Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Bone Miner Res ; 38(12): 1834-1845, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37737377

RESUMO

Patients with classical melorheostosis exhibit exuberant bone overgrowth in the appendicular skeleton, resulting in pain and deformity with no known treatment. Most patients have somatic, mosaic mutations in MAP2K1 (encoding the MEK1 protein) in osteoblasts and overlying skin. As with most rare bone diseases, lack of affected tissue has limited the opportunity to understand how the mutation results in excess bone formation. The aim of this study was to create a cellular model to study melorheostosis. We obtained patient skin cells bearing the MAP2K1 mutation (affected cells), and along with isogenic control normal fibroblasts reprogrammed them using the Sendai virus method into induced pluripotent stem cells (iPSCs). Pluripotency was validated by marker staining and embryoid body formation. iPSCs were then differentiated to mesenchymal stem cells (iMSCs) and validated by flow cytometry. We confirmed retention of the MAP2K1 mutation in iMSCs with polymerase chain reaction (PCR) and confirmed elevated MEK1 activity by immunofluorescence staining. Mutation-bearing iMSCs showed significantly elevated vascular endothelial growth factor (VEGF) secretion, proliferation and collagen I and IV secretion. iMSCs were then differentiated into osteoblasts, which showed increased mineralization at 21 days and increased VEGF secretion at 14 and 21 days of differentiation. Administration of VEGF to unaffected iMSCs during osteogenic differentiation was sufficient to increase mineralization. Blockade of VEGF by bevacizumab reduced mineralization in iMSC-derived affected osteoblasts and in affected primary patient-derived osteoblasts. These data indicate that patient-derived induced pluripotent stem cells recreate the elevated MEK1 activity, increased mineralization, and increased proliferation seen in melorheostosis patients. The increased bone formation is driven, in part, by abundant VEGF secretion. Modifying the activity of VEGF (a known stimulator of osteoblastogenesis) represents a promising treatment pathway to explore. iPSCs may have wide applications to other rare bone diseases. © 2023 American Society for Bone and Mineral Research (ASBMR).


Assuntos
Melorreostose , Osteogênese , Humanos , Osso e Ossos/metabolismo , Diferenciação Celular , MAP Quinase Quinase 1/genética , Melorreostose/genética , Osteogênese/genética , Fator A de Crescimento do Endotélio Vascular
2.
Mol Genet Genomic Med ; 10(10): e2043, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36004822

RESUMO

BACKGROUND: Melorheostosis (MEL) is an exceptionally rare sclerosing bone dysplasia with asymmetrically exuberant bone formation and soft tissue lesions in a segmental distribution. We aimed to summarize the clinical characteristics of Chinese MEL patients and identify their pathogenic cause. METHODS: In total, 10 Chinese MEL patients were recruited, and clinical manifestations and radiological characteristics were recorded. Sanger sequencing of the LEMD3 gene was performed on peripheral blood samples of all patients, while the exome sequencing of matched peripheral blood, melorheostotic bone, and skin lesion samples was conducted on one patient who provided affected bone and skin tissues. Micro-computed tomography (micro-CT) was also used to scan the melorheostotic bone tissue. RESULTS: We found the average age of the 10 MEL patients was 29.5 years (range 11-40 years), and the major symptoms were bone pain, restricted movement, and bone deformity. The lesions sites were mainly located in femur (8/10), tibia (8/10), fibula (6/10), and foot (7/10), the next was pelvis (4/10), and the last were patella (1/10), hand (1/10) and spine (1/10). Radiological examinations showed a mixture of hyperostosis consisting of classic "dripping candle wax," "osteoma-like," or "myositis ossificans-like" patterns in most patients. No germline pathogenic variants in the LEMD3 gene were found in all patients, but a disease-causing somatic variant of MAP2K1 (c.167A > C, p.Gln56Pro) was detected in melorheostotic bone from one patient. Moreover, the micro-CT analysis showed increased porosity in the melorheostotic bone with somatic MAP2K1 variant. CONCLUSION: This is a summary of the clinical characteristics of Chinese MEL patients and we first identify the somatic MAP2K1 variant in Chinese patients. Our findings validate the molecular genetic mechanism of MEL and broaden its phenotype spectrum in the Chinese population.


Assuntos
Melorreostose , Osso e Ossos/patologia , China , Humanos , MAP Quinase Quinase 1/genética , Melorreostose/diagnóstico por imagem , Melorreostose/genética , Melorreostose/patologia , Sequenciamento do Exoma , Microtomografia por Raio-X
3.
J Musculoskelet Neuronal Interact ; 22(2): 292-295, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35642708

RESUMO

Buschke-Ollendorff syndrome (BOS) is a rare, usually benign, autosomal dominant genetic disease affecting about 0.005% globally. BOS commonly manifests with asymptomatic connective tissue nevi, sometimes with sclerotic bone lesions like osteopoikilosis or melorheostosis. However, BOS may develop severe, symptomatic complications that require surgical intervention. Here we report a 9-year-8-month girl presenting with multiple nonpruritic, nonpainful skin plaques scattered around the trunk, buttocks, and bilateral legs. She had a history of right varus foot with inadequate plantar flexion. Upon visiting, obvious leg length discrepancy (LLD) was noted. Lesional biopsy revealed increased fibroblasts within dermal collagen bundles. Verhoeff-van Gieson stain revealed scattered foci of thickened elastic fibers between collagen fibers, especially in the mid-dermis. Radiographic examination of the lower extremities showed multiple small, round-to-oval shaped, radiopaque spots on the pelvic bones, femurs, tibiae, and both feet. Hyperostosis along the long axis with "dripping candle wax" appearance was characteristic of osteopoikilosis and melorheostosis. Genetic analysis showed heterozygous point mutation in exon 1 of LEMD3 gene (c.1323C>A, p.Y441X), confirming diagnosis of BOS. Sequential and epiphyseodesis were performed to correct LLD with a favorable outcome at 2-year follow-up. BOS associated with severe bone abnormalities is rare, but orthopedic surgical intervention can provide satisfactory outcome.


Assuntos
Melorreostose , Osteopecilose , Criança , Colágeno , Feminino , Humanos , Perna (Membro) , Melorreostose/diagnóstico , Melorreostose/genética , Osteopecilose/diagnóstico , Osteopecilose/genética , Osteopecilose/patologia , Dermatopatias Genéticas
4.
J Invest Dermatol ; 142(9): 2406-2414.e5, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35189151

RESUMO

Melorheostosis is a rare sclerosing bone disease with associated vascular abnormalities in skin and bone, which is caused by somatic mosaic single nucleotide variations in the MAP2K1 gene, which encodes MAPK/extracellular signal‒regulated kinase (ERK) kinase 1. However, disease pathogenesis is poorly understood. Using patient-derived cells, we found that affected skin fibroblasts carrying the single nucleotide variations have increased activation of ERK1/2, which results in increased expression and secretion of proangiogenic factors, including VEGF. VEGF secretion was strongly reduced in affected cells after treatment with MAPK/ERK kinase 1 inhibitor trametinib. Treatment of healthy endothelial cells on matrigel with conditioned medium from affected fibroblasts induces the adoption of a proangiogenic phenotype. Direct coculture of fibroblasts and endothelial cells further shows that both secreted factors and extracellular matrix are capable of inducing a proangiogenic phenotype in healthy endothelial cells. Blocking VEGF with bevacizumab reduces the proangiogenic effect of affected fibroblasts in both the matrigel and direct coculture angiogenesis models, indicating that elevated VEGF secretion is a key mediator of increased angiogenesis in melorheostosis tissue. In conclusion, this work identifies the role of several important molecular mediators in the pathogenesis of melorheostosis, including MAPK/ERK kinase 1, phosphorylated ERK1/2, and VEGF, all of which have clinically available pharmacologic inhibitors, which could be further explored as therapeutic targets.


Assuntos
Melorreostose , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Humanos , Melorreostose/genética , Neovascularização Patológica/patologia , Nucleotídeos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Bone ; 137: 115406, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32387835

RESUMO

Melorheostosis is a very rare sclerosing bone dysplasia characterized by asymmetrical and progressive cortical hyperostosis, usually with involvement of soft tissues surrounding the lesions. Recently Kang et al. identified somatic mosaicism for variants (p.Gln56Pro, p.Lys57Asn, or p.Lys57Glu) in the negative regulatory domain of MAP2K1, resulting in increased ERK1/2 signalling in affected tissues. In our study, we employed several sequencing technologies to unravel genetic variants (only present in affected tissues) from four sporadic melorheostosis patients. In the exome of two patients, we identified the same variants (p.K57N and p.K57E) as previously described by Kang et al. WGS and RNAseq analysis in a third patient demonstrated the presence of a novel variant (p.Cys121Ser) in the catalytic domain of MAP2K1. In addition, gene set enrichment analysis of the transcriptome data demonstrated upregulation of proliferative pathways. Interestingly, increased proliferation of MAP2K1 p.Lys57Asn-positive osteoblasts has been reported by Kang et al. The variants located in the hotspot region of the negative regulatory domain as well as this newly identified p.Cys121Ser variant have all been classified as MAP2K1 variants that can constitutively activate the downstream effector Erk. Finally, in a fourth patient with classical radiographic features of melorheostosis, no pathogenic variants could be identified in MAP2K1 or the other candidate genes for melorheostosis (SMAD3; LEMD3; KRAS). In conclusion, our study strongly suggests that not only somatic variants in the regulatory domain of MAP2K1 but also in the catalytic domain can cause melorheostosis. Our observations confirm that mutations in MAP2K1 are a major cause of melorheostosis and also suggest further locus heterogeneity for this disorder.


Assuntos
Melorreostose , Humanos , MAP Quinase Quinase 1 , Melorreostose/diagnóstico por imagem , Melorreostose/genética , Mutação/genética , Osteoblastos , Sequenciamento do Exoma
8.
J Exp Med ; 217(5)2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32232430

RESUMO

Melorheostosis is a rare sclerosing dysostosis characterized by asymmetric exuberant bone formation. Recently, we reported that somatic mosaicism for MAP2K1-activating mutations causes radiographical "dripping candle wax" melorheostosis. We now report somatic SMAD3 mutations in bone lesions of four unrelated patients with endosteal pattern melorheostosis. In vitro, the SMAD3 mutations stimulated the TGF-ß pathway in osteoblasts, enhanced nuclear translocation and target gene expression, and inhibited proliferation. Osteoblast differentiation and mineralization were stimulated by the SMAD3 mutation, consistent with higher mineralization in affected than in unaffected bone, but differing from MAP2K1 mutation-positive melorheostosis. Conversely, osteoblast differentiation and mineralization were inhibited when osteogenesis of affected osteoblasts was driven in the presence of BMP2. Transcriptome profiling displayed that TGF-ß pathway activation and ossification-related processes were significantly influenced by the SMAD3 mutation. Co-expression clustering illuminated melorheostosis pathophysiology, including alterations in ECM organization, cell growth, and interferon signaling. These data reveal antagonism of TGF-ß/SMAD3 activation by BMP signaling in SMAD3 mutation-positive endosteal melorheostosis, which may guide future therapies.


Assuntos
Melorreostose/genética , Mutação/genética , Transdução de Sinais , Proteína Smad3/genética , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima/genética , Animais , Proteína Morfogenética Óssea 2/metabolismo , Osso e Ossos/patologia , Calcificação Fisiológica , Diferenciação Celular , Linhagem Celular , Núcleo Celular/metabolismo , Proliferação de Células , Matriz Extracelular/metabolismo , Mutação com Ganho de Função , Regulação da Expressão Gênica , Humanos , MAP Quinase Quinase 1/genética , Melorreostose/diagnóstico por imagem , Melorreostose/patologia , Camundongos , Modelos Biológicos , Osteoblastos/metabolismo , Osteogênese , Transporte Proteico , Transcriptoma/genética
9.
J Exp Med ; 217(5)2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32289153

RESUMO

In the current issue of JEM, Kang et al. (https://doi.org/10.1084/jem.20191499) describe somatic mutations in the SMAD3 gene causing endosteal melorheostosis. Using osteoblast models, the identified mutations are demonstrated to exert a gain-of-function mechanism, augmenting transforming growth factor (TGF) ß signaling. These findings provide further insights into the genetic etiology of melorheostosis and consolidate the importance of the TGFß pathway in skeletal disorders.


Assuntos
Melorreostose , Fator de Crescimento Transformador beta , Humanos , Melorreostose/genética , Mutação/genética , Osteoblastos/metabolismo , Transdução de Sinais/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
10.
Calcif Tissue Int ; 105(2): 215-221, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31129707

RESUMO

Melorheostosis (MEL) is an uncommon, sclerosing disease, characterised by hyperostosis of long bones, resembling the flowing of candle wax. The disease is sporadic and the pathogenesis is still poorly understood. Occasionally, the same family can include individuals with MEL and Osteopoikilosis (OPK), a disease characterised by multiple round foci of increased bone density. LEMD3 gene mutations are related to OPK and Buschke-Ollendorff Syndrome, a genetic condition in which an association between MEL, OPK and skin lesions is observed. In rare cases, LEMD3 mutations and recently mosaic MAP2K1 gene mutations have been correlated to MEL suggesting that somatic mosaicism could be causative of the disease. In this study, we described the clinical, radiological and molecular findings of 19 individuals with MEL and 8 with OPK and compared the results to the medical literature. The molecular analyses of this case series corroborate the available data in the medical literature, indicating that LEMD3 germline mutations are not a major cause of isolated MEL and reporting five further cases of OPK caused by LEMD3 germline mutations.


Assuntos
Melorreostose/diagnóstico por imagem , Melorreostose/genética , Osteopecilose/diagnóstico por imagem , Osteopecilose/genética , Adolescente , Adulto , Criança , Proteínas de Ligação a DNA/genética , Feminino , Fêmur/patologia , Mutação em Linhagem Germinativa , Humanos , Itália/epidemiologia , MAP Quinase Quinase 1/genética , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Mutação Puntual , Adulto Jovem
11.
Calcif Tissue Int ; 104(5): 530-543, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30989250

RESUMO

Melorheostosis is an exceptionally rare sclerosing hyperostosis that typically affects the appendicular skeleton in a limited segmental fashion. It occasionally occurs on a background of another benign generalised sclerosing bone condition, known as osteopoikilosis caused by germline mutations in LEMD3, encoding the inner nuclear membrane protein MAN1, which modulates TGFß/bone morphogenetic protein signalling. Recent studies of melorheostosis lesional tissue indicate that most cases arise from somatic MAP2K1 mutations although a small number may arise from other genes in related pathways, such as KRAS. Those cases associated with MAP2K1 mutations are more likely to have the classic "dripping candle wax" appearance on radiographs. The relationship between these somatic mutations and those found in a variety of malignant conditions is discussed. There are also similar germline mutations involved in a group of genetic disorders known as the RASopathies (including Noonan syndrome, Costello syndrome and various cardiofaciocutaneous syndromes), successful treatments for which could be applied to melorheostosis. The diagnosis and management of melorheostosis are discussed; there are 4 distinct radiographic patterns of melorheostosis and substantial overlap with mixed sclerosing bone dysplasia. Medical treatments include bisphosphonates, but definitive guidance on their use is lacking given the small number of patients that have been studied. Surgical intervention may be required for those with large bone growths, nerve entrapments, joint impingement syndromes or major limb deformities. Bone regrowth is uncommon after surgery, but recurrent contractures represent a major issue in those with extensive associated soft tissue involvement.


Assuntos
Melorreostose/diagnóstico por imagem , Osteopecilose/diagnóstico por imagem , Proteínas Morfogenéticas Ósseas/metabolismo , Osso e Ossos/patologia , Proteínas de Ligação a DNA/genética , Diagnóstico Diferencial , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Humanos , MAP Quinase Quinase 1/genética , Melorreostose/genética , Proteínas de Membrana/genética , Osteopecilose/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
12.
J Bone Miner Res ; 34(5): 883-895, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30667555

RESUMO

Melorheostosis is a rare non-hereditary condition characterized by dense hyperostotic lesions with radiographic "dripping candle wax" appearance. Somatic activating mutations in MAP2K1 have recently been identified as a cause of melorheostosis. However, little is known about the development, composition, structure, and mechanical properties of the bone lesions. We performed a multi-method phenotype characterization of material properties in affected and unaffected bone biopsy samples from six melorheostosis patients with MAP2K1 mutations. On standard histology, lesions show a zone with intensively remodeled osteonal-like structure and prominent osteoid accumulation, covered by a shell formed through bone apposition, consisting of compact multi-layered lamellae oriented parallel to the periosteal surface and devoid of osteoid. Compared with unaffected bone, melorheostotic bone has lower average mineralization density measured by quantitative backscattered electron imaging (CaMean: -4.5%, p = 0.04). The lamellar portion of the lesion is even less mineralized, possibly because the newly deposited material has younger tissue age. Affected bone has higher porosity by micro-CT, due to increased tissue vascularity and elevated 2D-microporosity (osteocyte lacunar porosity: +39%, p = 0.01) determined on quantitative backscattered electron images. Furthermore, nano-indentation modulus characterizing material hardness and stiffness was strictly dependent on tissue mineralization (correlation with typical calcium concentration, CaPeak: r = 0.8984, p = 0.0150, and r = 0.9788, p = 0.0007, respectively) in both affected and unaffected bone, indicating that the surgical hardness of melorheostotic lesions results from their lamellar structure. The results suggest a model for pathophysiology of melorheostosis caused by somatic activating mutations in MAP2K1, in which the genetically induced gradual deterioration of bone microarchitecture triggers a periosteal reaction, similar to the process found to occur after bone infection or local trauma, and leads to an overall cortical outgrowth. The micromechanical properties of the lesions reflect their structural heterogeneity and correlate with local variations in mineral content, tissue age, and remodeling rates, in the same way as normal bone. © 2018 American Society for Bone and Mineral Research.


Assuntos
Densidade Óssea , MAP Quinase Quinase 1 , Melorreostose , Modelos Biológicos , Mutação , Periósteo , Microtomografia por Raio-X , Adulto , Feminino , Humanos , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , Masculino , Melorreostose/diagnóstico por imagem , Melorreostose/genética , Melorreostose/metabolismo , Melorreostose/fisiopatologia , Pessoa de Meia-Idade , Periósteo/diagnóstico por imagem , Periósteo/metabolismo , Periósteo/fisiopatologia
13.
J Bone Miner Res ; 34(1): 145-156, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30138550

RESUMO

Melorheostosis is a rare hyperostotic disease of the long bones classically characterized by a "dripping candle-wax" radiographic appearance. We recently described somatic activating mutations in MAP2K1 as a cause of melorheostosis. Here, we report distinguishing characteristics of patients with MAP2K1-positive melorheostosis. Fifteen unrelated patients with radiographic appearance of melorheostosis underwent paired biopsies of affected and unaffected bone for whole-exome sequencing, histology, and cell culture. Eight patients with mutations in MAP2K1 in affected bone were compared to the seven MAP2K1-negative patients to identify distinguishing characteristics. Patients with MAP2K1-positive melorheostosis had a distinct phenotype with classic "dripping candle-wax" appearance on radiographs (p = 0.01), characteristic vascular lesions on skin overlying affected bone (p = 0.01), and higher prevalence of extraosseous mineralization and joint involvement (p = 0.04 for both). Melorheostotic bone from both MAP2K1-positive and MAP2K1-negative patients showed two zones of distinct morphology-an outer segment of parallel layers of primary lamellar bone and a deeper zone of intensely remodeled highly porous osteonal-like bone. Affected bone from MAP2K1-positive patients showed excessive osteoid (p = 0.0012), increased number of osteoblasts (p = 0.012) and osteoclasts (p = 0.04), and increased vascularity on histology in comparison to paired unaffected bone which was not seen in affected bone in most MAP2K1-negative patients. The identification of a distinct phenotype of patients with MAP2K1-positive melorheostosis demonstrates clinical and genetic heterogeneity among patients with the disease. Further studies are needed to better understand the underlying pathophysiology and associated skin findings. © 2018 American Society for Bone and Mineral Research.


Assuntos
Osso e Ossos , Melorreostose , Mutação , Osteoblastos , Pele , Adulto , Idoso , Osso e Ossos/enzimologia , Osso e Ossos/patologia , Feminino , Humanos , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , Masculino , Melorreostose/enzimologia , Melorreostose/genética , Melorreostose/patologia , Pessoa de Meia-Idade , Osteoblastos/enzimologia , Osteoblastos/patologia , Pele/enzimologia , Pele/patologia , Sequenciamento do Exoma
14.
Nat Commun ; 9(1): 1390, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29643386

RESUMO

Melorheostosis is a sporadic disease of uncertain etiology characterized by asymmetric bone overgrowth and functional impairment. Using whole exome sequencing, we identify somatic mosaic MAP2K1 mutations in affected, but not unaffected, bone of eight unrelated patients with melorheostosis. The activating mutations (Q56P, K57E and K57N) cluster tightly in the MEK1 negative regulatory domain. Affected bone displays a mosaic pattern of increased p-ERK1/2 in osteoblast immunohistochemistry. Osteoblasts cultured from affected bone comprise two populations with distinct p-ERK1/2 levels by flow cytometry, enhanced ERK1/2 activation, and increased cell proliferation. However, these MAP2K1 mutations inhibit BMP2-mediated osteoblast mineralization and differentiation in vitro, underlying the markedly increased osteoid detected in affected bone histology. Mosaicism is also detected in the skin overlying bone lesions in four of five patients tested. Our data show that the MAP2K1 oncogene is important in human bone formation and implicate MEK1 inhibition as a potential treatment avenue for melorheostosis.


Assuntos
Osso e Ossos/metabolismo , MAP Quinase Quinase 1/genética , Melorreostose/genética , Mutação , Osteoblastos/metabolismo , Osteogênese/genética , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Osso e Ossos/patologia , Calcificação Fisiológica , Diferenciação Celular , Proliferação de Células , Regulação da Expressão Gênica , Humanos , MAP Quinase Quinase 1/metabolismo , Melorreostose/metabolismo , Melorreostose/patologia , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mosaicismo , Osteoblastos/patologia , Cultura Primária de Células , Transdução de Sinais , Pele/metabolismo , Pele/patologia , Sequenciamento do Exoma
15.
Curr Osteoporos Rep ; 16(3): 256-268, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29656376

RESUMO

PURPOSE OF REVIEW: The group of sclerosing bone disorders encompasses a variety of disorders all marked by increased bone mass. In this review, we give an overview of the genetic causes of this heterogeneous group of disorders and briefly touch upon the value of these findings for the development of novel therapeutic agents. RECENT FINDINGS: Advances in the next-generation sequencing technologies are accelerating the molecular dissection of the pathogenic mechanisms underlying skeletal dysplasias. Throughout the years, the genetic cause of these disorders has been extensively studied which resulted in the identification of a variety of disease-causing genes and pathways that are involved in bone formation by osteoblasts, bone resorption by osteoclasts, or both processes. Due to this rapidly increasing knowledge, the insights into the regulatory mechanisms of bone metabolism are continuously improving resulting in the identification of novel therapeutic targets for disorders with reduced bone mass and increased bone fragility.


Assuntos
Hiperostose/genética , Osteíte Deformante/genética , Osteosclerose/genética , Picnodisostose/genética , Anormalidades Múltiplas/genética , Doenças do Desenvolvimento Ósseo/genética , Remodelação Óssea/genética , Reabsorção Óssea/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Deficiência Intelectual/genética , Melorreostose/genética , Osteoblastos , Osteoclastos , Osteogênese/genética , Osteopetrose/genética , Osteopecilose/genética
16.
Curr Osteoporos Rep ; 15(4): 335-342, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28676968

RESUMO

PURPOSE OF REVIEW: Melorheostosis is a rare sclerosing bone dysplasia that affects both cortical bone and adjacent soft tissue structures in a sclerotomal distribution. In this review, we describe the natural history, radiological features, proposed pathogenesis, and management options for this debilitating condition. RECENT FINDINGS: Since its first description in 1922, about 400 cases of melorheostosis have been reported, either as single reports or in small case series. Melorheostosis affects the appendicular skeleton more commonly than the axial skeleton and usually presents with lower limb deformity. Diagnosis is based on a combination of clinical and radiological features that help differentiate this condition from other sclerosing bone dysplasias. LEM domain-containing protein 3 (LEMD3) gene mutations have been demonstrated in several familial cases, but these have been more strongly correlated with other hereditary dysplasias, such as osteopoikilosis, and are not thought to be the causative gene for melorheostosis. The exact etiology of classic sporadically occurring melorheostosis remains unknown, with possible causes being somatic LEMD3 mutations, somatic mutations in the bone morphogenetic protein/transforming growth factor-beta pathway, mutations in multiple genes, or other non-genetic causes. Management in recent years has involved nitrogen-containing bisphosphonates in addition to traditional orthopedic surgical approaches and physical therapy. Melorheostosis may present as mixed or atypical osseous involvement in addition to the classically described "dripping candle wax" appearance of hyperostosis. Some patients may have overlap with osteopoikilosis or Buschke-Ollendorff syndrome. In the future, better characterization of genetic and developmental factors predisposing to melorheostosis may lead to the development of targeted therapy for this condition, as well as for more commonly encountered skeletal abnormalities.


Assuntos
Osso e Ossos/diagnóstico por imagem , Melorreostose/diagnóstico por imagem , Osso e Ossos/patologia , Proteínas de Ligação a DNA , Humanos , Liberação da Cápsula Articular , Melorreostose/genética , Melorreostose/patologia , Melorreostose/terapia , Proteínas de Membrana/genética , Mutação , Proteínas Nucleares/genética , Osteotomia , Manejo da Dor , Radiografia , Doenças Raras/diagnóstico por imagem , Doenças Raras/genética , Doenças Raras/patologia , Doenças Raras/terapia , Tenotomia
17.
Bone ; 101: 145-155, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28434888

RESUMO

Melorheostosis (MEL) is the rare sporadic dysostosis characterized by monostotic or polyostotic osteosclerosis and hyperostosis often distributed in a sclerotomal pattern. The prevailing hypothesis for MEL invokes postzygotic mosaicism. Sometimes scleroderma-like skin changes, considered a representation of the pathogenetic process of MEL, overlie the bony changes, and sometimes MEL becomes malignant. Osteopoikilosis (OPK) is the autosomal dominant skeletal dysplasia that features symmetrically distributed punctate osteosclerosis due to heterozygous loss-of-function mutation within LEMD3. Rarely, radiographic findings of MEL occur in OPK. However, germline mutation of LEMD3 does not explain sporadic MEL. To explore if mosaicism underlies MEL, we studied a boy with polyostotic MEL and characteristic overlying scleroderma-like skin, a few bony lesions consistent with OPK, and a large epidermal nevus known to usually harbor a HRAS, FGFR3, or PIK3CA gene mutation. Exome sequencing was performed to ~100× average read depth for his two dermatoses, two areas of normal skin, and peripheral blood leukocytes. As expected for non-malignant tissues, the patient's mutation burden in his normal skin and leukocytes was low. He, his mother, and his maternal grandfather carried a heterozygous, germline, in-frame, 24-base-pair deletion in LEMD3. Radiographs of the patient and his mother revealed bony foci consistent with OPK, but she showed no MEL. For the patient, somatic variant analysis, using four algorithms to compare all 20 possible pairwise combinations of his five DNA samples, identified only one high-confidence mutation, heterozygous KRAS Q61H (NM_033360.3:c.183A>C, NP_203524.1:p.Gln61His), in both his dermatoses but absent in his normal skin and blood. Thus, sparing our patient biopsy of his MEL bone, we identified a heterozygous somatic KRAS mutation in his scleroderma-like dermatosis considered a surrogate for MEL. This implicates postzygotic mosaicism of mutated KRAS, perhaps facilitated by germline LEMD3 haploinsufficiency, causing his MEL.


Assuntos
Exoma/genética , Melorreostose/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Adolescente , Predisposição Genética para Doença/genética , Humanos , Masculino , Mosaicismo , Mutação , Nevo/genética , Osteopecilose/genética , Osteosclerose/genética
18.
Pediatr Dermatol ; 32(5): e219-20, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26135202

RESUMO

Buschke-Ollendorff syndrome is a rare autosomal dominant disorder caused by loss of function in LEMD3, resulting in connective tissue nevi and varying bone dysplasia. Although typically benign, we describe a novel LEMD3 splice site mutation (IVS12 + 1delG) in a 13-year-old boy with Buschke-Ollendorff syndrome presenting with severe skeletal deformities, polyostotic melorheostosis, and osteopoikilosis.


Assuntos
Predisposição Genética para Doença , Melorreostose/genética , Proteínas de Membrana/genética , Mutação , Proteínas Nucleares/genética , Osteopecilose/genética , Dermatopatias Genéticas/genética , Anormalidades Múltiplas/diagnóstico , Adolescente , Proteínas de Ligação a DNA , Diagnóstico Diferencial , Humanos , Masculino , Melorreostose/diagnóstico , Osteopecilose/diagnóstico , Prognóstico , Sítios de Splice de RNA/genética , Doenças Raras , Dermatopatias Genéticas/diagnóstico
19.
Eur J Dermatol ; 20(6): 693-7, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20732851

RESUMO

Buschke-Ollendorff syndrome refers to the concomitant occurrence of connective tissue nevi, composed of elastic fibers in most cases, with osteopoikilosis. This autosomal dominant inherited disorder is caused by mutations in the gene LEMD3 on chromosome 12q14, which induces a rather heterogeneous clinical phenotype. Here, we report on the most proximal germline mutation found to date in the LEMD3 gene, p.Val94fs, in a three-generation Swiss family. Quantitative RNA analyses in affected and non-affected skin tissue from the proband demonstrate a comparable nonsense-mediated decay of mutant LEMD3 mRNA in both tissues; however, different levels of tropoelastin expression suggest that additional factors are involved in the development of the cutaneous lesions.


Assuntos
Doenças do Tecido Conjuntivo/genética , Mutação da Fase de Leitura , Melorreostose/genética , Proteínas de Membrana/genética , Proteínas Nucleares/genética , Osteopecilose/genética , Tropoelastina/genética , Criança , Proteínas de Ligação a DNA , Humanos , Masculino , Linhagem , Fenótipo , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Suíça , Síndrome , Tropoelastina/metabolismo
20.
Arch Dermatol ; 146(1): 63-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20083694

RESUMO

BACKGROUND: Buschke-Ollendorff syndrome (BOS), an autosomal dominant disorder, features small, acquired, asymptomatic, symmetrical foci of osteosclerosis detected radiographically in epimetaphyseal bone (osteopoikilosis) (OPK) together with connective tissue nevi or juvenile elastomas. Heterozygous, loss-of-function, germline mutation in the LEMD3 gene (which encodes an inner nuclear membrane protein called LEMD3, or MAN1) has been repeatedly documented in patients with BOS or OPK. OBSERVATIONS: We describe a father and son with multiple yellowish papules and nodules coalescing into cobblestone nevoid plaques consistent with nevus elasticus. Radiographs of the father show multiple, small, bone islands within the hands, wrists, distal femurs, proximal tibias, and left distal fibula consistent with OPK. Although the clinical findings are diagnostic of Buschke-Ollendorf syndrome, analysis of the LEMD3 gene showed no exonic mutations. CONCLUSION: Absence of LEMD3 mutation in the exons and splice sites of a family with BOS suggests that there is genetic heterogeneity for this disorder.


Assuntos
DNA/genética , Melorreostose/genética , Proteínas de Membrana/genética , Mutação , Nevo/genética , Proteínas Nucleares/genética , Osteopecilose/genética , Pele/patologia , Autoantígenos , Biópsia , Criança , Proteínas de Ligação a DNA , Éxons , Família , Predisposição Genética para Doença , Humanos , Masculino , Melorreostose/sangue , Melorreostose/diagnóstico , Proteínas de Membrana/sangue , Nevo/sangue , Nevo/diagnóstico , Proteínas Nucleares/sangue , Osteopecilose/sangue , Osteopecilose/diagnóstico , Reação em Cadeia da Polimerase , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA