Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell Commun Signal ; 22(1): 264, 2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38734696

RESUMEN

BACKGROUND: Traditional biomarkers of chronic kidney disease (CKD) detect the disease in its late stages and hardly predict associated vascular damage. Integrin-linked kinase (ILK) is a scaffolding protein and a serine/threonine protein kinase that plays multiple roles in several pathophysiological processes during renal damage. However, the involvement of ILK as a biomarker of CKD and its associated vascular problems remains to be fully elucidated. METHODS: CKD was induced by an adenine-rich diet for 6 weeks in mice. We used an inducible ILK knockdown mice (cKD-ILK) model to decrease ILK expression. ILK content in mice's peripheral blood mononuclear cells (PBMCs) was determined and correlated with renal function parameters and with the expression of ILK and fibrosis and inflammation markers in renal and aortic tissues. Also, the expression of five miRNAs that target ILK was analyzed in whole blood of mice. RESULTS: The adenine diet increased ILK expression in PBMCs, renal cortex, and aortas, and creatinine and urea nitrogen concentrations in the plasma of WT mice, while these increases were not observed in cKD-ILK mice. Furthermore, ILK content in PBMCs directly correlated with renal function parameters and with the expression of renal and vascular ILK and fibrosis and inflammation markers. Finally, the expression of the five miRNAs increased in the whole blood of adenine-fed mice, although only four correlated with plasma urea nitrogen, and of those, three were downregulated in cKD-ILK mice. CONCLUSIONS: ILK, in circulating mononuclear cells, could be a potential biomarker of CKD and CKD-associated renal and vascular damage.


Asunto(s)
Biomarcadores , Riñón , Leucocitos Mononucleares , Proteínas Serina-Treonina Quinasas , ARN Mensajero , Insuficiencia Renal Crónica , Animales , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Leucocitos Mononucleares/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Biomarcadores/metabolismo , Biomarcadores/sangre , Ratones , Riñón/patología , Riñón/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/sangre , MicroARNs/metabolismo , Modelos Animales de Enfermedad , Fibrosis
2.
Bioorg Med Chem ; 44: 116295, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34246920

RESUMEN

Protein tyrosine phosphatase (PTP1B) is an interesting therapeutical target for diabetes, obesity, heart disease and cancer. As such, inhibition of PTP1B using orally administered drugs is still being pursued by academia and pharmaceutical companies. The failure of catalytic-site inhibitors led to the focus in this field being switched to allosteric inhibitors. To date, the non-competitive inhibitors that have reached clinical trials target the site formed by the α3/α6/α7 tunnel or the site found in a disordered C-terminal non-catalytic segment. Herein, pyrrolo[1,2-a]quinoxal-5-inium salts and 4,5-dihydropyrrolo[1,2-a]quinoxalines are synthesized from pyrrolo[1,2-a]quinoxalines by alkylation and reduction, respectively. These compounds showed no toxicity in HepG2 cells and exhibited inhibitory activity against PTP1B, with inhibition percentages of between 37% and 53% at 1 µM and activities (IC50) of between 0.25 and 1.90 µM. The inhibitory activity against T-cell protein tyrosine phosphatase (TC-TPT) was also assayed, with 4,5-dihydropyrrolo[1,2-a]quinoxalines being found to be slightly more active and selective. Compounds from the two series behave as insulin mimetics since they exhibit enhancement of glucose uptake in C2C12 cells. Computational docking studies provide information about the putative binding mode for both series and the preference for the α3/α6/α7 allosteric tunnel.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Simulación del Acoplamiento Molecular , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Pirroles/farmacología , Quinoxalinas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Pirroles/síntesis química , Pirroles/química , Quinoxalinas/síntesis química , Quinoxalinas/química , Sales (Química)/síntesis química , Sales (Química)/química , Sales (Química)/farmacología , Relación Estructura-Actividad
3.
Cell Physiol Biochem ; 54(1): 71-87, 2020 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-31972071

RESUMEN

BACKGROUND/AIMS: Diabetes type 2, metabolic syndrome or non-alcoholic fatty liver disease are insulin resistance-related metabolic disorders, which lack a better prognosis before their full establishment. We studied the importance of the intracellular scaffold protein integrin linked kinaes (ILK) as a key modulator in the initial pathogenesis and the early progression of those insulin resistance- related disorders. METHODS: Adult mice with a global transgenic downregulation of ILK expression (cKD-ILK) and littermates without that depletion (CT) were fed with either standard (STD) or high fat (HFD) diets during 2 and 6 weeks. Weights, blood glucose and other systemic biochemical parameters were determined in animals under fasting conditions and after glucose or pyruvate intraperitoneal injections to test their tolerance. In RNA or proteins extracted from insulin-sensitive tissues, we determined by reverse transcription-quantitative PCR and western blot the expression of ILK, metabolites transporters and other metabolism and inflammatory markers. Glucose uptake capacity was studied in freshly isolated tissues. RESULTS: HFD feeding was able to early and progressively increase glycaemia, insulinemia, circulating glycerol, body weight gain, liver-mediated gluconeogenesis along this time lapse, but cKD-ILK have all these systemic misbalances exacerbated compared to CT in the same HFD time lapse. Interestingly, the tisular expression of ILK in HFD-fed CT was dramatically downregulated in white adipose tissue (WAT), skeletal muscle and liver at the same extent of the original ILK downregulation of cKD-ILK. We previously published that basal STD-fed cKD-ILK compared to basal STD-CT have different expression of glucose transporters GLUT4 in WAT and skeletal muscle. In the same STD-fed cKD-ILK, we observed here the increased expressions of hepatic GLUT2 and WAT pro-inflammatory cytokines TNF-α and MCP-1. The administration of HFD exacerbated the expression changes in cKD-ILK of these and other markers related to the imbalanced metabolism observed, such as WAT lipolysis (HSL), hepatic gluconeogenesis (PCK-1) and glycerol transport (AQP9). CONCLUSION: ILK expression may be taken as a predictive determinant of metabolic disorders establishment, because its downregulation seems to correlate with the early imbalance of glucose and glycerol transport and the subsequent loss of systemic homeostasis of these metabolites.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Regulación hacia Abajo , Enfermedades Metabólicas/etiología , Proteínas Serina-Treonina Quinasas/genética , Animales , Femenino , Gluconeogénesis , Inflamación/etiología , Inflamación/genética , Resistencia a la Insulina , Lipólisis , Masculino , Enfermedades Metabólicas/genética , Ratones , Ratones Endogámicos BALB C
4.
FASEB J ; 32(2): 920-934, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29054855

RESUMEN

Ras proteins regulate cell survival, growth, differentiation, blood pressure, and fibrosis in some organs. We have demonstrated that H- ras gene deletion produces mice hypotension via a soluble guanylate cyclase-protein kinase G (PKG)-dependent mechanism. In this study, we analyzed the consequences of H- ras deletion on cardiac remodeling induced by continuous angiotensin II (AngII) infusion and the molecular mechanisms implied. Left ventricular posterior wall thickness and mass and cardiomyocyte cross-sectional area were similar between AngII-treated H-Ras knockout (H -ras-/-) and control wild-type (H -ras+/+) mice, as were extracellular matrix protein expression. Increased cardiac PKG-Iß protein expression in H -ras-/- mice suggests the involvement of this protein in heart protection. Ex vivo experiments on cardiac explants could support this mechanism, as PKG blockade blunted protection against AngII-induced cardiac hypertrophy and fibrosis markers in H -ras-/- mice. Genetic modulation studies in cardiomyocytes and cardiac and embryonic fibroblasts revealed that the lack of H-Ras down-regulates the B-RAF/MEK/ERK pathway, which induces the glycogen synthase kinase-3ß-dependent activation of the transcription factor, cAMP response element-binding protein, which is responsible for PKG-Iß overexpression in H -ras-/- mouse embryonic fibroblasts. This study demonstrates that H- ras deletion protects against AngII-induced cardiac remodeling, possibly via a mechanism in which PKG-Iß overexpression could play a partial role, and points to H-Ras and/or downstream proteins as potential therapeutic targets in cardiovascular disease.-Martín-Sánchez, P., Luengo, A., Griera, M., Orea, M. J., López-Olañeta, M., Chiloeches, A., Lara-Pezzi, E., de Frutos, S., Rodríguez-Puyol, M., Calleros, L., Rodríguez-Puyol, D. H- ras deletion protects against angiotensin II-induced arterial hypertension and cardiac remodeling through protein kinase G-Iß pathway activation.


Asunto(s)
Angiotensina II/efectos adversos , Cardiomegalia/enzimología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Hipertensión/enzimología , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas p21(ras)/deficiencia , Angiotensina II/farmacología , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/prevención & control , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/patología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Fibroblastos/enzimología , Fibroblastos/patología , Eliminación de Gen , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipertensión/inducido químicamente , Hipertensión/patología , Ratones , Ratones Noqueados
5.
Mol Med ; 21(1): 873-885, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26562149

RESUMEN

Soluble guanylyl cyclase (sGC) is activated by nitric oxide (NO) and produces cGMP, which activates cGMP-dependent protein kinases (PKG) and is hydrolyzed by specific phosphodiesterases (PDE). The vasodilatory and cytoprotective capacity of cGMP-axis activation results in a therapeutic strategy for several pathologies. Integrin-linked kinase (ILK), a major scaffold protein between the extracellular matrix and intracellular signaling pathways, may modulate the expression and functionality of the cGMP-axis-related proteins. We introduce ILK as a novel modulator in renal homeostasis as well as a potential target for cisplatin (CIS)-induced acute kidney injury (AKI) improvement. We used an adult mice model of depletion of ILK (cKD-ILK), which showed basal increase of sGC and PKG expressions and activities in renal cortex when compared with wildtype (WT) littermates. Twenty-four h activation of sGC activation with NO enhanced the filtration rate in cKD-ILK. During AKI, cKD-ILK maintained the cGMP-axis upregulation with consequent filtration rates enhancement and ameliorated CIS-dependent tubular epithelial-to-mesenchymal transition and inflammation and markers. To emphasize the role of cGMP-axis upregulation due to ILK depletion, we modulated the cGMP axis under AKI in vivo and in renal cultured cells. A suboptimal dose of the PDE inhibitor ZAP enhanced the beneficial effects of the ILK depletion in AKI mice. On the other hand, CIS increased contractility-related events in cultured glomerular mesangial cells and necrosis rates in cultured tubular cells; ILK depletion protected the cells while sGC blockade with ODQ fully recovered the damage.

6.
FASEB J ; 28(8): 3645-59, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24784577

RESUMEN

One of the clinical alterations observed in chronic renal disease (CRD) is the impaired urine concentration, known as diabetes insipidus (DI). Tubulointerstitial fibrosis of the kidney is also a pathological finding observed in CRD and involves composition of extracellular matrix (ECM). However, an association between these two events has not been elucidated. In this study, we showed that the extracellular-to-intracellular scaffold protein integrin-linked kinase (ILK) regulates expression of tubular water channel aquaporin-2 (AQP2) and its apical membrane presence in the renal tubule. Basally, polyuria and decreased urine osmolality were present in ILK conditional-knockdown (cKD-ILK) adult mice compared with nondepleted ILK littermates. No changes were observed in arginine-vasopressin (AVP) blood levels, renal receptor (V2R), or AQP3 expression. However, tubular AQP2 was decreased in expression and apical membrane presence in cKD-ILK mice, where the canonical V2R/cAMP axis activation is still functional, but independent of the absence of ILK. Thus, cKD-ILK constitutes a nephrogenic diabetes insipidus (NDI) model. AQP2 and ILK colocalize in cultured inner medullary collecting duct (mIMCD3) cells. Specific ILK siRNAs and collagen I (Col) decrease ILK and AQP2 levels and AQP2 presence on the membrane of tubular mIMCD3 cells, which impairs the capacity of the cells to transport water under hypotonic stress. The present work points to ILK as a therapeutic target in NDI.


Asunto(s)
Acuaporina 2/fisiología , Agua Corporal/metabolismo , Proteínas de la Matriz Extracelular/fisiología , Capacidad de Concentración Renal/fisiología , Túbulos Renales Colectores/metabolismo , Poliuria/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Acuaporina 2/biosíntesis , Acuaporina 2/genética , Acuaporina 3/biosíntesis , Acuaporina 3/genética , Arginina Vasopresina/sangre , Transporte Biológico Activo , Membrana Celular/química , Polaridad Celular , Células Cultivadas , Colágeno Tipo I/farmacología , Desamino Arginina Vasopresina/farmacología , Diabetes Insípida Nefrogénica/metabolismo , Modelos Animales de Enfermedad , Túbulos Renales Colectores/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Concentración Osmolar , Presión Osmótica/fisiología , Fosforilación , Poliuria/genética , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Receptores de Vasopresinas/biosíntesis , Receptores de Vasopresinas/genética
7.
Biomater Sci ; 11(14): 4916-4929, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37306667

RESUMEN

White adipose tissue (WAT) hypertrophy is caused by the excessive storage of triglycerides (TGs) and is associated with obesity. We previously demonstrated that extracellular matrix mediator integrin beta1 (INTB1) and its downstream effector integrin linked kinase (ILK) are implicated in obesity establishment. We also considered in our previous works that ILK upregulation is a therapeutical strategy to reduce WAT hypertrophy. Carbon based nanomaterials (CNMs) have interesting potential to modify cell differentiation but have been never studied to change the properties of adipocytes. METHODS: GMC is a new graphene-based CNM that was tested for biocompatibility and functionality in cultured adipocytes. MTT, TG content, lipolysis quantification, and transcriptional changes were determined. Specific INTB1 blocking antibody and ILK depletion with specific siRNA were used to study the intracellular signalling. We complemented the study using subcutaneous WAT (scWAT) explants from transgenic ILK knockdown mice (cKD-ILK). GMC was topically administrated in the dorsal area of high fat diet-induced obese rats (HFD) for 5 consecutive days. The scWAT weights and some intracellular markers were analyzed after the treatment. RESULTS: graphene presence was characterized in GMC. It was non-toxic and effective in reducing TG content in vitro in a dose-dependent manner. GMC rapidly phosphorylated INTB1 and increased the expression and activity of hormone sensitive lipase (HSL), the lipolysis subproduct glycerol, and the expression of glycerol and fatty acid transporters. GMC also reduced the expression of adipogenesis markers. Pro-inflammatory cytokines were unaffected. ILK was overexpressed, and INTB1 or ILK blockade avoided functional GMC effects. Topical administration of GMC in HFD rats overexpressed ILK in scWAT, and their weight gains were reduced, while systemic (renal, hepatic) toxicity parameters were unaffected. CONCLUSIONS: GMC is safe and effective in reducing hypertrophied scWAT weight when topically applied and it can be considered of interest in anti-obesogenic strategies. GMC increases lipolysis and reduces adipogenesis inside adipocytes by mechanisms that imply the activation of INTB1, the overexpression of ILK, and changes in the expression and activity of several markers related to fat metabolism.


Asunto(s)
Grafito , Lipólisis , Ratones , Ratas , Animales , Glicerol , Aumento de Peso , Obesidad/tratamiento farmacológico , Obesidad/etiología , Obesidad/metabolismo , Ratones Transgénicos , Hipertrofia/complicaciones , Integrinas
8.
Eur J Pharmacol ; 946: 175654, 2023 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-36930883

RESUMEN

Hypertension is the most common cause of left ventricular hypertrophy, contributing to heart failure progression. Candesartan (Cand) is an angiotensin receptor antagonist widely used for hypertension treatment. Structural modifications were previously performed by our group using Zinc (ZnCand) as a strategy for improving its pharmacological properties. The measurements showed that ZnCand exerts a stronger interaction with the angiotensin II receptor, type 1 (AT1 receptor), reducing oxidative stress and intracellular calcium flux, a mechanism implied in cell contraction. These results were accompanied by the reduction of the contractile capacity of mesangial cells. In vivo experiments showed that the complex causes a significant decrease in systolic blood pressure after 8 weeks of treatment in spontaneously hypertensive rats (SHR). The reduction of heart hypertrophy was evidenced by echocardiography, the histologic cross-sectional area of cardiomyocytes, collagen content, the B-type natriuretic peptide (BNP) marker and connective tissue growth factor (CTGF) and the matrix metalloproteinase 2 (MMP-2) expression. Besides, the complex restored the redox status. In this study, we demonstrated that the complexation with Zn(II) improves the antihypertensive and cardiac effects of the parental drug.


Asunto(s)
Antihipertensivos , Hipertensión , Hipertrofia Ventricular Izquierda , Zinc , Animales , Ratas , Antihipertensivos/química , Antihipertensivos/farmacología , Compuestos de Bifenilo/farmacología , Presión Sanguínea , Hipertensión/complicaciones , Hipertensión/tratamiento farmacológico , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Metaloproteinasa 2 de la Matriz , Miocitos Cardíacos , Ratas Endogámicas SHR , Tetrazoles/farmacología , Tetrazoles/uso terapéutico , Zinc/farmacología
9.
Arterioscler Thromb Vasc Biol ; 31(3): 567-73, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21164076

RESUMEN

OBJECTIVE: Hydrogen peroxide (H(2)O(2)) is an important mediator in the vasculature, but its role in the regulation of soluble guanylate cyclase (sGC) activity and expression is not completely understood. The aim of this study was to test the effect of H(2)O(2) on sGC expression and function and to explore the molecular mechanism involved. METHODS AND RESULTS: H(2)O(2) increased sGCß1 protein steady-state levels in rat aorta and aortic smooth muscle cells (RASMCs) in a time- and dose-dependent manner, and this effect was blocked by catalase. sGCα2 expression increased along with ß1 subunit, whereas α1 subunit remained unchanged. Vascular relaxation to an NO donor (sodium nitroprusside) was enhanced by H(2)O(2), and it was prevented by ODQ (sGC inhibitor). cGMP production in both freshly isolated vessels and RASMCs exposed to H(2)O(2) was greatly increased after sodium nitroprusside treatment. The H(2)O(2)-dependent sGCß1 upregulation was attributable to sGCß1 mRNA stabilization, conditioned by the translocation of the mRNA-binding protein HuR from the nucleus to the cytosol, and the increased mRNA binding of HuR to the sGCß1 3' untranslated region. HuR silencing reversed the effects of H(2)O(2) on sGCß1 levels and cGMP synthesis. CONCLUSIONS: Our results identify H(2)O(2) as an endogenous mediator contributing to the regulation of vascular tone and point to a key role of HuR in sGCß1 mRNA stabilization.


Asunto(s)
Antígenos de Superficie/metabolismo , Guanilato Ciclasa/genética , Peróxido de Hidrógeno/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Estabilidad del ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Regiones no Traducidas 3' , Animales , Antígenos de Superficie/genética , Aorta/efectos de los fármacos , Aorta/enzimología , Sitios de Unión , Catalasa/metabolismo , Células Cultivadas , GMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Proteínas ELAV , Proteína 1 Similar a ELAV , Inhibidores Enzimáticos/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Transporte de Proteínas , Estabilidad del ARN/efectos de los fármacos , Proteínas de Unión al ARN/genética , Ratas , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/metabolismo , Guanilil Ciclasa Soluble , Factores de Tiempo , Regulación hacia Arriba , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
10.
Exp Mol Med ; 54(3): 226-238, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35246616

RESUMEN

Cardiovascular disease is an important cause of death in patients with chronic kidney disease (CKD). Protein-bound uremic toxins, such as p-cresyl and indoxyl sulfate (IS), are poorly removed during hemodialysis, leading to vascular endothelial dysfunction and leukocyte extravasation. These processes can be related to dynamic adhesion structures called podosomes. Several studies have indicated the role of integrin-linked kinase (ILK) in the accumulation of integrin-associated proteins in podosomes. Here, we investigated the involvement of ILK and podosome formation in the adhesion and extravasation of monocytes under p-cresol (pc) and IS exposure. Incubation of THP-1 human monocyte cells with these toxins upregulated ILK kinase activity. Together, both toxins increased cell adhesion, podosome formation, extracellular matrix degradation, and migration of THP-1 cells, whereas ILK depletion with specific small interfering RNAs suppressed these processes. Interestingly, F-actin colocalized with cortactin in podosome cores, while ILK was colocalized in podosome rings under toxin stimulation. Podosome Wiskott-Aldrich syndrome protein (WASP)-interacting protein (WIP) and AKT protein depletion demonstrated that monocyte adhesion depends on podosome formation and that the ILK/AKT signaling pathway is involved in these processes. Ex vivo experiments showed that both toxins induced adhesion and podosome formation in leukocytes from wild-type mice, whereas these effects were not observed in leukocytes of conditional ILK-knockdown animals. In summary, under pc and IS stimulation, monocytes increase podosome formation and transmigratory capacity through an ILK/AKT signaling pathway-dependent mechanism, which could lead to vascular injury. Therefore, ILK could be a potential therapeutic target for the treatment of vascular damage associated with CKD.


Asunto(s)
Podosomas , Proteínas Serina-Treonina Quinasas , Animales , Adhesión Celular , Cresoles , Proteínas del Citoesqueleto/metabolismo , Humanos , Indicán/metabolismo , Indicán/farmacología , Ratones , Monocitos , Podosomas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células THP-1
11.
Am J Physiol Cell Physiol ; 300(4): C907-18, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21209362

RESUMEN

Glomerular diseases are characterized by a sustained synthesis and accumulation of abnormal extracellular matrix proteins, such as collagen type I. The extracellular matrix transmits information to cells through interactions with membrane components, which directly activate many intracellular signaling events. Moreover, accumulating evidence suggests that eicosanoids derived from cyclooxygenase (COX)-2 participate in a number of pathological processes in immune-mediated renal diseases, and it is known that protein kinase B (AKT) may act through different transcription factors in the regulation of the COX-2 promoter. The present results show that progressive accumulation of collagen I in the extracellular medium induces a significant increase of COX-2 expression in human mesangial cells, resulting in an enhancement in PGE(2) production. COX-2 overexpression is due to increased COX-2 mRNA levels. The study of the mechanism implicated in COX-2 upregulation by collagen I showed focal adhesion kinase (FAK) activation. Furthermore, we observed that the activation of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway by collagen I and collagen I-induced COX-2 overexpression was abolished by PI3K and AKT inhibitors. Additionally, we showed that the cAMP response element (CRE) transcription factor is implicated. Finally, we studied COX-2 expression in an animal model, N(G)-nitro-l-arginine methyl ester hypertensive rats. In renal tissue and vascular walls, COX-2 and collagen type I content were upregulated. In summary, our results provide evidence that collagen type I increases COX-2 expression via the FAK/PI3K/AKT/cAMP response element binding protein signaling pathway.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Matriz Extracelular/química , Regulación Enzimológica de la Expresión Génica , Células Mesangiales/enzimología , Animales , Células Cultivadas , Colágeno/química , Colágeno/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclooxigenasa 2/genética , Dinoprostona/metabolismo , Matriz Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Células Mesangiales/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Transducción de Señal/fisiología
12.
Am J Physiol Cell Physiol ; 300(3): C683-91, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21160032

RESUMEN

The nitric oxide (NO)-soluble guanylate cyclase (sGC) pathway exerts most of its cellular actions through the activation of the cGMP-dependent protein kinase (PKG). Accumulation of extracellular matrix is one of the main structural changes in pathological conditions characterized by a decreased activity of this pathway, such as hypertension, diabetes, or aging, and it is a well-known fact that extracellular matrix proteins modulate cell phenotype through the interaction with membrane receptors such as integrins. The objectives of this study were 1) to evaluate whether extracellular matrix proteins, particularly fibronectin (FN), modulate PKG expression in contractile cells, 2) to analyze the mechanisms involved, and 3) to evaluate the functional consequences. FN increased type I PKG (PKG-I) protein content in human mesangial cells, an effect dependent on the interaction with ß(1)-integrin. The FN upregulation of PKG-I protein content was due to increased mRNA expression, determined by augmented transcriptional activity of the PKG-I promoter region. Akt and the transcription factor CCAAT enhancer-binding protein (C/EBP) mediated the genesis of these changes. FN also increased PKG-I in another type of contractile cell, rat vascular smooth muscle cells (RVSMC). Tirofiban, a pharmacological analog of FN, increased PKG-I protein content in RVSMC and rat aortic walls and magnified the hypotensive effect of dibutyryl cGMP in conscious Wistar rats. The present results provide evidence of a mechanism able to increase PKG-I protein content in contractile cells. Elucidation of this novel mechanism provides a rationale for future pharmacotherapy in certain vascular diseases.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/biosíntesis , Fibronectinas/fisiología , Contracción Muscular/fisiología , Músculo Liso Vascular/fisiología , Activación Transcripcional/fisiología , Regulación hacia Arriba/fisiología , Animales , Aorta Torácica/enzimología , Aorta Torácica/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Células Cultivadas , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Fibronectinas/metabolismo , Humanos , Masculino , Células Mesangiales/citología , Células Mesangiales/enzimología , Células Mesangiales/fisiología , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
13.
ChemMedChem ; 16(18): 2895-2906, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34137509

RESUMEN

Protein tyrosine phosphatase 1B (PTP1B) is a very promising target for the treatment of metabolic disorders such as type II diabetes mellitus. Although it was validated as a promising target for this disease more than 30 years ago, as yet there is no drug in advanced clinical trials, and its biochemical mechanism and functions are still being studied. In the present study, based on our experience generating PTP1B inhibitors, we have developed and implemented a scaffold-hopping approach to vary the pyrrole ring of the pyrrolo[1,2-a]quinoxaline core, supported by extensive computational techniques aimed to explain the molecular interaction with PTP1B. Using a combination of docking, molecular dynamics and end-point free-energy calculations, we have rationally designed a hypothesis for new PTP1B inhibitors, supporting their recognition mechanism at a molecular level. After the design phase, we were able to easily synthesize proposed candidates and their evaluation against PTP1B was found to be in good concordance with our predictions. Moreover, the best candidates exhibited glucose uptake increments in cellulo model, thus confirming their utility for PTP1B inhibition and validating this approach for inhibitors design and molecules thus obtained.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Pirroles/farmacología , Quinoxalinas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Pirroles/síntesis química , Pirroles/química , Quinoxalinas/síntesis química , Quinoxalinas/química , Relación Estructura-Actividad
14.
ChemMedChem ; 15(19): 1788-1801, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32909701

RESUMEN

PTP1B dephosphorylates insulin receptor and substrates to modulate glucose metabolism. This enzyme is a validated therapeutic target for type 2 diabetes, but no current drug candidates have completed clinical trials. Pyrrolo[1,2-a]quinoxalines substituted at positions C1-C4 and/or C7-C8 were found to be nontoxic to cells and good inhibitors in the low- to sub-micromolar range, with the 4-benzyl derivative being the most potent inhibitor (0.24 µm). Some analogues bearing chlorine atoms at C7 and/or C8 kept potency and showed good selectivity compared to TCPTP (selectivity index >40). The most potent inhibitors behaved as insulin mimetics by increasing glucose uptake. The 4-benzyl derivative inhibited insulin receptor substrate 1 and AKT phosphorylation. Molecular docking and molecular dynamics simulations supported a putative binding mode for these compounds to the allosteric α3/α6/α7 pocket, but inconsistent results in enzyme inhibition kinetics were obtained due to the high tendency of these inhibitors to form stable aggregates. Computational calculations supported the druggability of inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Insulina/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Pirroles/farmacología , Quinoxalinas/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Glucosa/metabolismo , Células Hep G2 , Humanos , Ratones , Modelos Moleculares , Estructura Molecular , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Pirroles/síntesis química , Pirroles/química , Quinoxalinas/síntesis química , Quinoxalinas/química , Relación Estructura-Actividad
15.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1284-1297, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30726718

RESUMEN

Kidney fibrosis is one of the main pathological findings of progressive chronic kidney disease (CKD) although the pathogenesis of renal scar formation remains incompletely explained. Integrin-linked kinase (ILK), a major scaffold protein between the extracellular matrix (ECM) and intracellular signaling pathways, is involved in several pathophysiological processes during renal damage. However, ILK contribution in the CKD progress remains to be fully elucidated. In the present work, we studied 1) the renal functional and structural consequences of CKD genesis and progression when ILK is depleted and 2) the potential of ILK depletion as a therapeutic approach to delay CKD progression. We induced an experimental CKD model, based on an adenine-supplemented diet on adult wild-type (WT) and ILK-depleted mice, with a tubulointerstitial damage profile resembling that is observed in human CKD. The adenine diet induced in WT mice a progressive increase in plasma creatinine and urea concentrations. In the renal cortex it was also observed tubular damage, interstitial fibrosis and progressive increased ECM components, pro-inflammatory and chemo-attractant cytokines, EMT markers and TGF-ß1 expressions. These observations were highly correlated to a simultaneous increase of ILK expression and activity. In adenine-fed transgenic ILK-depleted mice, all these changes were prevented. Additionally, we evaluated the potential role of ILK depletion to be applied after the disease induction, as an effective approach to interventions in human CKD subjects. In this scenario, two weeks after the establishment of adenine-induced CKD, ILK was abrogated in WT mice and stabilized renal damage, avoiding CKD progression. We propose ILK to be a potential target to delay renal disease progression.


Asunto(s)
Adenina/administración & dosificación , Técnicas de Silenciamiento del Gen , Túbulos Renales/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Insuficiencia Renal Crónica/genética , Actinas/genética , Actinas/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Creatinina/sangre , Dieta , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibrosis , Regulación de la Expresión Génica , Humanos , Túbulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/metabolismo , Insuficiencia Renal Crónica/inducido químicamente , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Urea/sangre
16.
Nefrologia (Engl Ed) ; 38(6): 639-646, 2018.
Artículo en Inglés, Español | MEDLINE | ID: mdl-30337107

RESUMEN

BACKGROUND: Patients with chronic kidney disease present with an accumulation of uraemic toxins, which have been identified as pathogenic agents associated with cardiovascular mortality, which is very high is this patient group. A phenomenon common to the progressive renal dysfunction and associated vascular damage, is the abnormal accumulation of extracellular matrix (ECM) proteins in the renal or vascular structures. OBJECTIVE: To determine the contribution of uraemia or the uraemic toxins to the production of cytokinins and ECM in aortas of uraemic animals or human aortic smooth muscle cells (HASMCs). MATERIALS AND METHODS: Mice were used with uraemia induced by a diet rich in adenine (0.2%) for 2, 4 or 6 weeks. Kidney function was evaluated by means of urine volume, plasma levels of creatinine, urea, fractional excretion of sodium, and vascular damage using histology, as well as protein expression using RT-qPCR. The HASMCs were incubated in vitro with uraemic toxins: p-cresol 10-100 (µg/ml) and indoxyl-sulphate25-100 (µg/ml) alone or simultaneously. The protein expression was evaluated using Western blot and confocal microscopy. RESULTS: The administration of adenine produced progressive kidney damage in the mice, thickening of the aortic wall, and increasing the expression of TGF-ß1 and ECM proteins. The toxins at high doses and combined also induced the expression of TGF-ß1 and ECM proteins by the HASMCs. CONCLUSIONS: The uraemia produced by an adenine rich diet or high doses of uraemic toxins induced the abnormal deposit of ECM proteins in the vascular wall or its production by HASMCs. The understanding of the mechanisms that underlie this pathophysiological process may be useful in the prevention of cardiovascular damage associated with the progress of chronic kidney disease, a disease, at the moment that is irreversible and occasional silent until its diagnosis in advanced stages.


Asunto(s)
Vasos Sanguíneos/patología , Citocinas/fisiología , Proteínas de la Matriz Extracelular/fisiología , Insuficiencia Renal Crónica/complicaciones , Uremia/complicaciones , Adenina/administración & dosificación , Animales , Fibrosis/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Toxinas Biológicas/fisiología , Factor de Crecimiento Transformador beta1/fisiología
17.
Eur J Med Chem ; 157: 946-959, 2018 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-30165342

RESUMEN

A series of new azolopyrimidine-peptide hybrids and indolomethylideneimidazolones were obtained and evaluated as calpain inhibitors. The hybrid compounds were inactive, whereas some members of the initial azolomethylideneimidazolone series showed interesting calpain inhibitory activity. By using 4b as a hit compound, a new series of analogs were synthesized by an efficient synthetic procedure based on a multicomponent reaction followed by an unprecedented reaction at the methylene position of the molecule. The best inhibitor found for calpain I (IC50 = 20 nM) was about 20 times more potent than the hit compound. Studies on 4b showed that its inhibition is consistent with an uncompetitive inhibition mode. This compound did not exhibit cellular toxicity at any of the doses tested (0.1-10 µM) and further studies indicated that it was capable of blockading chemical ischemia induction of apoptosis by preventing sodium azide-dependent calpain activation in intact human kidney tubular epithelial cells. The results of molecular modeling studies rationalized the inhibitory activity found for this series and account, from a structural point of view, for the most active compound identified (4j).


Asunto(s)
Azoles/farmacología , Calpaína/antagonistas & inhibidores , Descubrimiento de Drogas , Glicoproteínas/química , Glicoproteínas/farmacología , Imidazolidinas/farmacología , Péptidos/farmacología , Apoptosis/efectos de los fármacos , Azoles/química , Calpaína/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Glicoproteínas/síntesis química , Humanos , Imidazolidinas/química , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Modelos Moleculares , Estructura Molecular , Péptidos/química , Relación Estructura-Actividad
18.
Circ Res ; 97(11): 1115-23, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16239590

RESUMEN

Many forms of vascular disease are characterized by increased transforming growth factor (TGF)-beta1 expression and endothelial dysfunction. Smad proteins are a key step in TGF-beta-initiated signal transduction. We hypothesized that NO may regulate endothelial TGF-beta-dependent gene expression. We show that NO inhibits TGF-beta/Smad-regulated gene transactivation in a cGMP-dependent manner. NO effects were mimicked by a soluble analogue of cGMP. Inhibition of cGMP-dependent protein kinase 1 (PKG-1) or overexpression of dominant-negative PKG-1alpha suppressed NO/cGMP inhibition of TGF-beta-induced gene expression. Inversely, overexpression of PKG-1alpha catalytic subunit blocked TGF-beta-induced gene transactivation. Furthermore NO delayed and reduced phosphorylated Smad2/3 nuclear translocation, an effect mediated by PKG-1, whereas NG-nitro-L-arginine methyl ester augmented Smad phosphorylation and gene expression in response to TGF-beta. Aortas from endothelial NO synthase-deficient mice showed enhanced basal TGF-beta1 and collagen type I expression; endothelial cells from these animals showed increased Smad phosphorylation and transcriptional activity. Proteasome inhibitors prevented the inhibitory effect of NO on TGF-beta signaling. NO reduced the metabolic life of ectopically expressed Smad2 and enhanced its ubiquitination. Taken together, these results suggest that the endothelial NO/cGMP/PKG pathway interferes with TGF-beta/Smad2 signaling by directing the proteasomal degradation of activated Smad.


Asunto(s)
Células Endoteliales/fisiología , Óxido Nítrico/fisiología , Transducción de Señal/fisiología , Proteína Smad2/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Bovinos , Células Cultivadas , GMP Cíclico/fisiología , Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , Humanos , Ratones , Complejo de la Endopetidasa Proteasomal/fisiología , Proteína smad3/fisiología , Activación Transcripcional
19.
Cardiovasc Res ; 69(2): 359-69, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16360131

RESUMEN

OBJECTIVES: Alterations in NO/cGMP signaling have been associated with vascular dysfunction. Here, we tested whether peptides containing arginine-glycine-aspartic acid (RGD) motifs, commonly found on the binding sites of extracellular matrix to integrins, could increase the expression and function of soluble guanylate cyclase (sGC) in human mesangial cell (HMC), and human aortic smooth muscle (HASMC) cells. METHODS AND RESULTS: Arginine-glycine-aspartic acid-serine (RGDS) promoted an up-regulation in the sGC beta1 subunit steady-state level, both in HMC and HASMC, in a time- and dose-dependent manner. The cellular effects of RGDS-stimulation of sGC expression was an enhanced cellular response to sodium nitroprusside, resulting in elevated cGMP levels and vasodilator-stimulated phosphoprotein (VASP) phosphorylation in both kinds of cells, and an increased NO relaxing effect on cells precontracted with H(2)O(2) or Angiotensin II. Moreover, RGDS was able to restore the sGC levels that had been previously decreased by long term exposure to NO donors. RGDS effects on sGC regulation were due to the specific interaction with alpha(5)beta(1) integrin. To investigate the intracellular mechanisms activated after RGDS cell treatment, pharmacological kinase inhibitors were used. The effect of RGDS on sGC protein content was completely abolished by treating the cells with c-Jun N-terminal kinase (JNK) inhibitors. In addition, c-fos and c-jun were found in the cell nuclei after RGDS treatment, suggesting that the RGDS effect could be mediated by the AP-1 transcription factor. CONCLUSION: Results provide evidence of a mechanism able to increase the sGC protein content linked to increased activity in contractile cells, not only in basal conditions, but also after the down-regulation of the receptor by its own substrate. Elucidation of this novel mechanism provides a rationale for future pharmacotherapy in certain vascular diseases.


Asunto(s)
Guanilato Ciclasa/metabolismo , Músculo Liso Vascular/enzimología , Oligopéptidos/metabolismo , Regulación hacia Arriba , Aorta , Células Cultivadas , Activación Enzimática , Expresión Génica , Humanos , Células Mesangiales , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Biochim Biophys Acta Gene Regul Mech ; 1860(9): 922-935, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28736155

RESUMEN

Two processes are associated with progressive loss of renal function: 1) decreased aquaporin-2 (AQP2) expression and urinary concentrating capacity (Nephrogenic Diabetes Insipidus, NDI); and 2) changes in extracellular matrix (ECM) composition, e.g. increased collagen I (Col I) deposition, characteristic of tubule-interstitial fibrosis. AQP2 expression is regulated by both the ECM-to-intracellular scaffold protein integrin-linked kinase (ILK) by NFATc/AP1 and other transcription factors. In the present work, we used in vivo and in vitro approaches to examine ILK participation in NFATc3/AP-1-mediated increases in AQP2 gene expression. Both NFATc3 knock-out mice and ILK conditional-knockdown mice (cKD-ILK) display symptoms of NDI (polyuria and reduced AQP2 expression). NFATc3 is upregulated in the renal medulla tubular cells of cKD-ILK mice but with reduced nuclear localization. Inner medullary collecting duct mIMCD3 cells were subjected to ILK depletion and transfected with reporter plasmids. Pharmacological activators or inhibitors determined the effect of ILK activity on NFATc/AP-1-dependent increases in transcription of AQP2. Finally, mIMCD3 cultured on Col I showed reduced activity of the ILK/GSK3ß/NFATc/AQP2 axis, suggesting this pathway is a potential target for therapeutic treatment of NDI.


Asunto(s)
Acuaporina 2/genética , Factores de Transcripción NFATC/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transcripción Genética/genética , Animales , Línea Celular , Diabetes Insípida Nefrogénica/genética , Diabetes Insípida Nefrogénica/metabolismo , Integrinas/metabolismo , Médula Renal/metabolismo , Túbulos Renales Colectores/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Poliuria/genética , Poliuria/metabolismo , Factor de Transcripción AP-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA