Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
Stem Cell Res Ther ; 15(1): 340, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39370505

RESUMEN

BACKGROUND: Ullrich congenital muscular dystrophy (UCMD) is caused by a deficiency in type 6 collagen (COL6) due to mutations in COL6A1, COL6A2, or COL6A3. COL6 deficiency alters the extracellular matrix structure and biomechanical properties, leading to mitochondrial defects and impaired muscle regeneration. Therefore, mesenchymal stromal cells (MSCs) that secrete COL6 have attracted attention as potential therapeutic targets. Various tissue-derived MSCs exert therapeutic effects in various diseases. However, no reports have compared the effects of MSCs of different origins on UCMD pathology. METHODS: To evaluate which MSC population has the highest therapeutic efficacy for UCMD, in vivo (transplantation of MSCs to Col6a1-KO/NSG mice) and in vitro experiments (muscle stem cell [MuSCs] co-culture with MSCs) were conducted using adipose tissue-derived MSCs, bone marrow-derived MSCs, and xeno-free-induced iPSC-derived MSCs (XF-iMSCs). RESULTS: In transplantation experiments on Col6a1-KO/NSG mice, the group transplanted with XF-iMSCs showed significantly enhanced muscle fiber regeneration compared to the other groups 1 week after transplantation. At 12 weeks after transplantation, only the XF-iMSCs transplantation group showed a significantly larger muscle fiber diameter than the other groups without inducing fibrosis, which was observed in the other transplantation groups. Similarly, in co-culture experiments, XF-iMSCs were found to more effectively promote the fusion and differentiation of MuSCs derived from Col6a1-KO/NSG mice than the other primary MSCs investigated in this study. Additionally, in vitro knockdown and supplementation experiments suggested that the IGF2 secreted by XF-iMSCs promoted MuSC differentiation. CONCLUSION: XF-iMSCs are promising candidates for promoting muscle regeneration while avoiding fibrosis, offering a safer and more effective therapeutic approach for UCMD than other potential therapies.


Asunto(s)
Colágeno Tipo VI , Modelos Animales de Enfermedad , Células Madre Pluripotentes Inducidas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Regeneración , Animales , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Colágeno Tipo VI/metabolismo , Colágeno Tipo VI/genética , Distrofias Musculares/terapia , Distrofias Musculares/patología , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Diferenciación Celular , Músculo Esquelético/metabolismo , Ratones Noqueados , Esclerosis
2.
J Hum Genet ; 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39014191

RESUMEN

Camurati-Engelmann disease (CED) is an autosomal dominant bone dysplasia characterized by progressive hyperostosis of the skull base and diaphyses of the long bones. CED is further divided into two subtypes, CED1 and CED2, according to the presence or absence of TGFB1 mutations, respectively. In this study, we used exome sequencing to investigate the genetic cause of CED2 in three pedigrees and identified two de novo heterozygous mutations in TGFB2 among the three patients. Both mutations were located in the region of the gene encoding the straitjacket subdomain of the latency-associated peptide (LAP) of pro-TGF-ß2. Structural simulations of the mutant LAPs suggested that the mutations could cause significant conformational changes and lead to a reduction in TGF-ß2 inactivation. An activity assay confirmed a significant increase in TGF-ß2/SMAD signaling. In vitro osteogenic differentiation experiment using iPS cells from one of the CED2 patients showed significantly enhanced ossification, suggesting that the pathogenic mechanism of CED2 is increased activation of TGF-ß2 by loss-of-function of the LAP. These results, in combination with the difference in hyperostosis patterns between CED1 and CED2, suggest distinct functions between TGFB1 and TGFB2 in human skeletal development and homeostasis.

3.
Nat Immunol ; 25(5): 764-777, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38609546

RESUMEN

The linear ubiquitin assembly complex (LUBAC) consists of HOIP, HOIL-1 and SHARPIN and is essential for proper immune responses. Individuals with HOIP and HOIL-1 deficiencies present with severe immunodeficiency, autoinflammation and glycogen storage disease. In mice, the loss of Sharpin leads to severe dermatitis due to excessive keratinocyte cell death. Here, we report two individuals with SHARPIN deficiency who manifest autoinflammatory symptoms but unexpectedly no dermatological problems. Fibroblasts and B cells from these individuals showed attenuated canonical NF-κB responses and a propensity for cell death mediated by TNF superfamily members. Both SHARPIN-deficient and HOIP-deficient individuals showed a substantial reduction of secondary lymphoid germinal center B cell development. Treatment of one SHARPIN-deficient individual with anti-TNF therapies led to complete clinical and transcriptomic resolution of autoinflammation. These findings underscore the critical function of the LUBAC as a gatekeeper for cell death-mediated immune dysregulation in humans.


Asunto(s)
Síndromes de Inmunodeficiencia , Proteínas del Tejido Nervioso , Ubiquitinas , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Femenino , Masculino , FN-kappa B/metabolismo , Ubiquitina-Proteína Ligasas/genética , Inflamación/inmunología , Inflamación/genética , Linfocitos B/inmunología , Mutación con Pérdida de Función , Fibroblastos/metabolismo , Fibroblastos/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Ratones , Alelos
4.
J Exp Med ; 221(6)2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38652464

RESUMEN

OTULIN-related autoinflammatory syndrome (ORAS), a severe autoinflammatory disease, is caused by biallelic pathogenic variants of OTULIN, a linear ubiquitin-specific deubiquitinating enzyme. Loss of OTULIN attenuates linear ubiquitination by inhibiting the linear ubiquitin chain assembly complex (LUBAC). Here, we report a patient who harbors two rare heterozygous variants of OTULIN (p.P152L and p.R306Q). We demonstrated accumulation of linear ubiquitin chains upon TNF stimulation and augmented TNF-induced cell death in mesenchymal stem cells differentiated from patient-derived iPS cells, which confirms that the patient has ORAS. However, although the de novo p.R306Q variant exhibits attenuated deubiquitination activity without reducing the amount of OTULIN, the deubiquitination activity of the p.P152L variant inherited from the mother was equivalent to that of the wild-type. Patient-derived MSCs in which the p.P152L variant was replaced with wild-type also exhibited augmented TNF-induced cell death and accumulation of linear chains. The finding that ORAS can be caused by a dominant-negative p.R306Q variant of OTULIN furthers our understanding of disease pathogenesis.


Asunto(s)
Ubiquitinación , Femenino , Humanos , Endopeptidasas/genética , Endopeptidasas/metabolismo , Enfermedades Autoinflamatorias Hereditarias/genética , Enfermedades Autoinflamatorias Hereditarias/patología , Enfermedades Autoinflamatorias Hereditarias/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mutación , Linaje , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Ubiquitina/metabolismo , Recién Nacido
5.
J Surg Oncol ; 129(7): 1364-1373, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38533914

RESUMEN

INTRODUCTION: Recycled bone autografts prepared using extracorporeal irradiation (ECIR) or liquid nitrogen freezing (LNF) methods have been used for the reconstruction of skeletal elements after wide resection of sarcomas involving bone tissues. Few reports include long-term follow-up data for histological analyses of recycled autografts, particularly in the case of ECIR autografts. MATERIALS: A total of 34 malignant bone and soft tissue tumors were resected and reconstructed using 11 ECIR- and 23 LNF-recycled autografts; the mean postoperative follow-ups were 14 and 8 years, respectively. ECIR was used for either osteosarcomas or Ewing sarcomas, whereas in addition to these tumors LNF was used for chondrosarcomas and soft tissue sarcomas involving bone tissues. Recycled bone was implanted as total bone, osteoarticular, or intercalary grafts, with or without prosthesis or vascularized fibular grafts. RESULTS: The 10-year graft survival rate was similar between groups, 81.8% using ECIR and 70.2% using LNF. There were no autograft-related tumor recurrences in either group. Graft survival was unrelated to type of graft or additional procedures. Complication rates tended to be higher using ECIR (64%) compared with LNF (52%) and the infection rate was significantly higher with ECIR (27%) versus LNF (0%). At the final assessment, plain radiographs revealed original recycled bone was present in 7 of 11 ECIR cases and in zero cases treated with LNF autografts, indicating that recycled bone treated with LNF autografts was remodeled into new bone. Histological examination of ECIR-treated bones revealed a delayed and incomplete endochondral ossification process, necrosis and empty lacunae. Conversely, LNF autografts showed remodeled bones with normal trabecular structures. CONCLUSIONS: ECIR and LNF treatment of autografts provided adequate tumor control with acceptable clinical results as a reconstruction method.


Asunto(s)
Neoplasias Óseas , Trasplante Óseo , Nitrógeno , Humanos , Neoplasias Óseas/cirugía , Neoplasias Óseas/radioterapia , Neoplasias Óseas/patología , Trasplante Óseo/métodos , Masculino , Femenino , Adulto , Adolescente , Persona de Mediana Edad , Niño , Adulto Joven , Condrosarcoma/cirugía , Condrosarcoma/radioterapia , Condrosarcoma/patología , Osteosarcoma/cirugía , Osteosarcoma/patología , Osteosarcoma/radioterapia , Supervivencia de Injerto , Estudios de Seguimiento , Sarcoma de Ewing/cirugía , Sarcoma de Ewing/radioterapia , Sarcoma de Ewing/patología , Autoinjertos , Sarcoma/cirugía , Sarcoma/radioterapia , Sarcoma/patología , Congelación , Neoplasias de los Tejidos Blandos/radioterapia , Neoplasias de los Tejidos Blandos/cirugía , Neoplasias de los Tejidos Blandos/patología
6.
Life Sci Alliance ; 7(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38365425

RESUMEN

Heterotopic ossification (HO) is a non-physiological bone formation where soft tissue progenitor cells differentiate into chondrogenic cells. In fibrodysplasia ossificans progressiva (FOP), a rare genetic disease characterized by progressive and systemic HO, the Activin A/mutated ACVR1/mTORC1 cascade induces HO in progenitors in muscle tissues. The relevant biological processes aberrantly regulated by activated mTORC1 remain unclear, however. RNA-sequencing analyses revealed the enrichment of genes involved in oxidative phosphorylation (OXPHOS) during Activin A-induced chondrogenesis of mesenchymal stem cells derived from FOP patient-specific induced pluripotent stem cells. Functional analyses showed a metabolic transition from glycolysis to OXPHOS during chondrogenesis, along with increased mitochondrial biogenesis. mTORC1 inhibition by rapamycin suppressed OXPHOS, whereas OXPHOS inhibitor IACS-010759 inhibited cartilage matrix formation in vitro, indicating that OXPHOS is principally involved in mTORC1-induced chondrogenesis. Furthermore, IACS-010759 inhibited the muscle injury-induced enrichment of fibro/adipogenic progenitor genes and HO in transgenic mice carrying the mutated human ACVR1. These data indicated that OXPHOS is a critical downstream mediator of mTORC1 signaling in chondrogenesis and therefore is a potential FOP therapeutic target.


Asunto(s)
Miositis Osificante , Osificación Heterotópica , Ratones , Animales , Humanos , Miositis Osificante/genética , Miositis Osificante/metabolismo , Fosforilación Oxidativa , Osificación Heterotópica/genética , Osificación Heterotópica/metabolismo , Transducción de Señal/genética , Ratones Transgénicos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo
7.
SLAS Technol ; 28(6): 433-441, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37562511

RESUMEN

Programmable liquid handling devices for cell culture systems have dramatically enhanced scalability and reproducibility. We previously reported a protocol to produce cell aggregates demonstrating growth plate-like structures containing hypertrophic chondrocytes from human induced pluripotent stem cells (hiPSCs). To apply this protocol to large-scale drug screening for growth plate-related diseases, we adapted it to the automated cell culture system (ACCS) consisting of programmable liquid handling devices connected to CO2 incubators, a refrigerator, and labware feeders, designed for up to 4 batches with several cell culture plates culturing for several months. We developed a new program preparing culture media with growth factors at final concentration immediately before dispensing them to each well and precisely positioning the tip for the medium change without damaging cell aggregates. Using these programs on the ACCS, we successfully cultured cell aggregates for 56 days, only needing to replenish the labware, medium, and growth factors twice a week. The size of cell aggregates in each well increased over time, with low well-to-well variability. Cell aggregates on day 56 showed histochemical, immunohistochemical, and gene expression properties of growth plate-like structures containing hypertrophic chondrocytes, indicating proper quality as materials for basic research and drug discovery of growth plate related diseases. The established program will be a suitable reference for making programs of experiments requiring long term and complex culture procedures using ACCS.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Reproducibilidad de los Resultados , Placa de Crecimiento , Técnicas de Cultivo de Célula/métodos , Células Cultivadas
9.
JBMR Plus ; 7(5): e10737, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37197316

RESUMEN

Collagen X is a non-fibril collagen produced by hypertrophic chondrocytes and was believed to associate with the calcification process of growth plate cartilage. The homozygous loss of Col10a1 gene in mice, however, demonstrated no remarkable effects on growth plate formation or skeletal development. To investigate the role of collagen X in human chondrocytes, we established human induced pluripotent stem cells (hiPSCs) with heterozygous (COL10A1 +/-) or homozygous (COL10A1 -/-) deletions of COL10A1 gene using the dual sgRNA CRISPR/Cas9 system. Several mutant clones were established and differentiated into hypertrophic chondrocytes by a previously reported 3D induction method. No remarkable differences were observed during the differentiation process between parental and mutant cell lines, which differentiated into cells with features of hypertrophic chondrocytes, indicating that collagen X is dispensable for the hypertrophic differentiation of human chondrocytes in vitro. To investigate the effects of collagen X deficiency in vivo, chondrocyte pellets at the proliferating or prehypertrophic stage were transplanted into immunodeficient mice. Proliferating pellet-derived tissues demonstrated the zonal distribution of chondrocytes with the transition to bone tissues mimicking growth plates, and the proportion of bone tended to be larger in COL10A1 -/- tissues. Prehypertrophic pellet-derived tissues produced trabecular bone structures with features of endochondral ossification, and there was no clear difference between parental- and mutant-derived tissues. A transcriptome analysis of chondrocyte pellets at the hypertrophic phase showed a lower expression of proliferating-phase genes and a higher expression of calcification-phase genes in COL10A1 -/- pellets compared with parental cell pellets. These in vitro and in vivo data suggested that collagen X is dispensable for the hypertrophic differentiation and endochondral ossification of human iPSC-derived chondrocytes, though it may facilitate the differentiation process. Thus, COL10A1 -/- iPSC lines are useful for investigating the physiological role of collagen X in chondrocyte differentiation. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

10.
Tomography ; 9(2): 768-775, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-37104133

RESUMEN

PURPOSE: To evaluate the usefulness of CT temporal subtraction (TS) images for detecting emerging or growing ectopic bone lesions in fibrodysplasia ossificans progressiva (FOP). MATERIALS AND METHODS: Four patients with FOP were retrospectively included in this study. TS images were produced by subtracting previously registered CT images from the current images. Two residents and two board-certified radiologists independently interpreted a pair of current and previous CT images for each subject with or without TS images. Changes in the visibility of the lesion, the usefulness of TS images for lesions with TS images, and the interpreter's confidence level in their interpretation of each scan were assessed on a semiquantitative 5-point scale (0-4). The Wilcoxon signed-rank test was used to compare the evaluated scores between datasets with and without TS images. RESULTS: The number of growing lesions tended to be larger than that of the emerging lesions in all cases. A higher sensitivity was found in residents and radiologists using TS compared to those not using TS. For all residents and radiologists, the dataset with TS tended to have more false-positive scans than the dataset without TS. All the interpreters recognized TS as useful, and confidence levels when using TS tended to be lower or the same as when not using TS for two residents and one radiologist. CONCLUSIONS: TS improved the sensitivity of all interpreters in detecting emerging or growing ectopic bone lesions in patients with FOP. TS could be applied further, including the areas of systematic bone disease.


Asunto(s)
Miositis Osificante , Tomografía Computarizada por Rayos X , Humanos , Tomografía Computarizada por Rayos X/métodos , Miositis Osificante/diagnóstico por imagen , Estudios Retrospectivos
11.
J Histochem Cytochem ; 71(3): 131-138, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36971322

RESUMEN

Giant cell tumors of bone (GCTBs) are locally aggressive tumors with the histological features of giant cells and stromal cells. Denosumab is a human monoclonal antibody that binds to the cytokine receptor activator of nuclear factor-kappa B ligand (RANKL). RANKL inhibition blocks tumor-induced osteoclastogenesis, and survival, and is used to treat unresectable GCTBs. Denosumab treatment induces osteogenic differentiation of GCTB cells. In this study, the expression of RANKL, special AT-rich sequence-binding protein 2 (SATB2, a marker of osteoblast differentiation), and sclerostin/SOST (a marker of mature osteocytes) was analyzed before and after treatment with denosumab in six cases of GCTB. Denosumab therapy was administered a mean of five times over a mean 93.5-day period. Before denosumab treatment, RANKL expression was observed in one of six cases. After denosumab therapy, spindle-like cells devoid of giant cell aggregation were RANKL-positive in four of six cases. Bone matrix-embedded osteocyte markers were observed, although RANKL was not expressed. Osteocyte-like cells were confirmed to have mutations, as identified using mutation-specific antibodies. Our study results suggest that treatment of GCTBs with denosumab results in osteoblast-osteocyte differentiation. Denosumab played a role in the suppression of tumor activity via inhibition of the RANK-RANKL pathway, which triggers osteoclast precursors to differentiate into osteoclasts.


Asunto(s)
Conservadores de la Densidad Ósea , Neoplasias Óseas , Tumor Óseo de Células Gigantes , Humanos , Denosumab/farmacología , Osteocitos/metabolismo , Tumor Óseo de Células Gigantes/genética , Tumor Óseo de Células Gigantes/metabolismo , Tumor Óseo de Células Gigantes/patología , Osteogénesis , FN-kappa B , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Conservadores de la Densidad Ósea/farmacología , Ligando RANK/metabolismo , Diferenciación Celular
12.
Sci Rep ; 13(1): 1094, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36658197

RESUMEN

Although the formation of bone-like nodules is regarded as the differentiation process from stem cells to osteogenic cells, including osteoblasts and osteocytes, the precise biological events during nodule formation are unknown. Here we performed the osteogenic induction of human induced pluripotent stem cells using a three-dimensional (3D) culture system using type I collagen gel and a rapid induction method with retinoic acid. Confocal and time-lapse imaging revealed the osteogenic differentiation was initiated with vigorous focal proliferation followed by aggregation, from which cells invaded the gel. Invading cells changed their morphology and expressed osteocyte marker genes, suggesting the transition from osteoblasts to osteocytes. Single-cell RNA sequencing analysis revealed that 3D culture-induced cells with features of periosteal skeletal stem cells, some of which expressed TGFß-regulated osteoblast-related molecules. The role of TGFß signal was further analyzed in the transition from osteoblasts to osteocytes, which revealed that modulation of the TGFß signal changed the morphology and motility of cells isolated from the 3D culture, suggesting that the TGFß signal maintains the osteoblastic phenotype and the transition into osteocytes requires down-regulation of the TGFß signal.


Asunto(s)
Células Madre Pluripotentes Inducidas , Osteocitos , Humanos , Factor de Crecimiento Transformador beta , Osteogénesis/genética , Osteoblastos , Diferenciación Celular/genética
13.
Cancer Sci ; 114(5): 1898-1911, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36661413

RESUMEN

Mesenchymal stem cell- or osteoblast-derived osteosarcoma is the most common malignant bone tumor. Its highly metastatic malignant phenotypes, which are often associated with a poor prognosis, have been correlated with the modulation of TP53- and cell-cycle-related pathways. MYC, which regulates the transcription of cell-cycle modulating genes, is used as a representative prognostic marker for osteosarcoma. Another member of the MYC oncoprotein family, MYCN, is highly expressed in a subset of osteosarcoma, however its roles in osteosarcoma have not been fully elucidated. Here, we attempted to create an in vitro tumorigenesis model using hiPSC-derived neural crest cells, which are precursors of mesenchymal stem cells, by overexpressing MYCN on a heterozygous TP53 hotspot mutation (c.733G>A; p.G245S) background. MYCN-expressing TP53 mutated transformed clones were isolated by soft agar colony formation, and administered subcutaneously into the periadrenal adipose tissue of immunodeficient mice, resulting in the development of chondroblastic osteosarcoma. MYCN suppression decreased the proliferation of MYCN-induced osteosarcoma cells, suggesting MYCN as a potential target for a subset of osteosarcoma treatment. Further, comprehensive analysis of gene expression and exome sequencing of MYCN-induced clones indicated osteosarcoma-specific molecular features, such as the activation of TGF-ß signaling and DNA copy number amplification of GLI1. The model of MYCN-expressing chondroblastic osteosarcoma was developed from hiPSC-derived neural crest cells, providing a useful tool for the development of new tumor models using hiPSC-derived progenitor cells with gene modifications and in vitro transformation.


Asunto(s)
Neuroblastoma , Osteosarcoma , Animales , Ratones , Regulación Neoplásica de la Expresión Génica , Proteína Proto-Oncogénica N-Myc/genética , Cresta Neural/metabolismo , Cresta Neural/patología , Neuroblastoma/patología , Proteínas Oncogénicas/genética , Osteosarcoma/patología
14.
J Pediatr Hematol Oncol ; 45(3): e356-e362, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35973000

RESUMEN

BACKGROUND: Patients with osteosarcoma who experience relapse or progression [R/P] have a poor prognosis. METHODS: Data from 30 patients who experienced R/P among 59 with a diagnosis of high-grade osteosarcoma, who were younger than 40 years old between 2000 and 2019, were retrospectively analyzed to identify prognostic and therapeutic factors influencing their outcomes. RESULTS: The 5-year overall survival [OS] rates after the last R/P of patients experiencing first [n=30], second [n=14], and third [n=9] R/P were 50.3%, 51.3%, and 46.7%, respectively. Multivariate analysis did not identify any independent risk factors affecting OS. The 5-year PFS rate of the 30 patients after first R/P was 22.4%, and multivariate analysis identified histologic subtype and curative local surgery as independent risk factors influencing PFS. Long [>6 mo] partial response was observed in three patients treated using temozolomide+etoposide, irinotecan+carboplatin, or regorafenib. CONCLUSIONS: OS rate in the patients with osteosarcoma experiencing R/P included in this study was markedly higher than that reported previously, mainly due to the surgical total removal of tumors, even after subsequent R/P. The recent establishment of salvage chemotherapy or molecular targeted therapy may also increase survival rates in a subgroup of patients.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , Adulto , Estudios Retrospectivos , Recurrencia Local de Neoplasia/patología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Pronóstico , Neoplasias Óseas/patología
15.
Orphanet J Rare Dis ; 17(1): 364, 2022 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-36131296

RESUMEN

BACKGROUND: Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease characterized by progressive heterotopic ossification (HO) in soft tissues due to a heterozygous mutation of the ACVR1A gene (FOP-ACVR1A), which erroneously transduces the BMP signal by Activin-A. Although inflammation is known to trigger HO in FOP, the role of FOP-ACVR1A on inflammatory cells remains to be elucidated. RESULTS: We generated immortalized monocytic cell lines from FOP-iPSCs (FOP-ML) and mutation rescued iPSCs (resFOP-ML). Cell morphology was evaluated during the monocyte induction and after immortalization. Fluorescence-activated cell sorting (FACS) was performed to evaluate the cell surface markers CD14 and CD16 on MLs. MLs were stimulated with lipopolysaccharide or Activin-A and the gene expression was evaluated by quantitative PCR and microarray analysis. Histological analysis was performed for HO tissue obtained from wild type mice and FOP-ACVR1A mice which conditionally express human mutant ACVR1A gene by doxycycline administration. Without any stimulation, FOP-ML showed the pro-inflammatory signature of CD16+ monocytes with an upregulation of INHBA gene, and treatment of resFOP-ML with Activin-A induced an expression profile mimicking that of FOP-ML at baseline. Treatment of FOP-ML with Activin-A further induced the inflammatory profile with an up-regulation of inflammation-associated genes, of which some, but not all, of which were suppressed by corticosteroid. Experiments using an inhibitor for TGFß or BMP signal demonstrated that Activin-A-induced genes such as CD16 and CCL7, were regulated by both signals, indicating Activin-A transduced dual signals in FOP-ML. A comparison with resFOP-ML identified several down-regulated genes in FOP-ML including LYVE-1, which is known to suppress matrix-formation in vivo. The down-regulation of LYVE-1 in HO tissues was confirmed in FOP model mice, verifying the significance of the in vitro experiments. CONCLUSION: These results indicate that FOP-ML faithfully recapitulated the phenotype of primary monocytes of FOP and the combination with resFOP-ML is a useful tool to investigate molecular events at the initial inflammation stage of HO in FOP.


Asunto(s)
Receptores de Activinas Tipo I/genética , Miositis Osificante , Osificación Heterotópica , Receptores de Activinas Tipo I/metabolismo , Activinas/genética , Activinas/metabolismo , Animales , Doxiciclina , Humanos , Inflamación/genética , Lipopolisacáridos , Ratones , Monocitos/metabolismo , Monocitos/patología , Mutación/genética , Miositis Osificante/patología , Osificación Heterotópica/genética , Osificación Heterotópica/patología , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
16.
Br J Cancer ; 127(8): 1487-1496, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35871234

RESUMEN

BACKGROUND: This randomised phase II/III trial aimed to determine whether perioperative chemotherapy with gemcitabine plus docetaxel (GD) is non-inferior to the standard Adriamycin plus ifosfamide (AI) in terms of overall survival (OS) in patients with soft tissue sarcoma (STS). METHODS: Patients with localised high-risk STS in the extremities or trunk were randomised to receive AI or GD. The treatments were repeated for three preoperative and two postoperative courses. The primary endpoint was OS. RESULTS: Among 143 enrolled patients who received AI (70 patients) compared to GD (73 patients), the estimated 3-year OS was 91.4% for AI and 79.2% for GD (hazard ratio 2.55, 95% confidence interval: 0.80-8.14, P = 0.78), exceeding the prespecified non-inferiority margin in the second interim analysis. The estimated 3-year progression-free survival was 79.1% for AI and 59.1% for GD. The most common Grade 3-4 adverse events in the preoperative period were neutropenia (88.4%), anaemia (49.3%), and febrile neutropenia (36.2%) for AI and neutropenia (79.5%) and febrile neutropenia (17.8%) for GD. CONCLUSIONS: Although GD had relatively mild toxicity, the regimen-as administered in this study-should not be considered a standard treatment of perioperative chemotherapy for high-risk STS in the extremities and trunk. CLINICAL TRIAL REGISTRATION: jRCTs031180003.


Asunto(s)
Neutropenia Febril , Sarcoma , Neoplasias de los Tejidos Blandos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Desoxicitidina/análogos & derivados , Docetaxel/uso terapéutico , Doxorrubicina , Humanos , Ifosfamida/efectos adversos , Sarcoma/tratamiento farmacológico , Sarcoma/cirugía , Gemcitabina
17.
Arch Rehabil Res Clin Transl ; 4(1): 100179, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35282152

RESUMEN

Objective: To assess the status of 10 patients with advanced osteonecrosis of the femoral head who underwent mesenchymal stromal cell transplants and a 12-week rehabilitation program 10 years earlier. Design: Retrospective study. Setting: University clinical research laboratory. Participants: Patients (N=10) who had undergone mesenchymal stromal cell transplantation and rehabilitation for a single hip osteonecrosis of the femoral head 10 years prior to the current study were recruited by telephone. The average age was 31.7 years and all participants were men; radiographic stages were 3A in 6 patients and 3B in 4 patients before treatment. Intervention: A 12-week rehabilitation program with follow-up once every 1 to 2 years was performed after mesenchymal stromal cell transplantation. Main Outcome Measures: Radiographic analysis, clinical score, timed Up and Go test, hip function (range of motion, muscle strength), and Short Form-36 scores were assessed before treatment and 1 and 10 years after treatment. Results: Upon imaging, 5 hips were found to be stable (stable group) and 5 had progressed (progressed group); 2 of the latter group required a total hip arthroplasty. The pretreatment radiographic stage of the progressed group was more advanced than that of the stable group. Body mass index was higher in the progressed group than in the stable group. Hip function and clinical score at 1 and 10 years after treatment improved in the hips of 8 patients without total hip arthroplasty. There were no severe adverse events during the rehabilitation. Conclusions: The 12-week rehabilitation program and annual follow-up after mesenchymal stromal cell transplantation for osteonecrosis of the femoral head was associated with pain reduction, maintaining hip muscle strength, widening range of motion, and improving quality of life. The level and timing of weight-bearing and social activity should be planned according to the individual's lifestyle and body composition.

18.
Oncogene ; 41(5): 683-691, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34803166

RESUMEN

Osteosarcoma (OS) in human patients is characterized by genetic alteration of TP53. Osteoprogenitor-specific p53-deleted mice (OS mice) have been widely used to study the process of osteosarcomagenesis. However, the molecular mechanisms responsible for the development of OS upon p53 inactivation remain largely unknown. In this study, we detected prominent RUNX3/Runx3 expression in human and mouse p53-deficient OS. Myc was aberrantly upregulated by Runx3 via mR1, a consensus Runx site in the Myc promoter, in a manner dependent on p53 deficiency. Reduction of the Myc level by disruption of mR1 or Runx3 knockdown decreased the tumorigenicity of p53-deficient OS cells and effectively suppressed OS development in OS mice. Furthermore, Runx inhibitors exerted therapeutic effects on OS mice. Together, these results show that p53 deficiency promotes osteosarcomagenesis in human and mouse by allowing Runx3 to induce oncogenic Myc expression.


Asunto(s)
Proteína p53 Supresora de Tumor
19.
Biofabrication ; 13(4)2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34380122

RESUMEN

Osteoarthritis is a leading cause of pain and joint immobility, the incidence of which is increasing worldwide. Currently, total joint replacement is the only treatment for end-stage disease. Scaffold-based tissue engineering is a promising alternative approach for joint repair but is subject to limitations such as poor cytocompatibility and degradation-associated toxicity. To overcome these limitations, a completely scaffold-free Kenzan method for bio-3D printing was used to fabricate cartilage constructs feasible for repairing large chondral defects. Human induced pluripotent stem cell (iPSC)-derived neural crest cells with high potential to undergo chondrogenesis through mesenchymal stem cell differentiation were used to fabricate the cartilage. Unified, self-sufficient, and functional cartilaginous constructs up to 6 cm2in size were assembled by optimizing fabrication time during chondrogenic induction. Maturation for 3 weeks facilitated the self-organisation of the cells, which improved the construct's mechanical strength (compressive and tensile properties) and induced changes in glycosaminoglycan and type II collagen expression, resulting in improved tissue function. The compressive modulus of the construct reached the native cartilage range of 0.88 MPa in the 5th week of maturation. This paper reports the fabrication of anatomically sized and shaped cartilage constructs, achieved by combining novel iPSCs and bio-3D printers using a Kenzan needle array technology, which may facilitate chondral resurfacing of articular cartilage defects.


Asunto(s)
Cartílago Articular , Células Madre Pluripotentes Inducidas , Diferenciación Celular , Condrocitos , Condrogénesis , Humanos , Impresión Tridimensional , Regeneración , Ingeniería de Tejidos , Andamios del Tejido
20.
J Orthop ; 26: 67-71, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34349396

RESUMEN

BACKGROUND: A prospective, open-label clinical trial, in which transplantation of cultured autologous bone marrow-derived multipotent mesenchymal stromal cells in combination with vascularised bone grafts for the treatment of post-collapse extensive osteonecrosis of the femoral head in ten patients, was conducted previously. The aim of this study was to assess the 10-year clinical and radiographic results of that study. METHODS: Patients were evaluated for radiographic progression of osteonecrosis of the femoral head using anteroposterior radiographs at 10 years postoperatively. Clinical score and hip function, including the timed up and go test, were also estimated. RESULTS: Osteoarthritic changes in the affected hip were found in five of the ten patients, two of whom had undergone total hip arthroplasty at 7 and 9 years postoperatively. Five of the six cases (83.3%) in which pre-operative femoral head collapse was less than 3 mm, had no further collapse. On the other hand, all four cases in which pre-operative femoral head collapse was ≥3 mm, showed osteoarthritic changes within 10 years. The average clinical score significantly improved postoperatively and was maintained at 10 years. CONCLUSIONS: Considering that eight of 10 post-collapse cases could avoid total hip arthroplasty conversion with good clinical results for 10 years and five of 6 post-collapse cases (collapse <3 mm) could avoid further collapse and osteoarthritic changes for 10 years, mesenchymal stromal cell transplantation in combination with vascularised bone grafts could be an effective treatment for post-collapse osteonecrosis of the femoral head.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA