Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 151(3)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38345329

RESUMEN

The cranial sutures are proposed to be a stem cell niche, harbouring skeletal stem cells that are directly involved in development, homeostasis and healing. Like the craniofacial bones, the sutures are formed from both mesoderm and neural crest. During cranial bone repair, neural crest cells have been proposed to be key players; however, neural crest contributions to adult sutures are not well defined, and the relative importance of suture proximity is unclear. Here, we use genetic approaches to re-examine the neural crest-mesoderm boundaries in the adult mouse skull. These are combined with calvarial wounding experiments suggesting that suture proximity improves the efficiency of cranial repair. Furthermore, we demonstrate that Gli1+ and Axin2+ skeletal stem cells are present in all calvarial sutures examined. We propose that the position of the defect determines the availability of neural crest-derived progenitors, which appear to be a key element in the repair of calvarial defects.


Asunto(s)
Suturas Craneales , Cráneo , Ratones , Animales , Células Madre , Cresta Neural , Mesodermo
2.
Int J Mol Sci ; 21(8)2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32326543

RESUMEN

Many Pasteurella multocida strains are carried as commensals, while some cause disease in animals and humans. Some type D strains cause atrophic rhinitis in pigs, where the causative agent is known to be the Pasteurella multocida toxin (PMT). PMT activates three families of G-proteins-Gq/11, G12/13, and Gi/o-leading to cellular mitogenesis and other sequelae. The effects of PMT on whole animals in vivo have been investigated previously, but only at the level of organ-specific pathogenesis. We report here the first study to screen all the organs targeted by the toxin by using the QE antibody that recognizes only PMT-modified G-proteins. Under our experimental conditions, short-term treatment of PMT is shown to have multiple in vivo targets, demonstrating G-alpha protein modification, stimulation of proliferation markers and expression of active ß-catenin in a tissue- and cell-specific manner. This highlights the usefulness of PMT as an important tool for dissecting the specific roles of different G-alpha proteins in vivo.


Asunto(s)
Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Proliferación Celular/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Pasteurella multocida/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Femenino , Inmunohistoquímica , Ratones , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Bazo/efectos de los fármacos , Bazo/metabolismo , Timo/efectos de los fármacos , Timo/metabolismo , Útero/efectos de los fármacos , Útero/metabolismo , beta Catenina/metabolismo
3.
Stem Cells ; 36(9): 1380-1392, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29726060

RESUMEN

The transcriptional profile induced by hypoxia plays important roles in the chondrogenic differentiation of marrow stromal/stem cells (MSC) and is mediated by the hypoxia inducible factor (HIF) complex. However, various compounds can also stabilize HIF's oxygen-responsive element, HIF-1α, at normoxia and mimic many hypoxia-induced cellular responses. Such compounds may prove efficacious in cartilage tissue engineering, where microenvironmental cues may mediate functional tissue formation. Here, we investigated three HIF-stabilizing compounds, which each have distinct mechanisms of action, to understand how they differentially influenced the chondrogenesis of human bone marrow-derived MSC (hBM-MSC) in vitro. hBM-MSCs were chondrogenically-induced in transforming growth factor-ß3-containing media in the presence of HIF-stabilizing compounds. HIF-1α stabilization was assessed by HIF-1α immunofluorescence staining, expression of HIF target and articular chondrocyte specific genes by quantitative polymerase chain reaction, and cartilage-like extracellular matrix production by immunofluorescence and histochemical staining. We demonstrate that all three compounds induced similar levels of HIF-1α nuclear localization. However, while the 2-oxoglutarate analog dimethyloxalylglycine (DMOG) promoted upregulation of a selection of HIF target genes, desferrioxamine (DFX) and cobalt chloride (CoCl2 ), compounds that chelate or compete with divalent iron (Fe2+ ), respectively, did not. Moreover, DMOG induced a more chondrogenic transcriptional profile, which was abolished by Acriflavine, an inhibitor of HIF-1α-HIF-ß binding, while the chondrogenic effects of DFX and CoCl2 were more limited. Together, these data suggest that HIF-1α function during hBM-MSC chondrogenesis may be regulated by mechanisms with a greater dependence on 2-oxoglutarate than Fe2+ availability. These results may have important implications for understanding cartilage disease and developing targeted therapies for cartilage repair. Stem Cells 2018;36:1380-1392.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Células Madre Mesenquimatosas/efectos de los fármacos , Aminoácidos Dicarboxílicos/farmacología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula/fisiología , Niño , Condrogénesis/efectos de los fármacos , Cobalto/farmacología , Deferoxamina/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo
4.
J Pathol ; 239(4): 438-49, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27172275

RESUMEN

Osteosarcoma (OS) is the most prevalent osseous tumour in children and adolescents and, within this, lung metastases remain one of the factors associated with a dismal prognosis. At present, the genetic determinants driving pulmonary metastasis are poorly understood. We adopted a novel strategy using robust filtering analysis of transcriptomic profiling in tumour osteoblastic cell populations derived from human chemo-naive primary tumours displaying extreme phenotypes (indolent versus metastatic) to uncover predictors associated with metastasis and poor survival. We identified MGP, encoding matrix-Gla protein (MGP), a non-collagenous matrix protein previously associated with the inhibition of arterial calcification. Using different orthotopic models, we found that ectopic expression of Mgp in murine and human OS cells led to a marked increase in lung metastasis. This effect was independent of the carboxylation of glutamic acid residues required for its physiological role. Abrogation of Mgp prevented lung metastatic activity, an effect that was rescued by forced expression. Mgp levels dramatically altered endothelial adhesion, trans-endothelial migration in vitro and tumour cell extravasation ability in vivo. Furthermore, Mgp modulated metalloproteinase activities and TGFß-induced Smad2/3 phosphorylation. In the clinical setting, OS patients who developed lung metastases had high serum levels of MGP at diagnosis. Thus, MGP represents a novel adverse prognostic factor and a potential therapeutic target in OS. Microarray datasets may be found at: http://bioinfow.dep.usal.es/osteosarcoma/ Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Neoplasias Óseas/patología , Proteínas de Unión al Calcio/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Neoplasias Pulmonares/secundario , Osteosarcoma/secundario , Animales , Neoplasias Óseas/metabolismo , Movimiento Celular/fisiología , Humanos , Neoplasias Pulmonares/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Desnudos , Osteosarcoma/metabolismo , Fosforilación , Pronóstico , Proteínas Smad/metabolismo , Proteína Gla de la Matriz
5.
Development ; 140(12): 2597-610, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23715552

RESUMEN

Osteoarthritis primarily affects the articular cartilage of synovial joints. Cell and/or cartilage replacement is a promising therapy, provided there is access to appropriate tissue and sufficient numbers of articular chondrocytes. Embryonic stem cells (ESCs) represent a potentially unlimited source of chondrocytes and tissues as they can generate a broad spectrum of cell types under appropriate conditions in vitro. Here, we demonstrate that mouse ESC-derived chondrogenic mesoderm arises from a Flk-1(-)/Pdgfrα(+) (F(-)P(+)) population that emerges in a defined temporal pattern following the development of an early cardiogenic F(-)P(+) population. Specification of the late-arising F(-)P(+) population with BMP4 generated a highly enriched population of chondrocytes expressing genes associated with growth plate hypertrophic chondrocytes. By contrast, specification with Gdf5, together with inhibition of hedgehog and BMP signaling pathways, generated a population of non-hypertrophic chondrocytes that displayed properties of articular chondrocytes. The two chondrocyte populations retained their hypertrophic and non-hypertrophic properties when induced to generate spatially organized proteoglycan-rich cartilage-like tissue in vitro. Transplantation of either type of chondrocyte, or tissue generated from them, into immunodeficient recipients resulted in the development of cartilage tissue and bone within an 8-week period. Significant ossification was not observed when the tissue was transplanted into osteoblast-depleted mice or into diffusion chambers that prevent vascularization. Thus, through stage-specific manipulation of appropriate signaling pathways it is possible to efficiently and reproducibly derive hypertrophic and non-hypertrophic chondrocyte populations from mouse ESCs that are able to generate distinct cartilage-like tissue in vitro and maintain a cartilage tissue phenotype within an avascular and/or osteoblast-free niche in vivo.


Asunto(s)
Cartílago Articular/citología , Condrocitos/citología , Condrogénesis , Células Madre Embrionarias/citología , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Cartílago Articular/metabolismo , Diferenciación Celular , Linaje de la Célula , Condrocitos/metabolismo , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Femenino , Factor 5 de Diferenciación de Crecimiento/genética , Factor 5 de Diferenciación de Crecimiento/metabolismo , Hipertrofia/metabolismo , Inmunohistoquímica , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Osteoblastos/metabolismo , Osteogénesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Factores de Tiempo
6.
Adv Healthc Mater ; : e2400472, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38809180

RESUMEN

Synthetic hydrogels provide controllable 3D environments, which can be used to study fundamental biological phenomena. The growing body of evidence that cell behavior depends upon hydrogel stress relaxation creates a high demand for hydrogels with tissue-like viscoelastic properties. Here, a unique platform of synthetic polyethylene glycol (PEG) hydrogels in which star-shaped PEG molecules are conjugated with alendronate and/or RGD peptides, attaining modifiable degradability as well as flexible cell adhesion, is created. Novel reversible ionic interactions between alendronate and calcium phosphate nanoparticles, leading to versatile viscoelastic properties with varying initial elastic modulus and stress relaxation time, are identified. This new crosslinking mechanism provides shear-thinning properties resulting in differential cellular responses between cancer cells and stem cells. The novel hydrogel system is an improved design to the other ionic crosslink platforms and opens new avenues for the development of pathologically relevant cancer models, as well as minimally invasive approaches for cell delivery for potential regenerative therapies.

7.
iScience ; 27(4): 109288, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38532886

RESUMEN

RNA-binding proteins (RBPs) are emerging as important regulators of cancer pathogenesis. We reveal that the RBPs LARP4A and LARP4B are differentially overexpressed in osteosarcoma and osteosarcoma lung metastases, as well as in prostate cancer. Depletion of LARP4A and LARP4B reduced tumor growth and metastatic spread in xenografts, as well as inhibiting cell proliferation, motility, and migration. Transcriptomic profiling and high-content multiparametric analyses unveiled a central role for LARP4B, but not LARP4A, in regulating cell cycle progression in osteosarcoma and prostate cancer cells, potentially through modulating key cell cycle proteins such as Cyclins B1 and E2, Aurora B, and E2F1. This first systematic comparison between LARP4A and LARP4B assigns new pro-tumorigenic functions to LARP4A and LARP4B in bone and prostate cancer, highlighting their similarities while also indicating distinct functional differences. Uncovering clear biological roles for these paralogous proteins provides new avenues for identifying tissue-specific targets and potential druggable intervention.

8.
Proc Natl Acad Sci U S A ; 107(29): 12913-8, 2010 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-20616007

RESUMEN

The neighbor of Brca1 gene (Nbr1) functions as an autophagy receptor involved in targeting ubiquitinated proteins for degradation. It also has a dual role as a scaffold protein to regulate growth-factor receptor and downstream signaling pathways. We show that genetic truncation of murine Nbr1 leads to an age-dependent increase in bone mass and bone mineral density through increased osteoblast differentiation and activity. At 6 mo of age, despite normal body size, homozygous mutant animals (Nbr1(tr/tr)) have approximately 50% more bone than littermate controls. Truncated Nbr1 (trNbr1) co-localizes with p62, a structurally similar interacting scaffold protein, and the autophagosome marker LC3 in osteoblasts, but unlike the full-length protein, trNbr1 fails to complex with activated p38 MAPK. Nbr1(tr/tr) osteoblasts and osteoclasts show increased activation of p38 MAPK, and significantly, pharmacological inhibition of the p38 MAPK pathway in vitro abrogates the increased osteoblast differentiation of Nbr1(tr/tr) cells. Nbr1 truncation also leads to increased p62 protein expression. We show a role for Nbr1 in bone remodeling, where loss of function leads to perturbation of p62 levels and hyperactivation of p38 MAPK that favors osteoblastogenesis.


Asunto(s)
Osteoblastos/enzimología , Osteogénesis , Proteínas/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Animales Recién Nacidos , Densidad Ósea , Células COS , Diferenciación Celular , Chlorocebus aethiops , Vesículas Citoplasmáticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Mutantes , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mutantes/metabolismo , Tamaño de los Órganos , Osteoblastos/citología , Estabilidad Proteica , Transporte de Proteínas , Proteínas/metabolismo , Fracciones Subcelulares/metabolismo , Factor de Transcripción TFIIH , Factores de Transcripción/metabolismo
9.
Int J Biochem Cell Biol ; 161: 106441, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37356415

RESUMEN

Recent developments have mounted a stunning body of evidence underlying the importance of RNA binding proteins (RBPs) in cancer research. In this minireview we focus on LARP4A and LARP4B, two paralogs belonging to the superfamily of La-related proteins, and provide a critical overview of current research, including their roles in cancer pathogenesis and cell proliferation, migration, cell cycle and apoptosis. We highlight current controversies surrounding LARP4A and LARP4B and conclude that their complex roles in tumorigenesis are cell-, tissue- and context-dependent, warning that caution must be exercised before categorising either protein as an oncoprotein or tumour-suppressor. We also reveal that LARP4A and LARP4B have often been confused with one another, adding uncertainty in delineating their functions. We suggest that further functional and mechanistic studies of LARP4 proteins present significant challenges for future investigations to recognise the vital contributions of these RBPs in cancer research.


Asunto(s)
Neoplasias , Ribonucleoproteínas , Humanos , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Autoantígenos/genética , Neoplasias/genética , Proteínas de Unión al ARN/genética , Genes Supresores de Tumor
10.
Lab Chip ; 23(4): 761-772, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36722906

RESUMEN

Creating vascularised cellular environments in vitro is a current challenge in tissue engineering and a bottleneck towards developing functional stem cell-derived microtissues for regenerative medicine and basic investigations. Here we have developed a new workflow to manufacture vasculature on chip (VoC) systems efficiently, quickly, and inexpensively. We have employed 3D printing for fast-prototyping of bespoke VoC and coupled them with a refined organotypic culture system (OVAA) to grow patent capillaries in vitro using tissue-specific endothelial and stromal cells. Furthermore, we have designed and implemented a pocket-size flow driver to establish physiologic perfusive flow throughout our VoC-OVAA with minimal medium use and waste. Using our platform, we have created vascularised microtissues and perfused them at physiologic flow rates for extended time (>2 weeks) observing flow-dependent vascular remodelling. Overall, we present for the first time a scalable and customisable system to grow vascularised and perfusable microtissues, a key initial step to grow mature and functional tissues in vitro. We envision that this technology will empower fast prototyping and validation of increasingly biomimetic in vitro systems, including interconnected multi-tissue systems.


Asunto(s)
Compuestos Orgánicos Volátiles , Ingeniería de Tejidos , Perfusión , Dispositivos Laboratorio en un Chip
11.
Am J Pathol ; 179(2): 1041-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21801875

RESUMEN

Podoplanin is a type-I transmembrane sialomucin-like protein, which is expressed in a wide range of cell types and is involved in platelet aggregation and tumor metastasis. Here, we investigated the function, regulation, and expression of podoplanin in osteosarcoma. Podoplanin expression was observed in three osteosarcoma cell lines (MG-63, HOS, and U-2 OS) with platelet aggregation-inducing ability, which was blocked by podoplanin small-interfering RNA or a neutralizing antibody. Overexpression of podoplanin in nonmetastatic Dunn osteosarcoma cells promoted cell migration without attenuating cell proliferation. Both podoplanin and TGF-ß1 were up-regulated by c-Fos induction in MC3T3-E1 osteoblastic cells, and were highly expressed in c-Fos transgenic mouse osteosarcomas and c-Fos-transformed osteosarcoma cell lines. Immunohistochemistry of human osteosarcoma tissue microarrays (n = 133) showed staining of tumor cells embedded in an excess of irregular neoplastic bone matrix in 100% of tumors undergoing so-called "normalization/maturation." Podoplanin was also expressed in osteosarcoma subtypes, with 65% of osteoblastic, 100% of chondroblastic, and 79% of fibroblastic tumors. CD44 and pERM immunohistochemistry showed coexpression with podoplanin in both mouse and human osteosarcoma. Podoplanin expression was significantly higher in metastatic osteosarcomas (n = 6) than in primary osteosarcomas (n = 10). Our data suggest that podoplanin, which is not expressed in normal osteoblasts but in osteocytes, is aberrantly expressed in transformed osteoblasts and in osteosarcoma, and is under AP-1 transcriptional control. Thus podoplanin is a candidate molecule for therapeutic targeting.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Osteosarcoma/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Agregación Plaquetaria , Interferencia de ARN , Factor de Crecimiento Transformador beta1/biosíntesis
12.
Blood ; 115(14): 2769-76, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20065292

RESUMEN

The directed differentiation of human pluripotent stem cells offers the unique opportunity to generate a broad spectrum of human cell types and tissues for transplantation, drug discovery, and studying disease mechanisms. Here, we report the stepwise generation of bone-resorbing osteoclasts from human embryonic and induced pluripotent stem cells. Generation of a primitive streak-like population in embryoid bodies, followed by specification to hematopoiesis and myelopoiesis by vascular endothelial growth factor and hematopoietic cytokines in serum-free media, yielded a precursor population enriched for cells expressing the monocyte-macrophage lineage markers CD14, CD18, CD11b, and CD115. When plated in monolayer culture in the presence of macrophage colony-stimulating factor and receptor activator of nuclear factor-kappaB ligand (RANKL), these precursors formed large, multinucleated osteoclasts that expressed tartrate-resistant acid phosphatase and were capable of resorption. No tartrate-resistant acid phosphatase-positive multinucleated cells or resorption pits were observed in the absence of RANKL. Molecular analyses confirmed the expression of the osteoclast marker genes NFATc1, cathepsin K, and calcitonin receptor in a RANKL-dependent manner, and confocal microscopy demonstrated the coexpression of the alphavbeta3 integrin, cathepsin K and F-actin rings characteristic of active osteoclasts. Generating hematopoietic and osteoclast populations from human embryonic and induced pluripotent stem cells will be invaluable for understanding embryonic bone development and postnatal bone disease.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Osteoclastos/metabolismo , Antígenos de Diferenciación/biosíntesis , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Madre Embrionarias/citología , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Factor Estimulante de Colonias de Macrófagos/farmacología , Mielopoyesis/efectos de los fármacos , Mielopoyesis/fisiología , Osteoclastos/citología , Ligando RANK/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
13.
Acta Biomater ; 149: 179-188, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35779773

RESUMEN

Successfully replacing damaged cartilage with tissue-engineered constructs requires integration with the host tissue and could benefit from leveraging the native tissue's intrinsic healing capacity; however, efforts are limited by a poor understanding of how cartilage repairs minor defects. Here, we investigated the conditions that foster natural cartilage tissue repair to identify strategies that might be exploited to enhance the integration of engineered/grafted cartilage with host tissue. We damaged porcine articular cartilage explants and using a combination of pulsed SILAC-based proteomics, ultrastructural imaging, and catabolic enzyme blocking strategies reveal that integration of damaged cartilage surfaces is not driven by neo-matrix synthesis, but rather local depletion of proteoglycans. ADAMTS4 expression and activity are upregulated in injured cartilage explants, but integration could be reduced by inhibiting metalloproteinase activity with TIMP3. These observations suggest that catabolic enzyme-mediated proteoglycan depletion likely allows existing collagen fibrils to undergo cross-linking, fibrillogenesis, or entanglement, driving integration. Catabolic enzymes are often considered pathophysiological markers of osteoarthritis. Our findings suggest that damage-induced upregulation of metalloproteinase activity may be a part of a healing response that tips towards tissue destruction under pathological conditions and in osteoarthritis, but could also be harnessed in tissue engineering strategies to mediate repair. STATEMENT OF SIGNIFICANCE: Cartilage tissue engineering strategies require graft integration with the surrounding tissue; however, how the native tissue repairs minor injuries is poorly understood. We applied pulsed SILAC-based proteomics, ultrastructural imaging, and catabolic enzyme blocking strategies to a porcine cartilage explant model and found that integration of damaged cartilage surfaces is driven by catabolic enzyme-mediated local depletion of proteoglycans. Although catabolic enzymes have been implicated in cartilage destruction in osteoarthritis, our findings suggest that damage-induced upregulation of metalloproteinase activity may be a part of a healing response that tips towards tissue destruction under pathological conditions. They also suggest that this natural cartilage tissue repair process could be harnessed in tissue engineering strategies to enhance the integration of engineered cartilage with host tissue.


Asunto(s)
Cartílago Articular , Osteoartritis , Animales , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Metaloproteasas/metabolismo , Osteoartritis/patología , Proteoglicanos/metabolismo , Porcinos , Ingeniería de Tejidos
14.
Cell Death Differ ; 29(12): 2459-2471, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36138226

RESUMEN

Oncohistones represent compelling evidence for a causative role of epigenetic perturbations in cancer. Giant cell tumours of bone (GCTs) are characterised by a mutated histone H3.3 as the sole genetic driver present in bone-forming osteoprogenitor cells but absent from abnormally large bone-resorbing osteoclasts which represent the hallmark of these neoplasms. While these striking features imply a pathogenic interaction between mesenchymal and myelomonocytic lineages during GCT development, the underlying mechanisms remain unknown. We show that the changes in the transcriptome and epigenome in the mesenchymal cells caused by the H3.3-G34W mutation contribute to increase osteoclast recruitment in part via reduced expression of the TGFß-like soluble factor, SCUBE3. Transcriptional changes in SCUBE3 are associated with altered histone marks and H3.3G34W enrichment at its enhancer regions. In turn, osteoclasts secrete unregulated amounts of SEMA4D which enhances proliferation of mutated osteoprogenitors arresting their maturation. These findings provide a mechanism by which GCTs undergo differentiation in response to denosumab, a drug that depletes the tumour of osteoclasts. In contrast, hTERT alterations, commonly found in malignant GCT, result in the histone-mutated neoplastic cells being independent of osteoclasts for their proliferation, predicting unresponsiveness to denosumab. We provide a mechanism for the initiation of GCT, the basis of which is dysfunctional cross-talk between bone-forming and bone-resorbing cells. The findings highlight the role of tumour/microenvironment bidirectional interactions in tumorigenesis and how this is exploited in the treatment of GCT.


Asunto(s)
Neoplasias Óseas , Tumor Óseo de Células Gigantes , Humanos , Tumor Óseo de Células Gigantes/genética , Tumor Óseo de Células Gigantes/tratamiento farmacológico , Tumor Óseo de Células Gigantes/patología , Histonas/genética , Histonas/metabolismo , Denosumab/metabolismo , Denosumab/uso terapéutico , Neoplasias Óseas/genética , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Osteoclastos/metabolismo , Remodelación Ósea/genética , Microambiente Tumoral , Proteínas de Unión al Calcio/metabolismo
15.
Tumour Biol ; 32(3): 611-22, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21360024

RESUMEN

S100A4, a 10-12 kDa calcium-binding protein, plays functional roles in tumor progression and metastasis. The present study aimed to investigate the function of S100A4 in osteosarcoma (OS) metastasis, using a loss-of-function approach. Our previous expression profiling analysis revealed that S100a4 was preferentially expressed in the highly metastatic mouse OS cell line, LM8. Introducing a short hairpin ribonucleic acid (shRNA) targeting S100a4 using a newly established vector containing insulators and transposons, we established stable LM8 subclones with almost 100% silencing of endogenous S100a4 protein. These transfectants showed a significant suppression of cell migration in vitro as well as a marked reduction in their ability to colonize the lung and form pulmonary metastases in vivo following intravenous inoculation, whereas there was no significant change in cell proliferation or cell attachment to fibronectin, laminin, and type I collagen. Expression and phosphorylation of ezrin, an emerging OS metastasis-associated factor, and expression of MMPs, remained the same in S100a4-shRNA clones. In 61 human OS, immunohistochemical analysis showed that lesional cells in 85.2% samples expressed S100A4 protein, and the immunoreactivity was primarily cytoplasmic, but it also showed occasional nuclear localization. Chondroblastic and osteoblastic OS subtypes expressed more S100A4 than fibroblastic subtypes. The causative role of S100A4 in OS lung metastasis shown in the murine xenograft model, together with the high proportion of primary human OS expressing S100A4, suggest that S100A4 protein represents an important potential target for future OS therapy.


Asunto(s)
Neoplasias Óseas/patología , Movimiento Celular , Osteosarcoma/patología , Proteínas S100/fisiología , Animales , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Neoplasias Pulmonares/secundario , Ratones , ARN Mensajero/análisis , Proteína de Unión al Calcio S100A4 , Proteínas S100/análisis , Proteínas S100/genética
16.
EMBO Mol Med ; 12(11): e11131, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33047515

RESUMEN

Sarcomas are heterogeneous and clinically challenging soft tissue and bone cancers. Although constituting only 1% of all human malignancies, sarcomas represent the second most common type of solid tumors in children and adolescents and comprise an important group of secondary malignancies. More than 100 histological subtypes have been characterized to date, and many more are being discovered due to molecular profiling. Owing to their mostly aggressive biological behavior, relative rarity, and occurrence at virtually every anatomical site, many sarcoma subtypes are in particular difficult-to-treat categories. Current multimodal treatment concepts combine surgery, polychemotherapy (with/without local hyperthermia), irradiation, immunotherapy, and/or targeted therapeutics. Recent scientific advancements have enabled a more precise molecular characterization of sarcoma subtypes and revealed novel therapeutic targets and prognostic/predictive biomarkers. This review aims at providing a comprehensive overview of the latest advances in the molecular biology of sarcomas and their effects on clinical oncology; it is meant for a broad readership ranging from novices to experts in the field of sarcoma.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Sarcoma , Neoplasias de los Tejidos Blandos , Adolescente , Niño , Humanos , Medicina Molecular , Sarcoma/genética , Sarcoma/terapia
17.
Dev Biol ; 314(2): 457-70, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18191120

RESUMEN

The GATA family of transcription factors are known to play multiple critical roles in vertebrate developmental processes, including erythropoiesis, endoderm formation and cardiogenesis. There have been no previous demonstrations of a functional role for any GATA family member being associated with musculoskeletal development but we now identify a possible role for GATA-6 in chondrogenesis. We detect abundant levels of GATA-6 mRNA in precartilaginous condensations (PCCs) in both the axial and appendicular skeleton of mouse embryos and in committed primary chondrocyte precursors. We also show that the G-protein coupled receptor, Gpr49, is a target of GATA-6 regulation in differentiating embryonal carcinoma cells and that, in vivo, the expression domains of the two genes overlap within PCCs. Finally, we have identified conserved, canonical GATA binding sites within the Gpr49 gene locus, and show by EMSAs that GATA-6 can bind to these sites in vitro. These data therefore suggest that GATA-6 also plays a role in chondrogenesis and that Gpr49 is a potential direct target of GATA regulation in this process.


Asunto(s)
Cartílago/embriología , Condrogénesis/fisiología , Factor de Transcripción GATA6/genética , Vertebrados/embriología , Animales , Núcleo Celular/fisiología , Cartilla de ADN , Embrión de Mamíferos/fisiología , Hibridación in Situ , Ratones , ARN Interferente Pequeño/genética , Receptores Acoplados a Proteínas G/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
18.
Mol Syndromol ; 10(1-2): 48-57, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30976279

RESUMEN

The craniofacial skeleton is formed from the neural crest and mesodermal lineages, both of which contribute mesenchymal precursors during formation of the skull bones. The large majority of cranial sutures also includes a proportion of neural crest-derived mesenchyme. While some studies have addressed the relative healing abilities of neural crest and mesodermal bone, relatively little attention has been paid to differences in intrinsic osteogenic potential. Here, we use mouse models to compare neural crest osteoblasts (from frontal bones or dura mater) to mesodermal osteoblasts (from parietal bones). Using in vitro culture approaches, we find that neural crest-derived osteoblasts readily generate bony nodules, while mesodermal osteoblasts do so less efficiently. Furthermore, we find that co-culture of neural crest-derived osteoblasts with mesodermal osteoblasts is sufficient to nucleate ossification centres. Altogether, this suggests that the intrinsic osteogenic abilities of neural crest-derived mesenchyme may be a primary driver behind craniosynostosis.

20.
Mol Cell Biol ; 39(5)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30559307

RESUMEN

Heterotrimeric G proteins are signal transduction proteins involved in regulating numerous signaling events. In particular, previous studies have demonstrated a role for G-proteins in regulating ß-catenin signaling. However, the link between G-proteins and ß-catenin signaling is controversial and appears to depend on G-protein specificity. We describe a detailed analysis of a link between specific G-alpha subunits and ß-catenin using G-alpha subunit genetic knockout and knockdown approaches. The Pasteurella multocida toxin was utilized as a unique tool to activate G-proteins, with LiCl treatment serving as a ß-catenin signaling agonist. The results show that Pasteurella multocida toxin (PMT) significantly enhanced LiCl-induced active ß-catenin levels in HEK293T cells and mouse embryo fibroblasts. Evaluation of the effect of specific G-alpha proteins on the regulation of ß-catenin showed that Gq/11 and G12/13 knockout cells had significantly higher levels of active and total ß-catenin than wild-type cells. The stimulation of active ß-catenin by PMT and LiCl was lost upon both constitutive and transient knockdown of G12 and G13 but not Gq Based on our results, we conclude that endogenous G-alpha proteins are negative regulators of active ß-catenin; however, PMT-activated G-alpha subunits positively regulate LiCl-induced ß-catenin expression in a G12/13-dependent manner. Hence, G-alpha subunit regulation of ß-catenin is context dependent.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , beta Catenina/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Fibroblastos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica/genética , Células HEK293 , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Ratones , Transducción de Señal , beta Catenina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA