Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 317(4): F874-F880, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31390230

RESUMEN

Parathyroid hormone-related protein (PTHrP) and its receptor are abundantly expressed throughout the renal parenchyma, where PTHrP exerts a modulatory action on renal function. PTHrP upregulation is a common event associated with the mechanism of renal injury and repair. However, no study has yet explored the putative excretion of PTHrP in urine, including its potential relationship with renal function. In the present study, we tested this hypothesis by studying the well-known rat model of acute renal injury induced by the chemotherapeutic agent cisplatin. Using Western blot analysis, we could detect a single protein band, corresponding to intact PTHrP, in the urine of both control and cisplatin-injected rats, whose levels were significantly higher in the latter group. PTHrP was detected in rat urine by dot blot, and its quantification with two specific ELISA kits showed that, compared with control rats, those treated with cisplatin displayed a significant increase in urinary PTHrP (expressed as the PTHrP-to-creatinine ratio or 24-h excretion). In addition, a positive correlation between urinary PTHrP excretion and serum creatinine was found in these animals. In conclusion, our data demonstrate that PTHrP is excreted in rat urine and that this excretion is higher with the decrease of renal function. This suggests that urinary PTHrP levels might be a renal function marker.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/orina , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Proteína Relacionada con la Hormona Paratiroidea/orina , Lesión Renal Aguda/patología , Animales , Biomarcadores/orina , Creatinina/orina , Riñón/patología , Pruebas de Función Renal , Masculino , Ratas , Ratas Wistar
2.
J Cell Physiol ; 232(4): 785-796, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27357344

RESUMEN

Oxidative damage is an important contributor to the morphological and functional changes in osteoporotic bone. Aging increases the levels of reactive oxygen species (ROS) that cause oxidative stress and induce osteoblast apoptosis. ROS modify several signaling responses, including mitogen-activated protein kinase (MAPK) activation, related to cell survival. Both parathyroid hormone (PTH) and its bone counterpart, PTH-related protein (PTHrP), can regulate MAPK activation by modulating MAPK phosphatase-1 (MKP1). Thus, we hypothesized that PTHrP might protect osteoblasts from ROS-induced apoptosis by targeting MKP1. In osteoblastic MC3T3-E1 and MG-63 cells, H2 O2 triggered p38, JNK, ERK and p66Shc phosphorylation, and cell apoptosis. Meanwhile, PTHrP (1-37) rapidly but transiently increased ERK and Akt phosphorylation without affecting p38, JNK, or p66Shc activation. H2 O2 -induced p38 and ERK phosphorylation and apoptosis were both decreased by pre-treatment with specific kinase inhibitors or PTHrP (1-37) in both osteoblastic cell types. These dephosphorylating and prosurvival actions of PTHrP (1-37) were prevented by a phosphatase inhibitor cocktail, the phosphatase MKP1 inhibitor sanguinarine or a MKP1 siRNA. PTHrP (1-37) promptly enhanced MKP1 protein and gene expression and MKP1-dependent catalase activity in osteoblastic cells. Furthermore, exposure to PTHrP (1-37) adsorbed in an implanted hydroxyapatite-based ceramic into a tibial defect in aging rats increased MKP1 and catalase gene expression in the healing bone area. Our findings demonstrate that PTHrP counteracts the pro-apoptotic actions of ROS by a mechanism dependent on MKP1-induced dephosphorylation of MAPKs in osteoblasts. J. Cell. Physiol. 232: 785-796, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Citoprotección/efectos de los fármacos , Fosfatasa 1 de Especificidad Dual/metabolismo , Osteoblastos/enzimología , Osteoblastos/patología , Estrés Oxidativo/efectos de los fármacos , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Animales , Apoptosis/efectos de los fármacos , Huesos/efectos de los fármacos , Catalasa/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Masculino , Ratones , Osteoblastos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas Wistar , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
J Cell Physiol ; 232(12): 3611-3621, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28138960

RESUMEN

Diabetes mellitus (DM) induces bone deterioration, while mechanical stimulation promotes osteocyte-driven bone formation. We aimed to evaluate the interaction of acute exposure (24 h) to high glucose (HG) with both the pro-survival effect conferred to osteocytic MLO-Y4 cells and osteoblastic MC3T3-E1 cells by mechanical stimulation and the interaction of these cells with osteoclast precursor RAW264.7 cells. We found that 24 h of HG (25 mM) pre-exposure prevented both cell survival and ERK and ß-catenin nuclear translocation upon mechanical stimulation by fluid flow (FF) (10 min) in both MLO-Y4 and MC3T3-E1 cells. However, migration of RAW 264.7 cells was inhibited by MLO-Y4 cell-conditioned medium (CM), but not by MC3T3-E1 cell-CM, with HG or FF. This inhibitory effect was associated with consistent changes in VEGF, RANTES, MIP-1α, MIP-1ß MCP-1, and GM-CSF in MLO-Y4 cell-CM. RAW264.7 proliferation was inhibited by MLO-Y4 CM under static or HG conditions, but it increased by FF-CM with or without HG. In addition, both FF and HG abrogated the capacity of RAW 264.7 cells to differentiate into osteoclasts, but in a different manner. Thus, HG-CM in static condition allowed formation of osteoclast-like cells, which were unable to resorb hydroxyapatite. In contrast, FF-CM prevented osteoclastogenesis even in HG condition. Moreover, HG did not affect basal RANKL or IL-6 secretion or their inhibition induced by FF in MLO-Y4 cells. In conclusion, this in vitro study demonstrates that HG exerts disparate effects on osteocyte mechanotransduction, and provides a novel mechanism by which DM disturbs skeletal metabolism through altered osteocyte-osteoclast communication.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Glucosa/farmacología , Mecanotransducción Celular/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteocitos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Células Madre/efectos de los fármacos , Células 3T3 , Animales , Comunicación Celular/efectos de los fármacos , Citocinas/metabolismo , Ratones , Osteoclastos/metabolismo , Osteocitos/metabolismo , Estimulación Física , Células RAW 264.7 , Células Madre/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Biochem Biophys Res Commun ; 482(4): 1019-1024, 2017 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-27908723

RESUMEN

Reactive Oxygen Species (ROS) increase during aging, potentially affecting many tissues including brain, heart, and bone. ROS alter signaling pathways and constitute potential therapeutic targets to limit oxidative damaging effects in aging-associated diseases. Parathyroid hormone receptors (PTHR) are widely expressed and PTH is the only anabolic therapy for osteoporosis. The effects of oxidative stress on PTHR signaling and trafficking have not been elucidated. Here, we used Fluorescence Resonance Energy Transfer (FRET)-based cAMP, ERK, and calcium fluorescent biosensors to analyze the effects of ROS on PTHR signaling and trafficking by live-cell imaging. PTHR internalization and recycling were measured in HEK-293 cells stably transfected with HA-PTHR. PTH increased cAMP production, ERK phosphorylation, and elevated intracellular calcium. Pre-incubation with H2O2 reduced all PTH-dependent signaling pathways. These inhibitory effects were not a result of PTH oxidation since PTH incubated with H2O2 triggered similar responses. PTH promoted internalization and recycling of the PTHR. Both events were significantly reduced by H2O2 pre-incubation. These findings highlight the role of oxidation on PTHR signaling and trafficking, and suggest the relevance of ROS as a putative target in diseases associated with oxidative stress such as age-related osteoporosis.


Asunto(s)
Estrés Oxidativo , Receptores de Hormona Paratiroidea/metabolismo , Transducción de Señal , AMP Cíclico/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Peróxido de Hidrógeno/metabolismo , Transporte de Proteínas
5.
Calcif Tissue Int ; 98(4): 359-69, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26259869

RESUMEN

The only bone anabolic agent currently available for osteoporosis treatment is parathyroid hormone (PTH)-either its N-terminal 1-34 fragment or the whole molecule of 1-84 aminoacids-whose intermittent administration stimulates new bone formation by targeting osteoblastogenesis and osteoblast survival. PTH-related protein (PTHrP) is an abundant factor in bone which shows N-terminal homology with PTH and thus exhibits high affinity for the same PTH type 1 receptor in osteoblasts. Therefore, it is not surprising that intermittently administered N-terminal PTHrP peptides induce bone anabolism in animals and humans. Furthermore, the C-terminal region of PTHrP also elicits osteogenic features in vitro in osteoblastic cells and in various animal models of osteoporosis. In this review, we discuss the current concepts about the cellular and molecular mechanisms whereby PTHrP may induce anabolic actions in bone. Pre-clinical studies and clinical data using N-terminal PTHrP analogs are also summarized, pointing to PTHrP as a promising alternative to current bone anabolic therapies.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Osteoporosis/tratamiento farmacológico , Proteína Relacionada con la Hormona Paratiroidea , Animales , Humanos , Osteogénesis/efectos de los fármacos
6.
J Cell Physiol ; 230(2): 278-85, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25102966

RESUMEN

Mechanical loading plays a key role in bone formation and maintenance. While unloading induces osteocyte apoptosis and bone loss in vivo, mechanical stimuli prevents osteocyte death through a mechanism involving ß-catenin accumulation and ERK nuclear translocation. Vascular endothelial growth factor (VEGF) has a crucial role in bone formation, but its interaction with osteocytes is not completely understood. Of interest, VEGF receptor 2 (VEGFR2) has recently been shown to mediate the mechanical response of endothelial cells. The present study aimed to evaluate the putative role of the VEGF system in osteocyte mechanosensing. We show that either short (10 min) mechanical stimulus by pulsatile fluid flow (FF) (10 dyn/cm(2), 8 Hz) or exogenous VEGF165 (6 ng/ml) similarly stimulated cell viability, ERK phosphorylation, and ß-catenin membrane translocation. A VEGFR2 antagonist (SU5416) or transfection with specific VEGFR2 siRNAs (siVEGFR2) decreased these events. FF for 10 min increased VEGFR2 phosphorylation at both Tyr-1059 and Tyr-1175; an effect that was mimicked by VEGF165 but was unaffected by a VEGF neutralizing antibody. Subsequently (at 6 h), this mechanical stimulus induced VEGF gene overexpression, which was prevented by siVEGFR2 transfection. Depletion of the structural protein caveolin-1 by using siRNA technology impaired FF-induced VEGFR2 phosphorylation. In conclusion, these in vitro findings point to caveolin-1-dependent VEGFR2 activation as an important mechanism whereby mechanical stimuli promote osteocyte viability.


Asunto(s)
Caveolina 1/metabolismo , Células Endoteliales/metabolismo , Mecanotransducción Celular/fisiología , Osteocitos/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Movimiento Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Activación Enzimática , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Osteocitos/citología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , beta Catenina/metabolismo
7.
Biochem J ; 455(3): 329-37, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23981124

RESUMEN

Autophagy is a highly regulated homoeostatic process involved in the lysosomal degradation of damaged cell organelles and proteins. This process is considered an important pro-survival mechanism under diverse stress conditions. A diabetic milieu is known to hamper osteoblast viability and function. In the present study, we explored the putative protective role of autophagy in osteoblastic cells exposed to an HG (high glucose) medium. HG was found to increase protein oxidation and triggered autophagy by a mechanism dependent on reactive oxygen species overproduction in osteoblastic MC3T3-E1 cells. MC3T3-E1 cell survival was impaired by HG and worsened by chemical or genetic inhibition of autophagy. These findings were mimicked by H2O2-induced oxidative stress in these cells. Autophagy impairment led to both defective mitochondrial morphology and decreased bioenergetic machinery and inhibited further osteoblast differentiation in MC3T3-E1 cells upon exposure to HG. These novel findings indicate that autophagy is an essential mechanism to maintain osteoblast viability and function in an HG environment.


Asunto(s)
Autofagia , Glucosa/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Animales , Diferenciación Celular , Línea Celular , Supervivencia Celular , Peróxido de Hidrógeno/metabolismo , Ratones , Oxidación-Reducción , Estrés Oxidativo/fisiología
8.
J Cell Biochem ; 114(6): 1404-13, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23444047

RESUMEN

Parathyroid hormone-related protein (PTHrP) stimulates osteoblastic function through its N- and C-terminal domains. Since the osteogenic action of the latter domain appears to depend at least in part on its interaction with the vascular endothelial growth factor (VEGF) system, we aimed to explore the putative mechanism underlying this interaction in osteoblasts. Using native conditions for protein extraction and immunoblotting, we found that both PTHrP (107-139) and the shorter PTHrP (107-111) peptide (known as osteostatin), at 100 nM, promoted the appearance of a VEGF receptor (VEGFR) 2 protein band of apparent Mr. wt. 230 kDa, which likely represents its activation by dimer formation, in mouse osteoblastic MC3T3-E1 cells. Moreover, osteostatin (100 nM) maximally increased VEGFR2 phosphorylation at Tyr-1059 within 5-10 min in both MC3T3-E1 and rat osteoblastic osteosarcoma UMR-106 cells. This phosphorylation elicited by osteostatin appears to be VEGF-independent, but prevented by the VEGFR2 activation inhibitor SU1498 and also by the Src kinase inhibitors SU6656 and PP1. Furthermore, osteostatin induced phosphorylation of Src, extracellular signal-regulated kinase (ERK) and Akt with a similar time course to that observed for VEGFR2 activation in these osteoblastic cells. This osteostatin-dependent induction of ERK and Akt activation was abrogated by SU6656. Up-regulation of VEGF and osteoprotegerin gene expression as well as the pro-survival effect induced by osteostatin treatment were all prevented by both SU1498 and SU6656 in these osteoblastic cells. Collectively, these findings demonstrate that the osteostatin domain of C-terminal PTHrP phosphorylates VEGFR2 through Src activation, which represents a mechanism for modulating osteoblastic function.


Asunto(s)
Osteoblastos/fisiología , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Fragmentos de Péptidos/fisiología , Activación Transcripcional , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Familia-src Quinasas/fisiología , Células 3T3 , Animales , Supervivencia Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones , Fosforilación , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
J Cell Biochem ; 114(8): 1908-16, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23494914

RESUMEN

Recent in vivo findings suggest that the bone sparing effect of parathyroid hormone-related protein (PTHrP) in diabetic mice might occur at least in part through targeting a suppressed Wnt/ß-catenin pathway in osteoblasts. We here aimed to examine the inhibitory action of a high glucose environment on specific components of the canonical Wnt pathway, and the putative compensatory effects of PTHrP, in osteoblastic cell cultures. Mouse osteoblastic MC3T3-E1 cells and primary cultures of fetal mouse calvaria were exposed to normal (5.5 mM) or high (25 mM) D-glucose (HG), with or without PTHrP (1-36) or PTHrP (107-139) for different times. In some experiments, MC3T3-E1 cells were incubated with the Wnt pathway activators Wnt3a and LiCl, or were transfected with plasmids encoding either a mutated ß-catenin that cannot be targeted for degradation or a human PTHrP (-36/+139) cDNA, or the corresponding empty plasmid, in the presence or absence of HG. The gene expression of Wnt3a and low density receptor-like proteins (LRP)-5 and 6, as well as ß-catenin protein stabilization and ß-catenin-dependent transcription activity were evaluated. Oxidative stress status under HG condition was also assessed. The present data demonstrate that HG can target different components of the canonical Wnt pathway, while ß-catenin degradation appears to be a key event leading to inhibition of Wnt/ß-catenin signaling in mouse osteoblastic cells. Both PTHrP peptides tested were able to counteract this deleterious action of HG. These in vitro findings also provide new clues to understand the underlying mechanisms whereby PTHrP can increase bone formation.


Asunto(s)
Glucosa/metabolismo , Osteoblastos/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Animales , Antimaníacos/farmacología , Línea Celular , Glucosa/genética , Humanos , Cloruro de Litio/farmacología , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Mutación , Osteoblastos/citología , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Estabilidad Proteica , Vía de Señalización Wnt/fisiología , Proteína Wnt3A/metabolismo , Proteína Wnt3A/farmacología , beta Catenina/genética , beta Catenina/metabolismo
10.
Kidney Int ; 83(5): 825-34, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23364519

RESUMEN

Runx2 is a key transcription factor in bone development regulating several processes, including osteoblast apoptosis. The antiapoptotic effects of parathyroid hormone (PTH) in osteoblasts depend on Runx2-mediated transcription of prosurvival genes. In the kidney, PTH-related protein (PTHrP) promotes tubulointerstitial cell survival by activating the PTH/PTHrP type 1 receptor. We found that Runx2 is expressed in renal tubuloepithelial MCT and HK2 cell lines in vitro and in the mouse kidney tubuloepithelium in vivo. The 1-36 amino-acid fragment of PTHrP was found to increase the expression and nuclear translocation of Runx2 in both cell lines in a dose- and time-dependent manner. PTHrP(1-36) protected renal tubuloepithelial cells from folic acid toxicity and serum deprivation, an effect inhibited by a dominant-negative Runx2 construct or a Runx2 siRNA. Furthermore, PTHrP(1-36) upregulated the antiapoptotic proteins Bcl-2 and osteopontin, and these effects were abolished by Runx2 siRNA. Runx2, osteopontin, and Bcl-2 were increased in tubuloepithelial cells from transgenic mice with PTHrP overexpression and in wild-type mice with acute or chronic renal failure. Thus, PTHrP regulates renal tubuloepithelial cell survival via Runx2 in the mammalian kidney.


Asunto(s)
Apoptosis , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Células Epiteliales/metabolismo , Túbulos Renales Proximales/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Fragmentos de Péptidos/metabolismo , Transporte Activo de Núcleo Celular , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Ácido Fólico/toxicidad , Humanos , Fallo Renal Crónico/genética , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/patología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Ratones , Ratones Transgénicos , Osteopontina/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección
11.
J Cell Physiol ; 227(4): 1752-60, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21702049

RESUMEN

We here compared the changes induced by subcutaneous injection of PTHrP (1-36) or PTHrP (107-139) (80 µg/kg/day, 5 days/week for 4 or 8 weeks) in bone histology and bone remodeling factors, and in bone marrow cells (BMCs) ex vivo, in ovariectomized (OVX) mice. We also examined the osteogenic effects of these peptides in mouse mesenchymal C3H10T1/2 cells under oxidative stress condition in vitro, which recapitulates the effects of OVX. We confirmed that PTHrP (1-36) exerts bone anabolic actions, as assessed by bone histology and osteoblast differentiation markers in the long bones and plasma from OVX mice. PTHrP (107-139) was also efficient in stimulating several bone formation parameters, and it dramatically decreased bone resorption markers. Moreover, both PTHrP peptides modulate DKK-1 and Sost/sclerostin in osteoblast-like UMR-106 cells highly expressing these Wnt pathway inhibitors, related to their osteogenic action in this in vivo scenario. Administration of either PTHrP peptide improved osteogenic differentiation in BMCs from OVX mice ex vivo and in mouse mesenchymal C3H10T1/2 cells under oxidative stress condition in vitro. These data demonstrate that PTHrP (1-36) and PTHrP (107-139) can exert similar osteogenic effects in the appendicular skeleton of OVX mice. Our results suggest that these effects might occur in part by modulating the Wnt pathway. These findings lend credence to the notion that the osteogenic action of PTHrP (107-139) is likely a consequence of its anti-resorptive and anabolic features, and further support the usefulness of PTHrP (1-36) as a bone anabolic peptide in the setting of estrogen-depletion.


Asunto(s)
Remodelación Ósea/efectos de los fármacos , Proteína Relacionada con la Hormona Paratiroidea/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Proteínas Adaptadoras Transductoras de Señales , Animales , Remodelación Ósea/genética , Resorción Ósea/genética , Resorción Ósea/prevención & control , Femenino , Glicoproteínas/genética , Inyecciones Subcutáneas , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Endogámicos C57BL , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Estrés Oxidativo , ARN Mensajero/genética , ARN Mensajero/metabolismo
12.
J Cell Physiol ; 227(5): 1980-7, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21732369

RESUMEN

Hypertrophy of human mesangial cells (HMC) is among the earliest characteristics in patients with diabetic nephropathy (DN). Recently, we observed the upregulation of parathyroid hormone (PTH)-related protein (PTHrP) in experimental DN, associated with renal hypertrophy. Herein, we first examined whether PTHrP was overexpressed in human DN, and next assessed the putative role of this protein on high glucose (HG)-induced HMC hypertrophy. As previously found in mice, kidneys from diabetic patients showed an increased tubular and glomerular immunostaining for PTHrP. In HMC, HG medium increased PTHrP protein expression associated with the development of hypertrophy as assessed by cell protein content. This effect was also induced by PTHrP(1-36). HG and PTHrP(1-36)-induced hypertrophy were associated with an increase in cyclin D1 and p27Kip1 protein expression, a decreased cyclin E expression, and the prevention of cyclin E/cdk2 complex activation. Both PTHrP neutralizing antiserum (α-PTHrP) and the PTH/PTHrP receptor antagonist (JB4250) were able to abolish HG induction of hypertrophy, the aforementioned changes in cell cycle proteins, and also TGF-ß1 up-regulation. Moreover, the capability of both HG and PTHrP(1-36) to induce HMC hypertrophy was abolished by α-TGFß1. These data show for the first time that PTHrP is upregulated in the kidney of patients with DN. Our findings also demonstrate that PTHrP acts as an important mediator of HG-induced HMC hypertrophy by modulating cell cycle regulatory proteins and TGF-ß1.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Riñón/metabolismo , Riñón/patología , Células Mesangiales/metabolismo , Células Mesangiales/patología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Humanos , Hipertrofia/metabolismo , Hipertrofia/patología , Riñón/citología , Masculino , Células Mesangiales/citología , Ratones , Persona de Mediana Edad , Proteína Relacionada con la Hormona Paratiroidea/genética , Factor de Crecimiento Transformador beta1/metabolismo
13.
Pharmaceutics ; 14(1)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35057097

RESUMEN

Throughout her impressive scientific career, Prof. María Vallet-Regí opened various research lines aimed at designing new bioceramics, including mesoporous bioactive glasses for bone tissue engineering applications. These bioactive glasses can be considered a spin-off of silica mesoporous materials because they are designed with a similar technical approach. Mesoporous glasses in addition to SiO2 contain significant amounts of other oxides, particularly CaO and P2O5 and therefore, they exhibit quite different properties and clinical applications than mesoporous silica compounds. Both materials exhibit ordered mesoporous structures with a very narrow pore size distribution that are achieved by using surfactants during their synthesis. The characteristics of mesoporous glasses made them suitable to be enriched with various osteogenic agents, namely inorganic ions and biopeptides as well as mesenchymal cells. In the present review, we summarize the evolution of mesoporous bioactive glasses research for bone repair, with a special highlight on the impact of Prof. María Vallet-Regí´s contribution to the field.

14.
J Biomed Biotechnol ; 2011: 290874, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21052497

RESUMEN

Parathyroid hormone- (PTH-) related protein (PTHrP) and its receptor, the PTH1 receptor (PTH1R), are widely expressed in the kidney, where PTHrP exerts a modulatory action on renal function. PTHrP is known to be upregulated in several experimental nephropathies such as acute renal failure (ARF), obstructive nephropathy (ON) as well as diabetic nephropathy (DN). In this paper, we will discuss the functional consequences of chronic PTHrP overexpression in the damaged kidney using a transgenic mouse strain overexpressing PTHrP in the renal proximal tubule. In both ARF and ON, PTHrP displays proinflammatory and profibrogenic actions including the induction of epithelia to mesenquima transition. Moreover, PTHrP participates in the mechanisms of renal hypertrophy as well as proteinuria in experimental DN. Angiotensin II (Ang II), a critical factor in the progression of renal injury, appears to be, at least in part, responsible for endogenous PTHrP upregulation in these pathophysiological settings. These findings provide novel insights into the well-known protective effects of Ang II antagonists in renal diseases, paving the way for new therapeutic approaches.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Renales/metabolismo , Ratones , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/metabolismo , Angiotensina II/antagonistas & inhibidores , Animales , Humanos , Hipertrofia/tratamiento farmacológico , Hipertrofia/metabolismo , Enfermedades Renales/tratamiento farmacológico , Ratones Transgénicos , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteinuria/tratamiento farmacológico , Proteinuria/metabolismo
15.
J Am Soc Nephrol ; 21(2): 237-48, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19959711

RESUMEN

Epithelial-mesenchymal transition (EMT) is an important process that contributes to renal fibrogenesis. TGF-beta1 and EGF stimulate EMT. Recent studies suggested that parathyroid hormone-related protein (PTHrP) promotes fibrogenesis in the damaged kidney, apparently dependent on its interaction with vascular endothelial growth factor (VEGF), but whether it also interacts with TGF-beta and EGF to modulate EMT is unknown. Here, PTHrP(1-36) increased TGF-beta1 in cultured tubuloepithelial cells and TGF-beta blockade inhibited PTHrP-induced EMT-related changes, including upregulation of alpha-smooth muscle actin and integrin-linked kinase, nuclear translocation of Snail, and downregulation of E-cadherin and zonula occludens-1. PTHrP(1-36) also induced EGF receptor (EGFR) activation; inhibition of protein kinase C and metalloproteases abrogated this activation. Inhibition of EGFR activation abolished these EMT-related changes, the activation of ERK1/2, and upregulation of TGF-beta1 and VEGF by PTHrP(1-36). Moreover, inhibition of ERK1/2 blocked EMT induced by either PTHrP(1-36), TGF-beta1, EGF, or VEGF. In vivo, obstruction of mouse kidneys led to changes consistent with EMT and upregulation of TGF-beta1 mRNA, p-EGFR protein, and PTHrP. Taken together, these data suggest that PTHrP, TGF-beta, EGF, and VEGF might cooperate through activation of ERK1/2 to induce EMT in renal tubuloepithelial cells.


Asunto(s)
Diferenciación Celular , Células Epiteliales/patología , Túbulos Renales/patología , Riñón/patología , Mesodermo/patología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/metabolismo , Receptores ErbB/metabolismo , Fibrosis , Humanos , Riñón/metabolismo , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Fragmentos de Péptidos/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Biomolecules ; 12(1)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-35053152

RESUMEN

The increase in fracture rates and/or problems associated with missing bones due to accidents or various pathologies generates socio-health problems with a very high impact. Tissue engineering aims to offer some kind of strategy to promote the repair of damaged tissue or its restoration as close as possible to the original tissue. Among the alternatives proposed by this specialty, the development of scaffolds obtained from recombinant proteins is of special importance. Furthermore, science and technology have advanced to obtain recombinant chimera's proteins. This review aims to offer a synthetic description of the latest and most outstanding advances made with these types of scaffolds, particularly emphasizing the main recombinant proteins that can be used to construct scaffolds in their own right, i.e., not only to impregnate them, but also to make scaffolds from their complex structure, with the purpose of being considered in bone regenerative medicine in the near future.


Asunto(s)
Huesos/metabolismo , Osteogénesis , Proteínas Recombinantes de Fusión , Medicina Regenerativa , Ingeniería de Tejidos , Andamios del Tejido/química , Humanos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo
17.
J Cell Physiol ; 225(2): 585-92, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20506394

RESUMEN

Glucagon-like peptide 1 (GLP-1) controls glucose metabolism in extrapancreatic tissues through receptors other than the pancreatic cAMP-linked GLP-1 receptor; also, GLP-1 induces an insulin- and PTH-independent bone anabolic action in insulin-resistant and type-2 diabetic rats. Here we searched for the presence and characteristics of GLP-1 receptors in osteoblastic MC3T3-E1 cells. [(125)I]-GLP-1 specific binding to MC3T3-E1 cells was time- and temperature-dependent, reaching maximal value at 30 min at 25 degrees C; in these conditions, [(125)I]-GLP-1 binding was dissociable, and displaced by GLP-1, partially by GLP-2, but not by exendin-4 (Ex-4), exendin-9 (Ex-9), glucagon or insulin; Scatchard analysis of the unlabeled GLP-1 data showed high and low affinity binding sites; cross-linking of GLP-1 binding revealed an estimated 70 kDa band, almost undetectable in the presence of 10(-6) M GLP-1. GLP-1, Ex-9, insulin or glucagon failed to modify cellular cAMP content, while GLP-2 and Ex-4 increased it. However, GLP-1 induced an immediate hydrolysis of glycosylphosphatidylinositols (GPIs) generating short-lived inositolphosphoglycans (IPGs), and an increase in phosphatidylinositol-3 kinase (PI3K) and mitogen activated protein kinase (MAPK) activities; Ex-4 also affected GPIs, but its action was delayed with respect to that of GLP-1. This incretin was found to decrease Runx2 but increased osteocalcin gene expression, without affecting that of osteoprotegerin or the canonical Wnt pathway activity in MC3T3-E1 cells which do not express the pancreatic GLP-1 receptor. Our data demonstrate for the first time that GLP-1 can directly and functionally interact with osteoblastic cells, possibly through a GPI/IPG-coupled receptor.


Asunto(s)
AMP Cíclico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Osteoblastos/metabolismo , Receptores de Glucagón/metabolismo , Células 3T3 , Adenilil Ciclasas/metabolismo , Animales , Colforsina/farmacología , Exenatida , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/farmacología , Péptido 2 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Humanos , Ratones , Osteoblastos/efectos de los fármacos , Hormona Paratiroidea/farmacología , Péptidos/farmacología , Unión Proteica , Ratas , Receptores de Glucagón/genética , Ponzoñas/farmacología
18.
J Cell Biochem ; 110(2): 304-10, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20225238

RESUMEN

The RANKL/RANK/OPG pathway is essential for bone remodeling regulation. Many hormones and cytokines are involved in regulating gene expression in most of the pathway components. Moreover, any deregulation of this pathway can alter bone metabolism, resulting in loss or gain of bone mass. Whether osteoblasts from osteoporotic and nonosteoporotic patients respond differently to cytokines is unknown. The aim of this study was to compare the effect of interleukin (IL)-1beta, proftaglandin E(2) (PGE(2)), and transforming growth factor-beta1 (TGF-beta1) treatments on OPG and RANKL gene expression in normal (n = 11) and osteoporotic (n = 8) primary osteoblasts. OPG and RANKL mRNA levels of primary human osteoblastic (hOB) cell cultures were assessed by real-time PCR. In all cultures, OPG mRNA increased significantly in response to IL-1beta treatment and decreased in response to TGF-beta1 whereas PGE(2) treatment had no effect. RANKL mRNA levels were significantly increased by all treatments. Differences in OPG and RANKL responses were observed between osteoporotic and nonosteoporotic hOB: in osteoporotic hOB, the OPG response to IL-1beta treatment was up to three times lower (P = 0.009), whereas that of RANKL response to TGF-beta1 was five times higher (P = 0.002) after 8 h of treatment, as compared with those in nonosteoporotic hOBs. In conclusion, osteoporotic hOB cells showed an anomalous response under cytokine stimulation, consistent with an enhanced osteoclastogenesis resulting in high levels of bone resorption.


Asunto(s)
Dinoprostona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-1beta/farmacología , Osteoblastos/efectos de los fármacos , Osteoporosis/genética , Osteoprotegerina/genética , Ligando RANK/genética , Factor de Crecimiento Transformador beta1/farmacología , Estudios de Casos y Controles , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Osteoblastos/citología , Osteoblastos/metabolismo , Reacción en Cadena de la Polimerasa
19.
Calcif Tissue Int ; 84(6): 453-61, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19219381

RESUMEN

It has been suggested that hormones released after nutrient absorption, such as glucose-dependent insulinotropic peptide (GIP) and glucagon-like peptide 2 (GLP-2), could be responsible for changes in bone resorption. However, information about the role of GLP-1 in this regard is scanty. Diabetes-related bone loss occurs as a consequence of poor control of glucose homeostasis, but the relationship between osteoporosis and type 2 diabetes remains unclear. Since GLP-1 is decreased in the latter condition, we evaluated some bone characteristics in streptozotocin-induced type 2 diabetic (T2D) and fructose-induced insulin-resistant (IR) rat models compared to normal (N) and the effect of GLP-1 or saline (control) treatment (3 days by osmotic pump). Blood was taken before and after treatment for plasma measurements; tibiae and femora were collected for gene expression of bone markers (RT-PCR) and structure (microCT) analysis. Compared to N, plasma glucose and insulin were, respectively, higher and lower in T2D; osteocalcin (OC) and tartrate-resistant alkaline phosphatase 5b were lower; phosphate in IR showed a tendency to be higher; PTH was not different in T2D and IR; all parameters were unchanged after GLP-1 infusion. Bone OC, osteoprotegerin (OPG) and RANKL mRNA were lower in T2D and IR; GLP-1 increased OC and OPG in all groups and RANKL in T2D. Compared to N, trabecular bone parameters showed an increased degree of anisotropy in T2D and IR, which was reduced after GLP-1. These findings show an insulin-independent anabolic effect of GLP-1 and suggest that GLP-1 could be a useful therapeutic agent for improving the deficient bone formation and bone structure associated with glucose intolerance.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Huesos/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Péptido 1 Similar al Glucagón/uso terapéutico , Resistencia a la Insulina , Fragmentos de Péptidos/uso terapéutico , Fosfatasa Ácida/sangre , Animales , Resorción Ósea/etiología , Resorción Ósea/patología , Huesos/efectos de los fármacos , Huesos/ultraestructura , Modelos Animales de Enfermedad , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Insulina/sangre , Isoenzimas/sangre , Masculino , Osteocalcina/sangre , Osteocalcina/metabolismo , Osteoprotegerina/metabolismo , Hormona Paratiroidea/sangre , Fragmentos de Péptidos/metabolismo , Ligando RANK/metabolismo , Ratas , Ratas Wistar , Fosfatasa Ácida Tartratorresistente
20.
Mol Oncol ; 13(4): 857-872, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30628165

RESUMEN

The potential involvement of type 2 diabetes mellitus (T2DM) as a risk factor for colon cancer (CC) has been previously reported. While several clinical studies show a higher incidence of CC and a lower survival rate in diabetics, others report no association. Our own experience indicates that diabetes does not seem to worsen the prognosis once the tumor is present. Despite this controversy, there are no wide-spectrum molecular studies that delve into the impact of T2DM-related mechanisms in colon carcinogenesis. Here, we present a transcriptomic and proteomic profiling of paired tumor and normal colon mucosa samples in a cohort of 42 CC patients, 23 of which have T2DM. We used gene set enrichment and network approaches to extract relevant pathways in diabetics, referenced them to current knowledge, and tested them using in vitro techniques. Through our transcriptomics approach, we identified an unexpected overlap of pathways overrepresented in diabetics compared to nondiabetics, in both tumor and normal mucosa, including diabetes-related metabolic and signaling processes. Proteomic approaches highlighted several cancer-related signaling routes in diabetics found only in normal mucosa, not in tumors. An integration of the transcriptome and proteome analyses suggested the deregulation of key pathways related to colon carcinogenesis which converged on tumor initiation axis TEAD/YAP-TAZ as a potential initiator of the process. In vitro studies confirmed upregulation of this pathway in nontumor colon cells under high-glucose conditions. In conclusion, T2DM associates with deregulation of cancer-related processes in normal colon mucosa adjacent to tissue which has undergone a malignant transformation. These data support that in diabetic patients, the local microenvironment in normal colon mucosa may be a factor driving field cancerization promoting carcinogenesis. Our results set a new framework to study links between diabetes and colon cancer, including a new role of the TEAD/YAP-TAZ complex as a potential driver.


Asunto(s)
Neoplasias del Colon/etiología , Neoplasias del Colon/genética , Diabetes Mellitus Tipo 2/complicaciones , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Estudios de Cohortes , Femenino , Regulación Neoplásica de la Expresión Génica , Genómica , Glucosa/metabolismo , Humanos , Hiperglucemia/complicaciones , Mucosa Intestinal/patología , Masculino , Ratones Desnudos , Transducción de Señal/genética , Transcriptoma/genética , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA