Network


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

Hotspot


Dive into the research topics where Maria C. Izquierdo is active.

Publication


Featured researches published by Maria C. Izquierdo.


Journal of The American Society of Nephrology | 2011

The Inflammatory Cytokines TWEAK and TNFα Reduce Renal Klotho Expression through NFκB

Juan Antonio Moreno; Maria C. Izquierdo; Maria Dolores Sanchez-Niño; Beatriz Suarez-Alvarez; Carlos López-Larrea; Aniela Jakubowski; Julià Blanco; Rafael Ramírez; Rafael Selgas; Marta Ruiz-Ortega; Jesús Egido; Alberto Ortiz; Ana Belen Sanz

Proinflammatory cytokines contribute to renal injury, but the downstream effectors within kidney cells are not well understood. One candidate effector is Klotho, a protein expressed by renal cells that has antiaging properties; Klotho-deficient mice have an accelerated aging-like phenotype, including vascular injury and renal injury. Whether proinflammatory cytokines, such as TNF and TNF-like weak inducer of apoptosis (TWEAK), modulate Klotho is unknown. In mice, exogenous administration of TWEAK decreased expression of Klotho in the kidney. In the setting of acute kidney injury induced by folic acid, the blockade or absence of TWEAK abrogated the injury-related decrease in renal and plasma Klotho levels. TWEAK, TNFα, and siRNA-mediated knockdown of IκBα all activated NFκB and reduced Klotho expression in the MCT tubular cell line. Furthermore, inhibition of NFκB with parthenolide prevented TWEAK- or TNFα-induced downregulation of Klotho. Inhibition of histone deacetylase reversed TWEAK-induced downregulation of Klotho, and chromatin immunoprecipitation showed that TWEAK promotes RelA binding to the Klotho promoter, inducing its deacetylation. In conclusion, inflammatory cytokines, such as TWEAK and TNFα, downregulate Klotho expression through an NFκB-dependent mechanism. These results may partially explain the relationship between inflammation and diseases characterized by accelerated aging of organs, including CKD.


Aids Research and Treatment | 2011

Tenofovir Nephrotoxicity: 2011 Update

Beatriz Fernandez-Fernandez; Ana Montoya-Ferrer; Ana Belen Sanz; Maria Dolores Sanchez-Niño; Maria C. Izquierdo; Jonay Poveda; Valeria Sainz-Prestel; Natalia Ortíz-Martín; Alejandro Parra-Rodríguez; Rafael Selgas; Marta Ruiz-Ortega; Jesús Egido; Alberto Ortiz

Tenofovir is an acyclic nucleotide analogue reverse-transcriptase inhibitor structurally similar to the nephrotoxic drugs adefovir and cidofovir. Tenofovir is widely used to treat HIV infection and approved for treatment of hepatitis B virus. Despite initial cell culture and clinical trials results supporting the renal safety of tenofovir, its clinical use is associated with a low, albeit significant, risk of kidney injury. Proximal tubular cell secretion of tenofovir explains the accumulation of the drug in these mitochondria-rich cells. Tenofovir nephrotoxicity is characterized by proximal tubular cell dysfunction that may be associated with acute kidney injury or chronic kidney disease. Withdrawal of the drug leads to improvement of analytical parameters that may be partial. Understanding the risk factors for nephrotoxicity and regular monitoring of proximal tubular dysfunction and serum creatinine in high-risk patients is required to minimize nephrotoxicity. Newer, structurally similar molecular derivatives that do not accumulate in proximal tubules are under study.


PLOS ONE | 2010

TWEAK Activates the Non-Canonical NFκB Pathway in Murine Renal Tubular Cells: Modulation of CCL21

Ana B. Sanz; Maria Dolores Sanchez-Niño; Maria C. Izquierdo; Aniela Jakubowski; Pilar Justo; Luis Miguel Blanco-Colio; Marta Ruiz-Ortega; Rafael Selgas; Jesús Egido; Alberto Ortiz

TWEAK is a member of the TNF superfamily of cytokines that contribute to kidney tubulointerstitial injury. It has previously been reported that TWEAK induces transient nuclear translocation of RelA and expression of RelA-dependent cytokines in renal tubular cells. Additionally, TWEAK induced long-lasting NFκB activation suggestive of engagement of the non-canonical NFκB pathway. We now explore TWEAK-induced activation of NFκB2 and RelB, as well as expression of CCL21, a T-cell chemotactic factor, in cultured murine tubular epithelial cells and in healthy kidneys in vivo. In cultured tubular cells, TWEAK and TNFα activated different DNA-binding NFκB complexes. TWEAK-induced sustained NFκB activation was associated with NFκB2 p100 processing to p52 via proteasome and nuclear translocation and DNA-binding of p52 and RelB. TWEAK, but not TNFα used as control), induced a delayed increase in CCL21a mRNA (3.5±1.22-fold over control) and CCL21 protein (2.5±0.8-fold over control), which was prevented by inhibition of the proteasome, or siRNA targeting of NIK or RelB, but not by RelA inhibition with parthenolide. A second NFκB2-dependent chemokine, CCL19, was upregulates by TWEAK, but not by TNFα. However, both cytokines promoted chemokine RANTES expression (3-fold mRNA at 24 h). In vivo, TWEAK induced nuclear NFκB2 and RelB translocation and CCL21a mRNA (1.5±0.3-fold over control) and CCL21 protein (1.6±0.5-fold over control) expression in normal kidney. Increased tubular nuclear RelB and tubular CCL21 expression in acute kidney injury were decreased by neutralization (2±0.9 vs 1.3±0.6-fold over healthy control) or deficiency of TWEAK (2±0.9 vs 0.8±0.6-fold over healthy control). Moreover, anti-TWEAK treatment prevented the recruitment of T cells to the kidney in this model (4.1±1.4 vs 1.8±1-fold over healthy control). Our results thus identify TWEAK as a regulator of non-canonical NFκB activation and CCL21 expression in tubular cells thus promoting lymphocyte recruitment to the kidney during acute injury.


Journal of Cellular and Molecular Medicine | 2009

Tweak induces proliferation in renal tubular epithelium: a role in uninephrectomy induced renal hyperplasia

Ana Belen Sanz; Maria Dolores Sanchez-Niño; Maria C. Izquierdo; Aniela Jakubowski; Pilar Justo; Luis Miguel Blanco-Colio; Marta Ruiz-Ortega; Jesús Egido; Alberto Ortiz

The tumour necrosis factor (TNF) family member TWEAK activates the Fn14 receptor and has pro‐apoptotic, proliferative and pro‐inflammatory actions that depend on the cell type and the microenvironment. We explored the proliferative actions of TWEAK on cultured tubular cells and in vivo on renal tubules. Additionally, we studied the role of TWEAK in compensatory proliferation following unilateral nephrectomy and in an inflammatory model of acute kidney injury (AKI) induced by a folic acid overdose. TWEAK increased the proliferation, cell number and cyclin D1 expression of cultured tubular cells, in vitro. Exposure to serum increased TWEAK and Fn14 expression and the proliferative response to TWEAK. TWEAK activated the mitogen‐activated protein kinases ERK and p38, the phosphatidyl‐inositol 3‐kinase (PI3K)/Akt pathway and NF‐κB. TWEAK‐induced proliferation was prevented by inhibitors of these protein kinases and by the NF‐κB inhibitor parthenolide. TWEAK‐induced tubular cell proliferation as assessed by PCNA and cyclin D1 expression in the kidneys of adult healthy mice in vivo. By contrast, TWEAK knock‐out mice displayed lower tubular cell proliferation in the remnant kidney following unilateral nephrectomy, a non‐inflammatory model. This is consistent with TWEAK‐induced proliferation on cultured tubular cells in the absence of inflammatory cytokines. Consistent with our previously published data, in the presence of inflammatory cytokines TWEAK promoted apoptosis, not proliferation, of cultured tubular cells. In this regard, TWEAK knock‐out mice with AKI displayed less tubular apoptosis and proliferation, as well as improved renal function. In conclusion, TWEAK actions in tubular cells are context dependent. In a non‐inflammatory milieu TWEAK induces proliferation of tubular epithelium. This may be relevant for compensatory renal hyperplasia following nephrectomy.


International Urology and Nephrology | 2014

Unilateral ureteral obstruction: beyond obstruction

Alvaro C. Ucero; Alberto Benito-Martin; Maria C. Izquierdo; Maria Dolores Sanchez-Niño; Ana Belen Sanz; Adrián M. Ramos; Sergio Berzal; Marta Ruiz-Ortega; Jesús Egido; Alberto Ortiz

Unilateral ureteral obstruction is a popular experimental model of renal injury. However, the study of the kidney response to urinary tract obstruction is only one of several advantages of this model. Unilateral ureteral obstruction causes subacute renal injury characterized by tubular cell injury, interstitial inflammation and fibrosis. For this reason, it serves as a model both of irreversible acute kidney injury and of events taking place during human chronic kidney disease. Being a unilateral disease, it is not useful to study changes in global kidney function, but has the advantage of a low mortality and the availability of an internal control (the non-obstructed kidney). Experimental unilateral ureteral obstruction has illustrated the molecular mechanisms of apoptosis, inflammation and fibrosis, all three key processes in kidney injury of any cause, thus providing information beyond obstruction. Recently this model has supported key concepts on the role in kidney fibrosis of epithelial–mesenchymal transition, tubular epithelial cell G2/M arrest, the anti-aging hormone Klotho and renal innervation. We now review the experimental model and its contribution to identifying novel therapeutic targets in kidney injury and fibrosis, independently of the noxa.


Mediators of Inflammation | 2010

TNF Superfamily: A Growing Saga of Kidney Injury Modulators

Maria Dolores Sanchez-Niño; Alberto Benito-Martin; Sara Gonçalves; Ana Belen Sanz; Alvaro C. Ucero; Maria C. Izquierdo; Adrián M. Ramos; Sergio Berzal; Rafael Selgas; Marta Ruiz-Ortega; Jesús Egido; Alberto Ortiz

Members of the TNF superfamily participate in kidney disease. Tumor necrosis factor (TNF) and Fas ligand regulate renal cell survival and inflammation, and therapeutic targeting improves the outcome of experimental renal injury. TNF-related apoptosis-inducing ligand (TRAIL and its potential decoy receptor osteoprotegerin are the two most upregulated death-related genes in human diabetic nephropathy. TRAIL activates NF-kappaB in tubular cells and promotes apoptosis in tubular cells and podocytes, especially in a high-glucose environment. By contrast, osteoprotegerin plays a protective role against TRAIL-induced apoptosis. Another family member, TNF-like weak inducer of apoptosis (TWEAK induces inflammation and tubular cell death or proliferation, depending on the microenvironment. While TNF only activates canonical NF-kappaB signaling, TWEAK promotes both canonical and noncanonical NF-kappaB activation in tubular cells, regulating different inflammatory responses. TWEAK promotes the secretion of MCP-1 and RANTES through NF-kappaB RelA-containing complexes and upregulates CCl21 and CCL19 expression through NF-kappaB inducing kinase (NIK-) dependent RelB/NF-kappaB2 complexes. In vivo TWEAK promotes postnephrectomy compensatory renal cell proliferation in a noninflammatory milieu. However, in the inflammatory milieu of acute kidney injury, TWEAK promotes tubular cell death and inflammation. Therapeutic targeting of TNF superfamily cytokines, including multipronged approaches targeting several cytokines should be further explored.


Nephrology Dialysis Transplantation | 2012

Klotho, phosphate and inflammation/ageing in chronic kidney disease

Maria C. Izquierdo; Maria Vanessa Perez-Gomez; Maria Dolores Sanchez-Niño; Ana Belen Sanz; Olga Ruiz-Andres; Jonay Poveda; Juan Antonio Moreno; Jesús Egido; Alberto Ortiz

Evidence is emerging for the inflammatory nature of many ageing-associated diseases, including atherosclerosis, vascular calcification, diabetes and chronic kidney disease (CKD), among others. Ageing itself results in chronic low-grade inflammation that promotes end-organ damage. Inflammatory organ damage, in turn, may contribute to inflammation. Recent research has identified the kidney-secreted hormone Klotho as a central player at the ageing-inflammation interface. Thus, systemic or local renal inflammation decreases kidney Klotho expression. Klotho down-regulation may be induced by specific cytokines such as tumour necrosis factor-α or TWEAK through the canonical activation of the inflammatory transcription factor nuclear factor kappa B (NFκB) and, specifically RelA. In addition, inflammatory cytokines lead to the epigenetic inactivation of Klotho transcription. Klotho itself has antioxidant and anti-inflammatory properties and the canonical NFκB component RelA is one of its targets. Klotho is a key regulator of phosphate balance and a role of phosphate in ageing has been shown. However, the potential relationship between phosphate and inflammation requires further clarification. A correct understanding of these interactions may lead to the design of novel therapeutic approaches to CKD and CKD-related inflammatory and ageing features as well as to inflammation/ageing in general.


Kidney International | 2012

TWEAK (tumor necrosis factor-like weak inducer of apoptosis) activates CXCL16 expression during renal tubulointerstitial inflammation

Maria C. Izquierdo; Ana Belen Sanz; Sergio Mezzano; Julià Blanco; Susana Carrasco; Maria Dolores Sanchez-Niño; Alberto Benito-Martin; Marta Ruiz-Ortega; Jesús Egido; Alberto Ortiz

TWEAK (tumor necrosis factor-like weak inducer of apoptosis) is a TNF superfamily cytokine that activates the fibroblast growth factor-inducible 14 (Fn14) receptor. Transcriptional analysis of experimental kidney tubulointerstitial inflammation showed a correlation between an upregulation of the mRNA for the transmembrane chemokine CXCL16, a T-cell chemoattractant, and Fn14 activation. Exogenous TWEAK increased mouse kidney CXCL16 expression and T-lymphocyte infiltration in vivo, processes inhibited by the NF-κB inhibitor parthenolide. Tubular cell CXCL16 was increased in a nephrotoxic tubulointerstitial inflammation model and neutralizing anti-TWEAK antibodies decreased this CXCL16 expression and lymphocyte infiltration. In human kidney biopsies with tubulointerstitial inflammation, tubular cell CXCL16 and Fn14 expressions were associated with inflammatory infiltrates. TWEAK upregulated CXCL16 mRNA expression in cultured renal tubular cells in an NF-κB-dependent manner and increased soluble and cellular CXCL16 protein. CXCL16 modestly promoted the expression of cytokines in tubular cells expressing its receptor (CXCR6) and appeared to synergize with TWEAK to promote an inflammatory response; however, it did not modulate tubular cell proliferation or survival. Thus, TWEAK upregulates the expression of the chemokine CXCL16 in tubular epithelium and this may contribute to kidney tubulointerstitial inflammation.


Cytokine & Growth Factor Reviews | 2013

MIF, CD74 and other partners in kidney disease: tales of a promiscuous couple.

Maria Dolores Sanchez-Niño; Ana Belen Sanz; O. Ruiz-Andres; J. Poveda; Maria C. Izquierdo; R. Selgas; Jesús Egido; Alberto Ortiz

Macrophage migration inhibitory factor (MIF) is increased in kidney and urine during kidney disease. MIF binds to and activates CD74 and chemokine receptors CXCR2 and CXCR4. CD74 is a protein trafficking regulator and a cell membrane receptor for MIF, D-dopachrome tautomerase (D-DT/MIF-2) and bacterial proteins. MIF signaling through CD74 requires CD44. CD74, CD44 and CXCR4 are upregulated in renal cells in diseased kidneys and MIF activation of CD74 in kidney cells promotes an inflammatory response. MIF or CXCR2 targeting protects from experimental kidney injury, CD44 deficiency modulates kidney injury and CXCR4 activation promotes glomerular injury. However, the contribution of MIF or MIF-2 to these actions of MIF receptors has not been explored. The safety and efficacy of strategies targeting MIF, CD74, CD44 and CXCR4 are under study in humans.


European Journal of Pharmacology | 2015

Translational value of animal models of kidney failure

Alberto Ortiz; Maria Dolores Sanchez-Niño; Maria C. Izquierdo; Catalina Martin-Cleary; Laura Garcia-Bermejo; Juan Antonio Moreno; Marta Ruiz-Ortega; Juliana Draibe; Josep M. Cruzado; Miguel A. Garcia-Gonzalez; José M. López-Novoa; Maria J. Soler; Ana Belen Sanz

Acute kidney injury (AKI) and chronic kidney disease (CKD) are associated with decreased renal function and increased mortality risk, while the therapeutic armamentarium is unsatisfactory. The availability of adequate animal models may speed up the discovery of biomarkers for disease staging and therapy individualization as well as design and testing of novel therapeutic strategies. Some longstanding animal models have failed to result in therapeutic advances in the clinical setting, such as kidney ischemia-reperfusion injury and diabetic nephropathy models. In this regard, most models for diabetic nephropathy are unsatisfactory in that they do not evolve to renal failure. Satisfactory models for additional nephropathies are needed. These include anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis, IgA nephropathy, anti-phospholipase-A2-receptor (PLA2R) membranous nephropathy and Fabry nephropathy. However, recent novel models hold promise for clinical translation. Thus, the AKI to CKD translation has been modeled, in some cases with toxins of interest for human CKD such as aristolochic acid. Genetically modified mice provide models for Alport syndrome evolving to renal failure that have resulted in clinical recommendations, polycystic kidney disease models that have provided clues for the development of tolvaptan, that was recently approved for the human disease in Japan; and animal models also contributed to target C5 with eculizumab in hemolytic uremic syndrome. Some ongoing trials explore novel concepts derived from models, such TWEAK targeting as tissue protection for lupus nephritis. We now review animal models reproducing diverse, genetic and acquired, causes of AKI and CKD evolving to kidney failure and discuss the contribution to clinical translation and prospects for the future.

Collaboration


Dive into the Maria C. Izquierdo's collaboration.

Top Co-Authors

Avatar

Jesús Egido

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Alberto Ortiz

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Marta Ruiz-Ortega

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Ana Belen Sanz

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alvaro C. Ucero

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Alberto Benito-Martin

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Adrián M. Ramos

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Rafael Selgas

Hospital Universitario La Paz

View shared research outputs
Top Co-Authors

Avatar

Jonay Poveda

Autonomous University of Madrid

View shared research outputs
Researchain Logo
Decentralizing Knowledge