Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 439
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Am J Med Genet A ; 194(10): e63709, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38801192

RESUMO

Osteopathia Striata with Cranial Sclerosis (OSCS) is a rare genetic condition primarily characterized by metaphyseal striations of long bones, bone sclerosis, macrocephaly, and other congenital anomalies. It is caused by pathogenic variants in AMER1, a tumor suppressor and a WNT signaling repressor gene with key roles in tissue regeneration, neurodevelopment, tumorigenesis, and other developmental processes. While somatic AMER1 pathogenic variants have frequently been identified in several tumor types (e.g., Wilms tumor and colorectal cancer), whether OSCS (i.e., with AMER1 germline variants) is a tumor predisposition syndrome is not clear, with only nine cases reported with tumors. We here report the first case of neuroblastoma diagnosed in a male child with OSCS, review all previously reported tumors diagnosed in individuals with OSCS, and discuss potential tumorigenic mechanisms of AMER1. Our report adds to the accumulating evidence suggesting OSCS is a tumor predisposition condition, highlighting the importance of maintaining a high index of suspicion for the associated tumors when evaluating patients with OSCS. Importantly, Wilms tumor stands out as the most commonly observed tumor in OSCS patients, underscoring the need for regular surveillance.


Assuntos
Predisposição Genética para Doença , Neuroblastoma , Osteosclerose , Humanos , Masculino , Osteosclerose/genética , Osteosclerose/patologia , Neuroblastoma/genética , Neuroblastoma/patologia , Lactente , Pré-Escolar , Proteínas Supressoras de Tumor , Proteínas Adaptadoras de Transdução de Sinal
2.
Am J Med Genet A ; 194(11): e63800, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38934054

RESUMO

We report three siblings homozygous for CSF1R variant c.1969 + 115_1969 + 116del to expand the phenotype of "brain abnormalities, neurodegeneration, and dysosteosclerosis" (BANDDOS) and discuss its link with "adult leukoencephalopathy with axonal spheroids and pigmented glia" (ALSP), caused by heterozygous CSF1R variants. We evaluated medical, radiological, and laboratory findings and reviewed the literature. Patients presented with developmental delay, therapy-resistant epilepsy, dysmorphic features, and skeletal abnormalities. Secondary neurological decline occurred from 23 years in sibling one and from 20 years in sibling two. Brain imaging revealed multifocal white matter abnormalities and calcifications during initial disease in siblings two and three. Developmental brain anomalies, seen in all three, were most severe in sibling two. During neurological decline in siblings one and two, the leukoencephalopathy was progressive and had the MRI appearance of ALSP. Skeletal survey revealed osteosclerosis, most severe in sibling three. Blood markers, monocytes, dendritic cell subsets, and T-cell proliferation capacity were normal. Literature review revealed variable initial disease and secondary neurological decline. BANDDOS presents with variable dysmorphic features, skeletal dysplasia, developmental delay, and epilepsy with on neuro-imaging developmental brain anomalies, multifocal white matter abnormalities, and calcifications. Secondary neurological decline occurs with a progressive leukoencephalopathy, in line with early onset ALSP. Despite the role of CSF1R signaling in myeloid development, immune deficiency is absent. Phenotype varies within families; skeletal and neurological manifestations may be disparate.


Assuntos
Encéfalo , Calcinose , Homozigoto , Leucoencefalopatias , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos , Irmãos , Humanos , Leucoencefalopatias/genética , Leucoencefalopatias/patologia , Leucoencefalopatias/diagnóstico por imagem , Masculino , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Feminino , Calcinose/genética , Calcinose/patologia , Encéfalo/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/anormalidades , Adulto , Imageamento por Ressonância Magnética , Osteosclerose/genética , Osteosclerose/patologia , Fenótipo , Adulto Jovem , Mutação/genética , Criança , Adolescente , Receptor de Fator Estimulador de Colônias de Macrófagos
3.
Am J Hum Genet ; 104(5): 925-935, 2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-30982609

RESUMO

Colony stimulating factor 1 receptor (CSF1R) plays key roles in regulating development and function of the monocyte/macrophage lineage, including microglia and osteoclasts. Mono-allelic mutations of CSF1R are known to cause hereditary diffuse leukoencephalopathy with spheroids (HDLS), an adult-onset progressive neurodegenerative disorder. Here, we report seven affected individuals from three unrelated families who had bi-allelic CSF1R mutations. In addition to early-onset HDLS-like neurological disorders, they had brain malformations and skeletal dysplasia compatible to dysosteosclerosis (DOS) or Pyle disease. We identified five CSF1R mutations that were homozygous or compound heterozygous in these affected individuals. Two of them were deep intronic mutations resulting in abnormal inclusion of intron sequences in the mRNA. Compared with Csf1r-null mice, the skeletal and neural phenotypes of the affected individuals appeared milder and variable, suggesting that at least one of the mutations in each affected individual is hypomorphic. Our results characterized a unique human skeletal phenotype caused by CSF1R deficiency and implied that bi-allelic CSF1R mutations cause a spectrum of neurological and skeletal disorders, probably depending on the residual CSF1R function.


Assuntos
Encéfalo/anormalidades , Leucoencefalopatias/etiologia , Mutação , Osteocondrodisplasias/etiologia , Osteosclerose/etiologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Adolescente , Adulto , Alelos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Pré-Escolar , Feminino , Humanos , Leucoencefalopatias/patologia , Masculino , Camundongos , Camundongos Knockout , Osteocondrodisplasias/patologia , Osteosclerose/patologia , Fenótipo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/fisiologia , Adulto Jovem
4.
PLoS Genet ; 15(6): e1008244, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31233501

RESUMO

Berardinelli-Seip congenital generalized lipodystrophy is associated with increased bone mass suggesting that fat tissue regulates the skeleton. Because there is little mechanistic information regarding this issue, we generated "fat-free" (FF) mice completely lacking visible visceral, subcutaneous and brown fat. Due to robust osteoblastic activity, trabecular and cortical bone volume is markedly enhanced in these animals. FF mice, like Berardinelli-Seip patients, are diabetic but normalization of glucose tolerance and significant reduction in circulating insulin fails to alter their skeletal phenotype. Importantly, the skeletal phenotype of FF mice is completely rescued by transplantation of adipocyte precursors or white or brown fat depots, indicating that adipocyte derived products regulate bone mass. Confirming such is the case, transplantation of fat derived from adiponectin and leptin double knockout mice, unlike that obtained from their WT counterparts, fails to normalize FF bone. These observations suggest a paucity of leptin and adiponectin may contribute to the increased bone mass of Berardinelli-Seip patients.


Assuntos
Adiponectina/genética , Leptina/genética , Lipodistrofia Generalizada Congênita/genética , Osteosclerose/genética , Adipócitos/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Densidade Óssea/genética , Modelos Animais de Doenças , Feminino , Glucose/genética , Glucose/metabolismo , Humanos , Insulina/genética , Gordura Intra-Abdominal/metabolismo , Lipodistrofia Generalizada Congênita/complicações , Lipodistrofia Generalizada Congênita/patologia , Camundongos , Camundongos Knockout , Osteosclerose/etiologia , Osteosclerose/metabolismo , Osteosclerose/patologia , Esqueleto/metabolismo , Esqueleto/patologia , Gordura Subcutânea/metabolismo
5.
Ann Diagn Pathol ; 56: 151878, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34953234

RESUMO

Charcot neuropathic arthropathy is a degenerative, debilitating disease that affects the foot and ankle in patients with diabetes and peripheral neuropathy, often resulting in destruction, amputation. Proposed etiologies include neurotraumatic, inflammatory, and neurovascular. There has been no previous animal model for Charcot. This study proposes a novel rodent model of induced neuropathic arthropathy to understand the earliest progressive pathologic changes of human Charcot. High-fat-diet-induced obese (DIO) Wild-type C57BL/6J mice (n = 8, diabetic) and age-matched low-fat-diet controls (n = 6) were run on an inclined high-intensity treadmill protocol four times per week for 7 weeks to induce mechanical neurotrauma to the hind-paw, creating Charcot neuropathic arthropathy. Sensory function and radiologic correlation were assessed; animals were sacrificed to evaluate hindpaw soft tissue and joint pathology. With this model, Charcot-DIO mice reveals early pathologic features of Charcot neuropathic arthropathy, a distinctive subchondral microfracture callus, perichondral/subchondral osseous hypertrophy/osteosclerosis, that precedes fragmentation/destruction observed in human surgical pathology specimens. There is intraneural vacuolar-myxoid change and arteriolosclerosis. The DIO mice demonstrated significant hot plate sensory neuropathy compared (P < 0.01), radiographic collapse of the longitudinal arch in DIO mice (P < 0.001), and diminished bone density in DIO, compared with normal controls. Despite exercise, high-fat-DIO mice increased body weight and percentage of body fat (P < 0.001). This murine model of diet-induced obesity and peripheral neuropathy, combined with repetitive mechanical trauma, simulates the earliest changes observed in human Charcot neuropathic arthropathy, of vasculopathic-neuropathic etiology. An understanding of early pathophysiology may assist early diagnosis and intervention and reduce patient morbidity and mortality in Charcot neuropathic arthropathy.


Assuntos
Artropatia Neurogênica/patologia , Cartilagem Articular/patologia , Fraturas de Estresse/patologia , Osteosclerose/patologia , Animais , Modelos Animais de Doenças , Camundongos , Obesidade/patologia
6.
J Hum Genet ; 66(4): 371-377, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33037392

RESUMO

Dysosteosclerosis (DOS) is a distinct form of sclerosing bone disease characterized by platyspondyly and progressive osteosclerosis. DOS is genetically heterogeneous. Three causal genes, SLC29A3, CSF1R, and TNFRSF11A are reported. TNFRSF11A-associated DOS has been identified in two patients; however, TNFRSF11A is also a causal gene for osteopetrosis, autosomal recessive 7 (OP-AR7). Whole-exome sequencing in a patient with sclerosing bone disease identified novel compound heterozygous variants (c.414_427 + 7del, c.1664del) in TNFRSF11A. We examined the impact of the two variants on five splicing isoforms of TNFRSF11A by RT-PCR. We found that c.1664del resulted in elongated proteins (p.S555Cfs*121, etc.), while c.414_427 + 7del generated two aberrant splicing products (p.A139Wfs*19 and p.E132Dfs*19) that lead to nonsense mediated mRNA decay (NMD). In the previous two cases of TNFRSF11A-associated DOS, their mutations produced truncated TNFRSF11A protein isoforms. The mutations in all three cases thus contrast with TNFRSF11A mutations reported in OP-AR7, which does not generated truncated or elongated TNFRSF11A proteins. Thus, we identified the third case of TNFRSF11A-associated DOS and reinforced the genotype-phenotype correlation that aberrant protein-producing TNFRSF11A mutations cause DOS.


Assuntos
Mutação , Osteosclerose/patologia , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Pré-Escolar , Feminino , Humanos , Osteosclerose/genética , Osteosclerose/metabolismo , Prognóstico , Sequenciamento do Exoma
7.
J Hum Genet ; 66(6): 607-611, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33402699

RESUMO

Dysosteosclerosis (DOS) is a rare sclerosing bone dysplasia characterized by osteosclerosis and platyspondyly. DOS is genetically heterogeneous and causally associated with mutations in three genes, SLC29A3, CSF1R, and TNFRSF11A. TNFRSF11A has been known as the causal gene for osteopetrosis, autosomal recessive 7, and is recently reported to cause DOS in three cases, which show a complex genotype-phenotype relationship. The phenotypic spectrum of TNFRSF11A-associated sclerosing bone dysplasia remains unclear and needs to be characterized further in more cases with molecular genetic diagnosis. Here, we report another TNFRSF11A-associated DOS case with a homozygous missense mutation (p.R129C). The mutation effect is different from the previous three cases, in which truncated or elongated RANK proteins were generated in isoform specific manner, thus enriching our understanding of the genotype-phenotype association in TNFRSF11A-associated sclerosing bone dysplasia. Besides DOS, our case presented with intracranial extramedullary hematopoiesis, which is an extremely rare condition and has not been identified in any other sclerosing bone dysplasias with molecular genetic diagnosis. Our findings provide the fourth case of TNFRSF11A-associated DOS and further expand its phenotypic spectrum.


Assuntos
Hematopoese/genética , Osteosclerose/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Doenças Ósseas , Criança , Pré-Escolar , Feminino , Estudos de Associação Genética , Heterogeneidade Genética , Homozigoto , Humanos , Lactente , Deficiência Intelectual , Mutação/genética , Proteínas de Transporte de Nucleosídeos/genética , Osteopetrose/genética , Osteopetrose/patologia , Osteosclerose/diagnóstico , Osteosclerose/patologia , Fenótipo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Esclerose
8.
Am J Med Genet A ; 185(12): 3793-3803, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34414661

RESUMO

Osteopathia striata with cranial sclerosis (OSCS; OMIM# 300373) is a rare X-linked disorder caused by mutations of the AMER1 gene. OSCS is traditionally considered a skeletal dysplasia, characterized by cranial sclerosis and longitudinal striations in the long bone metaphyses. However, OSCS affects many body systems and varies significantly in phenotypic severity between individuals. This case series focuses on the phenotypic presentation and development of individuals with OSCS. We provide an account of 12 patients with OSCS, ranging from 5 months to 38 years of age. These patients were diagnosed with OSCS after genetic testing confirmed pathogenic mutations in AMER1. Patient consent was obtained for photos and participation. Data were collected regarding perinatal history, dysmorphic features, and review of systems. This case series documents common facial dysmorphology, as well as rare extraskeletal features of OSCS, including two patients with intestinal malrotation and two patients with pyloric stenosis. We share four apparently nonmosaic males with OSCS (one de novo and three maternal variants). We also provide a clinical update on a patient who was previously published by Chénier et al. (2012). American Journal of Medical Genetics Part A, 158, 2946-2952. More research is needed to investigate the links between genotype and phenotype and assess the long-term comorbidities and overall quality of life of individuals with OSCS.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Predisposição Genética para Doença , Osteosclerose/genética , Crânio/patologia , Proteínas Supressoras de Tumor/genética , Adolescente , Adulto , Canadá , Criança , Pré-Escolar , Feminino , Genes Ligados ao Cromossomo X , Humanos , Lactente , Masculino , Anormalidades Musculoesqueléticas , Mutação/genética , Osteosclerose/diagnóstico , Osteosclerose/patologia , Fenótipo , Gravidez , Qualidade de Vida , Crânio/diagnóstico por imagem , Adulto Jovem
9.
Am J Med Genet A ; 185(7): 2271-2277, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33837634

RESUMO

Dysosteosclerosis is a group of sclerosing bone dysplasia characterized by short stature, increased bone fragility, osteosclerosis, and platyspondyly. It is a genetically heterogeneous disorder caused by biallelic mutations in the SLC29A3, TNFRSF11A, TCIRG1, and CSF1R genes. To date, four dysosteosclerosis patients with SLC29A3 mutations have been reported. Here, we report biallelic SLC29A3 (c.303_320dupCTACTTTGAGAGCTACCT) variant in a three-year-old girl. She had large anterior fontanelle, fracture history, short stature, camptodactyly, elbow contracture, and melanocytic nevus. Initial skeletal radiographs revealed platyspondyly, dense vertebral endplates (sandwich appearance of the vertebral bodies), diffuse sclerosis of the peripheral side of the pelvic bones, sclerosis of metaphysis and diaphysis of the long bones, metaphyseal widening, and diaphyseal cortical thickening. Mild sclerosis was also present in the skull base, maxilla, rib, scapula, and phalanges. Notably, we observed that sandwich vertebrae appearance significantly resolved and sclerosis of ribs, scapula, pelvis, and long bone metaphysis regressed over a 2.5-year period. However, platyspondyly, metaphyseal widening, and diaphyseal cortical thickening persisted. In conclusion, this study demonstrates spontaneous resolution of osteosclerosis, which was not described previously in patients with dysosteosclerosis.


Assuntos
Predisposição Genética para Doença , Proteínas de Transporte de Nucleosídeos/genética , Osteosclerose/genética , Pré-Escolar , Feminino , Humanos , Mutação/genética , Osteosclerose/diagnóstico por imagem , Osteosclerose/patologia , Costelas/diagnóstico por imagem , Costelas/patologia , Coluna Vertebral/diagnóstico por imagem , Coluna Vertebral/patologia , Turquia/epidemiologia
10.
Am J Med Genet A ; 185(6): 1888-1896, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33749994

RESUMO

Colony stimulating factor 1 receptor (CSF1R, MIM# 164770) encodes a tyrosine-kinase receptor playing an important role in development of osteoclasts and microglia. Heterozygous CSF1R variants have been known to cause hereditary diffuse leukoencephalopathy with spheroids (HDLS, MIM# 221820), an adult-onset leukoencephalopathy characterized by loss of motor functions and cognitive decline. Recently, a new phenotype characterized by brain abnormalities, neurodegeneration, and dysosteosclerosis (BANDDOS) with biallelic CSF1R pathogenic variants in the etiology has been described. BANDDOS differs from HDLS by early-onset neurodegenerative changes with additional structural brain abnormalities and skeletal findings resembling dysosteosclerosis (DOS). Described skeletal findings of the disease are highly variable ranging from absence of a skeletal phenotype and milder Pyle disease-like to osteopetrosis and DOS. To date, only a few patients carrying biallelic CSF1R variants have been reported. In this clinical report, we describe three siblings with variable skeletal findings along with neurological symptoms ranging from mild to severe in whom exome sequencing revealed a novel homozygous splice site variant in canonical splice donor site of intron 21 adjacent to an exon, which encoding part of kinase domain of CSF1R along with a review of the literature.


Assuntos
Encéfalo/anormalidades , Leucoencefalopatias/genética , Transtornos do Neurodesenvolvimento/genética , Osteosclerose/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Adolescente , Encéfalo/patologia , Criança , Feminino , Predisposição Genética para Doença , Homozigoto , Humanos , Íntrons/genética , Leucoencefalopatias/patologia , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mutação/genética , Degeneração Neural/genética , Degeneração Neural/patologia , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/patologia , Transtornos do Neurodesenvolvimento/patologia , Osteocondrodisplasias/genética , Osteocondrodisplasias/patologia , Osteosclerose/patologia , Fenótipo , Irmãos
11.
Int J Mol Sci ; 22(15)2021 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-34360805

RESUMO

FAM20C is a gene coding for a protein kinase that targets S-X-E/pS motifs on different phosphoproteins belonging to diverse tissues. Pathogenic variants of FAM20C are responsible for Raine syndrome (RS), initially described as a lethal and congenital osteosclerotic dysplasia characterized by generalized atherosclerosis with periosteal bone formation, characteristic facial dysmorphisms and intracerebral calcifications. The aim of this review is to give an overview of targets and variants of FAM20C as well as RS aspects. We performed a wide phenotypic review focusing on clinical aspects and differences between all lethal (LRS) and non-lethal (NLRS) reported cases, besides the FAM20C pathogenic variant description for each. As new targets of FAM20C kinase have been identified, we reviewed FAM20C targets and their functions in bone and other tissues, with emphasis on novel targets not previously considered. We found the classic lethal and milder non-lethal phenotypes. The milder phenotype is defined by a large spectrum ranging from osteonecrosis to osteosclerosis with additional congenital defects or intellectual disability in some cases. We discuss our current understanding of FAM20C deficiency, its mechanism in RS through classic FAM20C targets in bone tissue and its potential biological relevance through novel targets in non-bone tissues.


Assuntos
Anormalidades Múltiplas , Caseína Quinase I , Fissura Palatina , Exoftalmia , Proteínas da Matriz Extracelular , Variação Genética , Microcefalia , Osteosclerose , Fenótipo , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/mortalidade , Anormalidades Múltiplas/patologia , Caseína Quinase I/genética , Caseína Quinase I/metabolismo , Fissura Palatina/genética , Fissura Palatina/metabolismo , Fissura Palatina/mortalidade , Fissura Palatina/patologia , Exoftalmia/genética , Exoftalmia/metabolismo , Exoftalmia/mortalidade , Exoftalmia/patologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Humanos , Microcefalia/genética , Microcefalia/metabolismo , Microcefalia/mortalidade , Microcefalia/patologia , Osteosclerose/genética , Osteosclerose/metabolismo , Osteosclerose/mortalidade , Osteosclerose/patologia
12.
Ann Hum Genet ; 84(1): 102-106, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31571209

RESUMO

Osteosclerotic metaphyseal dysplasia (OSMD) is a very rare autosomal-recessive disorder and a distinctive type of osteopetrosis, characterized mainly by skeletal fractures and deformity, osteosclerosis, and sometimes hypotonia, developmental delay, and seizures. Sequence variants in the leucine-rich repeat kinase 1 (LRRK1) gene underlying OSMD have been reported previously. In the present study, we investigated a 14-year-old girl suspected with OSMD in a consanguineous family of Iranian origin segregating the disease in an autosomal-recessive manner. The patient had severe short stature, multiple sclerotic lesions, sandwich vertebrae, Erlenmeyer flask deformity, and looser zones. The multifocal active bony pathology suggested multifocal bony inflammation or multiple looser fractures. Whole-exome sequencing followed by Sanger sequencing confirmation revealed a novel homozygous stop gain mutation (c.G2785T, p.E929X) in the LRRK1 gene. This is the first mutation in the LRRK1 gene, underlying OSMD, in the Iranian population and the third case worldwide. The mutation is located in the C terminal of the Roc domain, distinct from domains affected in the previous two LRRK1 mutations. Additionally, a new group of clinical indications different from the two previous cases is discussed.


Assuntos
Mutação com Ganho de Função , Osteocondrodisplasias/genética , Osteosclerose/genética , Proteínas Serina-Treonina Quinases/genética , Adolescente , Sequência de Aminoácidos , Códon sem Sentido , Feminino , Humanos , Osteocondrodisplasias/patologia , Osteosclerose/patologia , Prognóstico , Homologia de Sequência
13.
Anal Chem ; 92(1): 749-757, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31793292

RESUMO

Osteosclerosis and myefibrosis are complications of myeloproliferative neoplasms. These disorders result in excess growth of trabecular bone and collagen fibers that replace hematopoietic cells, resulting in abnormal bone marrow function. Treatments using imatinib and JAK2 pathway inhibitors can be effective on osteosclerosis and fibrosis; therefore, accurate grading is critical for tracking treatment effectiveness. Current grading standards use a four-class system based on analysis of biopsies stained with three histological stains: hematoxylin and eosin (H&E), Masson's trichrome, and reticulin. However, conventional grading can be subjective and imprecise, impacting the effectiveness of treatment. In this Article, we demonstrate that mid-infrared spectroscopic imaging may serve as a quantitative diagnostic tool for quantitatively tracking disease progression and response to treatment. The proposed approach is label-free and provides automated quantitative analysis of osteosclerosis and collagen fibrosis.


Assuntos
Osteosclerose/diagnóstico , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Biópsia , Osso e Ossos/química , Osso e Ossos/patologia , Colágeno/análise , Progressão da Doença , Fibrose , Humanos , Osteosclerose/patologia
14.
J Exp Zool B Mol Dev Evol ; 334(6): 339-349, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32729176

RESUMO

Bowhead whales are among the longest-lived mammals with an extreme lifespan of about 211 years. During the first 25 years of their lives, rib bones increase in mineral density and the medulla transitions from compact to trabecular bone. Molecular drivers associated with these phenotypic changes in bone remain unknown. This study assessed expression levels of osteogenic genes from samples of rib bones of bowheads. Samples were harvested from prenatal to 86-year-old whales, representing the first third of the bowhead lifespan. Fetal to 2-year-old bowheads showed expression levels consistent with the rapid deposition of the bone extracellular matrix. Sexually mature animals showed expression levels associated with low rates of osteogenesis and increased osteoclastogenesis. After the first 25 years of life, declines in osteogenesis corresponded with increased expression of EZH2, an epigenetic regulator of osteogenesis. These findings suggest EZH2 may be at least one epigenetic modifier that contributes to the age-related changes in the rib bone phenotype along with the transition from compact to trabecular bone. Ancient cetaceans and their fossil relatives also display these phenotypes, suggesting EZH2 may have shaped the skeleton of whales in evolutionary history.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Osteosclerose/veterinária , Costelas/fisiologia , Baleias/crescimento & desenvolvimento , Baleias/genética , Envelhecimento , Animais , Epigênese Genética , Osteosclerose/genética , Osteosclerose/patologia , Costelas/metabolismo
15.
Calcif Tissue Int ; 107(6): 636-648, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32920683

RESUMO

Autosomal recessive hypophosphatemic rickets type 1 (ARHR1) was reported to be caused by homozygous mutation of dentin matrix protein 1 (DMP1). To date, very few cases have been reported. Here, we summarized clinical, laboratory and imaging findings of ARHR1 patients in our hospital. Literature review was performed to analyze genotype-phenotype correlation. Five Chinese patients from three unrelated pedigrees presented with lower extremity deformity and short stature. Hypophosphatemia, elevated alkaline phosphatase, high intact fibroblast growth factor 23 and sclerostin were found. X-ray uncovered coexistence of osteomalacia and osteosclerosis. Although areal bone mineral density (aBMD) of axial bone measured by dual-energy X-ray absorptiometry was relatively high in all patients, volumetric BMD (vBMD) and microstructure of one adult patient's peripheral bone detected by HR-pQCT were damaged. Mutation analyses of DMP1 revealed three homozygous mutations including two novel mutations, c.54 + 1G > C and c.94C > A (p.E32X), and a reported mutation c.184-1G > A. Genotype-phenotype correlation analysis including 30 cases (25 from literature review and 5 from our study) revealed that patients harboring mutations affecting C-terminal fragment of DMP1 presented with shorter stature (Z score of height = - 3.4 ± 1.6 vs - 1.0 ± 1.6, p = 0.001) and lower serum phosphate level (0.70 ± 0.15 vs 0.84 ± 0.16, p = 0.03) than those harboring mutations only affecting N-terminal fragment. In summary, we reported five Chinese ARHR1 patients and identified two novel DMP1 mutations. High aBMD and local osteosclerosis in axial bone with low vBMD and damaged microstructure in peripheral bone were featured. Genotype-phenotype correlation analysis confirmed the important role of C-terminal fragment of DMP1.


Assuntos
Osso e Ossos/patologia , Raquitismo Hipofosfatêmico Familiar/patologia , Adulto , Densidade Óssea , China , Humanos , Hipofosfatemia , Osteomalacia/patologia , Osteosclerose/patologia
16.
Am J Med Genet A ; 182(11): 2632-2640, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32888393

RESUMO

Robinow syndrome (RS) is a genetically heterogeneous skeletal dysplasia with recent reports suggesting an osteosclerotic form of the disease. We endeavored to investigate the full spectrum of skeletal anomalies in a genetically diverse cohort of RS patients with a focus on the bone micro-architecture. Seven individuals with molecularly confirmed RS, including four with DVL1 variants and single individuals with variants in WNT5A, ROR2, and GPC4 underwent a musculoskeletal focused physical examination, dual-energy X-ray absorptiometry (DEXA) scan, and high-resolution peripheral quantitative computed tomography (HR-pQCT). Skeletal examination revealed variability in limb shortening anomalies consistent with recent reports. DEXA scan measures revealed increased total body bone mineral density (BMD) (3/7), cranial BMD (5/7), and non-cranial BMD (1/7). Cranial osteosclerosis was only observed in DVL1-RS (4/4) and GPC4-RS (1/1) subjects and in one case was complicated by choanal atresia, bilateral conductive hearing loss, and cranial nerve III, VI, and VII palsy. HR-pQCT revealed a unique pattern of low cortical BMD, increased trabecular BMD, decreased number of trabeculations, and increased thickness of the trabeculations for the DVL1-RS subjects. The spectrum of skeletal anomalies including the micro-architecture of the bones observed in RS has considerable variability with some osteosclerosis genotype-phenotype correlations more frequent with DVL1 variants.


Assuntos
Osso e Ossos/ultraestrutura , Anormalidades Craniofaciais/genética , Nanismo/genética , Estudos de Associação Genética , Deformidades Congênitas dos Membros/genética , Osteosclerose/genética , Anormalidades Urogenitais/genética , Absorciometria de Fóton , Adolescente , Adulto , Densidade Óssea , Osso e Ossos/patologia , Criança , Estudos de Coortes , Anormalidades Craniofaciais/patologia , Proteínas Desgrenhadas/genética , Nanismo/patologia , Feminino , Fêmur , Variação Genética , Glipicanas/genética , Humanos , Deformidades Congênitas dos Membros/patologia , Masculino , Pessoa de Meia-Idade , Osteosclerose/patologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Tomografia Computadorizada por Raios X , Anormalidades Urogenitais/patologia , Proteína Wnt-5a/genética , Adulto Jovem
17.
Pediatr Blood Cancer ; 67(5): e28232, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32134181

RESUMO

OBJECTIVES: Pediatric myelofibrosis is a rare entity with the largest reported series of 19 cases. We describe here the clinicopathological spectrum and outcomes of 15 cases of pediatric myelofibrosis. METHODS: Case files of myelofibrosis of patients less than 18 years were retrieved from January 2016 to January 2019, and patients with idiopathic myelofibrosis after exhaustive work-up were studied. Their clinicopathological profiles were studied and then followed up for resolution and malignant transformation. RESULTS: Of the 15 cases of idiopathic myelofibrosis, transfusion-dependent anemia (14/15) was most common presentation. Only one patient showed leukoerythroblastosis with dacryocytes. Myeloid hyperplasia was seen in 13 of 15 patients and megakaryocytic hyperplasia in 10 patients. Dysmegakaryopoiesis was seen in 8 of 15 patients, and only three had small loose megakaryocytic clustering. None showed hyperchromatic megakaryocytes, intrasinusoidal hematopoiesis, or osteosclerosis. One patient with trisomy 8 tested positive for JAK2V617F. Bone marrow biopsy was hypercellular in 13, and 8 had world health organization (WHO) MF-3 fibrosis. None of the patients developed malignancy, one had spontaneous resolution, and one patient required allogenic stem cell transplant. CONCLUSIONS: Pediatric myelofibrosis is a distinct entity from primary myelofibrosis in adults and merits mention in the WHO manual as a distinct entity.


Assuntos
Transformação Celular Neoplásica , Janus Quinase 2 , Mutação de Sentido Incorreto , Proteínas de Neoplasias , Trombopoese , Adolescente , Adulto , Substituição de Aminoácidos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Masculino , Megacariócitos/metabolismo , Megacariócitos/patologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Osteosclerose/genética , Osteosclerose/metabolismo , Osteosclerose/patologia , Mielofibrose Primária/genética , Mielofibrose Primária/metabolismo , Mielofibrose Primária/patologia , Estudos Retrospectivos
18.
PLoS Genet ; 12(5): e1006037, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27187611

RESUMO

One to two percent of all children are born with a developmental disorder requiring pediatric hospital admissions. For many such syndromes, the molecular pathogenesis remains poorly characterized. Parallel developmental disorders in other species could provide complementary models for human rare diseases by uncovering new candidate genes, improving the understanding of the molecular mechanisms and opening possibilities for therapeutic trials. We performed various experiments, e.g. combined genome-wide association and next generation sequencing, to investigate the clinico-pathological features and genetic causes of three developmental syndromes in dogs, including craniomandibular osteopathy (CMO), a previously undescribed skeletal syndrome, and dental hypomineralization, for which we identified pathogenic variants in the canine SLC37A2 (truncating splicing enhancer variant), SCARF2 (truncating 2-bp deletion) and FAM20C (missense variant) genes, respectively. CMO is a clinical equivalent to an infantile cortical hyperostosis (Caffey disease), for which SLC37A2 is a new candidate gene. SLC37A2 is a poorly characterized member of a glucose-phosphate transporter family without previous disease associations. It is expressed in many tissues, including cells of the macrophage lineage, e.g. osteoclasts, and suggests a disease mechanism, in which an impaired glucose homeostasis in osteoclasts compromises their function in the developing bone, leading to hyperostosis. Mutations in SCARF2 and FAM20C have been associated with the human van den Ende-Gupta and Raine syndromes that include numerous features similar to the affected dogs. Given the growing interest in the molecular characterization and treatment of human rare diseases, our study presents three novel physiologically relevant models for further research and therapy approaches, while providing the molecular identity for the canine conditions.


Assuntos
Anormalidades Múltiplas/genética , Aracnodactilia/genética , Blefarofimose/genética , Fissura Palatina/genética , Contratura/genética , Exoftalmia/genética , Hiperostose Cortical Congênita/genética , Microcefalia/genética , Osteosclerose/genética , Anormalidades Múltiplas/patologia , Animais , Antiporters/genética , Aracnodactilia/patologia , Blefarofimose/patologia , Doenças Ósseas/genética , Doenças Ósseas/patologia , Caseína Quinase I/genética , Fissura Palatina/patologia , Contratura/patologia , Transtornos Craniomandibulares/genética , Transtornos Craniomandibulares/patologia , Modelos Animais de Doenças , Cães , Exoftalmia/patologia , Proteínas da Matriz Extracelular/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Hiperostose Cortical Congênita/patologia , Microcefalia/patologia , Osteosclerose/patologia , Receptores Depuradores Classe F/genética
19.
Br J Dermatol ; 178(1): 261-264, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28078666

RESUMO

Erdheim-Chester disease (ECD), a type of systemic non-Langerhans cell histiocytosis, is uncommon and characterized by the accumulation of CD68+ CD1a- foamy histiocytes. It is extremely rare in children. The skin lesions of paediatric ECD have not been systemically described before. We report a case of ECD in a 3·5-year-old Chinese boy. The patient presented with generalized skin and bone involvement of 3 years' duration. Marked generalized annular maculopapular lesions with central atrophy were seen. These differed from previously reported adult xanthoma-like papules or periorbital xanthelasma-like lesions. Computed tomography revealed diffuse pulmonary fibrosis and generalized skeletal involvement, including osteolysis and osteosclerosis. The presence of CD68+ CD1a- histiocytes allowed the diagnosis of ECD. According to our review of the literature, this is the paediatric case of ECD with the youngest age of onset. The generalized skin involvement made our case unique in comparison with those previously reported.


Assuntos
Doença de Erdheim-Chester/patologia , Dermatopatias/patologia , Pré-Escolar , Humanos , Masculino , Osteólise/patologia , Osteosclerose/patologia , Tomografia Computadorizada por Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA