Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
FASEB J ; 34(8): 11143-11167, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32627872

RESUMEN

Exercise modulates metabolism and the gut microbiome. Brief exposure to low mT-range pulsing electromagnetic fields (PEMFs) was previously shown to accentuate in vitro myogenesis and mitochondriogenesis by activating a calcium-mitochondrial axis upstream of PGC-1α transcriptional upregulation, recapitulating a genetic response implicated in exercise-induced metabolic adaptations. We compared the effects of analogous PEMF exposure (1.5 mT, 10 min/week), with and without exercise, on systemic metabolism and gut microbiome in four groups of mice: (a) no intervention; (b) PEMF treatment; (c) exercise; (d) exercise and PEMF treatment. The combination of PEMFs and exercise for 6 weeks enhanced running performance and upregulated muscular and adipose Pgc-1α transcript levels, whereas exercise alone was incapable of elevating Pgc-1α levels. The gut microbiome Firmicutes/Bacteroidetes ratio decreased with exercise and PEMF exposure, alone or in combination, which has been associated in published studies with an increase in lean body mass. After 2 months, brief PEMF treatment alone increased Pgc-1α and mitohormetic gene expression and after >4 months PEMF treatment alone enhanced oxidative muscle expression, fatty acid oxidation, and reduced insulin levels. Hence, short-term PEMF treatment was sufficient to instigate PGC-1α-associated transcriptional cascades governing systemic mitohormetic adaptations, whereas longer-term PEMF treatment was capable of inducing related metabolic adaptations independently of exercise.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Adaptación Fisiológica/fisiología , Animales , Bacteroidetes/crecimiento & desarrollo , Composición Corporal/fisiología , Ácidos Grasos/metabolismo , Femenino , Firmicutes/crecimiento & desarrollo , Estudios de Seguimiento , Expresión Génica/fisiología , Insulina/metabolismo , Campos Magnéticos , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Desarrollo de Músculos/fisiología , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/fisiología , Transcripción Genética/fisiología , Activación Transcripcional/fisiología
2.
PLoS One ; 12(7): e0181600, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28732055

RESUMEN

The role of mammalian high temperature requirement protease A1 (HTRA1) in somatic stem cell differentiation and mineralized matrix formation remains controversial, having been demonstrated to impart either anti- or pro-osteogenic effects, depending on the in vitro cell model used. The aim of this study was therefore to further evaluate the role of HTRA1 in regulating the differentiation potential and lineage commitment of murine mesenchymal stem cells in vitro, and to assess its influence on bone structure and regeneration in vivo. Our results demonstrated that short hairpin RNA-mediated ablation of Htra1 in the murine mesenchymal cell line C3H10T1/2 increased the expression of several osteogenic gene markers, and significantly enhanced matrix mineralization in response to BMP-2 stimulation. These effects were concomitant with decreases in the expression of chondrogenic gene markers, and increases in adipogenic gene expression and lipid accrual. Despite the profound effects of loss-of-function of HTRA1 on this in vitro osteochondral model, these were not reproduced in vivo, where bone microarchitecture and regeneration in 16-week-old Htra1-knockout mice remained unaltered as compared to wild-type controls. By comparison, analysis of femurs from 52-week-old mice revealed that bone structure was better preserved in Htra1-knockout mice than age-matched wild-type controls. These findings therefore provide additional insights into the role played by HTRA1 in regulating mesenchymal stem cell differentiation, and offer opportunities for improving our understanding of how this multifunctional protease may act to influence bone quality.


Asunto(s)
Condrogénesis/fisiología , Osteogénesis/fisiología , Regeneración/fisiología , Serina Endopeptidasas/metabolismo , Adipogénesis/fisiología , Animales , Proteína Morfogenética Ósea 2/metabolismo , Calcificación Fisiológica/fisiología , Diferenciación Celular/fisiología , Línea Celular , Expresión Génica/fisiología , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Osteoblastos/metabolismo
3.
Sci Rep ; 7(1): 2243, 2017 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-28533546

RESUMEN

The osteoinductive properties of prostaglandin E2 (PGE2) and its signaling pathways have led to suggestions that it may serve as a potential therapeutic strategy for bone loss. However, the prominence of PGE2 as an inducer of bone formation is attributed primarily to findings from studies using rodent models. In the current study, we investigated the effects of PGE2 on human bone marrow stromal cell (hBMSC) lineage commitment and determined its mode of action. We demonstrated that PGE2 treatment of hBMSCs significantly altered the expression profile of several genes associated with osteoblast differentiation (RUNX2 and ALP) and maturation (BGLAP and MGP). This was attributed to the activation of specific PGE2 receptors, and was associated with increases in cAMP production and sustained AKT phosphorylation. Pharmacological inhibition of exchange protein directly activated by cAMP (Epac), but not protein kinase A (PKA), recovered the mineralization functions of hBMSC-derived osteoblasts treated with PGE2 and restored AKT phosphorylation, along with the expression levels of RUNX2, ALP, BGLAP and MGP. Our findings therefore provide insights into how PGE2 influences hBMSC-mediated matrix mineralization, and should be taken into account when evaluating the role of PGE2 in human bone metabolism.


Asunto(s)
Calcificación Fisiológica , Dinoprostona/metabolismo , Matriz Extracelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Adipogénesis/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Dinoprostona/farmacología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Modelos Moleculares , Osteogénesis/efectos de los fármacos , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal
4.
Genes (Basel) ; 7(11)2016 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-27834918

RESUMEN

Although several tendon-selective genes exist, they are also expressed in other musculoskeletal tissues. As cell and tissue engineering is reliant on specific molecular markers to discriminate between cell types, tendon-specific genes need to be identified. In order to accomplish this, we have used RNA sequencing (RNA-seq) to compare gene expression between tendon, bone, cartilage and ligament from horses. We identified several tendon-selective gene markers, and established eyes absent homolog 2 (EYA2) and a G-protein regulated inducer of neurite outgrowth 3 (GPRIN3) as specific tendon markers using RT-qPCR. Equine tendon cells cultured as three-dimensional spheroids expressed significantly greater levels of EYA2 than GPRIN3, and stained positively for EYA2 using immunohistochemistry. EYA2 was also found in fibroblast-like cells within the tendon tissue matrix and in cells localized to the vascular endothelium. In summary, we have identified EYA2 and GPRIN3 as specific molecular markers of equine tendon as compared to bone, cartilage and ligament, and provide evidence for the use of EYA2 as an additional marker for tendon cells in vitro.

5.
Stem Cells Dev ; 25(9): 687-98, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26950191

RESUMEN

All-trans retinoic acid (ATRA) is a potent inducer of osteogenic differentiation in mouse adipose-derived stromal cells (mASCs), although the underlying mechanisms responsible for its mode of action have yet to be completely elucidated. High temperature requirement protease A1 (HtrA1) is a newly recognized modulator of human multipotent stromal cell (MSC) osteogenesis and as such, may play a role in regulating ATRA-dependent osteogenic differentiation of mASCs. In this study, we assessed the influence of small interfering RNA (siRNA)-induced repression of HtrA1 production on mASC osteogenesis and examined its effects on ATRA-mediated mammalian target of rapamycin (mTOR) signaling. Inhibition of HtrA1 production in osteogenic mASCs resulted in a significant reduction of alkaline phosphatase activity and mineralized matrix formation. Western blot analyses revealed the rapid activation of Akt (Ser473) and p70S6K (Thr389) in ATRA-treated mASCs, and that levels of phosphorylated p70S6K were noticeably reduced in HtrA1-deficient mASCs. Further studies using mTOR inhibitor rapamycin and siRNA specific for the p70S6K gene Rps6kb1 confirmed ATRA-mediated mASC osteogenesis as being dependent on p70S6K activation. Finally, transfection of cells with a constitutively active rapamycin-resistant p70S6K mutant could restore the mineralizing capacity of HtrA1-deficient mASCs. These findings therefore lend further support for HtrA1 as a positive mediator of MSC osteogenesis and provide new insights into the molecular mode of action of ATRA in regulating mASC lineage commitment.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina Endopeptidasas/deficiencia , Tretinoina/farmacología , Animales , Western Blotting , Activación Enzimática/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Serina Peptidasa A1 que Requiere Temperaturas Altas , Ratones , Modelos Biológicos , Mutación/genética , Serina Endopeptidasas/metabolismo , Sirolimus/farmacología , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/enzimología
6.
Stem Cells ; 34(6): 1601-14, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26864869

RESUMEN

Adipogenesis is the process by which mesenchymal stem cells (MSCs) develop into lipid-laden adipocytes. Being the dominant cell type within adipose tissue, adipocytes play a central role in regulating circulating fatty acid levels, which is considered to be of critical importance in maintaining insulin sensitivity. High temperature requirement protease A1 (HTRA1) is a newly recognized regulator of MSC differentiation, although its role as a mediator of adipogenesis has not yet been defined. The aim of this work was therefore to evaluate HTRA1's influence on human MSC (hMSC) adipogenesis and to establish a potential mode of action. We report that the addition of exogenous HTRA1 to hMSCs undergoing adipogenesis suppressed their ability to develop into lipid laden adipocytes. These effects were demonstrated as being reliant on both its protease and PDZ domain, and were mediated through the actions of c-Jun N-terminal kinase and matrix metalloproteinases (MMPs). The relevance of such findings with regards to HTRA1's potential influence on adipocyte function in vivo is made evident by the fact that HTRA1 and MMP-13 were readily identifiable within crown-like structures present in visceral adipose tissue samples from insulin resistant obese human subjects. These data therefore implicate HTRA1 as a negative regulator of MSC adipogenesis and are suggestive of its potential involvement in adipose tissue remodeling under pathological conditions. Stem Cells 2016;34:1601-1614.


Asunto(s)
Adipogénesis , Serina Peptidasa A1 que Requiere Temperaturas Altas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/enzimología , Regulación hacia Arriba , Activación Enzimática , Matriz Extracelular/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Grasa Intraabdominal/patología , Gotas Lipídicas/metabolismo , Obesidad/patología
7.
Sci Rep ; 6: 21131, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26883084

RESUMEN

While ADP-ribosyltransferase diphtheria toxin-like 1 (ARTD1, formerly PARP1) and its enzymatic activity have been shown to be important for reprogramming and differentiation of cells, such as during adipogenesis, their role and mechanism in regulating osteoclastogenesis and bone homeostasis are largely unknown. Here, in cell culture-based RANKL-induced osteoclastogenesis models, we show that silencing of ARTD1 or inhibition of its enzymatic activity enhances osteoclast differentiation and function. As a consequence of ARTD1 silencing or inhibition, the recruitment of p65/RelA to the IL-1ß promoter, which is associated with transcriptionally active histone marks, IL-1ß expression and inflammasome-dependent secretion of IL-1ß are enhanced. This subsequently promotes sustained induction of the transcription factor Nfatc1/A and osteoclastogenesis in an autocrine manner via the IL-1 receptor. In vivo, Artd1-deficient mice display significantly decreased bone mass as a consequence of increased osteoclast differentiation. Accordingly, the expression of osteoclast markers is enhanced in mutant compared to wild-type mice. Together, these results indicate that ARTD1 controls osteoclast development and bone remodelling via its enzymatic activity by modulating the epigenetic marks surrounding the IL-1ß promoter and expression of IL-1ß and subsequently also Nfatc1/A.


Asunto(s)
Resorción Ósea , Huesos/metabolismo , Homeostasis , Interleucina-1beta/genética , FN-kappa B/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Transcripción Genética , Animales , Comunicación Autocrina , Sitios de Unión , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , ADN-Topoisomerasas de Tipo II/metabolismo , Activación Enzimática , Regulación de la Expresión Génica , Silenciador del Gen , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Fenotipo , Poli(ADP-Ribosa) Polimerasa-1/genética , Regiones Promotoras Genéticas , Unión Proteica , Ligando RANK/metabolismo , Ligando RANK/farmacología , Transducción de Señal
8.
Biomaterials ; 69: 99-109, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26283157

RESUMEN

Tenocytes represent a valuable source of cells for the purposes of tendon tissue engineering and regenerative medicine and as such, should possess a high degree of tenogenic differentiation prior to their use in vivo in order to achieve maximal efficacy. In the current report, we identify an efficient means by which to maintain differentiated tenocytes in vitro by employing the hanging drop technique in combination with defined growth media supplements. Equine tenocytes retained a more differentiated state when cultured as scaffold-free microtissue spheroids in low serum-containing medium supplemented with L-ascorbic acid 2-phosphate, insulin and transforming growth factor (TGF)-ß1. This was made evident by significant increases in the expression levels of pro-tenogenic markers collagen type I (COL1A2), collagen type III (COL3A1), scleraxis (SCX) and tenomodulin (TNMD), as well as by enhanced levels of collagen type I and tenomodulin protein. Furthermore, tenocytes cultured under these conditions demonstrated a typical spindle-like morphology and when embedded in collagen gels, became highly aligned with respect to the orientation of the collagen structure following their migration out from the microtissue spheroids. Our findings therefore provide evidence to support the use of a biomimetic microtissue approach to culturing tenocytes and that in combination with the defined growth media described, can improve their differentiation status and functional repopulation of collagen matrix.


Asunto(s)
Colágeno/química , Medios de Cultivo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Tendones/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Ácido Ascórbico/análogos & derivados , Ácido Ascórbico/metabolismo , Biomimética , Diferenciación Celular , Células Cultivadas , Caballos , Regeneración , Esferoides Celulares , Tendones/fisiología , Factor de Crecimiento Transformador beta1/metabolismo
9.
Oncotarget ; 6(24): 20288-301, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25978030

RESUMEN

Two main features common to all solid tumors are tissue hypoxia and inflammation, both of which cause tumor progression, metastasis, therapy resistance and increased mortality. Chronic inflammation is associated with increased cancer risk, as demonstrated for inflammatory bowel disease patients developing colon cancer. However, the interplay between hypoxia and inflammation on the molecular level remains to be elucidated. We found that MC-38 mouse colon cancer cells contain functional hypoxic (HIF-1α) and inflammatory (p65/RelA) signaling pathways. In contrast to cells of the myeloid lineage, HIF-1α levels remained unaffected in MC-38 cells treated with LPS, and hypoxia failed to induce NF-κB. A similar regulation of canonical HIF and NF-κB target genes confirmed these results. RNA deep sequencing of HIF-1α and p65/RelA knock-down cells revealed that a surprisingly large fraction of HIF target genes required p65/RelA for hypoxic regulation and a number of p65/RelA target genes required HIF-1α for proinflammatory regulation, respectively. Hypoxia attenuated the inflammatory response to LPS by inhibiting nuclear translocation of p65/RelA independently of HIF-1α, which was associated with enhanced IκBα levels and decreased IKKß phosphorylation. These data demonstrate that the interaction between hypoxic and inflammatory signaling pathways needs to be considered when designing cancer therapies targeting HIF or NF-κB.


Asunto(s)
Proteínas I-kappa B/metabolismo , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , FN-kappa B/metabolismo , Hipoxia de la Célula , Neoplasias del Colon , Humanos , Lipopolisacáridos , Transducción de Señal
10.
Nucleic Acids Res ; 43(1): 129-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25452336

RESUMEN

PPARγ-dependent gene expression during adipogenesis is facilitated by ADP-ribosyltransferase D-type 1 (ARTD1; PARP1)-catalyzed poly-ADP-ribose (PAR) formation. Adipogenesis is accompanied by a dynamic modulation of the chromatin landscape at PPARγ target genes by ligand-dependent co-factor exchange. However, how endogenous PPARγ ligands, which have a low affinity for the receptor and are present at low levels in the cell, can induce sufficient co-factor exchange is unknown. Moreover, the significance of PAR formation in PPARγ-regulated adipose tissue function is also unknown. Here, we show that inhibition of PAR formation in mice on a high-fat diet reduces weight gain and cell size of adipocytes, as well as PPARγ target gene expression in white adipose tissue. Mechanistically, topoisomerase II activity induces ARTD1 recruitment to PPARγ target genes, and ARTD1 automodification enhances ligand binding to PPARγ, thus promoting sufficient transcriptional co-factor exchange in adipocytes. Thus, ARTD1-mediated PAR formation during adipogenesis is necessary to adequately convey the low signal of endogenous PPARγ ligand to effective gene expression. These results uncover a new regulatory mechanism of ARTD1-induced ADP-ribosylation and highlight its importance for nuclear factor-regulated gene expression.


Asunto(s)
Adipogénesis/genética , PPAR gamma/metabolismo , Poli Adenosina Difosfato Ribosa/biosíntesis , Poli(ADP-Ribosa) Polimerasas/metabolismo , Activación Transcripcional , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Línea Celular , Tamaño de la Célula/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/metabolismo , Dieta Alta en Grasa , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Activados del Proliferador del Peroxisoma/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1 , Elementos de Respuesta , Aumento de Peso/efectos de los fármacos
11.
Biomaterials ; 35(26): 7326-35, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24933514

RESUMEN

Adipose-derived stromal cells (ASCs) are increasingly being used for orthopedic-based tissue engineering approaches due to their ability to readily undergo osteogenic differentiation. In the present study, we used in vitro and in vivo approaches to evaluate the use of ASCs as a treatment strategy for age-related osteoporosis. Molecular, histological and micro-computed tomography (micro-CT) based approaches confirmed that ASCs isolated from 18-week-old osteoporotic senescence-accelerated mice (SAMP6) were capable of undergoing osteogenesis when cultured in either silk fibroin (SF) scaffolds or scaffold-free microtissues (ASC-MT). A single intratibial injection of CM-Dil-labeled isogeneic ASCs or ASC-MT into SAMP6 recipients significantly improved trabecular bone quality after 6 weeks in comparison to untreated contralateral bones, as determined by micro-CT. Injected ASCs could be observed in paraffin wax bone sections at 24 h and 6 weeks post treatment and induced a significant increase in several molecular markers of bone turnover. Furthermore, a significant improvement in the osteogenic potential of osteoporotic patient-derived human bone marrow stromal cells (BMSCs) was observed when differentiated in conditioned culture media harvested from osteoporotic patient-derived human ASCs. These findings therefore support the use of ASCs as an autologous cell-based approach for the treatment of osteoporosis.


Asunto(s)
Tejido Adiposo/citología , Osteogénesis , Osteoporosis/terapia , Células del Estroma/trasplante , Factores de Edad , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Osteoporosis/epidemiología , Osteoporosis/patología , Células del Estroma/citología , Tibia/citología , Tibia/patología
12.
Curr Protoc Stem Cell Biol ; 27: 2B.5.1-2B.5.12, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24510285

RESUMEN

In this unit, previously described methods are expanded upon, where procedures relating to the preparation, culturing, and osteogenic differentiation of scaffold-free mouse adipose-derived stromal cell microtissue spheroids (ASC-MT) are outlined. Not only is a detailed methodology of how to engineer such spheroids are presented, but a full account of how to induce and analyze osteogenesis in these ASC-MT constructs is given along with relevant figures to help better illustrate the methods described.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular , Osteogénesis , Esferoides Celulares/citología , Técnicas de Cultivo de Tejidos/métodos , Andamios del Tejido , Animales , Proliferación Celular , Separación Celular , Células Cultivadas , Masculino , Ratones , Células del Estroma/citología
13.
Stem Cells ; 30(10): 2271-82, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22865667

RESUMEN

Mammalian high-temperature requirement serine protease A1 (HTRA1) is a secreted member of the trypsin family of serine proteases which can degrade a variety of bone matrix proteins and as such has been implicated in musculoskeletal development. In this study, we have investigated the role of HTRA1 in mesenchymal stem cell (MSC) osteogenesis and suggest a potential mechanism through which it controls matrix mineralization by differentiating bone-forming cells. Osteogenic induction resulted in a significant elevation in the expression and secretion of HTRA1 in MSCs isolated from human bone marrow-derived MSCs (hBMSCs), mouse adipose-derived stromal cells (mASCs), and mouse embryonic stem cells. Recombinant HTRA1 enhanced the osteogenesis of hBMSCs as evidenced by significant changes in several osteogenic markers including integrin-binding sialoprotein (IBSP), bone morphogenetic protein 5 (BMP5), and sclerostin, and promoted matrix mineralization in differentiating bone-forming osteoblasts. These stimulatory effects were not observed with proteolytically inactive HTRA1 and were abolished by small interfering RNA against HTRA1. Moreover, loss of HTRA1 function resulted in enhanced adipogenesis of hBMSCs. HTRA1 Immunofluorescence studies showed colocalization of HTRA1 with IBSP protein in osteogenic mASC spheroid cultures and was confirmed as being a newly identified HTRA1 substrate in cell cultures and in proteolytic enzyme assays. A role for HTRA1 in bone regeneration in vivo was also alluded to in bone fracture repair studies where HTRA1 was found localized predominantly to areas of new bone formation in association with IBSP. These data therefore implicate HTRA1 as having a central role in osteogenesis through modification of proteins within the extracellular matrix.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Proteínas de la Matriz Extracelular/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proteína Morfogenética Ósea 5/genética , Proteína Morfogenética Ósea 5/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Glicoproteínas/metabolismo , Serina Peptidasa A1 que Requiere Temperaturas Altas , Humanos , Sialoproteína de Unión a Integrina/genética , Sialoproteína de Unión a Integrina/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Serina Endopeptidasas/genética , Serina Endopeptidasas/farmacología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
14.
J Biol Chem ; 287(25): 21335-45, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22556410

RESUMEN

Human HTRA1 is a highly conserved secreted serine protease that degrades numerous extracellular matrix proteins. We have previously identified HTRA1 as being up-regulated in osteoarthritic patients and as having the potential to regulate matrix metalloproteinase (MMP) expression in synovial fibroblasts through the generation of fibronectin fragments. In the present report, we have extended these studies and investigated the role of HTRA1 in the pathogenesis of intervertebral disc (IVD) degeneration. HTRA1 mRNA expression was significantly elevated in degenerated disc tissue and was associated with increased protein levels. However, these increases did not correlate with the appearance of rs11200638 single nucleotide polymorphism in the promoter region of the HTRA1 gene, as has previously been suggested. Recombinant HTRA1 induced MMP production in IVD cell cultures through a mechanism critically dependent on MEK but independent of IL-1ß signaling. The use of a catalytically inactive mutant confirmed these effects to be primarily due to HTRA1 serine protease activity. HTRA1-induced fibronectin proteolysis resulted in the generation of various sized fragments, which when added to IVD cells in culture, caused a significant increase in MMP expression. Furthermore, one of these fragments was identified as being the amino-terminal fibrin- and heparin-binding domain and was also found to be increased within HTRA1-treated IVD cell cultures as well as in disc tissue from patients with IVD degeneration. Our results therefore support a scenario in which HTRA1 promotes IVD degeneration through the proteolytic cleavage of fibronectin and subsequent activation of resident disc cells.


Asunto(s)
Colagenasas/biosíntesis , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Regulación Enzimológica de la Expresión Génica , Degeneración del Disco Intervertebral/enzimología , Proteolisis , Serina Endopeptidasas/biosíntesis , Línea Celular , Colagenasas/genética , Matriz Extracelular/genética , Matriz Extracelular/patología , Femenino , Fibronectinas/genética , Serina Peptidasa A1 que Requiere Temperaturas Altas , Humanos , Disco Intervertebral/enzimología , Disco Intervertebral/patología , Degeneración del Disco Intervertebral/genética , Degeneración del Disco Intervertebral/patología , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , Masculino , Polimorfismo de Nucleótido Simple , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Serina Endopeptidasas/genética , Serina Endopeptidasas/farmacología
15.
FASEB J ; 26(6): 2631-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22426118

RESUMEN

ADP-ribosyltransferase Diphtheria toxin-like 1 [ARTD1; formerly called poly-ADP-ribose polymerase 1 (PARP1)] is a chromatin-associated enzyme involved in regulating metabolic homeostasis. The liver is at the core of glucose and lipid metabolism and is significantly affected by obesity and the metabolic syndrome. Here, we show that when fed a high-fat diet (HFD), mice lacking ARTD1 developed exacerbated hepatic steatosis. ARTD1(-/-) mice had a 19% higher liver weight than wild-type (WT) animals and exhibited a significantly increased serum concentration of cholesterol (38%) and impaired glucose tolerance. In addition, adipocyte function and size were significantly reduced in ARTD1(-/-) mice fed an HFD (7794 µm(2) for WT and 5579 µm(2) for ARTD1(-/-) mice). The significantly reduced adipogenic differentiation of adipose-derived stromal cells (ASCs) isolated from ARTD1(-/-) mice (28 vs. 11% Oil red O-positive cells in WT and ARTD1(-/-) ASCs, respectively) suggested that impaired adipogenesis as the underlying cause for this adipose tissue malfunction. This function of ARTD1 was specific for adipogenesis, since osteogenic differentiation was not affected by the ARTD1 deletion. In summary, we show that ARTD1(-/-) mice fed an HFD display impaired adipogenesis and show exacerbated hepatic steatosis, which can have important implications for nonalcoholic fatty liver disease.


Asunto(s)
Hígado Graso/etiología , Hígado/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Adipocitos/metabolismo , Adipogénesis , Tejido Adiposo/metabolismo , Animales , Diferenciación Celular , Colesterol/sangre , Dieta Alta en Grasa , Intolerancia a la Glucosa/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , Osteogénesis , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/deficiencia
16.
J Tissue Eng Regen Med ; 6(5): 378-90, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21710574

RESUMEN

Adipose tissue provides for a rich and easily accessible source of multipotent stromal cells and thus offers the potential for autologous cell-based therapy for a number of degenerative diseases. Senile osteoporosis is characterized by a reduction in bone quality, which is associated with inadequacies in bone marrow stromal cell (BMSC) differentiation. In the present study, we have characterized adipose-derived stromal cells (ASCs) isolated from aged osteoporotic mice and evaluated their suitability as a source of osteogenic precursor cells. Significant reductions in both tibia bone quality and telomere length in liver tissue were observed in the senescence-accelerated mouse prone 6 strain (SAMP6), as compared to the control age-matched senescence-accelerated mouse resistant 1 strain (SAMR1), thus confirming osteoporosis and accelerated ageing traits in this model. ASCs isolated from inguinal fat expressed mesenchymal surface markers and were capable of differentiating along the osteoblast, adipocyte and chondrocyte lineages. Telomere length was not compromised in ASCs from SAMP6 mice but was actually found to be significantly increased as compared to control SAMR1 mice. Furthermore, ASCs from both strains were comparable in terms of telomerase activity, p21 mRNA expression, SA-ß-gal activity and proliferative capacity. The overall osteogenic and adipogenic potential of ASCs was comparable between SAMP6 and SAMR1 strains, as determined by quantitative molecular, biochemical and histological analyses. In conclusion, adipose tissue may represent a promising autologous cell source for the development of novel bone regenerative therapeutic strategies in the treatment of age-related osteoporosis.


Asunto(s)
Tejido Adiposo/metabolismo , Envejecimiento/metabolismo , Células de la Médula Ósea/metabolismo , Osteoporosis/metabolismo , Células Madre/metabolismo , Telomerasa/metabolismo , Telómero/metabolismo , Tejido Adiposo/patología , Envejecimiento/patología , Animales , Células de la Médula Ósea/patología , Ratones , Ratones Mutantes , Osteoporosis/patología , Células Madre/patología , Células del Estroma/metabolismo , Células del Estroma/patología , Telómero/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA