Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Carmen Herencia is active.

Publication


Featured researches published by Carmen Herencia.


PLOS ONE | 2014

Magnesium Inhibits Wnt/β-Catenin Activity and Reverses the Osteogenic Transformation of Vascular Smooth Muscle Cells

Addy Montes de Oca; Fatima Guerrero; Julio M. Martínez-Moreno; Juan Antonio Madueño; Carmen Herencia; Alan Peralta; Yolanda Almaden; Ignacio González López; Escolastico Aguilera-Tejero; Kristina Gundlach; Janine Büchel; Mirjam E. Peter; Jutta Passlick-Deetjen; Mariano Rodriguez; Juan R. Muñoz-Castañeda

Magnesium reduces vascular smooth muscle cell (VSMC) calcification in vitro but the mechanism has not been revealed so far. This work used only slightly increased magnesium levels and aimed at determining: a) whether inhibition of magnesium transport into the cell influences VSMC calcification, b) whether Wnt/β-catenin signaling, a key mediator of osteogenic differentiation, is modified by magnesium and c) whether magnesium can influence already established vascular calcification. Human VSMC incubated with high phosphate (3.3 mM) and moderately elevated magnesium (1.4 mM) significantly reduced VSMC calcification and expression of the osteogenic transcription factors Cbfa-1 and osterix, and up-regulated expression of the natural calcification inhibitors matrix Gla protein (MGP) and osteoprotegerin (OPG). The protective effects of magnesium on calcification and expression of osteogenic markers were no longer observed in VSMC cultured with an inhibitor of cellular magnesium transport (2-aminoethoxy-diphenylborate [2-APB]). High phosphate induced activation of Wnt/β-catenin pathway as demonstrated by the translocation of β-catenin into the nucleus, increased expression of the frizzled-3 gene, and downregulation of Dkk-1 gene, a specific antagonist of the Wnt/β-catenin signaling pathway. The addition of magnesium however inhibited phosphate-induced activation of Wnt/β-catenin signaling pathway. Furthermore, TRPM7 silencing using siRNA resulted in activation of Wnt/β-catenin signaling pathway. Additional experiments were performed to test the ability of magnesium to halt the progression of already established VSMC calcification in vitro. The delayed addition of magnesium decreased calcium content, down-regulated Cbfa-1 and osterix and up-regulated MGP and OPG, when compared with a control group. This effect was not observed when 2-APB was added. In conclusion, magnesium transport through the cell membrane is important to inhibit VSMC calcification in vitro. Inhibition of Wnt/β-catenin by magnesium is one potential intracellular mechanism by which this anti-calcifying effect is achieved.


American Journal of Physiology-renal Physiology | 2012

In vascular smooth muscle cells paricalcitol prevents phosphate-induced Wnt/β-catenin activation

Julio M. Martínez-Moreno; Juan R. Muñoz-Castañeda; Carmen Herencia; Addy Montes de Oca; J.C. Estepa; Rocio Canalejo; Maria E. Rodriguez-Ortiz; Pablo Perez-Martinez; Escolastico Aguilera-Tejero; Antonio Canalejo; Mariano Rodriguez; Yolanda Almaden

The present study investigates the differential effect of two vitamin D receptor agonists, calcitriol and paricalcitol, on human aortic smooth muscle cells calcification in vitro. Human vascular smooth muscle cells were incubated in a high phosphate (HP) medium alone or supplemented with either calcitriol 10(-8)M (HP + CTR) or paricalcitol 3·10(-8) M (HP + PC). HP medium induced calcification, which was associated with the upregulation of mRNA expression of osteogenic factors such as bone morphogenetic protein 2 (BMP2), Runx2/Cbfa1, Msx2, and osteocalcin. In these cells, activation of Wnt/β-catenin signaling was evidenced by the translocation of β-catenin into the nucleus and the increase in the expression of direct target genes as cyclin D1, axin 2, and VCAN/versican. Addition of calcitriol to HP medium (HP + CTR) further increased calcification and also enhanced the expression of osteogenic factors together with a significant elevation of nuclear β-catenin levels and the expression of cyclin D1, axin 2, and VCAN. By contrast, the addition of paricalcitol (HP + PC) not only reduced calcification but also downregulated the expression of BMP2 and other osteoblastic phenotype markers as well as the levels of nuclear β-catenin and the expression of its target genes. The role of Wnt/β-catenin on phosphate- and calcitriol-induced calcification was further demonstrated by the inhibition of calcification after addition of Dickkopf-related protein 1 (DKK-1), a specific natural antagonist of the Wnt/β-catenin signaling pathway. In conclusion, the differential effect of calcitriol and paricalcitol on vascular calcification appears to be mediated by a distinct regulation of the BMP and Wnt/β-catenin signaling pathways.


Nephrology Dialysis Transplantation | 2014

Magnesium modulates parathyroid hormone secretion and upregulates parathyroid receptor expression at moderately low calcium concentration

Maria E. Rodriguez-Ortiz; Antonio Canalejo; Carmen Herencia; Julio M. Martínez-Moreno; Alan Peralta-Ramírez; Pablo Perez-Martinez; Juan F. Navarro-González; Mariano Rodriguez; Mirjam E. Peter; Kristina Gundlach; Sonja Steppan; Jutta Passlick-Deetjen; Juan R. Muñoz-Castañeda; Yolanda Almaden

Background The interest on magnesium (Mg) has grown since clinical studies have shown the efficacy of Mg-containing phosphate binders. However, some concern has arisen for the potential effect of increased serum Mg on parathyroid hormone (PTH) secretion. Our objective was to evaluate the direct effect of Mg in the regulation of the parathyroid function; specifically, PTH secretion and the expression of parathyroid cell receptors: CaR, the vitamin D receptor (VDR) and FGFR1/Klotho. Methods The work was performed in vitro by incubating intact rat parathyroid glands in different calcium (Ca) and Mg concentrations. Results Increasing Mg concentrations from 0.5 to 2 mM produced a left shift of PTH–Ca curves. With Mg 5 mM, the secretory response was practically abolished. Mg was able to reduce PTH only if parathyroid glands were exposed to moderately low Ca concentrations; with normal–high Ca concentrations, the effect of Mg on PTH inhibition was minor or absent. After 6-h incubation at a Ca concentration of 1.0 mM, the expression of parathyroid CaR, VDR, FGFR1 and Klotho (at mRNA and protein levels) was increased with a Mg concentration of 2.0 when compared with 0.5 mM. Conclusions Mg reduces PTH secretion mainly when a moderate low calcium concentration is present; Mg also modulates parathyroid glands function through upregulation of the key cellular receptors CaR, VDR and FGF23/Klotho system.


PLOS ONE | 2012

Nuclear Translocation of β-Catenin during Mesenchymal Stem Cells Differentiation into Hepatocytes Is Associated with a Tumoral Phenotype

Carmen Herencia; Julio M. Martínez-Moreno; Concepción Herrera; Fernando J. Corrales; Raquel Santiago-Mora; Isabel Espejo; Monserrat Barco; Yolanda Almaden; Manuel de la Mata; Antonio Rodríguez-Ariza; Juan R. Muñoz-Castañeda

Wnt/β-catenin pathway controls biochemical processes related to cell differentiation. In committed cells the alteration of this pathway has been associated with tumors as hepatocellular carcinoma or hepatoblastoma. The present study evaluated the role of Wnt/β-catenin activation during human mesenchymal stem cells differentiation into hepatocytes. The differentiation to hepatocytes was achieved by the addition of two different conditioned media. In one of them, β-catenin nuclear translocation, up-regulation of genes related to the Wnt/β-catenin pathway, such as Lrp5 and Fzd3, as well as the oncogenes c-myc and p53 were observed. While in the other protocol there was a Wnt/β-catenin inactivation. Hepatocytes with nuclear translocation of β-catenin also had abnormal cellular proliferation, and expressed membrane proteins involved in hepatocellular carcinoma, metastatic behavior and cancer stem cells. Further, these cells had also increased auto-renewal capability as shown in spheroids formation assay. Comparison of both differentiation protocols by 2D-DIGE proteomic analysis revealed differential expression of 11 proteins with altered expression in hepatocellular carcinoma. Cathepsin B and D, adenine phosphoribosyltransferase, triosephosphate isomerase, inorganic pyrophosphatase, peptidyl-prolyl cis-trans isomerase A or lactate dehydrogenase β-chain were up-regulated only with the protocol associated with Wnt signaling activation while other proteins involved in tumor suppression, such as transgelin or tropomyosin β-chain were down-regulated in this protocol. In conclusion, our results suggest that activation of the Wnt/β-catenin pathway during human mesenchymal stem cells differentiation into hepatocytes is associated with a tumoral phenotype.


PLOS ONE | 2014

TGF-β Prevents Phosphate-Induced Osteogenesis through Inhibition of BMP and Wnt/β-Catenin Pathways

Fatima Guerrero; Carmen Herencia; Yolanda Almaden; Julio M. Martínez-Moreno; Addy Montes de Oca; Maria E. Rodriguez-Ortiz; Juan M. Díaz-Tocados; Antonio Canalejo; Monica Florio; Ignacio González López; William G. Richards; Mariano Rodriguez; Escolastico Aguilera-Tejero; Juan R. Muñoz-Castañeda

Background Transforming growth factor-β (TGF-β) is a key cytokine during differentiation of mesenchymal stem cells (MSC) into vascular smooth muscle cells (VSMC). High phosphate induces a phenotypic transformation of vascular smooth muscle cells (VSMC) into osteogenic-like cells. This study was aimed to evaluate signaling pathways involved during VSMC differentiation of MSC in presence or not of high phosphate. Results Our results showed that TGF-β induced nuclear translocation of Smad3 as well as the expression of vascular smooth muscle markers, such as smooth muscle alpha actin, SM22α, myocardin, and smooth muscle-myosin heavy chain. The addition of high phosphate to MSC promoted nuclear translocation of Smad1/5/8 and the activation of canonical Wnt/β-catenin in addition to an increase in BMP-2 expression, calcium deposition and alkaline phosphatase activity. The administration of TGF-β to MSC treated with high phosphate abolished all these effects by inhibiting canonical Wnt, BMP and TGF-β pathways. A similar outcome was observed in high phosphate-treated cells after the inhibition of canonical Wnt signaling with Dkk-1. Conversely, addition of both Wnt/β-catenin activators CHIR98014 and lithium chloride enhanced the effect of high phosphate on BMP-2, calcium deposition and alkaline phosphatase activity. Conclusions Full VSMC differentiation induced by TGF-β may not be achieved when extracellular phosphate levels are high. Moreover, TGF-β prevents high phosphate-induced osteogenesis by decreasing the nuclear translocation of Smad 1/5/8 and avoiding the activation of Wnt/β-catenin pathway.


The FASEB Journal | 2016

Vitamin D modulates tissue factor and protease-activated receptor 2 expression in vascular smooth muscle cells

Julio M. Martínez-Moreno; Carmen Herencia; Addy Montes de Oca; Juan R. Muñoz-Castañeda; M. Encarnación Rodríguez-Ortiz; Juan M. Díaz-Tocados; Esther Peralbo-Santaella; Antonio Camargo; Antonio Canalejo; Mariano Rodriguez; Francisco Velasco-Gimena; Yolanda Almaden

Clinical and epidemiologic studies reveal an association between vitamin D deficiency and increased risk of cardiovascular disease. Because vascular smooth muscle cell (VSMC)‐derived tissue factor (TF) is suggested to be critical for arterial thrombosis, we investigated whether the vitamin D molecules calcitriol and paricalcitol could reduce the expression of TF induced by the proinflammatory cytokine TNF‐α in human aortic VSMCs. We found that, compared with controls, incubation with TNF‐α increased TF expression and procoagulant activity in a NF‐κB‐dependent manner, as deduced from the increased nuclear translocation of nuclear factor κ‐light‐chain‐enhancer of activated B cells protein 65 (p65‐NF‐κB) and direct interaction of NF‐kB to the TF promoter. This was accompanied by the up‐regulation of TF signaling mediator protease‐activated receptor 2 (PAR‐2) expression and by the down‐regulation of vitamin D receptor expression in a miR‐346‐dependent way. However, addition of calcitriol or paricalcitol blunted the TNF‐α‐induced TF expression and activity (2.01 ± 0.24 and 1.32 ± 0.14 vs. 3.02 ± 0.39 pmol/mg protein, P < 0.05), which was associated with down‐regulation of NF‐kB signaling and PAR‐2 expression, as well as with restored levels of vitamin D receptor and enhanced expression of TF pathway inhibitor. Our data suggest that inflammation promotes a prothrombotic state through the up‐regulation of TF function in VSMCs and that the beneficial cardiovascular effects of vitamin D may be partially due to decreases in TF expression and its activity in VSMCs.—Martinez‐Moreno, J. M., Herencia, C., Montes de Oca, A., Muñoz‐Castañeda, J. R., Rodríguez‐Ortiz, M. E., Díaz‐Tocados, J. M., Peralbo‐Santaella, E., Camargo, A., Canalejo, A., Rodriguez, M., Velasco‐Gimena, F., Almaden, Y., Vitamin D modulates tissue factor and protease‐activated receptor 2 expression in vascular smooth muscle cells. FASEB J. 30, 1367–1376 (2016). www.fasebj.org


European Journal of Clinical Investigation | 2015

Angiotensin II prevents calcification in vascular smooth muscle cells by enhancing magnesium influx.

Carmen Herencia; M. Encarnación Rodríguez-Ortiz; Juan R. Muñoz-Castañeda; Julio M. Martínez-Moreno; Rocio Canalejo; Addy Montes de Oca; Juan M. Díaz-Tocados; Esther Peralbo-Santaella; Carmen Marin; Antonio Canalejo; Mariano Rodriguez; Yolanda Almaden

Vascular calcification (VC) is highly prevalent in patients with chronic kidney disease (CKD). Low magnesium levels are associated with VC, and recent in vitro studies confirm a protective role of magnesium, which is mediated by its entry into the VSMCs through the Transient Receptor Potential Melastatin 7 (TRPM7) channel. The role of Angiotensin II (Ang II) on VC is still unclear. As Ang II is able to stimulate TRPM7 activity, we hypothesize that it might prevent VC. Thus, the aim of this study was to dissect the direct effect of Ang II on VC.


The FASEB Journal | 2017

Differential regulation of renal Klotho and FGFR1 in normal and uremic rats.

Juan R. Muñoz-Castañeda; Carmen Herencia; María Pendón-Ruiz de Mier; Maria E. Rodriguez-Ortiz; Juan M. Díaz-Tocados; Noemi Vergara; Julio M. Martínez-Moreno; Maria Dolores Salmerón; William G. Richards; Arnold J. Felsenfeld; Makoto Kuro-o; Yolanda Almaden; Mariano Rodriguez

In renal failure, hyperphosphatemia occurs despite a marked elevation in serum fibroblast growth factor (FGF)‐23. Abnormal regulation of the FGFR1‐Klotho receptor complex may cause a resistance to the phosphaturic action of FGF23. The purpose of the present study was to investigate the regulation of renal Klotho and FGF receptor (FEFR)‐1 in healthy and uremic rats induced by 5/6 nephrectomy. In normal rats, the infusion of rat recombinant FGF23 enhanced phosphaturia and increased renal FGFR1 expression; however, Klotho expression was reduced. Uremic rats on a highphosphate (HP) diet presented hyperphosphatemia with marked elevation of FGF23 and an increased fractional excretion of phosphate (P) that was associated with a marked reduction of Klotho expression and an increase in FGFR1. After neutralization of FGF23 by anti‐FGF23 administration, phosphaturia was still abundant, Klotho expression remained low, and the FGFR1 level was reduced. These results suggest that the expression of renal Klotho is modulated by phosphaturia, whereas the FGFR1 expression is regulated by FGF23. Calcitriol (CTR) administration prevented a decrease in renal Klotho expression. In HEK293 cells HP produced nuclear translocation of β‐cateniv together with a reduction in Klotho. Wnt/β‐catenin inhibition with Dkk‐1 prevented the P‐induced down‐regulation of Klotho. The addition of CTR to HP medium was able to recover Klotho expression. In summary, high FGF23 levels increase FGFR1, whereas phosphaturia decreases Klotho expression through the activation of Wnt/β‐catenin pathway.—Muñoz‐Castañeda, J. R., Herencia, C., Pendón‐Ruiz de Mier M. V., Rodriguez‐Ortiz, M. E., Diaz‐Tocados, J. M., Vergara, N., Martínez‐Moreno, J. M., Salmerón M. D., Richards, W. G., Felsenfeld, A., Kuro‐O, M., Almadén Y., Rodríguez, M. Differential regulation of renal Klotho and FGFR1 in normal and uremic rats. FASEB J. 31, 3858–3867 (2017). www.fasebj.org—Muñoz‐Castañeda, Juan R., Herencia, Carmen, Pendón‐Ruiz de Mier, Maria Victoria, Rodriguez‐Ortiz, Maria Encarnación, Diaz‐Tocados, Juan M., Vergara, Noemi, Martínez‐Moreno, Julio M., Salmerón, Maria Dolores, Richards, William G., Felsenfeld, Arnold, Kuro‐O, Makoto, Almadén, Yolanda, Rodríguez, Mariano Differential regulation of renal Klotho and FGFR1 in normal and uremic rats. FASEB J. 31, 3858–3867 (2017)


PLOS ONE | 2016

Procaine Inhibits Osteo/Odontogenesis through Wnt/β-Catenin Inactivation

Carmen Herencia; Juan M. Díaz-Tocados; Lidia Jurado; Addy Montes de Oca; Maria E. Rodriguez-Ortiz; Carmen Martín-Alonso; Julio M. Martínez-Moreno; Noemi Vergara; Mariano Rodriguez; Yolanda Almaden; Juan R. Muñoz-Castañeda

Introduction Periodontitis is a complex pathology characterized by the loss of alveolar bone. The causes and the mechanisms that promote this bone resorption still remain unknown. The knowledge of the critical regulators involved in the alteration of alveolar bone homeostasis is of great importance for developing molecular therapies. Procaine is an anesthetic drug with demethylant properties, mainly used by dentists in oral surgeries. The inhibitor role of Wnt signaling of procaine was described in vitro in colon cancer cells. Methods In this work we evaluated the role of procaine (1 uM) in osteo/odontogenesis of rat bone marrow mesenchymal stem cells. Similarly, the mechanisms whereby procaine achieves these effects were also studied. Results Procaine administration led to a drastic decrease of calcium content, alkaline phosphatase activity, alizarin red staining and an increase in the expression of Matrix Gla Protein. With respect to osteo/odontogenic markers, procaine decreased early and mature osteo/odontogenic markers. In parallel, procaine inhibited canonical Wnt/β-catenin pathway, observing a loss of nuclear β-catenin, a decrease in Lrp5 and Frizzled 3, a significant increase of sclerostin and Gsk3β and an increase of phosphorylated β-catenin. The combination of osteo/odontogenic stimuli and Lithium Chloride decreased mRNA expression of Gsk3β, recovered by Procaine. Furthermore it was proved that Procaine alone dose dependently increases the expression of Gsk3β and β-catenin phosphorylation. These effects of procaine were also observed on mature osteoblast. Interestingly, at this concentration of procaine no demethylant effects were observed. Conclusions Our results demonstrated that procaine administration drastically reduced the mineralization and osteo/odontogenesis of bone marrow mesenchymal stem cells inhibiting Wnt/β-catenin pathway through the increase of Gsk3β expression and β-catenin phosphorylation.


Regenerative Medicine | 2015

Human mesenchymal stromal cell lysates as a novel strategy to recover liver function

Carmen Herencia; Yolanda Almaden; Julio M. Martínez-Moreno; Isabel Espejo; Concha Herrera; C. Perez-Sanchez; Fatima Guerrero; Rubén Ciria; Francisco Javier Briceño; Gustavo Ferrín; Manuel de la Mata; Juan R. Muñoz-Castañeda

AIM It is unknown if the beneficial effects of mesenchymal stromal cells (MSC) transplantation into the liver are dependent on their anchorage and differentiation into hepatocytes or rather the result of the release of stem cell intracellular content with hepatoprotector properties. MATERIALS & METHODS The effects of intact MSC transplantation were compared with the infusion of MSC lysates in an experimental rat model of acute liver failure. RESULTS A more powerful hepatoprotective and antiapoptotic effect was obtained after infusion of MSC lysates than intact MSC. Changes in IL-6 levels and miRNAs might explain the beneficial effects of MSC lysates. CONCLUSION Infusion of MSC lysates show a better hepatoprotective effect than the transplantation of intact MSC.

Collaboration


Dive into the Carmen Herencia's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yolanda Almaden

University of Córdoba (Spain)

View shared research outputs
Top Co-Authors

Avatar

Yolanda Almaden

University of Córdoba (Spain)

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rubén Ciria

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge