Network


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

Hotspot


Dive into the research topics where Diego Martin-Sanchez is active.

Publication


Featured researches published by Diego Martin-Sanchez.


Journal of The American Society of Nephrology | 2017

Ferroptosis, but Not Necroptosis, Is Important in Nephrotoxic Folic Acid–Induced AKI

Diego Martin-Sanchez; Olga Ruiz-Andres; Jonay Poveda; Susana Carrasco; Pablo Cannata-Ortiz; Maria Dolores Sanchez-Niño; Marta Ruiz Ortega; Jesús Egido; Andreas Linkermann; Alberto Ortiz; Ana Belen Sanz

AKI is histologically characterized by necrotic cell death and inflammation. Diverse pathways of regulated necrosis have been reported to contribute to AKI, but the molecular regulators involved remain unclear. We explored the relative contributions of ferroptosis and necroptosis to folic acid (FA)-induced AKI in mice. FA-AKI in mice associates with lipid peroxidation and downregulation of glutathione metabolism proteins, features that are typical of ferroptotic cell death. We show that ferrostatin-1 (Fer-1), an inhibitor of ferroptosis, preserved renal function and decreased histologic injury, oxidative stress, and tubular cell death in this model. With respect to the immunogenicity of ferroptosis, Fer-1 prevented the upregulation of IL-33, an alarmin linked to necroptosis, and other chemokines and cytokines and prevented macrophage infiltration and Klotho downregulation. In contrast, the pancaspase inhibitor zVAD-fmk did not protect against FA-AKI. Additionally, although FA-AKI resulted in increased protein expression of the necroptosis mediators receptor-interacting protein kinase 3 (RIPK3) and mixed lineage domain-like protein (MLKL), targeting necroptosis with the RIPK1 inhibitor necrostatin-1 or genetic deficiency of RIPK3 or MLKL did not preserve renal function. Indeed, compared with wild-type mice, MLKL knockout mice displayed more severe AKI. However, RIPK3 knockout mice with AKI had less inflammation than their wild-type counterparts, and this effect associated with higher IL-10 concentration and regulatory T cell-to-leukocyte ratio in RIPK3 knockout mice. These data suggest that ferroptosis is the primary cause of FA-AKI and that immunogenicity secondary to ferroptosis may further worsen the damage, although necroptosis-related proteins may have additional roles in AKI.


Cell Death and Disease | 2016

TWEAK favors phosphate-induced calcification of vascular smooth muscle cells through canonical and non-canonical activation of NFκB

L Hénaut; Ana Belen Sanz; Diego Martin-Sanchez; S Carrasco; R Villa-Bellosta; G Aldamiz-Echevarria; Z A Massy; Maria Dolores Sanchez-Niño; Alberto Ortiz

Vascular calcification (VC) is associated with increased cardiovascular mortality in aging, chronic kidney disease (CKD), type 2 diabetes mellitus (T2DM) and atherosclerosis. TNF-like weak inducer of apoptosis (TWEAK) recently emerged as a new biomarker for the diagnosis and prognosis of cardiovascular diseases. TWEAK binding to its functional receptor Fn14 was reported to promote several steps of atherosclerotic plaque progression. However, no information is currently available on the role of TWEAK/Fn14 on the development of medial calcification, which is highly prevalent in aging, CKD and T2DM. This study explored the involvement of TWEAK in human vascular smooth muscle cells (h-VSMCs) calcification in vitro. We report that TWEAK binding to Fn14 promotes inorganic phosphate-induced h-VSMCs calcification, favors h-VSMCs osteogenic transition, decreasing acta2 and myh11 and increasing bmp2 mRNA and tissue non-specific alkaline phosphatase (TNAP), and increases MMP9 activity. Blockade of the canonical NFκB pathway reduced by 80% TWEAK pro-calcific properties and decreased osteogenic transition, TNAP and MMP9 activity. Blockade of non-canonical NFκB signaling by a siRNA targeting RelB reduced by 20% TWEAK pro-calcific effects and decreased TWEAK-induced loss of h-VSMCs contractile phenotype and MMP9 activity, without modulating bmp2 mRNA or TNAP activity. Inhibition of ERK1/2 activation by a MAPK kinase inhibitor did not influence TWEAK pro-calcific properties. Our results suggest that TWEAK/Fn14 directly favors inorganic phosphate-induced h-VSMCs calcification by activation of both canonical and non-canonical NFκB pathways. Given the availability of neutralizing anti-TWEAK strategies, our study sheds light on the TWEAK/Fn14 axis as a novel therapeutic target in the prevention of VC.


Proceedings of the National Academy of Sciences of the United States of America | 2018

TWEAK and RIPK1 mediate a second wave of cell death during AKI

Diego Martin-Sanchez; Miguel Fontecha-Barriuso; Susana Carrasco; Maria Dolores Sanchez-Niño; Anne von Mässenhausen; Andreas Linkermann; Pablo Cannata-Ortiz; Marta Ruiz-Ortega; Jesús Egido; Alberto Ortiz; Ana Belen Sanz

Significance Acute kidney injury (AKI) has a mortality of 50%. There is no satisfactory therapy and the incidence is increasing. The etiology of AKI is heterogeneous, and this has therapeutic implications. Here we show that necroptosis plays a role in a second wave of tubular cell death in experimental toxic AKI. This second wave of death is triggered by TWEAK activation of the Fn14 receptor and contributes to persistence of injury. We previously observed that the initial wave of cell death was ferroptosis dependent and necroptosis independent. The identification of a pathway contributing to AKI persistence may facilitate the design of therapies, as exemplified by the protection afforded by RIPK1 inhibitors when administered after AKI had been induced. Acute kidney injury (AKI) is characterized by necrotic tubular cell death and inflammation. The TWEAK/Fn14 axis is a mediator of renal injury. Diverse pathways of regulated necrosis have recently been reported to contribute to AKI, but there are ongoing discussions on the timing or molecular regulators involved. We have now explored the cell death pathways induced by TWEAK/Fn14 activation and their relevance during AKI. In cultured tubular cells, the inflammatory cytokine TWEAK induces apoptosis in a proinflammatory environment. The default inhibitor of necroptosis [necrostatin-1 (Nec-1)] was protective, while caspase inhibition switched cell death to necroptosis. Additionally, folic acid-induced AKI in mice resulted in increased expression of Fn14 and necroptosis mediators, such as receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage domain-like protein (MLKL). Targeting necroptosis with Nec-1 or by genetic RIPK3 deficiency and genetic Fn14 ablation failed to be protective at early time points (48 h). However, a persistently high cell death rate and kidney dysfunction (72–96 h) were dependent on an intact TWEAK/Fn14 axis driving necroptosis. This was prevented by Nec-1, or MLKL, or RIPK3 deficiency and by Nec-1 stable (Nec-1s) administered before or after induction of AKI. These data suggest that initial kidney damage and cell death are amplified through recruitment of inflammation-dependent necroptosis, opening a therapeutic window to treat AKI once it is established. This may be relevant for clinical AKI, since using current diagnostic criteria, severe injury had already led to loss of renal function at diagnosis.


Nefrologia | 2017

Targeting of regulated necrosis in kidney disease

Diego Martin-Sanchez; Jonay Poveda; Miguel Fontecha-Barriuso; Olga Ruiz-Andres; Maria Dolores Sanchez-Niño; Marta Ruiz-Ortega; Alberto Ortiz; Ana Belen Sanz

The term acute tubular necrosis was thought to represent a misnomer derived from morphological studies of human necropsies and necrosis was thought to represent an unregulated passive form of cell death which was not amenable to therapeutic manipulation. Recent advances have improved our understanding of cell death in acute kidney injury. First, apoptosis results in cell loss, but does not trigger an inflammatory response. However, clumsy attempts at interfering with apoptosis (e.g. certain caspase inhibitors) may trigger necrosis and, thus, inflammation-mediated kidney injury. Second, and most revolutionary, the concept of regulated necrosis emerged. Several modalities of regulated necrosis were described, such as necroptosis, ferroptosis, pyroptosis and mitochondria permeability transition regulated necrosis. Similar to apoptosis, regulated necrosis is modulated by specific molecules that behave as therapeutic targets. Contrary to apoptosis, regulated necrosis may be extremely pro-inflammatory and, importantly for kidney transplantation, immunogenic. Furthermore, regulated necrosis may trigger synchronized necrosis, in which all cells within a given tubule die in a synchronized manner. We now review the different modalities of regulated necrosis, the evidence for a role in diverse forms of kidney injury and the new opportunities for therapeutic intervention.


Nephrology Dialysis Transplantation | 2018

Targeting epigenetic DNA and histone modifications to treat kidney disease

Miguel Fontecha-Barriuso; Diego Martin-Sanchez; Olga Ruiz-Andres; Jonay Poveda; Maria Dolores Sanchez-Niño; Lara Valiño-Rivas; Marta Ruiz-Ortega; Alberto Ortiz; Ana Belen Sanz

Epigenetics refers to heritable changes in gene expression patterns not caused by an altered nucleotide sequence, and includes non-coding RNAs and covalent modifications of DNA and histones. This review focuses on functional evidence for the involvement of DNA and histone epigenetic modifications in the pathogenesis of kidney disease and the potential therapeutic implications. There is evidence of activation of epigenetic regulatory mechanisms in acute kidney injury (AKI), chronic kidney disease (CKD) and the AKI-to-CKD transition of diverse aetiologies, including ischaemia-reperfusion injury, nephrotoxicity, ureteral obstruction, diabetes, glomerulonephritis and polycystic kidney disease. A beneficial in vivo effect over preclinical kidney injury has been reported for drugs that decrease DNA methylation by either inhibiting DNA methylation (e.g. 5-azacytidine and decitabine) or activating DNA demethylation (e.g. hydralazine), decrease histone methylation by inhibiting histone methyltransferases, increase histone acetylation by inhibiting histone deacetylases (HDACs, e.g. valproic acid, vorinostat, entinostat), increase histone crotonylation (crotonate) or interfere with histone modification readers [e.g. inhibits of bromodomain and extra-terminal proteins (BET)]. Most preclinical studies addressed CKD or the AKI-to-CKD transition. Crotonate administration protected from nephrotoxic AKI, but evidence is conflicting on DNA methylation inhibitors for preclinical AKI. Several drugs targeting epigenetic regulators are in clinical development or use, most of them for malignancy. The BET inhibitor apabetalone is in Phase 3 trials for atherosclerosis, kidney function being a secondary endpoint, but nephrotoxicity was reported for DNA and HDAC inhibitors. While research into epigenetic modulators may provide novel therapies for kidney disease, caution should be exercised based on the clinical nephrotoxicity of some drugs.


Scientific Reports | 2017

Deferasirox-induced iron depletion promotes BclxL downregulation and death of proximal tubular cells

Diego Martin-Sanchez; Angel Gallegos-Villalobos; Miguel Fontecha-Barriuso; Susana Carrasco; Maria Dolores Sanchez-Niño; Francisco J. López-Hernández; Marta Ruiz-Ortega; Jesús Egido; Alberto Ortiz; Ana Belen Sanz

Iron deficiency has been associated with kidney injury. Deferasirox is an oral iron chelator used to treat blood transfusion-related iron overload. Nephrotoxicity is the most serious and common adverse effect of deferasirox and may present as an acute or chronic kidney disease. However, scarce data are available on the molecular mechanisms of nephrotoxicity. We explored the therapeutic modulation of deferasirox-induced proximal tubular cell death in culture. Deferasirox induced dose-dependent tubular cell death and AnexxinV/7AAD staining showed features of apoptosis and necrosis. However, despite inhibiting caspase-3 activation, the pan-caspase inhibitor zVAD-fmk failed to prevent deferasirox-induced cell death. Moreover, zVAD increased deferasirox-induced cell death, a feature sometimes found in necroptosis. Electron microscopy identified mitochondrial injury and features of necrosis. However, neither necrostatin-1 nor RIP3 knockdown prevented deferasirox-induced cell death. Deferasirox caused BclxL depletion and BclxL overexpression was protective. Preventing iron depletion protected from BclxL downregulation and deferasirox cytotoxicity. In conclusion, deferasirox promoted iron depletion-dependent cell death characterized by BclxL downregulation. BclxL overexpression was protective, suggesting a role for BclxL downregulation in iron depletion-induced cell death. This information may be used to develop novel nephroprotective strategies. Furthermore, it supports the concept that monitoring kidney tissue iron depletion may decrease the risk of deferasirox nephrotoxicity.


Cell Death and Disease | 2018

Cell death-based approaches in treatment of the urinary tract-associated diseases: a fight for survival in the killing fields

Diego Martin-Sanchez; Miguel Fontecha-Barriuso; Maria Dolores Sanchez-Niño; Adrián M. Ramos; Ramiro Cabello; Carmen Gonzalez-Enguita; Andreas Linkermann; Ana Belen Sanz; Alberto Ortiz

Urinary tract-associated diseases comprise a complex set of disorders with a variety of etiologic agents and therapeutic approaches and a huge global burden of disease, estimated at around 1 million deaths per year. These diseases include cancer (mainly prostate, renal, and bladder), urinary tract infections, and urolithiasis. Cell death plays a key role in the pathogenesis and therapy of these conditions. During urinary tract infections, invading bacteria may either promote or prevent host cell death by interfering with cell death pathways. This has been studied in detail for uropathogenic E. coli (UPEC). Inhibition of host cell death may allow intracellular persistence of live bacteria, while promoting host cell death causes tissue damage and releases the microbes. Both crystals and urinary tract obstruction lead to tubular cell death and kidney injury. Among the pathomechanisms, apoptosis, necroptosis, and autophagy represent key processes. With respect to malignant disorders, traditional therapeutic efforts have focused on directly promoting cancer cell death. This may exploit tumor-specific characteristics, such as targeting Vascular Endothelial Growth Factor (VEGF) signaling and mammalian Target of Rapamycin (mTOR) activity in renal cancer and inducing survival factor deprivation by targeting androgen signaling in prostate cancer. An area of intense research is the use of immune checkpoint inhibitors, aiming at unleashing the full potential of immune cells to kill cancer cells. In the future, this may be combined with additional approaches exploiting intrinsic sensitivities to specific modes of cell death such as necroptosis and ferroptosis. Here, we review the contribution of diverse cell death mechanisms to the pathogenesis of urinary tract-associated diseases as well as the potential for novel therapeutic approaches based on an improved molecular understanding of these mechanisms.


Nephrology Dialysis Transplantation | 2018

FP215MOLECULAR MECHANISMS OF OMEPRAZOLE NEPHROTOXICITY

Miguel Fontecha-Barriuso; Diego Martin-Sanchez; Daniela E Cárdenas-Villacrés; Marta Ruiz-Ortega; Maria Dolores Sanchez-Niño; Alberto Ortiz; Ana Belen Sanz


Nephrology Dialysis Transplantation | 2017

MP244TWEAK/FN14 MEDIATES SECONDARY CELL DEATH DURING AKI

Diego Martin-Sanchez; Miguel Fontecha-Barriuso; Pablo Cannata-Ortiz; Maria Dolores Sanchez-Niño; Marta Ruiz-Ortega; Jesús Egido; Andreas Linkermann; Alberto Ortiz; Ana Belen Sanz


Nephrology Dialysis Transplantation | 2017

MP251THE DEFICIENCY OF THE TRANSCRIPTIONAL COACTIVATOR PGC-1α INCREASES RENAL INJURY IN ACUTE KIDNEY INJURY (AKI)

Miguel Fontecha-Barriuso; Olga Ruiz-Andres; María Monsalve; Diego Martin-Sanchez; Marta Ruiz-Ortega; Maria Dolores Sanchez-Niño; Alberto Ortiz; Ana Belen Sanz

Collaboration


Dive into the Diego Martin-Sanchez's collaboration.

Top Co-Authors

Avatar

Alberto Ortiz

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

Miguel Fontecha-Barriuso

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Marta Ruiz-Ortega

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Jesús Egido

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Olga Ruiz-Andres

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Andreas Linkermann

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Jonay Poveda

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Pablo Cannata-Ortiz

Autonomous University of Madrid

View shared research outputs
Researchain Logo
Decentralizing Knowledge