Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 590: 158-162, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-34974305

RESUMEN

The progression of chronic kidney disease (CKD) increases the risks of cardiovascular morbidity and end-stage kidney disease. Indoxyl sulfate (IS), which is derived from dietary l-tryptophan by the action of bacterial l-tryptophan indole-lyase (TIL) in the gut, serves as a uremic toxin that exacerbates CKD-related kidney disorder. A mouse model previously showed that inhibition of TIL by 2-aza-l-tyrosine effectively reduced the plasma IS level, causing the recovery of renal damage. In this study, we found that (+)-sesamin and related lignans, which occur abundantly in sesame seeds, inhibit intestinal bacteria TILs. Kinetic studies revealed that (+)-sesamin and sesamol competitively inhibited Escherichia coli TIL (EcTIL) with Ki values of 7 µM and 14 µM, respectively. These Ki values were smaller than that of 2-aza-l-tyrosine (143 µM). Molecular docking simulation of (+)-sesamin- (or sesamol-)binding to EcTIL predicted that these inhibitors potentially bind near the active site of EcTIL, where the cofactor pyridoxal 5'-phosphate is bound, consistent with the kinetic results. (+)-Sesamin is a phytochemical with a long history of consumption and is generally regarded as safe. Hence, dietary supplementation of (+)-sesamin encapsulated in enteric capsules could be a promising mechanism-based strategy to prevent CKD progression. Moreover, the present findings would provide a new structural basis for designing more potent TIL inhibitors for the development of mechanism-based therapeutic drugs to treat CKD.


Asunto(s)
Dioxoles/farmacología , Inhibidores Enzimáticos/farmacología , Microbioma Gastrointestinal , Lignanos/farmacología , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/etiología , Sesamum/química , Triptofanasa/antagonistas & inhibidores , Benzodioxoles/química , Benzodioxoles/farmacología , Dioxoles/química , Microbioma Gastrointestinal/efectos de los fármacos , Cinética , Lignanos/química , Simulación del Acoplamiento Molecular , Fenoles/química , Fenoles/farmacología , Triptofanasa/metabolismo
2.
Circ Res ; 127(3): 360-375, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32284030

RESUMEN

RATIONALE: Hypoxia promotes renal damage and progression of chronic kidney disease (CKD). The erythrocyte is the only cell type for oxygen (O2) delivery. Sphingosine 1-phosphate (S1P)-a highly enriched biolipid in erythrocytes-is recently reported to be induced under high altitude in normal humans to enhance O2 delivery. However, nothing is known about erythrocyte S1P in CKD. OBJECTIVE: To investigate the function and metabolic basis of erythrocyte S1P in CKD with a goal to explore potential therapeutics. METHODS AND RESULTS: Using erythrocyte-specific SphK1 (sphingosine kinase 1; the only enzyme to produce S1P in erythrocytes) knockout mice (eSphK1-/-) in an experimental model of hypertensive CKD with Ang II (angiotensin II) infusion, we found severe renal hypoxia, hypertension, proteinuria, and fibrosis in Ang II-infused eSphk1-/- mice compared with controls. Untargeted metabolomics profiling and in vivo U-13C6 isotopically labeled glucose flux analysis revealed that SphK1 is required for channeling glucose metabolism toward glycolysis versus pentose phosphate pathway, resulting in enhanced erythroid-specific Rapoport-Luebering shunt in Ang II-infused mice. Mechanistically, increased erythrocyte S1P functioning intracellularly activates AMPK (AMP-activated protein kinase) 1α and BPGM (bisphosphoglycerate mutase) by reducing ceramide/S1P ratio and inhibiting PP2A (protein phosphatase 2A), leading to increased 2,3-bisphosphoglycerate (an erythrocyte-specific metabolite negatively regulating Hb [hemoglobin]-O2-binding affinity) production and thus more O2 delivery to counteract kidney hypoxia and progression to CKD. Preclinical studies revealed that an AMPK agonist or a PP2A inhibitor rescued the severe CKD phenotype in Ang II-infused eSphK1-/- mice and prevented development of CKD in the control mice by inducing 2,3-bisphosphoglycerate production and thus enhancing renal oxygenation. Translational research validated mouse findings in erythrocytes of hypertensive CKD patients and cultured human erythrocytes. CONCLUSIONS: Our study elucidates the beneficial role of eSphk1-S1P in hypertensive CKD by channeling glucose metabolism toward Rapoport-Luebering shunt and inducing 2,3-bisphosphoglycerate production and O2 delivery via a PP2A-AMPK1α signaling pathway. These findings reveal the metabolic and molecular basis of erythrocyte S1P in CKD and new therapeutic avenues.


Asunto(s)
Reprogramación Celular , Metabolismo Energético , Eritrocitos/metabolismo , Riñón/metabolismo , Insuficiencia Renal Crónica/sangre , Adulto , Animales , Estudios de Casos y Controles , Hipoxia de la Célula , Modelos Animales de Enfermedad , Eritrocitos/enzimología , Femenino , Fibrosis , Humanos , Hipertensión/complicaciones , Riñón/patología , Masculino , Metaboloma , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/patología
3.
Circ Res ; 127(5): 593-606, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32418507

RESUMEN

RATIONALE: Kidney homeostasis is critically determined by the coordinated activity of the renin-angiotensin system (RAS), including the balanced synthesis of its main effector peptides Ang (angiotensin) II and Ang (1-7). The condition of enzymatic overproduction of Ang II relative to Ang (1-7) is termed RAS dysregulation and leads to cellular signals, which promote hypertension and organ damage, and ultimately progressive kidney failure. ACE2 (angiotensin-converting enzyme 2) and NEP (neprilysin) induce the alternative, and potentially reno-protective axis by enhancing Ang (1-7) production. However, their individual contribution to baseline RAS balance and whether their activities change in chronic kidney disease (CKD) has not yet been elucidated. OBJECTIVE: To examine whether NEP-mediated Ang (1-7) generation exceeds Ang II formation in the healthy kidney compared with diseased kidney. METHODS AND RESULTS: In this exploratory study, we used liquid chromatography-tandem mass spectrometry to measure Ang II and Ang (1-7) synthesis rates of ACE, chymase and NEP, ACE2, PEP (prolyl-endopeptidase), PCP (prolyl-carboxypeptidase) in kidney biopsy homogenates in 11 healthy living kidney donors, and 12 patients with CKD. The spatial expression of RAS enzymes was determined by immunohistochemistry. Healthy kidneys showed higher NEP-mediated Ang (1-7) synthesis than Ang II formation, thus displaying a strong preference towards the reno-protective alternative RAS axis. In contrast, in CKD kidneys higher levels of Ang II were recorded, which originated from mast cell chymase activity. CONCLUSIONS: Ang (1-7) is the dominant RAS peptide in healthy human kidneys with NEP rather than ACE2 being essential for its generation. Severe RAS dysregulation is present in CKD dictated by high chymase-mediated Ang II formation. Kidney RAS enzyme analysis might lead to novel therapeutic approaches for CKD.


Asunto(s)
Angiotensina II/metabolismo , Angiotensina I/metabolismo , Riñón/enzimología , Neprilisina/metabolismo , Fragmentos de Péptidos/metabolismo , Insuficiencia Renal Crónica/enzimología , Sistema Renina-Angiotensina , Anciano , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Estudios de Casos y Controles , Quimasas/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Riñón/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neprilisina/antagonistas & inhibidores
4.
J Am Soc Nephrol ; 32(3): 597-613, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33510039

RESUMEN

BACKGROUND: The ubiquitin-proteasome system (UPS) and the autophagy-lysosomal system (APLS) are major intracellular degradation procedures. The importance of the APLS in podocytes is established, but the role of the UPS is not well understood. METHODS: To investigate the role of the UPS in podocytes, mice were generated that had deletion of Rpt3 (Rpt3pdKO), which encodes an essential regulatory subunit required for construction of the 26S proteasome and its deubiquitinating function. RESULTS: Rpt3pdKO mice showed albuminuria and glomerulosclerosis, leading to CKD. Impairment of proteasome function caused accumulation of ubiquitinated proteins and of oxidative modified proteins, and it induced podocyte apoptosis. Although impairment of proteasome function normally induces autophagic activity, the number of autophagosomes was lower in podocytes of Rpt3pdKO mice than in control mice, suggesting the autophagic activity was suppressed in podocytes with impairment of proteasome function. In an in vitro study, antioxidant apocynin and autophagy activator rapamycin suppressed podocyte apoptosis induced by proteasome inhibition. Moreover, rapamycin ameliorated the glomerular injury in the Rpt3pdKO mice. The accumulation of ubiquitinated proteins and of oxidative modified proteins, which were detected in the podocytes of Rpt3pdKO mice, is a characteristic feature of aging. An aging marker was increased in the podocytes of Rpt3pdKO mice, suggesting that impairment of proteasome function promoted signs of aging in podocytes. CONCLUSIONS: Impairment of proteasome function in podocytes led to CKD, and antioxidants and autophagy activators can be therapeutic agents for age-dependent CKD.


Asunto(s)
Podocitos/enzimología , Complejo de la Endopetidasa Proteasomal/deficiencia , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/etiología , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Apoptosis/efectos de los fármacos , Autofagia , Bortezomib/farmacología , Células Cultivadas , Glomeruloesclerosis Focal y Segmentaria/enzimología , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/patología , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Podocitos/efectos de los fármacos , Podocitos/patología , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma/farmacología , Agregado de Proteínas , Insuficiencia Renal Crónica/patología , Sirolimus/farmacología , Ubiquitinación
5.
J Am Soc Nephrol ; 32(11): 2834-2850, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34716244

RESUMEN

BACKGROUND: CKD, characterized by retained uremic solutes, is a strong and independent risk factor for thrombosis after vascular procedures . Urem ic solutes such as indoxyl sulfate (IS) and kynurenine (Kyn) mediate prothrombotic effect through tissue factor (TF). IS and Kyn biogenesis depends on multiple enzymes, with therapeutic implications unexplored. We examined the role of indoleamine 2,3-dioxygenase-1 (IDO-1), a rate-limiting enzyme of kynurenine biogenesis, in CKD-associated thrombosis after vascular injury. METHODS: IDO-1 expression in mice and human vessels was examined. IDO-1-/- mice, IDO-1 inhibitors, an adenine-induced CKD, and carotid artery injury models were used. RESULTS: Both global IDO-1-/- CKD mice and IDO-1 inhibitor in wild-type CKD mice showed reduced blood Kyn levels, TF expression in their arteries, and thrombogenicity compared with respective controls. Several advanced IDO-1 inhibitors downregulated TF expression in primary human aortic vascular smooth muscle cells specifically in response to uremic serum. Further mechanistic probing of arteries from an IS-specific mouse model, and CKD mice, showed upregulation of IDO-1 protein, which was due to inhibition of its polyubiquitination and degradation by IS in vascular smooth muscle cells. In two cohorts of patients with advanced CKD, blood IDO-1 activity was significantly higher in sera of study participants who subsequently developed thrombosis after endovascular interventions or vascular surgery. CONCLUSION: Leveraging genetic and pharmacologic manipulation in experimental models and data from human studies implicate IS as an inducer of IDO-1 and a perpetuator of the thrombotic milieu and supports IDO-1 as an antithrombotic target in CKD.


Asunto(s)
Indicán/fisiología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/sangre , Quinurenina/fisiología , Terapia Molecular Dirigida , Complicaciones Posoperatorias/enzimología , Insuficiencia Renal Crónica/enzimología , Trombosis/enzimología , Procedimientos Quirúrgicos Vasculares/efectos adversos , Animales , Aorta , Traumatismos de las Arterias Carótidas/complicaciones , Trombosis de las Arterias Carótidas/etiología , Trombosis de las Arterias Carótidas/prevención & control , Medios de Cultivo/farmacología , Inducción Enzimática/efectos de los fármacos , Retroalimentación Fisiológica , Femenino , Células HEK293 , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/deficiencia , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Quinurenina/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/efectos de los fármacos , Complicaciones Posoperatorias/sangre , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/prevención & control , Insuficiencia Renal Crónica/tratamiento farmacológico , Tromboplastina/metabolismo , Trombosis/sangre , Trombosis/etiología , Trombosis/prevención & control , Triptófano/metabolismo , Uremia/sangre
6.
Am J Physiol Renal Physiol ; 320(5): F706-F718, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33719570

RESUMEN

Cellular metabolic rates in the kidney are critical for maintaining normal renal function. In a hypoxic milieu, cells rely on glycolysis to meet energy needs, resulting in the generation of pyruvate and NADH. In the absence of oxidative phosphorylation, the continuation of glycolysis is dependent on the regeneration of NAD+ from NADH accompanied by the fermentation of pyruvate to lactate. This reaction is catalyzed by lactate dehydrogenase (LDH) isoform A (LDHA), whereas LDH isoform B (LDHB) catalyzes the opposite reaction. LDH is widely used as a potential injury marker as it is released from damaged cells into the urine and serum; however, the precise isoform-specific cellular localization of the enzyme along the nephron has not been characterized. By combining immunohistochemistry results and single-cell RNA-sequencing data on healthy mouse kidneys, we identified that LDHA is primarily expressed in proximal segments, whereas LDHB is expressed in the distal parts of the nephron. In vitro experiments in mouse and human renal proximal tubule cells showed an increase in LDHA following hypoxia with no change in LDHB. Using immunofluorescence, we observed that the overall expression of both LDHA and LDHB proteins decreased following renal ischemia-reperfusion injury as well as in the adenine-diet-induced model of chronic kidney disease. Single-nucleus RNA-sequencing analyses of kidneys following ischemia-reperfusion injury revealed a significant decline in the number of cells expressing detectable levels of Ldha and Ldhb; however, cells that were positive showed increased average expression postinjury, which subsided during the recovery phase. These data provide information on the cell-specific expression of LDHA and LDHB in the normal kidney as well as following acute and chronic kidney disease.NEW & NOTEWORTHY Cellular release of lactate dehydrogenase (LDH) is being used as an injury marker; however, the exact localization of LDH within the nephron remains unclear. We show that LDH isoform A is expressed proximally, whereas isoform B is expressed distally. Both subunit expressions were significantly altered in models of acute kidney injury and chronic kidney disease. Our study provides new insights into basal and postinjury renal lactate metabolism.


Asunto(s)
Lesión Renal Aguda/enzimología , Riñón/enzimología , L-Lactato Deshidrogenasa/metabolismo , Insuficiencia Renal Crónica/enzimología , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Animales , Biomarcadores/metabolismo , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Humanos , Isoenzimas , Riñón/patología , L-Lactato Deshidrogenasa/genética , Masculino , Ratones Endogámicos C57BL , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Factores de Tiempo
7.
Am J Physiol Renal Physiol ; 320(3): F285-F296, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33346727

RESUMEN

This study investigated the molecular mechanisms underlying the antiproteinuric effect of DPP4 inhibition in 5/6 renal ablation rats and tested the hypothesis that the urinary activity of DPP4 correlates with chronic kidney disease (CKD) progression. Experiments were conducted in male Wistar rats who underwent 5/6 nephrectomy (Nx) or sham operation followed by 8 wk of treatment with the DPP4 inhibitor (DPP4i) sitagliptin or vehicle. Proteinuria increased progressively in Nx rats throughout the observation period. This increase was remarkably mitigated by sitagliptin. Higher levels of proteinuria in Nx rats compared to control rats were accompanied by higher urinary excretion of retinol-binding protein 4, a marker of tubular proteinuria, as well as higher urinary levels of podocin, a marker of glomerular proteinuria. Retinol-binding protein 4 and podocin were not detected in the urine of Nx + DPP4i rats. Tubular and glomerular proteinuria was associated with the reduced expression of megalin and podocin in the renal cortex of Nx rats. Sitagliptin treatment partially prevented this decrease. Besides, the angiotensin II renal content was significantly reduced in the Nx rats that received sitagliptin compared to vehicle-treated Nx rats. Interestingly, both urinary DPP4 activity and abundance increased progressively in Nx rats. Additionally, urinary DPP4 activity correlated positively with serum creatinine levels, proteinuria, and blood pressure. Collectively, these results suggest that DPP4 inhibition ameliorated both tubular and glomerular proteinuria and prevented the reduction of megalin and podocin expression in CKD rats. Furthermore, these findings suggest that urinary DPP4 activity may serve as a biomarker of renal disease and progression.


Asunto(s)
Dipeptidil Peptidasa 4/orina , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón/efectos de los fármacos , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteínas de la Membrana/metabolismo , Proteinuria/prevención & control , Insuficiencia Renal Crónica/prevención & control , Fosfato de Sitagliptina/farmacología , Angiotensina II/metabolismo , Animales , Biomarcadores/orina , Modelos Animales de Enfermedad , Fibrosis , Riñón/enzimología , Riñón/patología , Masculino , Proteinuria/enzimología , Proteinuria/patología , Proteinuria/orina , Ratas Wistar , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/orina , Proteínas Plasmáticas de Unión al Retinol/orina , Transducción de Señal
8.
Am J Physiol Renal Physiol ; 320(1): F61-F73, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33196323

RESUMEN

Oxidative stress is a key concept in basic, translational, and clinical research to understand the pathophysiology of various disorders, including cardiovascular and renal diseases. Although attempts to directly reduce oxidative stress with redox-active substances have until now largely failed to prove clinical benefit, indirect approaches to combat oxidative stress enzymatically have gained further attention as potential therapeutic strategies. The pantetheinase Vanin-1 is expressed on kidney proximal tubular cells, and its reaction product cysteamine is described to negatively affect redox homeostasis by inhibiting the replenishment of cellular antioxidative glutathione stores. Vanin-1-deficient mice were shown to be protected against oxidative stress damage. The aim of this study was to elucidate whether pharmacological inhibition of Vanin-1 protects mice from oxidative stress-related acute or chronic kidney injury as well. By studying renal ischemia-reperfusion injury in Col4α3-/- (Alport syndrome) mice and in vitro hypoxia-reoxygenation in human proximal tubular cells we found that treatment with a selective and potent Vanin-1 inhibitor resulted in ample inhibition of enzymatic activity in vitro and in vivo. However, surrogate parameters of metabolic and redox homeostasis were only partially and insufficiently affected. Consequently, apoptosis and reactive oxygen species level in tubular cells as well as overall kidney function and fibrotic processes were not improved by Vanin-1 inhibition. We thus conclude that Vanin-1 functionality in the context of cardiovascular diseases needs further investigation and the biological relevance of pharmacological Vanin-1 inhibition for the treatment of kidney diseases remains to be proven.


Asunto(s)
Lesión Renal Aguda/prevención & control , Amidohidrolasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Nefritis Hereditaria/prevención & control , Estrés Oxidativo/efectos de los fármacos , Insuficiencia Renal Crónica/prevención & control , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/enzimología , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autoantígenos/genética , Autoantígenos/metabolismo , Línea Celular , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacocinética , Fibrosis , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis Hereditaria/enzimología , Nefritis Hereditaria/genética , Nefritis Hereditaria/patología , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética , Daño por Reperfusión/patología
9.
Clin Sci (Lond) ; 135(21): 2483-2502, 2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34643227

RESUMEN

Vascular calcification is highly prevalent in chronic kidney disease (CKD), and is characterized by transdifferentiation from contractile vascular smooth muscle cells (VSMCs) into an osteogenic phenotype. However, no effective and therapeutic option to prevent vascular calcification is yet available. Dihydromyricetin (DMY), a bioactive flavonoid isolated from Ampelopsis grossedentata, has been found to inhibit VSMCs proliferation and the injury-induced neointimal formation. However, whether DMY has an effect on osteogenic differentiation of VSMCs and vascular calcification is still unclear. In the present study, we sought to investigate the effect of DMY on vascular calcification in CKD and the underlying mechanism. DMY treatment significantly attenuated calcium/phosphate-induced calcification of rat and human VSMCs in a dose-dependent manner, as shown by Alizarin Red S staining and calcium content assay, associated with down-regulation of osteogenic markers including type I collagen (COL I), Runt-related transcription factor 2 (RUNX2), bone morphogenetic protein 2 (BMP2) and osteocalcin (OCN). These results were further confirmed in aortic rings ex vivo. Moreover, DMY ameliorated vascular calcification in rats with CKD. Additionally, we found that AKT signaling was activated during vascular calcification, whereas significantly inhibited by DMY administration. DMY treatment significantly reversed AKT activator-induced vascular calcification. Furthermore, inhibition of AKT signaling efficiently attenuated calcification, which was similar to that after treatment with DMY alone, and DMY had a better inhibitory effect on calcification as compared with AKT inhibitor. The present study demonstrated that DMY has a potent inhibitory role in vascular calcification partially by inhibiting AKT activation, suggesting that DMY may act as a promising therapeutic candidate for patients suffering from vascular calcification.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Flavonoles/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Insuficiencia Renal Crónica/tratamiento farmacológico , Calcificación Vascular/prevención & control , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Aorta/patología , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/patología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Masculino , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/patología , Transducción de Señal , Calcificación Vascular/enzimología , Calcificación Vascular/etiología , Calcificación Vascular/patología
10.
Pharmacol Res ; 170: 105629, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34089864

RESUMEN

Endoplasmic reticulum (ER) stress-mediated phenotypic switching of vascular smooth muscle cells (VSMCs) is key to vascular calcification (VC) in patients with chronic kidney disease (CKD). Studies have shown that activation/upregulation of SIRT1 has a protective effect on CKD-VC. Meanwhile, although terpinen-4-ol has been shown to exert a protective effect against cardiovascular disease, its role and underlying mechanism in VC remain unclear. Herein, we explored whether terpinen-4-ol alleviates ER stress-mediated VC through sirtuin 1 (SIRT1) and elucidated its mechanism to provide evidence for its application in the clinical prevention and treatment of VC. To this end, a CKD-related VC animal model and ß-glycerophosphate (ß-GP)-induced VSMC calcification model were established to investigate the role of terpinen-4-ol in ER stress-induced VC, in vitro and in vivo. Additionally, to evaluate the involvement of SIRT1, mouse and VSMC Sirt1-knockdown models were established. Results show that terpinen-4-ol inhibits calcium deposition, phenotypic switching, and ER stress in VSMCs in vitro and in vivo. Furthermore, pre-incubation of VSMCs with terpinen-4-ol or a SIRT1 agonist, decreased ß-GP-induced calcium salt deposition, increased SIRT1 protein level, and inhibited PERK-eIF2α-ATF4 pathway activation, thus, alleviating VC. Similar results were observed in VSMCs induced to overexpress SIRT1 via lentivirus transcription. Meanwhile, the opposite results were obtained in SIRT1-knockdown models. Further, results suggest that SIRT1 physically interacts with, and deacetylates PERK. Specifically, mass spectrometry analysis identified lysine K889 as the acetylation site of SIRT1, which regulates PERK. Finally, inhibition of SIRT1 reduced the effect of terpinen-4-ol on the deacetylation of PERK in vitro and in vivo and weakened the inhibitory effect of terpinen-4-ol against ER stress-mediated VC. Cumulatively, terpinen-4-ol was found to inhibit post-translational modification of PERK at the K889 acetylation site by upregulating SIRT1 expression, thereby ameliorating VC by regulating ER stress. This study provides insights into the underlying molecular mechanism of terpinen-4-ol, supporting its development as a promising therapeutic agent for CKD-VC.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Insuficiencia Renal Crónica/tratamiento farmacológico , Sirtuina 1/metabolismo , Terpenos/farmacología , Calcificación Vascular/prevención & control , eIF-2 Quinasa/metabolismo , Acetilación , Factor de Transcripción Activador 4/metabolismo , Animales , Modelos Animales de Enfermedad , Factor 2 Eucariótico de Iniciación/metabolismo , Ratones , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Fenotipo , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/patología , Sirtuina 1/genética , Calcificación Vascular/enzimología , Calcificación Vascular/etiología , Calcificación Vascular/patología
11.
J Am Soc Nephrol ; 31(5): 946-961, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32253273

RESUMEN

BACKGROUND: The serine/threonine kinases MST1 and MST2 are core components of the Hippo pathway, which has been found to be critically involved in embryonic kidney development. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are the pathway's main effectors. However, the biologic functions of the Hippo/YAP pathway in adult kidneys are not well understood, and the functional role of MST1 and MST2 in the kidney has not been studied. METHODS: We used immunohistochemistry to examine expression in mouse kidneys of MST1 and MST2, homologs of Hippo in Drosophila. We generated mice with tubule-specific double knockout of Mst1 and Mst2 or triple knockout of Mst1, Mst2, and Yap. PCR array and mouse inner medullary collecting duct cells were used to identify the primary target of Mst1/Mst2 deficiency. RESULTS: MST1 and MST2 were predominantly expressed in the tubular epithelial cells of adult kidneys. Deletion of Mst1/Mst2 in renal tubules increased activity of YAP but not TAZ. The kidneys of mutant mice showed progressive inflammation, tubular and glomerular damage, fibrosis, and functional impairment; these phenotypes were largely rescued by deletion of Yap in renal tubules. TNF-α expression was induced via both YAP-dependent and YAP-independent mechanisms, and TNF-α and YAP amplified the signaling activities of each other in the tubules of kidneys with double knockout of Mst1/Mst2. CONCLUSIONS: Our findings show that tubular Mst1/Mst2 deficiency leads to CKD through both the YAP and non-YAP pathways and that tubular YAP activation induces renal fibrosis. The pathogenesis seems to involve the reciprocal stimulation of TNF-α and YAP signaling activities.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas de Ciclo Celular/fisiología , Túbulos Renales/enzimología , Proteínas Serina-Treonina Quinasas/deficiencia , Insuficiencia Renal Crónica/enzimología , Animales , Células Cultivadas , Fibrosis , Regulación de la Expresión Génica , Vía de Señalización Hippo , Etiquetado Corte-Fin in Situ , Riñón/embriología , Riñón/enzimología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Serina-Treonina Quinasa 3 , Transducción de Señal , Transactivadores/fisiología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/fisiología , Proteínas Señalizadoras YAP
12.
Am J Physiol Renal Physiol ; 318(2): F475-F485, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31841390

RESUMEN

Tobacco smoking has been identified as a risk factor in the progression of chronic kidney disease (CKD). In previous studies, we showed that nicotine induces cyclooxygenase (COX)-2 expression in vivo and in vitro and that the administration of nicotine in vivo worsens the severity of renal injury in a model of subtotal renal ablation. In the present study, we tested the role of COX-2-derived prostaglandins on the deleterious effects of nicotine in CKD. Sham and 5/6 nephrectomy (5/6Nx) rats received tap water or nicotine (100 µg/mL) in the drinking water for 12 wk. Additional groups also systemically received the COX-2 inhibitor NS-398 (1.5 mg·kg-1·day-1 via osmotic minipump). The administration of nicotine worsened renal injury and proteinuria in 5/6Nx rats and increased proteinuria in sham rats. 5/6Nx rats had increased cortical production of the prostaglandins PGE2, PGI2, PGD2, and PGF2α and of thromboxane A2. In these rats, nicotine reduced the production of all prostaglandins examined except thromboxane A2. Treatment with the COX-2 inhibitor NS-398 resulted in complete inhibition of all prostaglandins studied and ameliorated renal injury and proteinuria in 5/6Nx rats on nicotine but not in 5/6 Nx rats on tap water. Nicotine also reduced the expression of megalin in all groups examined, and this was partially prevented by COX-2 inhibition. In the present study, we showed that in CKD, nicotine worsens renal injury at least in part by producing an imbalance in the production of prostaglandins. This imbalance in the production of prostaglandins likely plays a role in the deleterious effects of smoking on the progression of CKD.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Riñón/efectos de los fármacos , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Prostaglandinas/metabolismo , Insuficiencia Renal Crónica/inducido químicamente , Animales , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprost/metabolismo , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Epoprostenol/metabolismo , Riñón/enzimología , Riñón/patología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Nefrectomía , Prostaglandina D2/metabolismo , Proteinuria/inducido químicamente , Proteinuria/enzimología , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/prevención & control , Transducción de Señal , Tromboxano A2/metabolismo
13.
Am J Physiol Renal Physiol ; 319(4): F603-F611, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32830538

RESUMEN

The acyl-CoA synthetase medium-chain family member 2 (Acsm2) gene was first identified and cloned by our group as a kidney-specific "KS" gene. However, its expression pattern and function remain to be clarified. In the present study, we found that the Acsm2 gene was expressed specifically and at a high level in normal adult kidneys. Expression of Acsm2 in kidneys followed a maturational pattern: it was low in newborn mice and increased with kidney development and maturation. In situ hybridization and immunohistochemistry revealed that Acsm2 was expressed specifically in proximal tubular cells of adult kidneys. Data from the Encyclopedia of DNA Elements database revealed that the Acsm2 gene locus in the mouse has specific histone modifications related to the active transcription of the gene exclusively in kidney cells. Following acute kidney injury, partial unilateral ureteral obstruction, and chronic kidney diseases, expression of Acsm2 in the proximal tubules was significantly decreased. In human samples, the expression pattern of ACSM2A, a homolog of mouse Acsm2, was similar to that in mice, and its expression decreased with several types of renal injuries. These results indicate that the expression of Acsm2 parallels the structural and functional maturation of proximal tubular cells. Downregulation of its expression in several models of kidney disease suggests that Acms2 may serve as a novel marker of proximal tubular injury and/or dysfunction.


Asunto(s)
Coenzima A Ligasas/metabolismo , Células Epiteliales/metabolismo , Túbulos Renales Proximales/metabolismo , Proteínas Mitocondriales/metabolismo , Lesión Renal Aguda/enzimología , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Animales , Coenzima A Ligasas/genética , Modelos Animales de Enfermedad , Células Epiteliales/patología , Fibrosis , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Túbulos Renales Proximales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Renina/genética , Renina/metabolismo , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética , Daño por Reperfusión/patología
14.
Am J Physiol Renal Physiol ; 318(2): F388-F401, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31841388

RESUMEN

Cardiovascular disease (CVD) is the main cause of death in patients with kidney disease. Hypoxia plays a crucial role in the progression of chronic kidney disease (CKD) and cardiovascular disease, which is associated with fibrosis, inflammation, and oxidative injury. Previous studies have indicated that prolyl hydroxylase (PHD) inhibitors, stabilizers of hypoxia-inducible factors (HIFs), can be used to treat acute organ injuries such as renal ischemia-reperfusion, myocardial infarction, and, in some contexts, CKD. However, the effects of PHD inhibitors on cardiovascular complications in CKD remain unknown. In the present study, we investigated whether HIF activation has a beneficial effect on kidney and cardiovascular outcomes in the remnant kidney model. We used the 5/6 nephrectomy model with the nitric oxide synthase inhibitor Nω-nitro-l-arginine (20 mg/L in the drinking water). Rats received diet with 0.005% enarodustat (PHD inhibitor) or vehicle for 8 wk starting 2 wk before 5/6 nephrectomy. Activation of HIF by the PHD inhibitor reduced cardiac hypertrophy and ameliorated myocardial fibrosis in association with restored capillary density and improvement in mitochondrial morphology. With regard to kidneys, enarodustat ameliorated fibrosis in association with reduced proinflammatory cytokine expression, reduced apoptosis, and restored capillary density, even though renal endpoints such as proteinuria and serum creatinine levels were not significantly affected by enarodustat, except for blood urea nitrogen levels at 4 wk. In addition, cardiac hypertrophy marker genes, including atrial natriuretic peptide, were suppressed in P19CL6 cells treated with enarodustat. These findings suggest that PHD inhibitors might show beneficial effects in cardiovascular complications caused by CKD.


Asunto(s)
Hipertrofia Ventricular Izquierda/prevención & control , Riñón/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Glicinas N-Sustituídas/farmacología , Prolil Hidroxilasas/metabolismo , Inhibidores de Prolil-Hidroxilasa/farmacología , Piridinas/farmacología , Insuficiencia Renal Crónica/tratamiento farmacológico , Triazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mediadores de Inflamación/metabolismo , Riñón/enzimología , Riñón/patología , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/enzimología , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/ultraestructura , Neovascularización Fisiológica/efectos de los fármacos , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/patología , Transducción de Señal , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
15.
Curr Opin Nephrol Hypertens ; 29(1): 4-15, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31725015

RESUMEN

PURPOSE OF REVIEW: In chronic kidney disease (CKD), disturbance of several metabolic regulatory mechanisms cause premature ageing, accelerated cardiovascular disease (CVD), and mortality. Single-target interventions have repeatedly failed to improve the prognosis for CKD patients. Epigenetic interventions have the potential to modulate several pathogenetic processes simultaneously. Alkaline phosphatase (ALP) is a robust predictor of CVD and all-cause mortality and implicated in pathogenic processes associated with CVD in CKD. RECENT FINDINGS: In experimental studies, epigenetic modulation of ALP by microRNAs or bromodomain and extraterminal (BET) protein inhibition has shown promising results for the treatment of CVD and other chronic metabolic diseases. The BET inhibitor apabetalone is currently being evaluated for cardiovascular risk reduction in a phase III clinical study in high-risk CVD patients, including patients with CKD (ClinicalTrials.gov Identifier: NCT02586155). Phase II studies demonstrate an ALP-lowering potential of apabetalone, which was associated with improved cardiovascular and renal outcomes. SUMMARY: ALP is a predictor of CVD and mortality in CKD. Epigenetic modulation of ALP has the potential to affect several pathogenetic processes in CKD and thereby improve cardiovascular outcome.


Asunto(s)
Fosfatasa Alcalina/genética , Enfermedades Cardiovasculares/tratamiento farmacológico , Epigénesis Genética , Insuficiencia Renal Crónica/enzimología , Fosfatasa Alcalina/antagonistas & inhibidores , Fosfatasa Alcalina/fisiología , Enfermedades Cardiovasculares/etiología , Regulación Enzimológica de la Expresión Génica , Humanos , Quinazolinonas/uso terapéutico , Insuficiencia Renal Crónica/complicaciones
16.
Pharmacol Res ; 159: 105019, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32553713

RESUMEN

Diabetes Mellitus (DM) is a chronic and severe metabolic disease, characterized by chronic hyperglycemia due to insulin resistance and/or reduced insulin secretion. Concerning the non-insulin glucose-lowering therapy for diabetes, Dipeptidyl-peptidase-4 (DPP-4) inhibitors, members of the incretin family, represent new agents, capable of a glycemic control improvement with an advantageous safety profile, given the absence of weight gain, the low incidence of hypoglycemia and the good renal tolerance in patients suffering from chronic renal failure. In addition to demonstrating efficacy in glycemic control through inhibition of GLP-1 degradation, DPP-4 inhibitors (DPP-4is) seem to demonstrate pleiotropic effects, which also make them interesting in both diabetic and non-diabetic nephropathies, especially for their capacity of reducing proteinuria. Several studies about diabetic nephropathy on patients' cohorts and murine models have demonstrated a solid direct relationship between DPP-4 activity and urinary albumin excretion (UAE), thus confirming the capacity of DPP-4is to reduce proteinuria; the mechanism responsible for that effect was studied to assess if it was the result of a direct action on renal impairment or a secondary consequence of the better glycemic control related to these agents. As a result of these more in-depth studies, DPP-4is have demonstrated an improvement of renal inflammation markers and consequent proteinuria reduction, regardless of glucose concentrations. Considering the nephroprotective effects of DPP-4is might be glycemic independent, several studies were conducted to prove the validity of the same effects in non-diabetic nephropathies. Among these studies, DPP-4is demonstrated an improvement of various renal inflammatory markers on several models of non-diabetes dependent renal impairment, confirming their capacity to reduce proteinuria, independently from the action on glucose metabolism. The objective of this review is to present and discuss the so far demonstrated antiproteinuric effect of DPP-4is and their effects on diabetic and non-diabetic nephropathies.


Asunto(s)
Albuminuria/tratamiento farmacológico , Nefropatías Diabéticas/tratamiento farmacológico , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Riñón/efectos de los fármacos , Insuficiencia Renal Crónica/tratamiento farmacológico , Albuminuria/enzimología , Albuminuria/fisiopatología , Albuminuria/orina , Animales , Biomarcadores/sangre , Biomarcadores/orina , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/fisiopatología , Nefropatías Diabéticas/orina , Inhibidores de la Dipeptidil-Peptidasa IV/efectos adversos , Humanos , Riñón/enzimología , Riñón/fisiopatología , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/orina
17.
J Pathol ; 249(4): 461-471, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31397884

RESUMEN

Vascular calcification is prevalent in patients with chronic kidney disease (CKD) and a major risk factor of cardiovascular disease. Vascular calcification is now recognised as a biological process similar to bone formation involving osteogenic differentiation of vascular smooth muscle cells (VSMCs). Cell division cycle 42 (CDC42), a Rac1 family member GTPase, is essential for cartilage development during endochondral bone formation. However, whether CDC42 affects osteogenic differentiation of VSMCs and vascular calcification remains unknown. In the present study, we observed a significant increase in the expression of CDC42 both in rat VSMCs and in calcified arteries during vascular calcification. Alizarin red staining and calcium content assay revealed that adenovirus-mediated CDC42 overexpression led to an apparent VSMC calcification in the presence of calcifying medium, accompanied with up-regulation of bone-related molecules including RUNX2 and BMP2. By contrast, inhibition of CDC42 by ML141 significantly blocked calcification of VSMCs in vitro and aortic rings ex vivo. Moreover, ML141 markedly attenuated vascular calcification in rats with CKD. Furthermore, pharmacological inhibition of AKT signal was shown to block CDC42-induced VSMC calcification. These findings demonstrate for the first time that CDC42 contributes to vascular calcification through a mechanism involving AKT signalling; this uncovered a new function of CDC42 in regulating vascular calcification. This may provide a potential therapeutic target for the treatment of vascular calcification in the context of CKD. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Osteogénesis , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/etiología , Proteína de Unión al GTP cdc42/metabolismo , Anciano , Animales , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/farmacología , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/patología , Transducción de Señal , Sulfonamidas/farmacología , Técnicas de Cultivo de Tejidos , Calcificación Vascular/sangre , Calcificación Vascular/enzimología , Calcificación Vascular/patología
18.
BMC Nephrol ; 21(1): 77, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32131757

RESUMEN

BACKGROUND: Renalase is a flavoprotein that plays a protective role in chronic kidney disease (CKD) and cardiovascular diseases. The secretion and way of action of this protein are still discussed. The aim of our study was to estimate the balance between serum and urine renalase in healthy individuals and CKD patients, using two parameters: fractional excretion (FE) and serum-to-urine renalase ratio (StURR). METHODS: Our study involved 28 healthy volunteers and 62 patients with CKD in stages I to IV. The concentration of renalase in serum and urine was measured using an enzyme-linked immunosorbent assay (ELISA) kit (EIAab, Wuhan, China). We analyzed associations between renalase levels in urine and serum, and other parameters: sex, age, GFR, presence of hypertension, diabetes, and proteinuria, and determined the serum-to-urine renalase ratio and fractional excretion of renalase. RESULTS: Renalase and serum-to-urine ratio were significantly higher in CKD patients in comparison with the control group. Fractional excretion was lower in CKD patients but this difference did not reach the statistical significance (p = 0.092). Multivariate analysis performed in the CKD group showed, that from mentioned parameters, serum renalase was the only significant independent factor strongly positively associated with urinary renalase concentration. CONCLUSIONS: The serum-to-urine ratio is significantly and about 6.5-fold higher in CKD patients, and the fractional excretion of renalase is 3-fold, but not significantly lower in CKD patients. Renalase levels in both serum and urine are not related to the glomerular filtration rate and not associated with blood pressure.


Asunto(s)
Monoaminooxidasa/sangre , Monoaminooxidasa/orina , Insuficiencia Renal Crónica/enzimología , Adulto , Factores de Edad , Presión Sanguínea , Complicaciones de la Diabetes , Femenino , Tasa de Filtración Glomerular , Humanos , Hipertensión/complicaciones , Masculino , Persona de Mediana Edad , Proteinuria/complicaciones , Valores de Referencia , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/fisiopatología , Factores Sexuales
19.
Int J Mol Sci ; 21(22)2020 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-33207603

RESUMEN

Stearoyl-CoA Desaturase-2 (SCD2) is a member of the Stearoyl-CoA Desaturase (SCD) family of enzymes that catalyze the rate-limiting step in monounsaturated fatty acid (MUFA) synthesis. The MUFAs palmitoleoyl-CoA (16:1n7) and oleoyl-CoA (18:1n9) are the major products of SCD2. Palmitoleoyl-CoA and oleoyl-CoA have various roles, from being a source of energy to signaling molecules. Under normal feeding conditions, SCD2 is ubiquitously expressed and is the predominant SCD isoform in the brain. However, obesogenic diets highly induce SCD2 in adipose tissue, lung, and kidney. Here we provide a comprehensive review of SCD2 in mouse development, metabolism, and various diseases, such as obesity, chronic kidney disease, Alzheimer's disease, multiple sclerosis, and Parkinson's disease. In addition, we show that bone mineral density is decreased in SCD2KO mice under high-fat feeding conditions and that SCD2 is not required for preadipocyte differentiation or the expression of PPARγ in vivo despite being required in vitro.


Asunto(s)
Adipocitos/enzimología , Diferenciación Celular , Ácidos Grasos Monoinsaturados/metabolismo , Enfermedades Neurodegenerativas/enzimología , Obesidad/enzimología , Insuficiencia Renal Crónica/enzimología , Estearoil-CoA Desaturasa/metabolismo , Acilcoenzima A/biosíntesis , Acilcoenzima A/genética , Animales , Dieta Alta en Grasa/efectos adversos , Ratones , Ratones Noqueados , Enfermedades Neurodegenerativas/genética , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/metabolismo , Palmitoil Coenzima A/biosíntesis , Palmitoil Coenzima A/genética , Insuficiencia Renal Crónica/genética , Estearoil-CoA Desaturasa/genética
20.
J Cell Physiol ; 234(4): 4825-4839, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30207376

RESUMEN

Vascular calcification (VC) is the pathological accumulation of calcium phosphate crystals in one of the layers of blood vessels, leading to loss of elasticity and causing severe calcification in vessels. Medial calcification is mostly seen in patients with chronic kidney disease (CKD) and diabetes. Identification of key enzymes and their actions during calcification will contribute to understand the onset of pathological calcification. Phospholipase D (PLD1, PLD2) is active at the earlier steps of mineralization in osteoblasts and chondrocytes. In this study, we aimed to determine their effects during high-phosphate treatment in mouse vascular smooth muscle cell line MOVAS, in the ex vivo model of the rat aorta, and in the in vivo model of adenine-induced CKD. We observed an early increase in PLD1 gene and protein expression along with the increase in the PLD activity in vascular muscle cell line, during calcification induced by ascorbic acid and ß-glycerophosphate. Inhibition of PLD1 by the selective inhibitor VU0155069, or the pan-PLD inhibitor, halopemide, prevented calcification. The mechanism of PLD activation is likely to be protein kinase C (PKC)-independent since bisindolylmaleimide X hydrochloride, a pan-PKC inhibitor, did not affect the PLD activity. In agreement, we found an increase in Pld1 gene expression and PLD activity in aortic explant cultures treated with high phosphate, whereas PLD inhibition by halopemide decreased calcification. Finally, an increase in both Pld1 and Pld2 expression occurred simultaneously with the appearance of VC in a rat model of CKD. Thus, PLD, especially PLD1, promotes VC in the context of CKD and could be an important target for preventing onset or progression of VC.


Asunto(s)
Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Fosfolipasa D/metabolismo , Fósforo Dietético , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/etiología , Animales , Calcio de la Dieta , Línea Celular , Transdiferenciación Celular , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Inhibidores de Fosfodiesterasa/farmacología , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/genética , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/enzimología , Transducción de Señal , Técnicas de Cultivo de Tejidos , Calcificación Vascular/enzimología , Calcificación Vascular/patología , Calcificación Vascular/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA