skeletal development. Deficiency of Nipbl remarkably compromised cell growth and survival, and osteogenic differentiation of mammalian osteoblast precursors. Furthermore, Nipbl depletion impaired the cell cycle process, and caused DNA damage accumulation and cellular senescence. In addition, nucleolar fibrillarin expression, global rRNA biogenesis, and protein translation were defective in the Nipbl-depleted osteoblast precursors. Interestingly, an integrated stress response inhibitor (ISRIB), partially rescued Nipbl depletion-induced cellular defects in proliferation and apoptosis, osteogenesis, and nucleolar function. Simultaneously, we performed transcriptome analysis of Nipbl deficiency on human neural crest cells and mouse embryonic fibroblasts in combination with Nipbl ChIP-Seq. We found that Nipbl deficiency caused thousands of differentially expressed genes including some important genes in bone and cartilage development. In conclusion, Nipbl deficiency compromised skeleton development through impairing osteoblast precursor cell proliferation and survival, and osteogenic differentiation, and also disturbing the expression of some osteogenesis-regulatory genes. Our study elucidated that Nipbl played a pivotal role in skeleton development, and supported the fact that treatment of ISRIB may provide an early intervention strategy to alleviate the bone dysplasia of CdLS.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Síndrome de Cornélia de Lange/genética , Síndrome de Cornélia de Lange/metabolismo , Fibroblastos/metabolismo , Osteogênese/genética , Animais , Doenças do Desenvolvimento Ósseo/metabolismo , Segregação de Cromossomos/genética , Heterozigoto , Mutação/genética , Fenótipo , Transcrição Gênica/genética , Peixe-Zebra/genética
4.
Nat Rev Endocrinol ; 17(6): 336-349, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33948016

RESUMO

The major mineralized tissues are bone and teeth, which share several mechanisms governing their development and mineralization. This crossover includes the hormones that regulate circulating calcium and phosphate concentrations, and the genes that regulate the differentiation and transdifferentiation of cells. In developing endochondral bone and in developing teeth, parathyroid hormone-related protein (PTHrP) acts in chondrocytes to delay terminal differentiation, thereby increasing the pool of precursor cells. Chondrocytes and (in specific circumstances) pre-odontoblasts can also transdifferentiate into osteoblasts. Moreover, bone and teeth share outcomes when affected by systemic disorders of mineral homeostasis or of the extracellular matrix, and by adverse effects of treatments such as bisphosphonates and fluoride. Unlike bone, teeth have more permanent effects from systemic disorders because they are not remodelled after they are formed. This Review discusses the normal processes of bone and tooth development, followed by disorders that have effects on both bone and teeth, versus disorders that have effects in one without affecting the other. The takeaway message is that bone specialists should know when to screen for dental disorders, just as dental specialists should recognize when a tooth disorder should raise suspicions about a possible underlying bone disorder.


Assuntos
Biomineralização/fisiologia , Desenvolvimento Ósseo/fisiologia , Doenças do Desenvolvimento Ósseo/metabolismo , Odontogênese/fisiologia , Doenças Dentárias/metabolismo , Animais , Doenças do Desenvolvimento Ósseo/patologia , Humanos , Doenças Dentárias/patologia
5.
Cells ; 10(1)2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33467654

RESUMO

The transient receptor potential vanilloid 4 channel (TRPV4) belongs to the mammalian TRP superfamily of cation channels. TRPV4 is ubiquitously expressed, activated by a disparate array of stimuli, interacts with a multitude of proteins, and is modulated by a range of post-translational modifications, the majority of which we are only just beginning to understand. Not surprisingly, a great number of physiological roles have emerged for TRPV4, as have various disease states that are attributable to the absence, or abnormal functioning, of this ion channel. This review will highlight structural features of TRPV4, endogenous and exogenous activators of the channel, and discuss the reported roles of TRPV4 in health and disease.


Assuntos
Perfilação da Expressão Gênica , Canais de Cátion TRPV/metabolismo , Animais , Doenças do Desenvolvimento Ósseo/metabolismo , Regulação da Expressão Gênica , Estudos de Associação Genética , Humanos , Ligantes , Lipídeos , Camundongos , Mutação , Fosforilação , Mapeamento de Interação de Proteínas , Isoformas de Proteínas , Temperatura
6.
EBioMedicine ; 62: 103075, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33242826

RESUMO

BACKGROUND: Beyond its structural role in the skeleton, the extracellular matrix (ECM), particularly basement membrane proteins, facilitates communication with intracellular signaling pathways and cell to cell interactions to control differentiation, proliferation, migration and survival. Alterations in extracellular proteins cause a number of skeletal disorders, yet the consequences of an abnormal ECM on cellular communication remains less well understood METHODS: Clinical and radiographic examinations defined the phenotype in this unappreciated bent bone skeletal disorder. Exome analysis identified the genetic alteration, confirmed by Sanger sequencing. Quantitative PCR, western blot analyses, immunohistochemistry, luciferase assay for WNT signaling were employed to determine RNA, proteins levels and localization, and dissect out the underlying cell signaling abnormalities.  Migration and wound healing assays examined cell migration properties. FINDINGS: This bent bone dysplasia resulted from biallelic mutations in LAMA5, the gene encoding the alpha-5 laminin basement membrane protein. This finding uncovered a mechanism of disease driven by ECM-cell interactions between alpha-5-containing laminins, and integrin-mediated focal adhesion signaling, particularly in cartilage. Loss of LAMA5 altered ß1 integrin signaling through the non-canonical kinase PYK2 and the skeletal enriched SRC kinase, FYN. Loss of LAMA5 negatively impacted the actin cytoskeleton, vinculin localization, and WNT signaling. INTERPRETATION: This newly described mechanism revealed a LAMA5-ß1 Integrin-PYK2-FYN focal adhesion complex that regulates skeletogenesis, impacted WNT signaling and, when dysregulated, produced a distinct skeletal disorder. FUNDING: Supported by NIH awards R01 AR066124, R01 DE019567, R01 HD070394, and U54HG006493, and Czech Republic grants INTER-ACTION LTAUSA19030, V18-08-00567 and GA19-20123S.


Assuntos
Alelos , Doenças do Desenvolvimento Ósseo/etiologia , Doenças do Desenvolvimento Ósseo/metabolismo , Adesão Celular/genética , Laminina/genética , Laminina/metabolismo , Mutação , Transdução de Sinais , Doenças do Desenvolvimento Ósseo/diagnóstico , Osso e Ossos/anormalidades , Osso e Ossos/diagnóstico por imagem , Condrócitos/metabolismo , Análise Mutacional de DNA , Quinase 2 de Adesão Focal/genética , Quinase 2 de Adesão Focal/metabolismo , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Fenótipo , Via de Sinalização Wnt , Quinases da Família src/metabolismo
10.
Dis Model Mech ; 13(6)2020 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-32430393

RESUMO

Human disorders of the post-squalene cholesterol biosynthesis pathway frequently result in skeletal abnormalities, yet our understanding of the mechanisms involved is limited. In a forward-genetic approach, we have found that a late-onset skeletal mutant, named kolibernu7 , is the result of a cis-acting regulatory mutation leading to loss of methylsterol monooxygenase 1 (msmo1) expression within pre-hypertrophic chondrocytes. Generated msmo1nu81 knockdown mutation resulted in lethality at larval stage. We demonstrated that this is a result of both cholesterol deprivation and sterol intermediate accumulation by creating a mutation eliminating activity of Lanosterol synthase (Lss). Our results indicate that double lssnu60;msmo1nu81 and single lssnu60 mutants survive significantly longer than msmo1nu81 homozygotes. Liver-specific restoration of either Msmo1 or Lss in corresponding mutant backgrounds suppresses larval lethality. Rescued mutants develop dramatic skeletal abnormalities, with a loss of Msmo1 activity resulting in a more-severe patterning defect of a near-complete loss of hypertrophic chondrocytes marked by col10a1a expression. Our analysis suggests that hypertrophic chondrocytes depend on endogenous cholesterol synthesis, and blocking C4 demethylation exacerbates the cholesterol deficiency phenotype. Our findings offer new insight into the genetic control of bone development and provide new zebrafish models for human disorders of the cholesterol biosynthesis pathway.


Assuntos
Doenças do Desenvolvimento Ósseo/metabolismo , Osso e Ossos/metabolismo , Colesterol/biossíntese , Condrócitos/metabolismo , Fígado/metabolismo , Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/patologia , Osso e Ossos/patologia , Condrócitos/patologia , Colágeno Tipo X/genética , Colágeno Tipo X/metabolismo , Modelos Animais de Doenças , Predisposição Genética para Doença , Transferases Intramoleculares/genética , Transferases Intramoleculares/metabolismo , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Mutação , Fenótipo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
11.
Sci Rep ; 10(1): 7408, 2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32366894

RESUMO

ANKH mutations are associated with calcium pyrophosphate deposition disease and craniometaphyseal dysplasia. This study investigated the effects of these ANKH mutants on cellular localisation and associated biochemistry. We generated four ANKH overexpression-plasmids containing either calcium pyrophosphate deposition disease or craniometaphyseal dysplasia linked mutations: P5L, E490del and S375del, G389R. They were transfected into CH-8 articular chondrocytes and HEK293 cells. The ANKH mutants dynamic differential localisations were imaged and we investigated the interactions with the autophagy marker LC3. Extracellular inorganic pyrophosphate, mineralization, ENPP1 activity expression of ENPP1, TNAP and PIT-1 were measured. P5L delayed cell membrane localisation but once recruited into the membrane it increased extracellular inorganic pyrophosphate, mineralization, and ENPP1 activity. E490del remained mostly cytoplasmic, forming punctate co-localisations with LC3, increased mineralization, ENPP1 and ENPP1 activity with an initial but unsustained increase in TNAP and PIT-1. S375del trended to decrease extracellular inorganic pyrophosphate, increase mineralization. G389R delayed cell membrane localisation, trended to decrease extracellular inorganic pyrophosphate, increased mineralization and co-localised with LC3. Our results demonstrate a link between pathological localisation of ANKH mutants with different degrees in mineralization. Furthermore, mutant ANKH functions are related to synthesis of defective proteins, inorganic pyrophosphate transport, ENPP1 activity and expression of ENPP1, TNAP and PIT-1.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Condrocalcinose/genética , Anormalidades Craniofaciais/genética , Hiperostose/genética , Hipertelorismo/genética , Mutação , Proteínas de Transporte de Fosfato/genética , Fosfatase Alcalina , Autofagia , Doenças do Desenvolvimento Ósseo/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Condrocalcinose/metabolismo , Condrócitos/metabolismo , Anormalidades Craniofaciais/metabolismo , Difosfatos/metabolismo , Células HEK293 , Humanos , Hiperostose/metabolismo , Hipertelorismo/metabolismo , Microscopia Confocal , Proteínas de Transporte de Fosfato/metabolismo , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Domínios Proteicos , Pirofosfatases/genética , Pirofosfatases/metabolismo , Fator de Transcrição Pit-1/genética , Fator de Transcrição Pit-1/metabolismo
12.
Int J Mol Sci ; 21(8)2020 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-32295296

RESUMO

Proteoglycans (PGs) are macromolecules present on the cell surface and in the extracellular matrix that confer specific mechanical, biochemical, and physical properties to tissues. Sulfate groups present on glycosaminoglycans, linear polysaccharide chains attached to PG core proteins, are fundamental for correct PG functions. Indeed, through the negative charge of sulfate groups, PGs interact with extracellular matrix molecules and bind growth factors regulating tissue structure and cell behavior. The maintenance of correct sulfate metabolism is important in tissue development and function, particularly in cartilage where PGs are fundamental and abundant components of the extracellular matrix. In chondrocytes, the main sulfate source is the extracellular space, then sulfate is taken up and activated in the cytosol to the universal sulfate donor to be used in sulfotransferase reactions. Alteration in each step of sulfate metabolism can affect macromolecular sulfation, leading to the onset of diseases that affect mainly cartilage and bone. This review presents a panoramic view of skeletal dysplasias caused by mutations in genes encoding for transporters or enzymes involved in macromolecular sulfation. Future research in this field will contribute to the understanding of the disease pathogenesis, allowing the development of targeted therapies aimed at alleviating, preventing, or modifying the disease progression.


Assuntos
Doenças do Desenvolvimento Ósseo/etiologia , Doenças do Desenvolvimento Ósseo/metabolismo , Suscetibilidade a Doenças , Processamento de Proteína Pós-Traducional , Sulfatos/metabolismo , Animais , Cartilagem/metabolismo , Metabolismo Energético/genética , Matriz Extracelular , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Glicosaminoglicanos/metabolismo , Humanos , Redes e Vias Metabólicas , Fenótipo , Proteoglicanas/metabolismo
13.
Commun Biol ; 3(1): 144, 2020 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-32214226

RESUMO

The Creeper (Cp) chicken is characterized by chondrodystrophy in Cp/+ heterozygotes and embryonic lethality in Cp/Cp homozygotes. However, the genes underlying the phenotypes have not been fully known. Here, we show that a 25 kb deletion on chromosome 7, which contains the Indian hedgehog (IHH) and non-homologous end-joining factor 1 (NHEJ1) genes, is responsible for the Cp trait in Japanese bantam chickens. IHH is essential for chondrocyte maturation and is downregulated in the Cp/+ embryos and completely lost in the Cp/Cp embryos. This indicates that chondrodystrophy is caused by the loss of IHH and that chondrocyte maturation is delayed in Cp/+ heterozygotes. The Cp/Cp homozygotes exhibit impaired DNA double-strand break (DSB) repair due to the loss of NHEJ1, resulting in DSB accumulation in the vascular and nervous systems, which leads to apoptosis and early embryonic death.


Assuntos
Doenças do Desenvolvimento Ósseo/veterinária , Osso e Ossos/embriologia , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Deleção de Genes , Proteínas Hedgehog/genética , Doenças das Aves Domésticas/genética , Animais , Apoptose , Doenças do Desenvolvimento Ósseo/embriologia , Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/metabolismo , Osso e Ossos/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Embrião de Galinha , Enzimas Reparadoras do DNA/deficiência , Proteínas de Ligação a DNA/deficiência , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Proteínas Hedgehog/deficiência , Heterozigoto , Homozigoto , Fenótipo , Doenças das Aves Domésticas/embriologia , Doenças das Aves Domésticas/metabolismo
14.
FEBS Open Bio ; 10(1): 127-134, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31725956

RESUMO

Osteosarcoma (OS) is the primary bone malignancy in children and adolescents, with a high incidence of lung metastasis and poor prognosis. Here, we report that growth hormone receptor (GHR) is overexpressed in OS samples compared with osteofibrous dysplasia. We subsequently demonstrated that GHR knockdown inhibited colony formation, promoted cell apoptosis and decreased the number of cells at G2/M phase in 143B and U2OS cells. Furthermore, knockdown of GHR inhibited tumor growth in vivo. Together, these findings indicate that GHR modulates cell proliferation and metastasis through the phosphoinositide 3-kinase/AKT signaling pathway and may be suitable for use as a putative biomarker of OS.


Assuntos
Doenças do Desenvolvimento Ósseo/metabolismo , Neoplasias Ósseas/metabolismo , Osteossarcoma/metabolismo , Receptores da Somatotropina/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Doenças do Desenvolvimento Ósseo/tratamento farmacológico , Doenças do Desenvolvimento Ósseo/patologia , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Proliferação de Células/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , RNA Interferente Pequeno/farmacologia , Receptores da Somatotropina/antagonistas & inibidores , Receptores da Somatotropina/genética , Células Tumorais Cultivadas
15.
Toxicol Lett ; 321: 122-130, 2020 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-31874197

RESUMO

Our previous studies confirmed that prenatal caffeine exposure (PCE) could induce susceptibility to osteoarthritis in adult offspring rats due to poor chondrocyte differentiation, but its mechanism remains to be further investigated. This study aimed to explore whether subchondral bone dysplasia mediates susceptibility to osteoarthritis in adult offspring rats induced by PCE. Pregnant Wistar rats were treated with caffeine (120 mg/kg.d) or saline from gestational day (GD) 9 to 20. The female offspring were euthanized to collect femurs at GD20, postnatal week (PW) 6, and PW28 (non-ovariectomy and ovariectomy groups) to detect osteoarthritis-like phenotype, subchondral bone mass, ossification center development, and other evidence. The results showed that PCE increased the Mankin score of pathological articular cartilage, but decreased articular cartilage thickness and subchondral bone mass, which were more obvious after ovariectomy. Meanwhile, the correlation analysis results demonstrated that the Mankin score of articular cartilage was significantly negatively correlated with subchondral bone mass, and the thickness of articular cartilage was significantly positively correlated with subchondral bone mass. Further, the length and area of the primary and secondary ossification centers, the number of osteoblasts, and the related genes' expression of osteogenic differentiation (e.g., Runx2, BSP, ALP, and OCN) were all significantly decreased in the PCE group before and after birth. Taken together, PCE induced susceptibility to osteoarthritis in adult female offspring, which was likely related to the subchondral bone dysplasia and reduction of subchondral bone mass production due to developmental disorder of primary and secondary ossification centers caused by osteoblast differentiation disability before and after birth.


Assuntos
Doenças do Desenvolvimento Ósseo/induzido quimicamente , Cafeína/toxicidade , Cartilagem Articular/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/toxicidade , Osteoartrite/induzido quimicamente , Osteogênese/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Fatores Etários , Animais , Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/metabolismo , Doenças do Desenvolvimento Ósseo/patologia , Cartilagem Articular/patologia , Diferenciação Celular/efeitos dos fármacos , Feminino , Fêmur/efeitos dos fármacos , Fêmur/metabolismo , Fêmur/patologia , Idade Gestacional , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite/patologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteogênese/genética , Ovariectomia , Gravidez , Ratos Wistar , Fatores Sexuais , Tíbia/efeitos dos fármacos , Tíbia/metabolismo , Tíbia/patologia
16.
Mol Genet Genomic Med ; 7(9): e844, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31350823

RESUMO

BACKGROUND: Geleophysic dysplasia (GPHYSD) is a disorder characterized by dysmorphic features, stiff joints and cardiac involvement due to defects of TGF-ß signaling. GPHYSD can be caused by mutations in FBN1, ADAMTLS2, and LTBP3 genes. METHODS AND RESULTS: Consistent with previous reports, we found intracellular inclusions of unknown material by electron microscopy (EM) in skin fibroblasts of two GPHYSD individuals carrying FBN1 mutations. Moreover, we found that the storage material is enclosed within lysosomes and is associated with the upregulation of several lysosomal genes. Treatment of GPHYSD fibroblasts carrying FBN1 mutations with the angiotensin II receptor type 1 inhibitor losartan that inhibits TGF-ß signaling did not reduce the storage but improved the extracellular deposition of fibrillin-1 microfibrils. CONCLUSION: Losartan is a promising candidate drug for treatment of GPHYSD due to FBN1 defects.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/metabolismo , Fibrilina-1/genética , Fibroblastos/metabolismo , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/metabolismo , Losartan/farmacologia , Lisossomos/metabolismo , Microfibrilas/metabolismo , Pele/metabolismo , Pele/patologia , Adolescente , Doenças do Desenvolvimento Ósseo/patologia , Criança , Pré-Escolar , Matriz Extracelular , Feminino , Fibroblastos/ultraestrutura , Humanos , Lactente , Deformidades Congênitas dos Membros/patologia , Masculino , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
17.
Am J Hum Genet ; 105(1): 108-121, 2019 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31204009

RESUMO

Pediatric acute liver failure (ALF) is life threatening with genetic, immunologic, and environmental etiologies. Approximately half of all cases remain unexplained. Recurrent ALF (RALF) in infants describes repeated episodes of severe liver injury with recovery of hepatic function between crises. We describe bi-allelic RINT1 alterations as the cause of a multisystem disorder including RALF and skeletal abnormalities. Three unrelated individuals with RALF onset ≤3 years of age have splice alterations at the same position (c.1333+1G>A or G>T) in trans with a missense (p.Ala368Thr or p.Leu370Pro) or in-frame deletion (p.Val618_Lys619del) in RINT1. ALF episodes are concomitant with fever/infection and not all individuals have complete normalization of liver function testing between episodes. Liver biopsies revealed nonspecific liver damage including fibrosis, steatosis, or mild increases in Kupffer cells. Skeletal imaging revealed abnormalities affecting the vertebrae and pelvis. Dermal fibroblasts showed splice-variant mediated skipping of exon 9 leading to an out-of-frame product and nonsense-mediated transcript decay. Fibroblasts also revealed decreased RINT1 protein, abnormal Golgi morphology, and impaired autophagic flux compared to control. RINT1 interacts with NBAS, recently implicated in RALF, and UVRAG, to facilitate Golgi-to-ER retrograde vesicle transport. During nutrient depletion or infection, Golgi-to-ER transport is suppressed and autophagy is promoted through UVRAG regulation by mTOR. Aberrant autophagy has been associated with the development of similar skeletal abnormalities and also with liver disease, suggesting that disruption of these RINT1 functions may explain the liver and skeletal findings. Clarifying the pathomechanism underlying this gene-disease relationship may inform therapeutic opportunities.


Assuntos
Autofagia , Doenças do Desenvolvimento Ósseo/etiologia , Proteínas de Ciclo Celular/genética , Fibroblastos/patologia , Falência Hepática Aguda/etiologia , Mutação , Idade de Início , Alelos , Sequência de Aminoácidos , Doenças do Desenvolvimento Ósseo/metabolismo , Doenças do Desenvolvimento Ósseo/patologia , Proteínas de Ciclo Celular/metabolismo , Criança , Pré-Escolar , Feminino , Fibroblastos/metabolismo , Complexo de Golgi/metabolismo , Complexo de Golgi/patologia , Humanos , Lactente , Falência Hepática Aguda/metabolismo , Falência Hepática Aguda/patologia , Masculino , Linhagem , Transporte Proteico , Recidiva , Homologia de Sequência
18.
Am J Med Genet A ; 179(7): 1270-1275, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31148362

RESUMO

PIGQ (OMIM *605754) encodes phosphatidylinositol glycan biosynthesis class Q (PIGQ) and is required for proper functioning of an N-acetylglucosamine transferase complex in a similar manner to the more established PIGA, PIGC, and PIGH. There are two previous patients reported with homozygous and apparently deleterious PIGQ mutations. Here, we provide the first detailed clinical report of a patient with heterozygous deleterious mutations associated with glycosylphosphatidylinositol-anchored protein (GPI-AP) biosynthesis deficiency. Our patient died at 10 months of age. The rare skeletal findings in this disorder expand the differential diagnosis of long bone radiolucent lesions and sphenoid wing dysplasia. This clinical report describes a new and rare disorder-PIGQ GPI-AP biosynthesis deficiency syndrome.


Assuntos
Anormalidades Múltiplas/genética , Doenças do Desenvolvimento Ósseo/genética , Glicosilfosfatidilinositóis/deficiência , Proteínas de Membrana/genética , Hipotonia Muscular/genética , Mutação , Convulsões/genética , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Doenças do Desenvolvimento Ósseo/diagnóstico , Doenças do Desenvolvimento Ósseo/metabolismo , Doenças do Desenvolvimento Ósseo/patologia , Evolução Fatal , Expressão Gênica , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/metabolismo , Heterozigoto , Humanos , Lactente , Masculino , Proteínas de Membrana/deficiência , Hipotonia Muscular/diagnóstico , Hipotonia Muscular/metabolismo , Hipotonia Muscular/patologia , Fenótipo , Convulsões/diagnóstico , Convulsões/metabolismo , Convulsões/patologia , Osso Esfenoide/metabolismo , Osso Esfenoide/patologia , Síndrome , Sequenciamento do Exoma
19.
Hum Mol Genet ; 28(15): 2573-2588, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31009951

RESUMO

Mutations in subunits of the cilia-specific cytoplasmic dynein-2 (CD2) complex cause short-rib thoracic dystrophy syndromes (SRTDs), characterized by impaired bone growth and life-threatening perinatal respiratory complications. Different SRTD mutations result in varying disease severities. It remains unresolved whether this reflects the extent of retained hypomorphic protein functions or relative importance of the affected subunits for the activity of the CD2 holoenzyme. To define the contribution of the LC8-type dynein light chain subunit to the CD2 complex, we have generated Dynll1-deficient mouse strains, including the first-ever conditional knockout (KO) mutant for any CD2 subunit. Germline Dynll1 KO mice exhibit a severe ciliopathy-like phenotype similar to mice lacking another CD2 subunit, Dync2li1. Limb mesoderm-specific loss of Dynll1 results in severe bone shortening similar to human SRTD patients. Mechanistically, loss of Dynll1 leads to a partial depletion of other SRTD-related CD2 subunits, severely impaired retrograde intra-flagellar transport, significant thickening of primary cilia and cilia signaling defects. Interestingly, phenotypes of Dynll1-deficient mice are very similar to entirely cilia-deficient Kif3a/Ift88-null mice, except that they never present with polydactyly and retain relatively higher signaling outputs in parts of the hedgehog pathway. Compared to complete loss of Dynll1, maintaining very low DYNLL1 levels in mice lacking the Dynll1-transcription factor ASCIZ (ATMIN) results in significantly attenuated phenotypes and improved CD2 protein levels. The results suggest that primary cilia can maintain some functionality in the absence of intact CD2 complexes and provide a viable animal model for the analysis of the underlying bone development defects of SRTDs.


Assuntos
Doenças do Desenvolvimento Ósseo/metabolismo , Cílios/metabolismo , Ciliopatias/metabolismo , Dineínas do Citoplasma/genética , Osteogênese , Animais , Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/fisiopatologia , Células Cultivadas , Cílios/fisiologia , Ciliopatias/genética , Ciliopatias/fisiopatologia , Dineínas do Citoplasma/metabolismo , Dineínas do Citoplasma/fisiologia , Extremidades/patologia , Extremidades/fisiopatologia , Proteínas Hedgehog/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Transdução de Sinais , Fatores de Transcrição/metabolismo
20.
Clin Calcium ; 29(3): 323-328, 2019.
Artigo em Japonês | MEDLINE | ID: mdl-30814377

RESUMO

Identification of responsible genes for skeletal dysplasias evidences their critical roles in the skeletal development and maintenance. Mutations in the genes encoding the components of Wnt canonical pathway, which include WNT1, LRP5, LRP4, SOST and WTX, cause the disorders characterized by abnormal in bone mass. On the other hand, mutations in the genes for the components of Wnt non-canonical pathway such as WNT5A, ROR2, DVL1 and DVL3 are associated with dysmorphic skeletal disorders which manifest short limbs and facial anomalies. Thus, both canonical and non-canonical pathways of Wnt signaling play substantial roles in the human skeletons, and it is suggested that the former mainly controls bone mass while the latter regulates skeletal morphogenesis.


Assuntos
Doenças do Desenvolvimento Ósseo/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Doenças do Desenvolvimento Ósseo/genética , Humanos , Mutação , Proteínas Wnt/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA