Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(6): 112578, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37267108

RESUMEN

Chondrosarcomas are the most common malignancy of cartilage and are associated with somatic mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 genes. Somatic IDH mutations are also found in its benign precursor lesion, enchondromas, suggesting that IDH mutations are early events in malignant transformation. Human mutant IDH chondrosarcomas and mutant Idh mice that develop enchondromas investigated in our studies display glycogen deposition exclusively in mutant cells from IDH mutant chondrosarcomas and Idh1 mutant murine growth plates. Pharmacologic blockade of glycogen utilization induces changes in tumor cell behavior, downstream energetic pathways, and tumor burden in vitro and in vivo. Mutant IDH1 interacts with hypoxia-inducible factor 1α (HIF1α) to regulate expression of key enzymes in glycogen metabolism. Here, we show a critical role for glycogen in enchondromas and chondrosarcomas, which is likely mediated through an interaction with mutant IDH1 and HIF1α.


Asunto(s)
Condroma , Condrosarcoma , Isocitrato Deshidrogenasa , Animales , Humanos , Ratones , Neoplasias Óseas/metabolismo , Cartílago/metabolismo , Condrosarcoma/genética , Condrosarcoma/metabolismo , Condrosarcoma/patología , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Mutación/genética
2.
J Bone Miner Res ; 37(5): 983-996, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35220602

RESUMEN

Enchondromas and chondrosarcomas are common cartilage neoplasms that are either benign or malignant, respectively. The majority of these tumors harbor mutations in either IDH1 or IDH2. Glutamine metabolism has been implicated as a critical regulator of tumors with IDH mutations. Using genetic and pharmacological approaches, we demonstrated that glutaminase-mediated glutamine metabolism played distinct roles in enchondromas and chondrosarcomas with IDH1 or IDH2 mutations. Glutamine affected cell differentiation and viability in these tumors differently through different downstream metabolites. During murine enchondroma-like lesion development, glutamine-derived α-ketoglutarate promoted hypertrophic chondrocyte differentiation and regulated chondrocyte proliferation. Deletion of glutaminase in chondrocytes with Idh1 mutation increased the number and size of enchondroma-like lesions. In contrast, pharmacological inhibition of glutaminase in chondrosarcoma xenografts reduced overall tumor burden partially because glutamine-derived non-essential amino acids played an important role in preventing cell apoptosis. This study demonstrates that glutamine metabolism plays different roles in tumor initiation and cancer maintenance. Supplementation of α-ketoglutarate and inhibiting GLS may provide a therapeutic approach to suppress enchondroma and chondrosarcoma tumor growth, respectively. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Neoplasias Óseas , Condroma , Condrosarcoma , Glutamina , Isocitrato Deshidrogenasa , Mutación , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Cartílago/metabolismo , Condroma/genética , Condroma/metabolismo , Condroma/patología , Condrosarcoma/genética , Condrosarcoma/metabolismo , Condrosarcoma/patología , Glutaminasa/genética , Glutaminasa/metabolismo , Glutamina/genética , Glutamina/metabolismo , Humanos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Ácidos Cetoglutáricos , Ratones
3.
JCI Insight ; 6(22)2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34618689

RESUMEN

Sarcomas contain a subpopulation of tumor-propagating cells (TPCs) with enhanced tumor-initiating and self-renewal properties. However, it is unclear whether the TPC phenotype in sarcomas is stable or a dynamic cell state that can derive from non-TPCs. In this study, we utilized a mouse model of undifferentiated pleomorphic sarcoma (UPS) to trace the lineage relationship between sarcoma side population (SP) cells that are enriched for TPCs and non-SP cells. By cotransplanting SP and non-SP cells expressing different endogenous fluorescent reporters, we show that non-SP cells can give rise to SP cells with enhanced tumor-propagating potential in vivo. Lineage trajectory analysis using single-cell RNA sequencing from SP and non-SP cells supports the notion that non-SP cells can assume the SP cell phenotype de novo. To test the effect of eradicating SP cells on tumor growth and self-renewal, we generated mouse sarcomas in which the diphtheria toxin receptor is expressed in the SP cells and their progeny. Ablation of the SP population using diphtheria toxin did not impede tumor growth or self-renewal. Altogether, we show that the sarcoma SP represent a dynamic cell state and targeting TPCs alone is insufficient to eliminate tumor progression.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Sarcoma/inmunología , Células de Población Lateral/metabolismo , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos NOD , Sarcoma/patología
4.
Nat Cell Biol ; 22(1): 49-59, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31907410

RESUMEN

Osteoclasts are multinucleated cells of the monocyte/macrophage lineage that degrade bone. Here, we used lineage tracing studies-labelling cells expressing Cx3cr1, Csf1r or Flt3-to identify the precursors of osteoclasts in mice. We identified an erythromyeloid progenitor (EMP)-derived osteoclast precursor population. Yolk-sac macrophages of EMP origin produced neonatal osteoclasts that can create a space for postnatal bone marrow haematopoiesis. Furthermore, EMPs gave rise to long-lasting osteoclast precursors that contributed to postnatal bone remodelling in both physiological and pathological settings. Our single-cell RNA-sequencing data showed that EMP-derived osteoclast precursors arose independently of the haematopoietic stem cell (HSC) lineage and the data from fate tracking of EMP and HSC lineages indicated the possibility of cell-cell fusion between these two lineages. Cx3cr1+ yolk-sac macrophage descendants resided in the adult spleen, and parabiosis experiments showed that these cells migrated through the bloodstream to the remodelled bone after injury.


Asunto(s)
Hematopoyesis/fisiología , Homeostasis/fisiología , Osteoclastos/metabolismo , Saco Vitelino/metabolismo , Animales , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Células Madre Hematopoyéticas/metabolismo , Macrófagos/metabolismo , Ratones
5.
JCI Insight ; 4(18)2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31534056

RESUMEN

Age is a well-established risk factor for impaired bone fracture healing. Here, we identify a role for apolipoprotein E (ApoE) in age-associated impairment of bone fracture healing and osteoblast differentiation, and we investigate the mechanism by which ApoE alters these processes. We identified that, in both humans and mice, circulating ApoE levels increase with age. We assessed bone healing in WT and ApoE-/- mice after performing tibial fracture surgery: bone deposition was higher within fracture calluses from ApoE-/- mice. In vitro recombinant ApoE (rApoE) treatment of differentiating osteoblasts decreased cellular differentiation and matrix mineralization. Moreover, this rApoE treatment decreased osteoblast glycolytic activity while increasing lipid uptake and fatty acid oxidation. Using parabiosis models, we determined that circulating ApoE plays a strong inhibitory role in bone repair. Using an adeno-associated virus-based siRNA system, we decreased circulating ApoE levels in 24-month-old mice and demonstrated that, as a result, fracture calluses from these aged mice displayed enhanced bone deposition and mechanical strength. Our results demonstrate that circulating ApoE as an aging factor inhibits bone fracture healing by altering osteoblast metabolism, thereby identifying ApoE as a new therapeutic target for improving bone repair in the elderly.


Asunto(s)
Envejecimiento/sangre , Apolipoproteínas E/sangre , Apolipoproteínas E/genética , Curación de Fractura/fisiología , Osteoblastos/fisiología , Fracturas de la Tibia/fisiopatología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/fisiología , Animales , Apolipoproteínas E/antagonistas & inhibidores , Callo Óseo/diagnóstico por imagen , Callo Óseo/efectos de los fármacos , Callo Óseo/fisiopatología , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Estudios de Cohortes , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Curación de Fractura/efectos de los fármacos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Ratones , Ratones Noqueados para ApoE , Persona de Mediana Edad , Osteoblastos/efectos de los fármacos , Cultivo Primario de Células , ARN Interferente Pequeño/genética , Proteínas Recombinantes/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Fracturas de la Tibia/diagnóstico por imagen , Fracturas de la Tibia/cirugía , Microtomografía por Rayos X
6.
Nat Commun ; 9(1): 5191, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518764

RESUMEN

The pace of repair declines with age and, while exposure to a young circulation can rejuvenate fracture repair, the cell types and factors responsible for rejuvenation are unknown. Here we report that young macrophage cells produce factors that promote osteoblast differentiation of old bone marrow stromal cells. Heterochronic parabiosis exploiting young mice in which macrophages can be depleted and fractionated bone marrow transplantation experiments show that young macrophages rejuvenate fracture repair, and old macrophage cells slow healing in young mice. Proteomic analysis of the secretomes identify differential proteins secreted between old and young macrophages, such as low-density lipoprotein receptor-related protein 1 (Lrp1). Lrp1 is produced by young cells, and depleting Lrp1 abrogates the ability to rejuvenate fracture repair, while treating old mice with recombinant Lrp1 improves fracture healing. Macrophages and proteins they secrete orchestrate the fracture repair process, and young cells produce proteins that rejuvenate fracture repair in mice.


Asunto(s)
Curación de Fractura , Fracturas Óseas/fisiopatología , Macrófagos/metabolismo , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Femenino , Fracturas Óseas/genética , Fracturas Óseas/metabolismo , Fracturas Óseas/terapia , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis , Receptores de LDL/genética , Rejuvenecimiento , Células del Estroma/citología , Células del Estroma/metabolismo , Células del Estroma/trasplante , Proteínas Supresoras de Tumor/genética
7.
Development ; 145(13)2018 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-29899135

RESUMEN

During enchondral ossification, mesenchymal cells express genes regulating the intracellular biosynthesis of cholesterol and lipids. Here, we have investigated conditional deletion of Scap or of Insig1 and Insig2 (Scap inhibits intracellular biosynthesis and Insig proteins activate intracellular biosynthesis). Mesenchymal condensation and chondrogenesis was disrupted in mice lacking Scap in mesenchymal progenitors, whereas mice lacking the Insig genes in mesenchymal progenitors had short limbs, but normal chondrogenesis. Mice lacking Scap in chondrocytes showed severe dwarfism, with ectopic hypertrophic cells, whereas deletion of Insig genes in chondrocytes caused a mild dwarfism and shortening of the hypertrophic zone. In vitro studies showed that intracellular cholesterol in chondrocytes can derive from exogenous and endogenous sources, but that exogenous sources cannot completely overcome the phenotypic effect of Scap deficiency. Genes encoding cholesterol biosynthetic proteins are regulated by Hedgehog (Hh) signaling, and Hh signaling is also regulated by intracellular cholesterol in chondrocytes, suggesting a feedback loop in chondrocyte differentiation. Precise regulation of intracellular biosynthesis is required for chondrocyte homeostasis and long bone growth, and these data support pharmacological modulation of cholesterol biosynthesis as a therapy for select cartilage pathologies.


Asunto(s)
Desarrollo Óseo/fisiología , Colesterol/biosíntesis , Condrocitos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Diferenciación Celular/fisiología , Colesterol/genética , Condrocitos/citología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Transducción de Señal/fisiología
8.
J Biol Chem ; 293(7): 2466-2475, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29196603

RESUMEN

Sarcomas, and the mesenchymal precursor cells from which they arise, express chondroitin sulfate proteoglycan 4 (NG2/CSPG4). However, NG2/CSPG4's function and its capacity to serve as a therapeutic target in this tumor type are unknown. Here, we used cells from human tumors and a genetically engineered autochthonous mouse model of soft-tissue sarcomas (STSs) to determine NG2/CSPG4's role in STS initiation and growth. Inhibiting NG2/CSPG4 expression in established murine and human STSs decreased tumor volume by almost two-thirds and cell proliferation rate by 50%. NG2/CSPG4 antibody immunotherapy in human sarcomas established as xenografts in mice similarly decreased tumor volume, and expression of a lentivirus blocking NG2/CSPG4 expression inhibited tumor cell proliferation and increased the latency of engraftment. Gene profiling showed that Ng2/Cspg4 deletion altered the expression of genes regulating cell proliferation and apoptosis. Surprisingly, Ng2/Cspg4 deletion at the time of tumor initiation resulted in larger tumors. Gene expression profiling indicated substantial down-regulation of insulin-like growth factor binding protein (Igfbp) genes when Ng2/Cspg4 is depleted at tumor initiation, but not when Ng2/Cspg4 is depleted after tumor initiation. Such differences may have clinical significance, as therapeutic targeting of a signaling pathway such as NG2/CSPG4 may have different effects on cell behavior with tumor progression. NG2/CSPG4 depletion has divergent effects, depending on the developmental stage of sarcoma. In established tumors, IGF signaling is active, and NG2 inhibition targets cell proliferation and apoptosis.


Asunto(s)
Antígenos/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteínas de la Membrana/metabolismo , Proteoglicanos/metabolismo , Sarcoma/metabolismo , Sarcoma/fisiopatología , Animales , Antígenos/genética , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Proteoglicanos Tipo Condroitín Sulfato/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Estadificación de Neoplasias , Proteoglicanos/genética , Sarcoma/genética , Sarcoma/patología
9.
Bone ; 98: 31-36, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28254468

RESUMEN

Patients with Neurofibromatosis type 1 display delayed fracture healing and the increased deposition of fibrous tissue at the fracture site. Severe cases can lead to non-union and even congenital pseudarthrosis. Neurofibromatosis type 1 is caused by a mutation in the NF1 gene and mice lacking the Nf1 gene show a fracture repair phenotype similar to that seen in patients. Tissue from the fracture site of patients with Neurofibromatosis type 1 and from mice deficient in the Nf1 gene both show elevated levels of ß-catenin protein and activation of ß-catenin mediated signaling. Constitutively elevated ß-catenin leads to a delayed and fibrous fracture repair process, and (RS)-5-methyl-1-phenyl-1,3,4,6-tetrahydro-2,5-benzoxazocine (Nefopam, a centrally-acting, non-narcotic analgesic agent) inhibits ß-catenin mediated signaling during skin wound repair. Here we investigate Nefopam's potential as a modulator of bone repair in mice deficient in Nf1. Mice were treated with Nefopam and investigated for bone fracture repair. Bone marrow stromal cells flushed from the long bones of unfractured mice were treated with Nefopam and investigated for osteogenic potential. Treatment with Nefopam was able to lower the ß-catenin level and the Axin2 transcript level in the fracture calluses of Nf1 deficient mice. Cultures from the bone marrow of Nf1-/- mice had significantly lower osteoblastic colonies and mineralized nodules, which was increased when cells were cultured in the presence of Nefopam. Fracture calluses were harvested and analyzed 14days and 21days after injury. Nf1-/- calluses had less bone, less cartilage, and higher fibrous tissue content than control calluses. Treatment with Nefopam increased the bone and cartilage content and decreased the fibrous tissue content in Nf1-/- calluses. These findings present a potential treatment for patients with Neurofibromatosis 1 in the context of bone repair. Since Nefopam is already in use in patient care, it could be rapidly translated to the clinical setting.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Curación de Fractura/efectos de los fármacos , beta Catenina/metabolismo , Animales , Western Blotting , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Curación de Fractura/fisiología , Fracturas Óseas/metabolismo , Genes de Neurofibromatosis 1 , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurofibromina 1/deficiencia , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
11.
Nat Commun ; 6: 7131, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25988592

RESUMEN

The capacity for tissues to repair and regenerate diminishes with age. We sought to determine the age-dependent contribution of native mesenchymal cells and circulating factors on in vivo bone repair. Here we show that exposure to youthful circulation by heterochronic parabiosis reverses the aged fracture repair phenotype and the diminished osteoblastic differentiation capacity of old animals. This rejuvenation effect is recapitulated by engraftment of young haematopoietic cells into old animals. During rejuvenation, ß-catenin signalling, a pathway important in osteoblast differentiation, is modulated in the early repair process and required for rejuvenation of the aged phenotype. Temporal reduction of ß-catenin signalling during early fracture repair improves bone healing in old mice. Our data indicate that young haematopoietic cells have the capacity to rejuvenate bone repair and this is mediated at least in part through ß-catenin, raising the possibility that agents that modulate ß-catenin can improve the pace or quality of fracture repair in the ageing population.


Asunto(s)
Curación de Fractura , Regeneración , beta Catenina/metabolismo , Alelos , Animales , Sitios de Unión , Médula Ósea/patología , Huesos/patología , Diferenciación Celular , Medios de Cultivo Condicionados , Femenino , Fluoresceínas/química , Fracturas Óseas/patología , Células Madre Hematopoyéticas/citología , Células Madre Mesenquimatosas/citología , Ratones , Ratones Transgénicos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis , Parabiosis , Fenotipo , Transducción de Señal , Tibia/patología , Factores de Tiempo , Proteínas Wnt/metabolismo
12.
Proc Natl Acad Sci U S A ; 112(9): 2829-34, 2015 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-25730874

RESUMEN

Enchondromas are benign cartilage tumors and precursors to malignant chondrosarcomas. Somatic mutations in the isocitrate dehydrogenase genes (IDH1 and IDH2) are present in the majority of these tumor types. How these mutations cause enchondromas is unclear. Here, we identified the spectrum of IDH mutations in human enchondromas and chondrosarcomas and studied their effects in mice. A broad range of mutations was identified, including the previously unreported IDH1-R132Q mutation. These mutations harbored enzymatic activity to catalyze α-ketoglutarate to d-2-hydroxyglutarate (d-2HG). Mice expressing Idh1-R132Q in one allele in cells expressing type 2 collagen showed a disordered growth plate, with persistence of type X-expressing chondrocytes. Chondrocyte cell cultures from these animals or controls showed that there was an increase in proliferation and expression of genes characteristic of hypertrophic chondrocytes with expression of Idh1-R132Q or 2HG treatment. Col2a1-Cre;Idh1-R132Q mutant knock-in mice (mutant allele expressed in chondrocytes) did not survive after the neonatal stage. Col2a1-Cre/ERT2;Idh1-R132 mutant conditional knock-in mice, in which Cre was induced by tamoxifen after weaning, developed multiple enchondroma-like lesions. Taken together, these data show that mutant IDH or d-2HG causes persistence of chondrocytes, giving rise to rests of growth-plate cells that persist in the bone as enchondromas.


Asunto(s)
Condrocitos , Encondromatosis , Regulación Enzimológica de la Expresión Génica , Isocitrato Deshidrogenasa , Mutación Missense , Sustitución de Aminoácidos , Animales , Condrocitos/enzimología , Condrocitos/patología , Colágeno Tipo II/biosíntesis , Colágeno Tipo II/genética , Encondromatosis/enzimología , Encondromatosis/genética , Encondromatosis/patología , Glutaratos/efectos adversos , Glutaratos/farmacología , Humanos , Isocitrato Deshidrogenasa/biosíntesis , Isocitrato Deshidrogenasa/genética , Ratones , Ratones Mutantes
13.
J Clin Invest ; 124(6): 2599-610, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24837430

RESUMEN

A ß-catenin/T cell factor-dependent transcriptional program is critical during cutaneous wound repair for the regulation of scar size; however, the relative contribution of ß-catenin activity and function in specific cell types in the granulation tissue during the healing process is unknown. Here, cell lineage tracing revealed that cells in which ß-catenin is transcriptionally active express a gene profile that is characteristic of the myeloid lineage. Mice harboring a macrophage-specific deletion of the gene encoding ß-catenin exhibited insufficient skin wound healing due to macrophage-specific defects in migration, adhesion to fibroblasts, and ability to produce TGF-ß1. In irradiated mice, only macrophages expressing ß-catenin were able to rescue wound-healing deficiency. Evaluation of scar tissue collected from patients with hypertrophic and normal scars revealed a correlation between the number of macrophages within the wound, ß-catenin levels, and cellularity. Our data indicate that ß-catenin regulates myeloid cell motility and adhesion and that ß-catenin-mediated macrophage motility contributes to the number of mesenchymal cells and ultimate scar size following cutaneous injury.


Asunto(s)
Células Mieloides/fisiología , Cicatrización de Heridas/fisiología , beta Catenina/fisiología , Animales , Adhesión Celular/genética , Adhesión Celular/fisiología , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Cicatriz/metabolismo , Cicatriz/patología , Cicatriz Hipertrófica/metabolismo , Cicatriz Hipertrófica/patología , Humanos , Macrófagos/citología , Macrófagos/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/citología , Piel/lesiones , Piel/patología , Piel/fisiopatología , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/fisiología , Transcriptoma , Factor de Crecimiento Transformador beta1/biosíntesis , Cicatrización de Heridas/genética , beta Catenina/deficiencia , beta Catenina/genética
14.
Mol Cancer Ther ; 13(5): 1259-69, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24634412

RESUMEN

Hedgehog (Hh) pathway inhibition in cancer has been evaluated in both the ligand-independent and ligand-dependent settings, where Hh signaling occurs either directly within the cancer cells or within the nonmalignant cells of the tumor microenvironment. Chondrosarcoma is a malignant tumor of cartilage in which there is ligand-dependent activation of Hh signaling. IPI-926 is a potent, orally delivered small molecule that inhibits Hh pathway signaling by binding to Smoothened (SMO). Here, the impact of Hh pathway inhibition on primary chondrosarcoma xenografts was assessed. Mice bearing primary human chondrosarcoma xenografts were treated with IPI-926. The expression levels of known Hh pathway genes, in both the tumor and stroma, and endpoint tumor volumes were measured. Gene expression profiling of tumors from IPI-926-treated mice was conducted to identify potential novel Hh target genes. Hh target genes were studied to determine their contribution to the chondrosarcoma neoplastic phenotype. IPI-926 administration results in downmodulation of the Hh pathway in primary chondrosarcoma xenografts, as demonstrated by evaluation of the Hh target genes GLI1 and PTCH1, as well as inhibition of tumor growth. Chondrosarcomas exhibited autocrine and paracrine Hh signaling, and both were affected by IPI-926. Decreased tumor growth is accompanied by histopathologic changes, including calcification and loss of tumor cells. Gene profiling studies identified genes differentially expressed in chondrosarcomas following IPI-926 treatment, one of which, ADAMTSL1, regulates chondrosarcoma cell proliferation. These studies provide further insight into the role of the Hh pathway in chondrosarcoma and provide a scientific rationale for targeting the Hh pathway in chondrosarcoma.


Asunto(s)
Condrosarcoma/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Alcaloides de Veratrum/farmacología , Proteínas ADAMTS , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Calcinosis/tratamiento farmacológico , Calcinosis/genética , Calcinosis/metabolismo , Línea Celular Tumoral , Proliferación Celular , Condrosarcoma/genética , Condrosarcoma/patología , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Humanos , Ratones , Receptor Smoothened , Carga Tumoral/efectos de los fármacos , Alcaloides de Veratrum/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Orthop Res ; 32(4): 581-6, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24347536

RESUMEN

Fracture repair is a well orchestrated process involving various cell types and signaling molecules. The hedgehog signaling pathway is activated in chondrocytes during fracture repair and is known to regulate chondrogenesis however, its role in osteoblasts during injury is yet unknown. In this study we observed tibial fracture repair in mice in which hedgehog signaling was modulated through genetic alterations of the pathway activator, smoothened. Levels of the hedgehog target genes Gli1 and Ptch1 in wildtype mice were upregulated in fracture calluses throughout the repair process. Forced activation of the hedgehog pathway in ubiquitous fashion resulted in increased matrix deposition in the fracture callus. Interestingly, inhibition in chondrocytes did not alter the fracture repair phenotype, while activation of hedgehog in osteoblasts was a requirement for normal fracture repair. In vitro, transcript levels of Gli1 and Ptch1 were elevated during osteoblastogenesis. Activation of hedgehog signaling positively affected osteoblastic differentiation and mineralization as detected using alkaline phosphatase and Von Kossa staining and Alp and Col1 expression. Here we show that the hedgehog signaling pathway plays a critical role in osteoblasts during fracture repair: inhibition of the pathway in osteoblasts leads to decreased matrix at the fracture site while activation increased matrix deposition.


Asunto(s)
Condrocitos/metabolismo , Curación de Fractura , Proteínas Hedgehog/fisiología , Osteoblastos/metabolismo , Animales , Células Cultivadas , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Transgénicos , Transducción de Señal/fisiología
16.
PLoS One ; 5(9)2010 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-20927391

RESUMEN

Lasers have in principle the capability to cut at the level of a single cell, the fundamental limit to minimally invasive procedures and restructuring biological tissues. To date, this limit has not been achieved due to collateral damage on the macroscale that arises from thermal and shock wave induced collateral damage of surrounding tissue. Here, we report on a novel concept using a specifically designed Picosecond IR Laser (PIRL) that selectively energizes water molecules in the tissue to drive ablation or cutting process faster than thermal exchange of energy and shock wave propagation, without plasma formation or ionizing radiation effects. The targeted laser process imparts the least amount of energy in the remaining tissue without any of the deleterious photochemical or photothermal effects that accompanies other laser wavelengths and pulse parameters. Full thickness incisional and excisional wounds were generated in CD1 mice using the Picosecond IR Laser, a conventional surgical laser (DELight Er:YAG) or mechanical surgical tools. Transmission and scanning electron microscopy showed that the PIRL laser produced minimal tissue ablation with less damage of surrounding tissues than wounds formed using the other modalities. The width of scars formed by wounds made by the PIRL laser were half that of the scars produced using either a conventional surgical laser or a scalpel. Aniline blue staining showed higher levels of collagen in the early stage of the wounds produced using the PIRL laser, suggesting that these wounds mature faster. There were more viable cells extracted from skin using the PIRL laser, suggesting less cellular damage. ß-catenin and TGF-ß signalling, which are activated during the proliferative phase of wound healing, and whose level of activation correlates with the size of wounds was lower in wounds generated by the PIRL system. Wounds created with the PIRL systsem also showed a lower rate of cell proliferation. Direct comparison of wound healing responses to a conventional surgical laser, and standard mechanical instruments shows far less damage and near absence of scar formation by using PIRL laser. This new laser source appears to have achieved the long held promise of lasers in minimally invasive surgery.


Asunto(s)
Procedimientos Quirúrgicos Dermatologicos , Terapia por Láser/instrumentación , Procedimientos Quirúrgicos Mínimamente Invasivos/instrumentación , Transducción de Señal , Animales , Terapia por Láser/métodos , Ratones , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Piel/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas , beta Catenina/metabolismo
17.
Cancer Res ; 70(19): 7690-8, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20841474

RESUMEN

The cellular origins from which most tumors arise are poorly defined, especially in mesenchymal neoplasms. Aggressive fibromatosis, also known as desmoid tumor, is a locally invasive soft tissue tumor that has mesenchymal characteristics. We found that aggressive fibromatosis tumors express genes and cell surface markers characteristic of mesenchymal stem cells (MSC). In mice that are genetically predisposed to develop aggressive fibromatosis tumors (Apc(wt/1638N)), we found that the number of tumors formed was proportional to the number of MSCs present. Sca-1(-/-) mice, which develop fewer MSCs, were crossed with Apc(wt/1638N) mice. Doubly mutant mice deficient in Sca-1 developed substantially fewer aggressive fibromatosis tumors than wild-type (WT) littermates, but Sca-1 deficiency had no effect on the formation of epithelial-derived intestinal polyps. MSCs isolated from Apc(wt/1638N) mice (or mice expressing a stabilized form of ß-catenin) induced aberrant cellular growth reminiscent of aggressive fibromatosis tumors after engraftment to immunocompromised mice, but WT cells and mature fibroblasts from the same animals did not. Taken together, our findings indicate that aggressive fibromatosis is derived from MSCs, and that ß-catenin supports tumorigenesis by maintaining mesenchymal progenitor cells in a less differentiated state. Protecting this progenitor cell population might prevent tumor formation in patients harboring a germline APC mutation, where fibromatosis is currently the leading cause of mortality.


Asunto(s)
Fibroma/patología , Células Madre Mesenquimatosas/patología , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/patología , Adolescente , Adulto , Alelos , Animales , Niño , Femenino , Fibroma/genética , Fibroma/metabolismo , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Adulto Joven , beta Catenina/biosíntesis
18.
Cancer Res ; 67(17): 8216-22, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17804735

RESUMEN

Although many cancers are maintained by tumor-initiating cells, this has not been shown for mesenchymal tumors, in part due to the lack of unique surface markers that identify mesenchymal progenitors. An alternative technique to isolate stem-like cells is to isolate side population (SP) cells based on efflux of Hoechst 33342 dye. We examined 29 mesenchymal tumors ranging from benign to high-grade sarcomas and identified SP cells in all but six samples. There was a positive correlation between the percentage of SP cells and the grade of the tumor. SP cells preferentially formed tumors when grafted into immunodeficient mice, and only cells from tumors that developed from the SP cells had the ability to initiate tumor formation upon serial transplantation. Although SP cells are able to efflux rhodamine dye in addition to Hoechst 33342, we found that the ability to efflux rhodamine dye did not identify a population of cells enriched for tumor-initiating capacity. Here, we identify a subpopulation of cells within a broad range of benign and malignant mesenchymal tumors with tumor-initiating capacity. In addition, our data suggest that the proportion of SP cells could be used as a prognostic factor and that therapeutically targeting this subpopulation of cells could be used to improve patient outcome.


Asunto(s)
Neoplasias de los Tejidos Conjuntivo y Blando/patología , Células Madre Neoplásicas/citología , Animales , Proliferación Celular , Progresión de la Enfermedad , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Trasplante de Neoplasias/patología , Células Madre Neoplásicas/trasplante , Trasplante Heterólogo
19.
PLoS Med ; 4(7): e249, 2007 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-17676991

RESUMEN

BACKGROUND: Delayed fracture healing causes substantial disability and usually requires additional surgical treatments. Pharmacologic management to improve fracture repair would substantially improve patient outcome. The signaling pathways regulating bone healing are beginning to be unraveled, and they provide clues into pharmacologic management. The beta-catenin signaling pathway, which activates T cell factor (TCF)-dependent transcription, has emerged as a key regulator in embryonic skeletogenesis, positively regulating osteoblasts. However, its role in bone repair is unknown. The goal of this study was to explore the role of beta-catenin signaling in bone repair. METHODS AND FINDINGS: Western blot analysis showed significant up-regulation of beta-catenin during the bone healing process. Using a beta-Gal activity assay to observe activation during healing of tibia fractures in a transgenic mouse model expressing a TCF reporter, we found that beta-catenin-mediated, TCF-dependent transcription was activated in both bone and cartilage formation during fracture repair. Using reverse transcription-PCR, we observed that several WNT ligands were expressed during fracture repair. Treatment with DKK1 (an antagonist of WNT/beta-catenin pathway) inhibited beta-catenin signaling and the healing process, suggesting that WNT ligands regulate beta-catenin. Healing was significantly repressed in mice conditionally expressing either null or stabilized beta-catenin alleles induced by an adenovirus expressing Cre recombinase. Fracture repair was also inhibited in mice expressing osteoblast-specific beta-catenin null alleles. In stark contrast, there was dramatically enhanced bone healing in mice expressing an activated form of beta-catenin, whose expression was restricted to osteoblasts. Treating mice with lithium activated beta-catenin in the healing fracture, but healing was enhanced only when treatment was started subsequent to the fracture. CONCLUSIONS: These results demonstrate that beta-catenin functions differently at different stages of fracture repair. In early stages, precise regulation of beta-catenin is required for pluripotent mesenchymal cells to differentiate to either osteoblasts or chondrocytes. Once these undifferentiated cells have become committed to the osteoblast lineage, beta-catenin positively regulates osteoblasts. This is a different function for beta-catenin than has previously been reported during development. Activation of beta-catenin by lithium treatment has potential to improve fracture healing, but only when utilized in later phases of repair, after mesenchymal cells have become committed to the osteoblast lineage.


Asunto(s)
Fracturas Óseas/fisiopatología , Transducción de Señal/fisiología , Cicatrización de Heridas/fisiología , beta Catenina/fisiología , Adenoviridae/genética , Animales , Western Blotting , Fracturas Óseas/genética , Fracturas Óseas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Integrasas/genética , Integrasas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Litio/farmacología , Masculino , Ratones , Ratones Transgénicos , Modelos Animales , Osteoblastos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Transfección/métodos , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas Wnt/fisiología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética , beta Catenina/genética , beta Catenina/metabolismo
20.
Cancer Res ; 67(15): 7124-31, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17671179

RESUMEN

Aggressive fibromatosis (also called desmoid tumor) is a benign, locally invasive, soft tissue tumor composed of cells with mesenchymal characteristics. These tumors are characterized by increased levels of beta-catenin-mediated T-cell factor (TCF)-dependent transcriptional activation. We found that type 1 IFN signaling is activated in human and murine aggressive fibromatosis tumors and that the expression of associated response genes is regulated by beta-catenin. When mice deficient for the type 1 IFN receptor (Ifnar1-/-) were crossed with mice predisposed to developing aggressive fibromatosis tumors (Apc/Apc1638N), a significant decrease in aggressive fibromatosis tumor formation was observed compared with littermate controls, showing a novel role for type 1 IFN signaling in promoting tumor formation. Type 1 IFN activation inhibits cell proliferation but does not alter cell apoptosis or the level of beta-catenin-mediated TCF-dependent transcriptional activation in aggressive fibromatosis cell cultures. Thus, these changes cannot explain our in vivo results. Intriguingly, Ifnar1-/- mice have smaller numbers of mesenchymal progenitor cells compared with littermate controls, and treatment of aggressive fibromatosis cell cultures with IFN increases the proportion of cells that exclude Hoechst dye and sort to the side population, raising the possibility that type 1 IFN signaling regulates the number of precursor cells present that drive aggressive fibromatosis tumor formation and maintenance. This study identified a novel role for IFN type 1 signaling as a positive regulator of neoplasia and suggests that IFN treatment is a less than optimal therapy for this tumor type.


Asunto(s)
Fibromatosis Agresiva/metabolismo , Genes APC/fisiología , Interferón beta/fisiología , Receptor de Interferón alfa y beta/metabolismo , Receptor de Interferón alfa y beta/fisiología , Transducción de Señal/fisiología , Animales , Western Blotting , Proliferación Celular , Transformación Celular Neoplásica , Ensayo de Unidades Formadoras de Colonias , Femenino , Fibroblastos/metabolismo , Fibromatosis Agresiva/patología , Citometría de Flujo , Humanos , Masculino , Células Madre Mesenquimatosas , Ratones , Invasividad Neoplásica/patología , Receptor de Interferón alfa y beta/genética , Factor 1 de Transcripción de Linfocitos T/metabolismo , Transcripción Genética , Transgenes/fisiología , Células Tumorales Cultivadas , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA