19 results on '"Raul R. Rodrigues-Diez"'
Search Results
2. Acute Kidney Injury is Aggravated in Aged Mice by the Exacerbation of Proinflammatory Processes
- Author
-
Laura Marquez-Exposito, Lucia Tejedor-Santamaria, Laura Santos-Sanchez, Floris A. Valentijn, Elena Cantero-Navarro, Sandra Rayego-Mateos, Raul R. Rodrigues-Diez, Antonio Tejera-Muñoz, Vanessa Marchant, Ana B. Sanz, Alberto Ortiz, Roel Goldschmeding, and Marta Ruiz-Ortega
- Subjects
aging ,necroptosis ,apoptosis ,cellular senescence ,inflammation ,immunosenescence ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Acute kidney injury (AKI) is more frequent in elderly patients. Mechanisms contributing to AKI (tubular cell death, inflammatory cell infiltration, impaired mitochondrial function, and prolonged cell-cycle arrest) have been linked to cellular senescence, a process implicated in regeneration failure and progression to fibrosis. However, the molecular and pathological basis of the age-related increase in AKI incidence is not completely understood. To explore these mechanisms, experimental AKI was induced by folic acid (FA) administration in young (3-months-old) and old (1-year-old) mice, and kidneys were evaluated in the early phase of AKI, at 48 h. Tubular damage score, KIM-1 expression, the recruitment of infiltrating immune cells (mainly neutrophils and macrophages) and proinflammatory gene expression were higher in AKI kidneys of old than of young mice. Tubular cell death in FA-AKI involves several pathways, such as regulated necrosis and apoptosis. Ferroptosis and necroptosis cell-death pathways were upregulated in old AKI kidneys. In contrast, caspase-3 activation was only found in young but not in old mice. Moreover, the antiapoptotic factor BCL-xL was significantly overexpressed in old, injured kidneys, suggesting an age-related apoptosis suppression. AKI kidneys displayed evidence of cellular senescence, such as increased levels of cyclin dependent kinase inhibitors p16ink4a and p21cip1, and of the DNA damage response marker γH2AX. Furthermore, p21cip1 mRNA expression and nuclear staining for p21cip1 and γH2AX were higher in old than in young FA-AKI mice, as well as the expression of senescence-associated secretory phenotype (SASP) components (Il-6, Tgfb1, Ctgf, and Serpine1). Interestingly, some infiltrating immune cells were p21 or γH2AX positive, suggesting that molecular senescence in the immune cells (“immunosenescence”) are involved in the increased severity of AKI in old mice. In contrast, expression of renal protective factors was dramatically downregulated in old AKI mice, including the antiaging factor Klotho and the mitochondrial biogenesis driver PGC-1α. In conclusion, aging resulted in more severe AKI after the exposure to toxic compounds. This increased toxicity may be related to magnification of proinflammatory-related pathways in older mice, including a switch to a proinflammatory cell death (necroptosis) instead of apoptosis, and overactivation of cellular senescence of resident renal cells and infiltrating inflammatory cells.
- Published
- 2021
- Full Text
- View/download PDF
3. Oxidative Stress and Cellular Senescence Are Involved in the Aging Kidney
- Author
-
Laura Marquez-Exposito, Lucia Tejedor-Santamaria, Floris A. Valentijn, Antonio Tejera-Muñoz, Sandra Rayego-Mateos, Vanessa Marchant, Raul R. Rodrigues-Diez, Irene Rubio-Soto, Sebastiaan N. Knoppert, Alberto Ortiz, Adrian M. Ramos, Roel Goldschmeding, and Marta Ruiz-Ortega
- Subjects
cellular senescence ,NRF2 ,aging kidney ,oxidation ,inflammaging ,fibrosis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Chronic kidney disease (CKD) can be considered as a clinical model for premature aging. However, non-invasive biomarkers to detect early kidney damage and the onset of a senescent phenotype are lacking. Most of the preclinical senescence studies in aging have been done in very old mice. Furthermore, the precise characterization and over-time development of age-related senescence in the kidney remain unclear. To address these limitations, the age-related activation of cellular senescence-associated mechanisms and their correlation with early structural changes in the kidney were investigated in 3- to 18-month-old C57BL6 mice. Inflammatory cell infiltration was observed by 12 months, whereas tubular damage and collagen accumulation occurred later. Early activation of cellular-senescence-associated mechanisms was found in 12-month-old mice, characterized by activation of the DNA-damage-response (DDR), mainly in tubular cells; activation of the antioxidant NRF2 pathway; and klotho downregulation. However, induction of tubular-cell-cycle-arrest (CCA) and overexpression of renal senescent-associated secretory phenotype (SASP) components was only found in 18-month-old mice. In aging mice, both inflammation and oxidative stress (marked by elevated lipid peroxidation and NRF2 inactivation) remained increased. These findings support the hypothesis that prolonged DDR and CCA, loss of nephroprotective factors (klotho), and dysfunctional redox regulatory mechanisms (NRF2/antioxidant defense) can be early drivers of age-related kidney-damage progression.
- Published
- 2022
- Full Text
- View/download PDF
4. Epigenetic Modulation of Gremlin-1/NOTCH Pathway in Experimental Crescentic Immune-Mediated Glomerulonephritis
- Author
-
Lucia Tejedor-Santamaria, Jose Luis Morgado-Pascual, Laura Marquez-Exposito, Beatriz Suarez-Alvarez, Raul R. Rodrigues-Diez, Antonio Tejera-Muñoz, Vanessa Marchant, Sergio Mezzano, Carlos Lopez-Larrea, Anna Sola, Gema Maria Fernandez-Juarez, Alberto Ortiz, Sandra Rayego-Mateos, and Marta Ruiz-Ortega
- Subjects
crescentic glomerulonephritis ,chronic kidney disease ,bromodomain ,BET proteins ,Gremlin ,NOTCH ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
Crescentic glomerulonephritis is a devastating autoimmune disease that without early and properly treatment may rapidly progress to end-stage renal disease and death. Current immunosuppressive treatment provides limited efficacy and an important burden of adverse events. Epigenetic drugs are a source of novel therapeutic tools. Among them, bromodomain and extraterminal domain (BET) inhibitors (iBETs) block the interaction between bromodomains and acetylated proteins, including histones and transcription factors. iBETs have demonstrated protective effects on malignancy, inflammatory disorders and experimental kidney disease. Recently, Gremlin-1 was proposed as a urinary biomarker of disease progression in human anti-neutrophil cytoplasmic antibody (ANCA)-associated crescentic glomerulonephritis. We have now evaluated whether iBETs could regulate Gremlin-1 in experimental anti-glomerular basement membrane nephritis induced by nephrotoxic serum (NTS) in mice, a model resembling human crescentic glomerulonephritis. In NTS-injected mice, the iBET JQ1 inhibited renal Gremlin-1 overexpression and diminished glomerular damage, restoring podocyte numbers. Chromatin immunoprecipitation assay demonstrated BRD4 enrichment of the Grem-1 gene promoter in injured kidneys, consistent with Gremlin-1 epigenetic regulation. Moreover, JQ1 blocked BRD4 binding and inhibited Grem-1 gene transcription. The beneficial effect of iBETs was also mediated by modulation of NOTCH pathway. JQ1 inhibited the gene expression of the NOTCH effectors Hes-1 and Hey-1 in NTS-injured kidneys. Our results further support the role for epigenetic drugs, such as iBETs, in the treatment of rapidly progressive crescentic glomerulonephritis.
- Published
- 2022
- Full Text
- View/download PDF
5. Interleukin 17A Participates in Renal Inflammation Associated to Experimental and Human Hypertension
- Author
-
Macarena Orejudo, Raul R. Rodrigues-Diez, Raquel Rodrigues-Diez, Ana Garcia-Redondo, Laura Santos-Sánchez, Javier Rández-Garbayo, Pablo Cannata-Ortiz, Adrian M. Ramos, Alberto Ortiz, Rafael Selgas, Sergio Mezzano, Carolina Lavoz, and Marta Ruiz-Ortega
- Subjects
IL-17A ,hypertension ,renal pathology ,IL-17A neutralization ,inflammation ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Hypertension is now considered as an inflammatory disease, and the kidney is a key end-organ target. Experimental and clinical studies suggest that interleukin 17A (IL-17A) is a promising therapeutic target in immune and chronic inflammatory diseases, including hypertension and kidney disease. Elevated circulating IL-17A levels have been observed in hypertensive patients. Our aim was to investigate whether chronically elevated circulating IL-17A levels could contribute to kidney damage, using a murine model of systemic IL-17A administration. Blood pressure increased after 14 days of IL-17A infusion in mice when compared with that in control mice, and this was associated to kidney infiltration by inflammatory cells, including CD3+ and CD4+ lymphocytes and neutrophils. Moreover, proinflammatory factors and inflammatory-related intracellular mechanisms were upregulated in kidneys from IL-17A-infused mice. In line with these findings, in the model of angiotensin II infusion in mice, IL-17A blockade, using an anti-IL17A neutralizing antibody, reduced kidney inflammatory cell infiltrates and chemokine overexpression. In kidney biopsies from patients with hypertensive nephrosclerosis, IL-17A positive cells, mainly Th17 and γδ T lymphocytes, were found. Overall, the results support a pathogenic role of IL-17A in hypertensive kidney disease-associated inflammation. Therapeutic approaches targeting this cytokine should be explored to prevent hypertension-induced kidney injury.
- Published
- 2019
- Full Text
- View/download PDF
6. IL-17A as a Potential Therapeutic Target for Patients on Peritoneal Dialysis
- Author
-
Vanessa Marchant, Antonio Tejera-Muñoz, Laura Marquez-Expósito, Sandra Rayego-Mateos, Raul R. Rodrigues-Diez, Lucia Tejedor, Laura Santos-Sanchez, Jesús Egido, Alberto Ortiz, Jose M. Valdivielso, Donald J. Fraser, Manuel López-Cabrera, Rafael Selgas, and Marta Ruiz-Ortega
- Subjects
Interleukin-17A ,peritoneal dialysis ,chronic kidney disease ,inflammation ,membrane failure ,mesothelial ,Microbiology ,QR1-502 - Abstract
Chronic kidney disease (CKD) is a health problem reaching epidemic proportions. There is no cure for CKD, and patients may progress to end-stage renal disease (ESRD). Peritoneal dialysis (PD) is a current replacement therapy option for ESRD patients until renal transplantation can be achieved. One important problem in long-term PD patients is peritoneal membrane failure. The mechanisms involved in peritoneal damage include activation of the inflammatory and immune responses, associated with submesothelial immune infiltrates, angiogenesis, loss of the mesothelial layer due to cell death and mesothelial to mesenchymal transition, and collagen accumulation in the submesothelial compact zone. These processes lead to fibrosis and loss of peritoneal membrane function. Peritoneal inflammation and membrane failure are strongly associated with additional problems in PD patients, mainly with a very high risk of cardiovascular disease. Among the inflammatory mediators involved in peritoneal damage, cytokine IL-17A has recently been proposed as a potential therapeutic target for chronic inflammatory diseases, including CKD. Although IL-17A is the hallmark cytokine of Th17 immune cells, many other cells can also produce or secrete IL-17A. In the peritoneum of PD patients, IL-17A-secreting cells comprise Th17 cells, γδ T cells, mast cells, and neutrophils. Experimental studies demonstrated that IL-17A blockade ameliorated peritoneal damage caused by exposure to PD fluids. This article provides a comprehensive review of recent advances on the role of IL-17A in peritoneal membrane injury during PD and other PD-associated complications.
- Published
- 2020
- Full Text
- View/download PDF
7. Gremlin Regulates Tubular Epithelial to Mesenchymal Transition via VEGFR2: Potential Role in Renal Fibrosis
- Author
-
Laura Marquez-Exposito, Carolina Lavoz, Raul R. Rodrigues-Diez, Sandra Rayego-Mateos, Macarena Orejudo, Elena Cantero-Navarro, Alberto Ortiz, Jesús Egido, Rafael Selgas, Sergio Mezzano, and Marta Ruiz-Ortega
- Subjects
gremlin ,VEGFR2 ,notch ,EMT ,tubular cells ,fibrosis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Chronic kidney disease (CKD) is emerging as an important health problem due to the increase number of CKD patients and the absence of an effective curative treatment. Gremlin has been proposed as a novel therapeutic target for renal inflammatory diseases, acting via Vascular Endothelial Growth Factor Receptor-2 (VEGFR2). Although many evidences suggest that Gremlin could regulate renal fibrosis, the receptor involved has not been yet clarified. Gremlin, as other TGF-β superfamily members, regulates tubular epithelial to mesenchymal transition (EMT) and, therefore, could contribute to renal fibrosis. In cultured tubular epithelial cells Gremlin binding to VEGFR2 is linked to proinflammatory responses. Now, we have found out that in these cells VEGFR2 is also involved in the profibrotic actions of Gremlin. VEGFR2 blockade by a pharmacological kinase inhibitor or gene silencing diminished Gremlin-mediated gene upregulation of profibrotic factors and restored changes in EMT-related genes. Moreover, VEGFR2 inhibition blocked EMT phenotypic changes and dampened the rate of wound healing in response to Gremlin. The role of VEGFR2 in experimental fibrosis was evaluated in experimental unilateral ureteral obstruction. VEFGR2 inhibition diminished the upregulation of profibrotic genes and EMT changes, as well as the accumulation of extracellular matrix proteins, such as fibronectin and collagens in the obstructed kidneys. Notch pathway activation participates in renal damage progression by regulating cell growth/proliferation, regeneration and inflammation. In cultured tubular epithelial cells, Notch inhibition markedly downregulated Gremlin-induced EMT changes and wound healing speed. These results show that Gremlin regulates the EMT process via VEGFR2 and Notch pathway activation, suggesting that the Gremlin/VEGFR2 axis could be a potential therapeutic target for CKD.
- Published
- 2018
- Full Text
- View/download PDF
8. Interleuquina-17A: posible mediador y diana terapéutica en la hipertensión
- Author
-
Raúl R. Rodrigues-Diez, Antonio Tejera-Muñoz, Macarena Orejudo, Laura Marquez-Exposito, Laura Santos, Sandra Rayego-Mateos, Elena Cantero-Navarro, Lucia Tejedor-Santamaria, Vanessa Marchant, Alberto Ortiz, Jesús Egido, Sergio Mezzano, Rafael Selgas, Juan F. Navarro-González, Jose M. Valdivielso, Carolina Lavoz, and Marta Ruiz-Ortega
- Subjects
Hypertension ,IL-17A ,Immune response ,Inflammation ,Cytokines ,Chronic kidney disease ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Resumen: La interleuquina 17A (IL-17A) es una citoquina proinflamatoria producida por células del sistema inmune, sobre todo por los linfocitos Th17 y los linfocitos γδ. En este trabajo, revisamos el papel de IL-17A en la patogenia de la hipertensión y de la lesión en órganos diana. Estudios en ratones han demostrado que la IL-17A aumenta la presión arterial, probablemente por acciones a varios niveles. Además, las concentraciones plasmáticas de IL-17A están ya aumentadas en pacientes con hipertensión arterial ligera o moderada. Estudios preclínicos sobre hipertensión arterial han detectado células productoras de IL-17A en órganos diana, como corazón, vasos y riñón. En pacientes con nefroesclerosis hipertensiva existe infiltración del riñón por linfocitos Th17 y linfocitos γδ que expresan IL-17A. Además, en modelos experimentales de hipertensión el bloqueo de IL-17A, mediante estrategias génicas, o utilizando anticuerpos neutralizantes, disminuye la presión arterial por acciones sobre la pared vascular y el transporte tubular de sodio y disminuye la lesión en órganos diana. En conjunto, los datos presentados en esta revisión sugieren que la IL-17A participa en la regulación de la presión arterial y en la génesis y mantenimiento de la hipertensión arterial, pudiendo constituir una diana terapéutica en el futuro. Abstract: Interleukin-17A (IL-17A) is a proinflammatory cytokine produced by cells of the immune system, predominantly Th17 lymphocytes and γδ lymphocytes. In this paper, we review the role of IL-17A in the pathogenesis of hypertension and target organ damage. Studies in mice have shown that IL-17A increases blood pressure, probably by acting on multiple levels. Furthermore, IL-17A plasma concentrations are already elevated in patients with mild or moderate hypertension. Preclinical studies on arterial hypertension have detected IL-17A-producing cells in target organs such as the heart, vessels and kidneys. Patients with hypertensive nephrosclerosis show kidney infiltration by Th17 lymphocytes and γδ lymphocytes that express IL-17A. In addition, in experimental models of hypertension, blocking IL-17A by genetic strategies, or using neutralising antibodies, lowers blood pressure by acting on the vascular wall and tubule sodium transport and reduces damage to target organs. As a whole, the data presented in this review suggest that IL-17A participates in the regulation of blood pressure and in the genesis and maintenance of arterial hypertension, and may constitute a therapeutic target in the future.
- Published
- 2021
- Full Text
- View/download PDF
9. Interleukin-17A: Potential mediator and therapeutic target in hypertension
- Author
-
Raúl R. Rodrigues-Diez, Antonio Tejera-Muñoz, Macarena Orejudo, Laura Marquez-Exposito, Laura Santos-Sanchez, Sandra Rayego-Mateos, Elena Cantero-Navarro, Lucia Tejedor-Santamaria, Vanessa Marchant, Alberto Ortiz, Jesús Egido, Sergio Mezzano, Rafael Selgas, Juan F. Navarro-González, Jose M. Valdivielso, Carolina Lavoz, and Marta Ruiz-Ortega
- Subjects
Hipertensión ,IL-17A ,Respuesta inmune ,Inflamación ,Citoquinas ,Enfermedad renal crónica ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Interleukin-17A (IL-17A) is a proinflammatory cytokine produced by cells of the immune system, predominantly Th17 and γδ lymphocytes. In this paper, we review the role of IL-17A in the pathogenesis of hypertension and in target organ damage. Preclinical studies in mice have shown that systemic adminstration of IL-17A increases blood pressure, probably by acting on multiple levels. Furthermore, IL-17A plasma concentrations are already elevated in patients with mild or moderate hypertension. Many studies in hypertensive mice models have detected IL-17A-producing cells in target organs such as the heart, vessels and kidneys. Patients with hypertensive nephrosclerosis show kidney infiltration by Th17 lymphocytes and γδ lymphocytes that express IL-17A. In addition, in experimental models of hypertension, the blockade of IL-17A by genetic strategies or using neutralizing antibodies, disminished blood pressure, probablyby acting on the small mesenteric arteries as well as in the regulation of tubule sodium transport. Moreover, IL-17A inhibition reduces end-organs damage. As a whole, the data presented in this review suggest that IL-17A participates in the regulation of blood pressure and in the genesis and maintenance of arterial hypertension, and may constitute a therapeutic target of hypertension-related pathologies in the future. Resume: La interleuquina 17A (IL-17A) es una citoquina proinflamatoria producida por células del sistema inmune, sobre todo por los linfocitos Th17 y los linfocitos γδ. En este trabajo, revisamos el papel de IL-17A en la patogenia de la hipertensión y de la lesión en órganos diana. Estudios en ratones han demostrado que la IL-17A aumenta la presión arterial, probablemente por acciones a varios niveles. Además, las concentraciones plasmáticas de IL-17A están ya aumentadas en pacientes con hipertensión arterial ligera o moderada. Estudios preclínicos sobre hipertensión arterial han detectado células productoras de IL-17A en órganos diana, como corazón, vasos y riñón. En pacientes con nefroesclerosis hipertensiva existe infiltración del riñón por linfocitos Th17 y linfocitos γδ que expresan IL-17A. Además, en modelos experimentales de hipertensión el bloqueo de IL-17A, mediante estrategias génicas, o utilizando anticuerpos neutralizantes, disminuye la presión arterial por acciones sobre la pared vascular y el transporte tubular de sodio y disminuye la lesión en órganos diana. En conjunto, los datos presentados en esta revisión sugieren que la IL-17A participa en la regulación de la presión arterial y en la génesis y mantenimiento de la hipertensión arterial, pudiendo constituir una diana terapéutica en el futuro.
- Published
- 2021
- Full Text
- View/download PDF
10. Kidney microRNA Expression Pattern in Type 2 Diabetic Nephropathy in BTBR Ob/Ob Mice
- Author
-
Lucas Opazo-Ríos, Antonio Tejera-Muñoz, Manuel Soto Catalan, Vanessa Marchant, Carolina Lavoz, Sebastián Mas Fontao, Juan Antonio Moreno, Marta Fierro Fernandez, Ricardo Ramos, Beatriz Suarez-Alvarez, Carlos López-Larrea, Marta Ruiz-Ortega, Jesús Egido, and Raúl R. Rodrigues-Díez
- Subjects
miRNA ,inflammation ,diabetes ,type 2 diabetes ,diabetic nephropaty ,chronic kidney disease ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Diabetic nephropathy (DN) is the main leading cause of chronic kidney disease worldwide. Although remarkable therapeutic advances have been made during the last few years, there still exists a high residual risk of disease progression to end-stage renal failure. To further understand the pathogenesis of tissue injury in this disease, by means of the Next-Generation Sequencing, we have studied the microRNA (miRNA) differential expression pattern in kidneys of Black and Tan Brachyury (BTBR) ob/ob (leptin deficiency mutation) mouse. This experimental model of type 2 diabetes and obesity recapitulates the key histopathological features described in advanced human DN and therefore can provide potential useful translational information. The miRNA-seq analysis, performed in the renal cortex of 22-week-old BTBR ob/ob mice, pointed out a set of 99 miRNAs significantly increased compared to non-diabetic, non-obese control mice of the same age, whereas no miRNAs were significantly decreased. Among them, miR-802, miR-34a, miR-132, miR-101a, and mir-379 were the most upregulated ones in diabetic kidneys. The in silico prediction of potential targets for the 99 miRNAs highlighted inflammatory and immune processes, as the most relevant pathways, emphasizing the importance of inflammation in the pathogenesis of kidney damage associated to diabetes. Other identified top canonical pathways were adipogenesis (related with ectopic fatty accumulation), necroptosis (an inflammatory and regulated form of cell death), and epithelial-to-mesenchymal transition, the latter supporting the importance of tubular cell phenotype changes in the pathogenesis of DN. These findings could facilitate a better understanding of this complex disease and potentially open new avenues for the design of novel therapeutic approaches to DN.
- Published
- 2022
- Full Text
- View/download PDF
11. Role of Macrophages and Related Cytokines in Kidney Disease
- Author
-
Elena Cantero-Navarro, Sandra Rayego-Mateos, Macarena Orejudo, Lucía Tejedor-Santamaria, Antonio Tejera-Muñoz, Ana Belén Sanz, Laura Marquez-Exposito, Vanessa Marchant, Laura Santos-Sanchez, Jesús Egido, Alberto Ortiz, Teresa Bellon, Raúl R. Rodrigues-Diez, and Marta Ruiz-Ortega
- Subjects
macrophages ,cytokines ,kidney disease ,CCL18 ,inflammation ,CCL8 ,Medicine (General) ,R5-920 - Abstract
Inflammation is a key characteristic of kidney disease, but this immune response is two-faced. In the acute phase of kidney injury, there is an activation of the immune cells to fight against the insult, contributing to kidney repair and regeneration. However, in chronic kidney diseases (CKD), immune cells that infiltrate the kidney play a deleterious role, actively participating in disease progression, and contributing to nephron loss and fibrosis. Importantly, CKD is a chronic inflammatory disease. In early CKD stages, patients present sub-clinical inflammation, activation of immune circulating cells and therefore, anti-inflammatory strategies have been proposed as a common therapeutic target for renal diseases. Recent studies have highlighted the plasticity of immune cells and the complexity of their functions. Among immune cells, monocytes/macrophages play an important role in all steps of kidney injury. However, the phenotype characterization between human and mice immune cells showed different markers; therefore the extrapolation of experimental studies in mice could not reflect human renal diseases. Here we will review the current information about the characteristics of different macrophage phenotypes, mainly focused on macrophage-related cytokines, with special attention to the chemokine CCL18, and its murine functional homolog CCL8, and the macrophage marker CD163, and their role in kidney pathology.
- Published
- 2021
- Full Text
- View/download PDF
12. CCN2 Binds to Tubular Epithelial Cells in the Kidney
- Author
-
Sandra Rayego-Mateos, José Luis Morgado-Pascual, Carolina Lavoz, Raúl R. Rodrigues-Díez, Laura Márquez-Expósito, Antonio Tejera-Muñoz, Lucía Tejedor-Santamaría, Irene Rubio-Soto, Vanessa Marchant, and Marta Ruiz-Ortega
- Subjects
CCN2 ,CTGF ,EGFR ,kidney damage ,Microbiology ,QR1-502 - Abstract
Cellular communication network-2 (CCN2), also called connective tissue growth factor (CTGF), is considered a fibrotic biomarker and has been suggested as a potential therapeutic target for kidney pathologies. CCN2 is a matricellular protein with four distinct structural modules that can exert a dual function as a matricellular protein and as a growth factor. Previous experiments using surface plasmon resonance and cultured renal cells have demonstrated that the C-terminal module of CCN2 (CCN2(IV)) interacts with the epidermal growth factor receptor (EGFR). Moreover, CCN2(IV) activates proinflammatory and profibrotic responses in the mouse kidney. The aim of this paper was to locate the in vivo cellular CCN2/EGFR binding sites in the kidney. To this aim, the C-terminal module CCN2(IV) was labeled with a fluorophore (Cy5), and two different administration routes were employed. Both intraperitoneal and direct intra-renal injection of Cy5-CCN2(IV) in mice demonstrated that CCN2(IV) preferentially binds to the tubular epithelial cells, while no signal was detected in glomeruli. Moreover, co-localization of Cy5-CCN2(IV) binding and activated EGFR was found in tubules. In cultured tubular epithelial cells, live-cell confocal microscopy experiments showed that EGFR gene silencing blocked Cy5-CCN2(IV) binding to tubuloepithelial cells. These data clearly show the existence of CCN2/EGFR binding sites in the kidney, mainly in tubular epithelial cells. In conclusion, these studies show that circulating CCN2(IV) can directly bind and activate tubular cells, supporting the role of CCN2 as a growth factor involved in kidney damage progression.
- Published
- 2022
- Full Text
- View/download PDF
13. Could the Notch signaling pathway be a potential therapeutic option in renal diseases?
- Author
-
Laura Marquez-Exposito, Elena Cantero-Navarro, Carolina Lavoz, Marta Fierro-Fernández, Jonay Poveda, Sandra Rayego-Mateos, Raúl R. Rodrigues-Diez, José Luis Morgado-Pascual, Macarena Orejudo, Sergio Mezzano, and Marta Ruiz-Ortega
- Subjects
Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
The Notch pathway regulates key processes in the kidney involved in embryonic development and tissue damage. Local activation of the Notch pathway has been described in many human chronic renal diseases, and has been suggested that several Notch pathway components could be considered as biomarkers of renal damage. Experimental studies done using Notch-components genetic modified mice as well as pharmacological approaches, such as γ-secretase inhibitors that block Notch pathway activation, have demonstrated the role of this pathway in renal regeneration, podocyte apoptosis, proliferation and fibroblasts activation, and induction of epithelial to mesenchymal transition of tubular epithelial cells. Recent studies suggest an interaction between Notch and NF-κB pathways in the regulation of the renal inflammatory process. Additionally, some miRNAs could regulate Notch components and down-stream responses. All these data suggest that blockade of the Notch signaling pathway could be a novel therapeutic option for renal diseases. Resumen: La vía de Notch regula procesos importantes en el riñón implicados en el desarrollo embrionario y en situaciones de agresión tisular. Así, en una gran variedad de nefropatías crónicas humanas se ha descrito una activación local de este sistema, sugiriendo que algunos de sus componentes podrían ser biomarcadores de daño renal. Los estudios realizados en modelos experimentales, modulando genéticamente componentes de la vía Notch o mediante su bloqueo farmacológico con inhibidores de la γ-secretasa, han demostrado la participación de esta vía en la regeneración renal, en la apoptosis de podocitos, en la proliferación y activación de fibroblastos y en la transición epitelio-mesenquimal de las células tubuloepiteliales. Estudios recientes sugieren una interacción entre las vías Notch y NF-κB, la cual podría jugar un papel relevante en el proceso inflamatorio renal. Por otra parte, en los últimos años se han descrito miRNA que son capaces de regular componentes de la vía Notch y modular sus respuestas. Todos estos datos indican que el bloqueo de la vía de señalización Notch podría representar una nueva opción terapéutica para la enfermedad renal. Keywords: Notch, Renal damage, Mechanisms, Fibrosis, Inflammation, Palabras clave: Notch, Daño renal, Mecanismos, Fibrosis, Inflamación
- Published
- 2018
- Full Text
- View/download PDF
14. Análisis de la vía Notch como una posible diana terapéutica en la patología renal
- Author
-
Laura Marquez-Exposito, Elena Cantero-Navarro, Carolina Lavoz, Marta Fierro-Fernández, Jonay Poveda, Sandra Rayego-Mateos, Raúl R. Rodrigues-Diez, José Luis Morgado-Pascual, Macarena Orejudo, Sergio Mezzano, and Marta Ruiz-Ortega
- Subjects
Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Resumen: La vía de Notch regula procesos importantes en el riñón implicados en el desarrollo embrionario y en situaciones de agresión tisular. Así, en una gran variedad de nefropatías crónicas humanas se ha descrito una activación local de este sistema, sugiriendo que algunos de sus componentes podrían ser biomarcadores de daño renal. Los estudios realizados en modelos experimentales, modulando genéticamente componentes de la vía Notch o mediante su bloqueo farmacológico con inhibidores de la γ-secretasa, han demostrado la participación de esta vía en la regeneración renal, en la apoptosis de podocitos, en la proliferación y activación de fibroblastos y en la transición epitelio-mesenquimal de las células tubuloepiteliales. Estudios recientes sugieren una interacción entre las vías Notch y NF-κB, la cual podría jugar un papel relevante en el proceso inflamatorio renal. Por otra parte, en los últimos años se han descrito miRNA que son capaces de regular componentes de la vía Notch y modular sus respuestas. Todos estos datos indican que el bloqueo de la vía de señalización Notch podría representar una nueva opción terapéutica para la enfermedad renal. Abstract: Notch pathway regulates key processes in the kidney, involved in embryonic development and tissue damage. In many human chronic renal diseases a local activation of Notch pathway has been described, suggesting that several components of Notch pathway could be considered as biomarkers of renal damage. Experimental studies by genetic modulation of Notch components or pharmacological approaches by γ-secretase inhibitors have demonstrated the role of this pathway in renal regeneration renal, podocyte apoptosis, proliferation and fibroblasts activation, and induction of epithelial to mesenchymal transition of tubular epithelial cells. Recent studies suggest an interaction between Notch and NF-κB pathway involved in the regulation of renal inflammatory process. On the other hand, there are some miRNAs that could regulate Notch components and down-stream responses. All these data suggest that Notch blockade could be a novel therapeutic option for renal diseases. Palabras clave: Notch, Daño renal, Mecanismos, Fibrosis, Inflamación, Keywords: Notch, Renal damage, Mechanisms, Fibrosis, Inflammation
- Published
- 2018
- Full Text
- View/download PDF
15. CCN2 Aggravates the Immediate Oxidative Stress–DNA Damage Response following Renal Ischemia–Reperfusion Injury
- Author
-
Floris A. Valentijn, Sebastiaan N. Knoppert, Georgios Pissas, Raúl R. Rodrigues-Diez, Laura Marquez-Exposito, Roel Broekhuizen, Michal Mokry, Lennart A. Kester, Lucas L. Falke, Roel Goldschmeding, Marta Ruiz-Ortega, Theodoros Eleftheriadis, and Tri Q. Nguyen
- Subjects
acute kidney injury ,ischemia–reperfusion injury ,oxidative stress response ,DNA damage response ,cell cycle arrest ,cellular communication network factor 2 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
AKI, due to the fact of altered oxygen supply after kidney transplantation, is characterized by renal ischemia–reperfusion injury (IRI). Recent data suggest that AKI to CKD progression may be driven by cellular senescence evolving from prolonged DNA damage response (DDR) following oxidative stress. Cellular communication factor 2 (CCN2, formerly called CTGF) is a major contributor to CKD development and was found to aggravate DNA damage and the subsequent DDR–cellular senescence–fibrosis sequence following renal IRI. We therefore investigated the impact of CCN2 inhibition on oxidative stress and DDR in vivo and in vitro. Four hours after reperfusion, full transcriptome RNA sequencing of mouse IRI kidneys revealed CCN2-dependent enrichment of several signaling pathways, reflecting a different immediate stress response to IRI. Furthermore, decreased staining for γH2AX and p-p53 indicated reduced DNA damage and DDR in tubular epithelial cells of CCN2 knockout (KO) mice. Three days after IRI, DNA damage and DDR were still reduced in CCN2 KO, and this was associated with reduced oxidative stress, marked by lower lipid peroxidation, protein nitrosylation, and kidney expression levels of Nrf2 target genes (i.e., HMOX1 and NQO1). Finally, silencing of CCN2 alleviated DDR and lipid peroxidation induced by anoxia-reoxygenation injury in cultured PTECs. Together, our observations suggest that CCN2 inhibition might mitigate AKI by reducing oxidative stress-induced DNA damage and the subsequent DDR. Thus, targeting CCN2 might help to limit post-IRI AKI.
- Published
- 2021
- Full Text
- View/download PDF
16. Gremlin Regulates Tubular Epithelial to Mesenchymal Transition via VEGFR2: Potential Role in Renal Fibrosis
- Author
-
Laura Marquez-Exposito, Carolina Lavoz, Raul R. Rodrigues-Diez, Sandra Rayego-Mateos, Macarena Orejudo, Elena Cantero-Navarro, Alberto Ortiz, Jesús Egido, Rafael Selgas, Sergio Mezzano, Marta Ruiz-Ortega, UAM. Departamento de Medicina, and Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD)
- Subjects
0301 basic medicine ,Notch ,Medicina ,Notch signaling pathway ,Tubular cells ,Proinflammatory cytokine ,03 medical and health sciences ,Downregulation and upregulation ,Fibrosis ,Renal fibrosis ,Medicine ,Pharmacology (medical) ,Epithelial–mesenchymal transition ,Renal ,Original Research ,Pharmacology ,Gremlin ,business.industry ,lcsh:RM1-950 ,fibrosis ,EMT ,medicine.disease ,tubular cells ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,VEGFR2 ,Cancer research ,renal ,business ,Gremlin (protein) ,Wound healing - Abstract
Chronic kidney disease (CKD) is emerging as an important health problem due to the increase number of CKD patients and the absence of an effective curative treatment. Gremlin has been proposed as a novel therapeutic target for renal inflammatory diseases, acting via Vascular Endothelial Growth Factor Receptor-2 (VEGFR2). Although many evidences suggest that Gremlin could regulate renal fibrosis, the receptor involved has not been yet clarified. Gremlin, as other TGF-β superfamily members, regulates tubular epithelial to mesenchymal transition (EMT) and, therefore, could contribute to renal fibrosis. In cultured tubular epithelial cells Gremlin binding to VEGFR2 is linked to proinflammatory responses. Now, we have found out that in these cells VEGFR2 is also involved in the profibrotic actions of Gremlin. VEGFR2 blockade by a pharmacological kinase inhibitor or gene silencing diminished Gremlin-mediated gene upregulation of profibrotic factors and restored changes in EMT-related genes. Moreover, VEGFR2 inhibition blocked EMT phenotypic changes and dampened the rate of wound healing in response to Gremlin. The role of VEGFR2 in experimental fibrosis was evaluated in experimental unilateral ureteral obstruction. VEFGR2 inhibition diminished the upregulation of profibrotic genes and EMT changes, as well as the accumulation of extracellular matrix proteins, such as fibronectin and collagens in the obstructed kidneys. Notch pathway activation participates in renal damage progression by regulating cell growth/proliferation, regeneration and inflammation. In cultured tubular epithelial cells, Notch inhibition markedly downregulated Gremlin-induced EMT changes and wound healing speed. These results show that Gremlin regulates the EMT process via VEGFR2 and Notch pathway activation, suggesting that the Gremlin/VEGFR2 axis could be a potential therapeutic target for CKD., This work was supported by grants from the Instituto de Salud Carlos III (ISCIII) and Fondos FEDER European Union (PI16/02057, PI17/00119, PI17/01495, and Red de Investigación Renal REDINREN: RD16/0009), Sociedad Española de Nefrologia, “NOVELREN-CM: Enfermedad renal crónica: nuevas Estrategias para la prevención, Diagnóstico y tratamiento”; B2017/BMD-3751, B2017/BMD-3686 CIFRA2-CM, PAI 82140017, and FONDECYT 1160465 (Chile) and Bayer HealthCare AG (Grants4Targets initiative, Berlin, Germany).
17. Role of Epidermal Growth Factor Receptor (EGFR) and Its Ligands in Kidney Inflammation and Damage.
- Author
-
Rayego-Mateos S, Rodrigues-Diez R, Morgado-Pascual JL, Valentijn F, Valdivielso JM, Goldschmeding R, and Ruiz-Ortega M
- Subjects
- Animals, Connective Tissue Growth Factor metabolism, ErbB Receptors genetics, Humans, Inflammation genetics, Kidney immunology, Kidney pathology, Signal Transduction genetics, Signal Transduction physiology, ErbB Receptors metabolism, Inflammation immunology, Inflammation metabolism, Kidney metabolism
- Abstract
Chronic kidney disease (CKD) is characterized by persistent inflammation and progressive fibrosis, ultimately leading to end-stage renal disease. Although many studies have investigated the factors involved in the progressive deterioration of renal function, current therapeutic strategies only delay disease progression, leaving an unmet need for effective therapeutic interventions that target the cause behind the inflammatory process and could slow down or reverse the development and progression of CKD. Epidermal growth factor receptor (EGFR) (ERBB1), a membrane tyrosine kinase receptor expressed in the kidney, is activated after renal damage, and preclinical studies have evidenced its potential as a therapeutic target in CKD therapy. To date, seven official EGFR ligands have been described, including epidermal growth factor (EGF) (canonical ligand), transforming growth factor- α , heparin-binding epidermal growth factor, amphiregulin, betacellulin, epiregulin, and epigen. Recently, the connective tissue growth factor (CTGF/CCN2) has been described as a novel EGFR ligand. The direct activation of EGFR by its ligands can exert different cellular responses, depending on the specific ligand, tissue, and pathological condition. Among all EGFR ligands, CTGF/CCN2 is of special relevance in CKD. This growth factor, by binding to EGFR and downstream signaling pathway activation, regulates renal inflammation, cell growth, and fibrosis. EGFR can also be "transactivated" by extracellular stimuli, including several key factors involved in renal disease, such as angiotensin II, transforming growth factor beta (TGFB), and other cytokines, including members of the tumor necrosis factor superfamily, showing another important mechanism involved in renal pathology. The aim of this review is to summarize the contribution of EGFR pathway activation in experimental kidney damage, with special attention to the regulation of the inflammatory response and the role of some EGFR ligands in this process. Better insights in EGFR signaling in renal disease could improve our current knowledge of renal pathology contributing to therapeutic strategies for CKD development and progression.
- Published
- 2018
- Full Text
- View/download PDF
18. Epigenetic Modification Mechanisms Involved in Inflammation and Fibrosis in Renal Pathology.
- Author
-
Morgado-Pascual JL, Marchant V, Rodrigues-Diez R, Dolade N, Suarez-Alvarez B, Kerr B, Valdivielso JM, Ruiz-Ortega M, and Rayego-Mateos S
- Subjects
- Animals, DNA Methylation genetics, DNA Methylation physiology, Epigenesis, Genetic physiology, Fibrosis genetics, Histone Code genetics, Histone Code physiology, Humans, Kidney metabolism, Kidney pathology, Kidney Failure, Chronic, MicroRNAs, Renal Insufficiency, Chronic genetics, Epigenesis, Genetic genetics, Inflammation genetics
- Abstract
The growing incidence of obesity, hypertension, and diabetes, coupled with the aging of the population, is increasing the prevalence of renal diseases in our society. Chronic kidney disease (CKD) is characterized by persistent inflammation, fibrosis, and loss of renal function leading to end-stage renal disease. Nowadays, CKD treatment has limited effectiveness underscoring the importance of the development of innovative therapeutic options. Recent studies have identified how epigenetic modifications participate in the susceptibility to CKD and have explained how the environment interacts with the renal cell epigenome to contribute to renal damage. Epigenetic mechanisms regulate critical processes involved in gene regulation and downstream cellular responses. The most relevant epigenetic modifications that play a critical role in renal damage include DNA methylation, histone modifications, and changes in miRNA levels. Importantly, these epigenetic modifications are reversible and, therefore, a source of potential therapeutic targets. Here, we will explain how epigenetic mechanisms may regulate essential processes involved in renal pathology and highlight some possible epigenetic therapeutic strategies for CKD treatment.
- Published
- 2018
- Full Text
- View/download PDF
19. Inhibition of Bromodomain and Extraterminal Domain Family Proteins Ameliorates Experimental Renal Damage.
- Author
-
Suarez-Alvarez B, Morgado-Pascual JL, Rayego-Mateos S, Rodriguez RM, Rodrigues-Diez R, Cannata-Ortiz P, Sanz AB, Egido J, Tharaux PL, Ortiz A, Lopez-Larrea C, and Ruiz-Ortega M
- Subjects
- Animals, Chromosomal Proteins, Non-Histone physiology, Disease Models, Animal, Kidney Diseases etiology, Male, Mice, Mice, Inbred C57BL, Nuclear Proteins physiology, Transcription Factors physiology, Azepines pharmacology, Azepines therapeutic use, Chromosomal Proteins, Non-Histone antagonists & inhibitors, Kidney Diseases drug therapy, Nuclear Proteins antagonists & inhibitors, Transcription Factors antagonists & inhibitors, Triazoles pharmacology, Triazoles therapeutic use
- Abstract
Renal inflammation has a key role in the onset and progression of immune- and nonimmune-mediated renal diseases. Therefore, the search for novel anti-inflammatory pharmacologic targets is of great interest in renal pathology. JQ1, a small molecule inhibitor of bromodomain and extraterminal (BET) proteins, was previously found to preserve renal function in experimental polycystic kidney disease. We report here that JQ1-induced BET inhibition modulated the in vitro expression of genes involved in several biologic processes, including inflammation and immune responses. Gene silencing of BRD4, an important BET protein, and chromatin immunoprecipitation assays showed that JQ1 alters the direct association of BRD4 with acetylated histone-packaged promoters and reduces the transcription of proinflammatory genes (IL-6, CCL-2, and CCL-5). In vivo, JQ1 abrogated experimental renal inflammation in murine models of unilateral ureteral obstruction, antimembrane basal GN, and infusion of Angiotensin II. Notably, JQ1 downregulated the expression of several genes controlled by the NF-κB pathway, a key inflammatory signaling pathway. The RelA NF-κB subunit is activated by acetylation of lysine 310. In damaged kidneys and cytokine-stimulated renal cells, JQ1 reduced the nuclear levels of RelA NF-κB. Additionally, JQ1 dampened the activation of the Th17 immune response in experimental renal damage. Our results show that inhibition of BET proteins reduces renal inflammation by several mechanisms: chromatin remodeling in promoter regions of specific genes, blockade of NF-κB pathway activation, and modulation of the Th17 immune response. These results suggest that inhibitors of BET proteins could have important therapeutic applications in inflammatory renal diseases., (Copyright © 2017 by the American Society of Nephrology.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.