Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.015
Filtrar
1.
Tunis Med ; 102(4): 241-244, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38746965

RESUMEN

INTRODUCTION: Toll-like- receptors (TLR) control important aspects of innate and adaptive immune responses. Renal cells are among the non-immune cells that express (TLR). Therefore, their activation might be implicated in renal tubulo-interstitial injury. AIM: The study aimed to compare TLR9 expression in patients with primary membranous nephropathy (MN) to patients with lupus membranous nephropathy. METHODS: Kidney sections from 10 Lupus nephritis (LN) patients and ten patients with primary MN were analyzed by immunohistochemistry using anti-human TLR9 antibody. RESULTS: Results showed that TLR9 expression was weak and exclusively tubular in primary MN patients' biopsies. There was a significant difference between LN patients' biopsies and primary MN patients' biopsies. TLR9 expression was more diffused in LN patients' specimen than in those with primary MN. CONCLUSION: This study focuses on molecular level pathogenesis of MN. The data suggest that the receptors TLR9 may play role in tubulointerstitial injury in the pathogenesis of LN but not primary membranous nephropathy.


Asunto(s)
Glomerulonefritis Membranosa , Nefritis Lúpica , Receptor Toll-Like 9 , Humanos , Receptor Toll-Like 9/metabolismo , Receptor Toll-Like 9/biosíntesis , Glomerulonefritis Membranosa/metabolismo , Glomerulonefritis Membranosa/patología , Glomerulonefritis Membranosa/inmunología , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Nefritis Lúpica/inmunología , Femenino , Adulto , Masculino , Persona de Mediana Edad , Túbulos Renales/patología , Túbulos Renales/metabolismo , Biopsia , Inmunohistoquímica , Adulto Joven
3.
Front Immunol ; 15: 1342350, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38720901

RESUMEN

Dyslipidemia is the most prevalent independent risk factor for patients with chronic kidney disease (CKD). Lipid-induced NLRP3 inflammasome activation in kidney-resident cells exacerbates renal injury by causing sterile inflammation. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that modulates the cellular redox balance; however, the exact role of Nrf2 signaling and its regulation of the NLRP3 inflammasome in hyperlipidemia-induced kidney injury are poorly understood. In this study, we demonstrated that activation of the mtROS-NLRP3 inflammasome pathway is a critical contributor to renal tubular epithelial cell (RTEC) apoptosis under hyperlipidemia. In addition, the Nrf2/ARE signaling pathway is activated in renal tubular epithelial cells under hyperlipidemia conditions both in vivo and in vitro, and Nrf2 silencing accelerated palmitic acid (PA)-induced mtROS production, mitochondrial injury, and NLRP3 inflammasome activation. However, the activation of Nrf2 with tBHQ ameliorated mtROS production, mitochondrial injury, NLRP3 inflammasome activation, and cell apoptosis in PA-induced HK-2 cells and in the kidneys of HFD-induced obese rats. Furthermore, mechanistic studies showed that the potential mechanism of Nrf2-induced NLRP3 inflammasome inhibition involved reducing mtROS generation. Taken together, our results demonstrate that the Nrf2/ARE signaling pathway attenuates hyperlipidemia-induced renal injury through its antioxidative and anti-inflammatory effects through the downregulation of mtROS-mediated NLRP3 inflammasome activation.


Asunto(s)
Células Epiteliales , Hiperlipidemias , Inflamasomas , Túbulos Renales , Factor 2 Relacionado con NF-E2 , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamasomas/metabolismo , Hiperlipidemias/metabolismo , Hiperlipidemias/complicaciones , Hiperlipidemias/inmunología , Células Epiteliales/metabolismo , Ratas , Humanos , Túbulos Renales/patología , Túbulos Renales/metabolismo , Masculino , Línea Celular , Apoptosis , Elementos de Respuesta Antioxidante , Mitocondrias/metabolismo , Modelos Animales de Enfermedad , Ratas Sprague-Dawley
4.
Ren Fail ; 46(1): 2347446, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38695335

RESUMEN

This study is intended to explore the effect of hypoxia-inducible factor-1α (HIF-1α) activation on lipid accumulation in the diabetic kidney. A type 1 diabetic rat model was established by STZ intraperitoneal injection. Cobalt chloride (CoCl2) and YC-1 were used as the HIF-1α activator and antagonist, respectively. CoCl2 treatment significantly increased HIF-1α expression, accelerated lipid deposition, and accelerated tubular injury in diabetic kidneys. In vitro, CoCl2 effectively stabilized HIF-1α and increased its transportation from the cytoplasm to the nucleus, which was accompanied by significantly increased lipid accumulation in HK-2 cells. Furthermore, results obtained in vivo showed that HIF-1α protein expression in the renal tubules of diabetic rats was significantly downregulated by YC-1 treatment. Meanwhile, lipid accumulation in the tubules of the DM + YC-1 group was markedly decreased in comparison to the DM + DMSO group. Accordingly, PAS staining revealed that the pathological injury caused to the tubular epithelial cells was alleviated by YC-1 treatment. Furthermore, the blood glucose level, urine albumin creatinine ratio, and NAG creatinine ratio in the DM + YC-1 group were significantly decreased compared to the DM + DMSO group. Moreover, the protein expression levels of transforming growth factor ß1 (TGF-ß1) and connective tissue growth factor (CTGF) in diabetic kidneys were decreased by YC-1 treatment. Our findings demonstrate that the activation of HIF-1α contributed to interstitial injury in a rat model of diabetic nephropathy and that the underlying mechanism involved the induction of lipid accumulation.


Asunto(s)
Cobalto , Diabetes Mellitus Experimental , Nefropatías Diabéticas , Subunidad alfa del Factor 1 Inducible por Hipoxia , Animales , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratas , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Masculino , Ratas Sprague-Dawley , Túbulos Renales/patología , Túbulos Renales/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Indazoles/farmacología , Humanos , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Línea Celular
5.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38697845

RESUMEN

Defective mitophagy in renal tubular epithelial cells is one of the main drivers of renal fibrosis in diabetic kidney disease. Our gene sequencing data showed the expression of PINK1 and BNIP3, two key molecules of mitophagy, was decreased in renal tissues of VDR-knockout mice. Herein, streptozotocin (STZ) was used to induce renal interstitial fibrosis in mice. VDR deficiency exacerbated STZ-induced renal impairment and defective mitophagy. Paricalcitol (pari, a VDR agonist) and the tubular epithelial cell-specific overexpression of VDR restored the expression of PINK1 and BNIP3 in the renal cortex and attenuated STZ-induced kidney fibrosis and mitochondrial dysfunction. In HK-2 cells under high glucose conditions, an increased level of α-SMA, COL1, and FN and a decreased expression of PINK1 and BNIP3 with severe mitochondrial damage were observed, and these alterations could be largely reversed by pari treatment. ChIP-qPCR and luciferase reporter assays showed VDR could positively regulate the transcription of Pink1 and Bnip3 genes. These findings reveal that VDR could restore mitophagy defects and attenuate STZ-induced fibrosis in diabetic mice through regulation of PINK1 and BNIP3.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Ergocalciferoles , Proteínas de la Membrana , Ratones Noqueados , Mitofagia , Proteínas Quinasas , Receptores de Calcitriol , Estreptozocina , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/genética , Ratones , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Receptores de Calcitriol/metabolismo , Receptores de Calcitriol/genética , Mitofagia/genética , Mitofagia/efectos de los fármacos , Proteínas Quinasas/metabolismo , Proteínas Quinasas/genética , Humanos , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/genética , Masculino , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Fibrosis , Túbulos Renales/metabolismo , Túbulos Renales/patología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Ratones Endogámicos C57BL , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos
6.
Nefrologia (Engl Ed) ; 44(2): 180-193, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38697696

RESUMEN

BACKGROUND: Contrast agents can directly or indirectly induce renal tubular ischemia and hypoxic damage. Given that cobalt chloride (CoCl2) can protect renal tubules, the protective effect and potential mechanism of action of CoCl2 on contrast-induced nephropathy (CIN) warrant investigation. METHODS: A CIN mouse model was established to determine the protective effect of CoCl2 on renal injury in vivo. Then, TMT-based proteomics was performed to determine the differentially expressed proteins (DEPs), following which, enrichment analyses of gene ontology and the KEGG pathway were performed. In vitro, a CIN model was constructed with renal tubular epithelial cells (HK-2) to determine the effect of CoCl2 on potential targets and the role of the key protein identified from the in vivo experiments. RESULTS: CoCl2 treatment decreased the levels of BUN and serum creatinine (sCr), while increasing the levels of urea and creatinine (Cr) in the urine of mice after CIN injury. Damage to the renal tubules in the CoCl2 treatment group was significantly less than in the CIN model group. We identified 79 DEPs after treating the in vivo model with CoCl2, and frequently observed ferroptosis-related GO and KEGG pathway terms. Of these, Hp (haptoglobin) was selected and found to have a strong renoprotective effect, even though its expression level in kidney tissue decreased after CoCl2 treatment. In HK-2 cells, overexpression of Hp reduced the ferroptosis caused by erastin, while knocking down Hp negated the attenuation effect of CoCl2 on HK-2 cell ferroptosis. CONCLUSION: CoCl2 attenuated kidney damage in the CIN model, and this effect was associated with the decrease in ferroptosis mediated by Hp.


Asunto(s)
Cobalto , Medios de Contraste , Ferroptosis , Ferroptosis/efectos de los fármacos , Animales , Ratones , Medios de Contraste/efectos adversos , Masculino , Enfermedades Renales/inducido químicamente , Enfermedades Renales/prevención & control , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Humanos , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología
7.
PeerJ ; 12: e17260, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38680884

RESUMEN

Chronic kidney disease (CKD) represents a significant global health concern, with renal fibrosis emerging as a prevalent and ultimate manifestation of this condition. The absence of targeted therapies presents an ongoing and substantial challenge. Accumulating evidence suggests that the integrity and functionality of mitochondria within renal tubular epithelial cells (RTECs) often become compromised during CKD development, playing a pivotal role in the progression of renal fibrosis. Mitophagy, a specific form of autophagy, assumes responsibility for eliminating damaged mitochondria to uphold mitochondrial equilibrium. Dysregulated mitophagy not only correlates with disrupted mitochondrial dynamics but also contributes to the advancement of renal fibrosis in CKD. While numerous studies have examined mitochondrial metabolism, ROS (reactive oxygen species) production, inflammation, and apoptosis in kidney diseases, the precise pathogenic mechanisms underlying mitophagy in CKD remain elusive. The exact mechanisms through which modulating mitophagy mitigates renal fibrosis, as well as its influence on CKD progression and prognosis, have not undergone systematic investigation. The role of mitophagy in AKI has been relatively clear, but the role of mitophagy in CKD is still rare. This article presents a comprehensive review of the current state of research on regulating mitophagy as a potential treatment for CKD. The objective is to provide fresh perspectives, viable strategies, and practical insights into CKD therapy, thereby contributing to the enhancement of human living conditions and patient well-being.


Asunto(s)
Mitocondrias , Mitofagia , Insuficiencia Renal Crónica , Humanos , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/metabolismo , Mitocondrias/metabolismo , Mitocondrias/patología , Fibrosis/patología , Fibrosis/metabolismo , Progresión de la Enfermedad , Especies Reactivas de Oxígeno/metabolismo , Animales , Túbulos Renales/patología , Túbulos Renales/metabolismo
9.
Chem Biol Interact ; 394: 110990, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38579922

RESUMEN

Swainsonine (SW) is the main toxic component of locoweed. Previous studies have shown that kidney damage is an early pathologic change in locoweed poisoning in animals. Trehalose induces autophagy and alleviates lysosomal damage, while its protective effect and mechanism against the toxic injury induced by SW is not clear. Based on the published literature, we hypothesize that transcription factor EB(TFEB) -regulated is targeted by SW and activating TFEB by trehalose would reverse the toxic effects. In this study, we investigate the mechanism of protective effects of trehalose using renal tubular epithelial cells. The results showed that SW induced an increase in the expression level of microtubule-associated protein light chain 3-II and p62 proteins and a decrease in the expression level of ATPase H+ transporting V1 Subunit A, Cathepsin B, Cathepsin D, lysosome-associated membrane protein 2 and TFEB proteins in renal tubular epithelial cells in a time and dose-dependent manner suggesting TFEB-regulated lysosomal pathway is adversely affected by SW. Conversely, treatment with trehalose, a known activator of TFEB promote TFEB nuclear translocation suggesting that TFEB plays an important role in protection against SW toxicity. We demonstrated in lysosome staining that SW reduced the number of lysosomes and increased the luminal pH, while trehalose could counteract these SW-induced effects. In summary, our results demonstrated for the first time that trehalose could alleviate the autophagy degradation disorder and lysosomal damage induced by SW. Our results provide an interesting method for reversion of SW-induced toxicity in farm animals and furthermore, activation of TFEB by trehalose suggesting novel mechanism of treating lysosomal storage diseases.


Asunto(s)
Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Células Epiteliales , Túbulos Renales , Lisosomas , Swainsonina , Trehalosa , Trehalosa/farmacología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Lisosomas/metabolismo , Lisosomas/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Autofagia/efectos de los fármacos , Animales , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Túbulos Renales/metabolismo , Túbulos Renales/citología , Swainsonina/farmacología , Línea Celular
10.
Int Immunopharmacol ; 133: 111955, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38626544

RESUMEN

Renal tubular injury is an important pathological change associated with diabetic nephropathy (DN), in which ferroptosis of renal tubular epithelial cells is critical to its pathogenesis. Inhibition of the glutathione/glutathione peroxidase 4 (GSH/GPX4) axis is the most important mechanism in DN tubular epithelial cell ferroptosis, but the underlying reason for this is unclear. Our biogenic analysis showed that a zinc-dependent metalloproteinase, dipeptidase 1 (DPEP1), is associated with DN ferroptosis. Here, we investigated the role and mechanism of DPEP1 in DN tubular epithelial cell ferroptosis. DPEP1 upregulation was observed in the renal tubular epithelial cells of DN patients and model mice, as well as in HK-2 cells stimulated with high glucose. Furthermore, the level of DPEP1 upregulation was associated with the degree of tubular injury in DN patients and HK-2 cell ferroptosis. Mechanistically, knocking down DPEP1 expression could alleviate the inhibition of GSH/GPX4 axis and reduce HK-2 cell ferroptosis levels in a high glucose environment. HK-2 cells with stable DPEP1 overexpression also showed GSH/GPX4 axis inhibition and ferroptosis, but blocking the GSH/GPX4 axis could mitigate these effects. Additionally, treatment with cilastatin, a DPEP1 inhibitor, could ameliorate GSH/GPX4 axis inhibition and relieve ferroptosis and DN progression in DN mice. These results revealed that DPEP1 can promote ferroptosis in DN renal tubular epithelial cells via inhibition of the GSH/GPX4 axis.


Asunto(s)
Nefropatías Diabéticas , Dipeptidasas , Células Epiteliales , Ferroptosis , Glutatión , Túbulos Renales , Ratones Endogámicos C57BL , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Animales , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/metabolismo , Humanos , Dipeptidasas/metabolismo , Dipeptidasas/genética , Células Epiteliales/metabolismo , Túbulos Renales/patología , Ratones , Masculino , Línea Celular , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Glutatión/metabolismo , Diabetes Mellitus Experimental/metabolismo , Glucosa/metabolismo , Proteínas Ligadas a GPI
11.
Transl Res ; 269: 14-30, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38453052

RESUMEN

The progression of chronic kidney disease (CKD) often involves renal interstitial fibrosis (RIF) and subsequent loss of peritubular capillaries (PTCs), which enhances disease severity. Despite advancements in our understanding of fibrosis, effective interventions for reversing capillary loss remain elusive. Notably, RIF exhibits reduced capillary density, whereas renal cell carcinoma (RCC) shows robust angiogenesis under hypoxic conditions. Using RNA sequencing and bioinformatics, we identified differentially expressed genes (DEGs) in hypoxic human renal tubular epithelial cells (HK-2) and renal cancer cells (786-0). Analysis of altered Ras and PI3K/Akt pathways coupled with hub gene investigation revealed RAS protein activator-like 2 (RASAL2) as a key candidate. Subsequent in vitro and in vivo studies confirmed RASAL2's early-stage response in RIF, which reduced with fibrosis progression. RASAL2 suppression in HK-2 cells enhanced angiogenesis, as evidenced by increased proliferation, migration, and branching of human umbilical vein endothelial cells (HUVECs) co-cultured with HK-2 cells. In mice, RASAL2 knockdown improved Vascular endothelial growth factor A (VEGFA) and Proliferating cell nuclear antigen (PCNA) levels in unilateral ureteral occlusion (UUO)-induced fibrosis (compared to wild type). Hypoxia-inducible factor 1 alpha (HIF-1α) emerged as a pivotal mediator, substantiated by chromatin immunoprecipitation (ChIP) sequencing, with its induction linked to activation. Hypoxia increased the production of RASAL2-enriched extracellular vesicles (EVs) derived from tubular cells, which were internalized by endothelial cells, contributing to the exacerbation of PTC loss. These findings underscore RASAL2's role in mediating reduced angiogenesis in RIF and reveal a novel EV-mediated communication between hypoxic tubular- and endothelial cells, demonstrating a complex interplay between angiogenesis and fibrosis in CKD pathogenesis.


Asunto(s)
Fibrosis , Humanos , Animales , Ratones , Masculino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Rarefacción Microvascular/metabolismo , Rarefacción Microvascular/patología , Rarefacción Microvascular/genética , Ratones Endogámicos C57BL , Riñón/irrigación sanguínea , Riñón/patología , Riñón/metabolismo , Hipoxia/patología , Hipoxia/metabolismo , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/genética , Hipoxia de la Célula , Túbulos Renales/patología , Túbulos Renales/metabolismo , Línea Celular , Neovascularización Patológica/metabolismo , Neovascularización Patológica/genética
12.
J Mol Med (Berl) ; 102(5): 679-692, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38453697

RESUMEN

Chronic kidney disease (CKD) is the 16th leading cause of mortality worldwide. Clinical studies have raised that long-term use of omeprazole (OME) is associated with the morbidity of CKD. OME is commonly used in clinical practice to treat peptic ulcers and gastroesophageal reflux disease. However, the mechanism underlying renal failure following OME treatment remains mostly unknown and the rodent model of OME-induced CKD is yet to be established. We described the process of renal injury after exposure to OME in mice; the early renal injury markers were increased in renal tubular epithelial cells (RTECs). And after long-term OME treatment, the OME-induced CKD mice model was established. Herein, aryl hydrocarbon receptor (AHR) translocation appeared after exposure to OME in HK-2 cells. Then for both in vivo and in vitro, we found that Ahr-knockout (KO) and AHR small interfering RNA (siRNA) substantially alleviated the OME-induced renal function impairment and tubular cell damage. Furthermore, our data demonstrate that antagonists of AHR and CYP1A1 could attenuate OME-induced tubular cell impairment in HK-2 cells. Taken together, these data indicate that OME induces CKD through the activation of the AHR-CYP axis in RTECs. Our findings suggest that blocking the AHR-CYP1A1 pathway acts as a potential strategy for the treatment of CKD caused by OME. KEY MESSAGES: We provide an omeprazole-induced chronic kidney disease (CKD) mice model. AHR activation and translocation process was involved in renal tubular damage and promoted the occurrence of CKD. The process of omeprazole nephrotoxicity can be ameliorated by blockade of the AHR-CYP1A1 axis.


Asunto(s)
Citocromo P-450 CYP1A1 , Ratones Endogámicos C57BL , Ratones Noqueados , Omeprazol , Receptores de Hidrocarburo de Aril , Insuficiencia Renal Crónica , Omeprazol/farmacología , Omeprazol/uso terapéutico , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Hidrocarburo de Aril/genética , Animales , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/inducido químicamente , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A1/genética , Humanos , Ratones , Línea Celular , Masculino , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Túbulos Renales/patología , Túbulos Renales/metabolismo , Túbulos Renales/efectos de los fármacos
13.
Toxicol Appl Pharmacol ; 485: 116892, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38492675

RESUMEN

Metabolic syndrome (MetS) is largely coupled with chronic kidney disease (CKD). Glycogen synthase kinase-3ß (GSK-3ß) pathway drives tubular injury in animal models of acute kidney injury; but its contribution in CKD is still elusive. This study investigated the effect empagliflozin and/or pirfenidone against MetS-induced kidney dysfunction, and to clarify additional underpinning mechanisms particularly the GSK-3ß signaling pathway. Adult male rats received 10%w/v fructose in drinking water for 20 weeks to develop MetS, then treated with either drug vehicle, empagliflozin (30 mg/kg/day) and/or pirfenidone (100 mg/kg/day) via oral gavage for subsequent 4 weeks, concurrently with the high dietary fructose. Age-matched rats receiving normal drinking water were used as controls. After 24 weeks, blood and kidneys were harvested for subsequent analyses. Rats with MetS showed signs of kidney dysfunction, structural changes and interstitial fibrosis. Activation of GSK-3ß, decreased cyclinD1 expression and enhanced apoptotic signaling were found in kidneys of MetS rats. There was abundant alpha-smooth muscle actin (α-SMA) expression along with up-regulation of TGF-ß1/Smad3 in kidneys of MetS rats. These derangements were almost alleviated by empagliflozin or pirfenidone, with evidence that the combined therapy was more effective than either individual drug. This study emphasizes a novel mechanism underpinning the beneficial effects of empagliflozin and pirfenidone on kidney dysfunction associated with MetS through targeting GSK-3ß signaling which can mediate the regenerative capacity, anti-apoptotic effects and anti-fibrotic properties of such drugs. These findings recommend the possibility of using empagliflozin and pirfenidone as promising therapies for management of CKD in patients with MetS.


Asunto(s)
Compuestos de Bencidrilo , Glucósidos , Glucógeno Sintasa Quinasa 3 beta , Túbulos Renales , Síndrome Metabólico , Piridonas , Animales , Piridonas/farmacología , Masculino , Glucósidos/farmacología , Glucósidos/uso terapéutico , Compuestos de Bencidrilo/farmacología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ratas , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/complicaciones , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Túbulos Renales/metabolismo , Regeneración/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
14.
BMJ Open Diabetes Res Care ; 12(2)2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38442987

RESUMEN

INTRODUCTION: We previously reported the significant upregulation of eight circulating exosomal microRNAs (miRNAs) in patients with diabetic kidney disease (DKD). However, their specific roles and molecular mechanisms in the kidney remain unknown. Among the eight miRNAs, we evaluated the effects of miR-5010-5p on renal tubular epithelial cells under diabetic conditions in this study. RESEARCH DESIGN AND METHODS: We transfected the renal tubular epithelial cell line, HK-2, with an miR-5010-5p mimic using recombinant plasmids. The target gene of hsa-miR-5010-5p was identified using a dual-luciferase assay. Cell viability was assessed via the 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay. Moreover, mRNA and protein expression levels were determined via real-time PCR and western blotting, respectively. RESULTS: High glucose levels did not significantly affect the intracellular expression of miR-5010-5p in HK-2 cells. Transfection of the miR-5010-5p mimic caused no change in cell viability. However, miR-5010-5p-transfected HK-2 cells exhibited significantly decreased expression levels of inflammatory cytokines, such as the monocyte chemoattractant protein-1, interleukin-1ß, and tumor necrosis factor-ɑ, under high-glucose conditions. These changes were accompanied by the restored expression of phosphorylated AMP-activated protein kinase (AMPK) and decreased phosphorylation of nuclear factor-kappa B. Dual-luciferase assay revealed that miR-5010-5p targeted the gene, protein phosphatase 2 regulatory subunit B delta (PPP2R2D), a subunit of protein phosphatase 2A, which modulates AMPK phosphorylation. CONCLUSIONS: Our findings suggest that increased miR-5010-5p expression reduces high glucose-induced inflammatory responses in renal tubular epithelial cells via the regulation of the target gene, PPP2R2D, which modulates AMPK phosphorylation. Therefore, miR-5010-5p may be a promising therapeutic target for DKD.


Asunto(s)
Proteínas Quinasas Activadas por AMP , MicroARNs , Proteína Fosfatasa 2 , Humanos , Proteínas Quinasas Activadas por AMP/metabolismo , Células Epiteliales , Glucosa/metabolismo , Inflamación/metabolismo , Luciferasas , MicroARNs/metabolismo , Proteína Fosfatasa 2/metabolismo , Túbulos Renales/metabolismo , Túbulos Renales/patología
15.
Int Immunopharmacol ; 132: 111956, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38554447

RESUMEN

BACKGROUND: While recent studies have suggested a potential involvement of circRNAs in acute kidney injury (AKI) after ischemia, mmu_circ_003062 role is undetermined. METHODS: The levels of mmu_circ_003062, miR-490-3p, CACNA1H, GRP78, CHOP and hsa_circ_0075663 were detected by Relative qPCR in Boston University mouse proximal tubule (BUMPT) cells, mouse kidneys, and human renal tubular epithelial (HK-2) cells. Moreover, the levels of hsa_circ_0075663 in serum and urine of patients with AKI following cardiopulmonary resuscitation (CPR) were detected by absolute quantitative PCR. Western blot was used to detect the relative expression of the protein. The function and regulatory mechanism of mmu_circ_003062 and hsa_circ_0075663 were investigated through a series of in vitro and in vivo experiments, including bioinformatic prediction, luciferase reporter assays, FISH, FCM, TUNEL staining, and H&E staining. RESULTS: It was found that mmu_circ_003062, hsa_circ_0075663 mediated apoptosis after ischemia/reperfusion (I/R) by interaction with miR-490-3p to enhance CACNA1H expression, thereby leading to the upregulation of endoplasmic reticulum stress (ERS)-relevant proteins GRP78 and CHOP. Ultimately, mmu_circ_003062 downregulation significantly ameliorated ischemic AKI by modulating the miR-490-3p/CACNA1H/GRP78 and CHOP pathway. Furthermore, the plasma and urinary levels of hsa_circ_0075663 in patients with AKI following CPR were significantly higher than non-AKI patients, exhibited a strongly correlation with serum creatinine. CONCLUSION: The involvement of mmu_circ_003062, hsa_circ_0075663/miR-490-3p/CACNA1H/GRP78 and CHOP axis is significant in the development of ischemic AKI. Moreover, hsa_circ_0075663 has potential as an early diagnostic biomarker.


Asunto(s)
Lesión Renal Aguda , Apoptosis , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , MicroARNs , ARN Circular , Animales , Humanos , Masculino , Ratones , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Línea Celular , Isquemia/metabolismo , Túbulos Renales/patología , Túbulos Renales/metabolismo , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Daño por Reperfusión/metabolismo , ARN Circular/genética , ARN Circular/metabolismo , Transducción de Señal , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética
16.
Mol Ther ; 32(5): 1526-1539, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38414248

RESUMEN

The Hippo/YAP pathway plays a critical role in tissue homeostasis. Our previous work demonstrated that renal tubular YAP activation induced by double knockout (dKO) of the upstream Hippo kinases Mst1 and Mst2 promotes tubular injury and renal inflammation under basal conditions. However, the importance of tubular YAP activation remains to be established in injured kidneys in which many other injurious pathways are simultaneously activated. Here, we show that tubular YAP was already activated 6 h after unilateral ureteral obstruction (UUO). Tubular YAP deficiency greatly attenuated tubular cell overproliferation, tubular injury, and renal inflammation induced by UUO or cisplatin. YAP promoted the transcription of the transcription factor KLF5. Consistent with this, the elevated expression of KLF5 and its target genes in Mst1/2 dKO or UUO kidneys was blocked by ablation of Yap in tubular cells. Inhibition of KLF5 prevented tubular cell overproliferation, tubular injury, and renal inflammation in Mst1/2 dKO kidneys. Therefore, our results demonstrate that tubular YAP is a key player in kidney injury. YAP and KLF5 form a transcriptional cascade, where tubular YAP activation induced by kidney injury promotes KLF5 transcription. Activation of this cascade induces tubular cell overproliferation, tubular injury, and renal inflammation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Túbulos Renales , Factores de Transcripción de Tipo Kruppel , Ratones Noqueados , Proteínas Señalizadoras YAP , Animales , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Túbulos Renales/metabolismo , Túbulos Renales/patología , Túbulos Renales/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Serina-Treonina Quinasa 3 , Transducción de Señal , Proliferación Celular , Regulación de la Expresión Génica , Modelos Animales de Enfermedad , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología , Cisplatino/farmacología
17.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167022, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38216068

RESUMEN

BACKGROUND: CAMK1 has been shown to be involved in human disease progression via regulating mitochondrial dynamics. However, whether CAMK1 mediates mitochondrial dynamics to regulate diabetic nephropathy (DN) process remains unclear. METHODS: Mice were injected with streptozotocin (STZ) to mimic diabetic mice models in vivo, and mice with proximal tubule-specific knockout of CAMK1 (CAMK1-KO) were generated. HK-2 cells were treated with high-glucose (HG) to mimic DN cell model in vitro. Histopathological analysis was performed to confirm kidney injury in mice. ROS production and apoptosis were assessed by DHE staining and TUNEL staining. Mitochondria morphology was observed and analyzed by electron microscopy. Mitochondrial membrane potential was detected by JC-1 staining, and cell proliferation was measured by EdU assay. The mRNA and protein expression were examined by qRT-PCR, western blot and immunostaining. RNA interaction was confirmed by RIP assay and dual-luciferase reporter assay. The mRNA stability was tested by actinomycin D treatment, and m6A level was examined by MeRIP assay. RESULTS: CAMK1 was reduced in DN patients and STZ-induced diabetic mice. Conditional deletion of CAMK1 aggravated kidney injury and promoted mitochondrial fission in diabetic mice. CAMK1 overexpression inhibited mitochondrial fission to alleviate HG-induced HK-2 cell apoptosis. IGF2BP3 promoted the stability of CAMK1 mRNA by m6A modification. IGF2BP3 inhibited mitochondrial fission to repress cell apoptosis in vitro and kidney injury in vivo by increasing CAMK1 expression. CONCLUSION: IGF2BP3-mediated CAMK1 mRNA stability alleviated DN progression by inhibiting mitochondria fission.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Animales , Humanos , Ratones , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/patología , Túbulos Renales/patología , Dinámicas Mitocondriales/fisiología , ARN Mensajero/metabolismo
18.
Apoptosis ; 29(5-6): 620-634, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38281282

RESUMEN

Maleic acid (MA) induces renal tubular cell dysfunction directed to acute kidney injury (AKI). AKI is an increasing global health burden due to its association with mortality and morbidity. However, targeted therapy for AKI is lacking. Previously, we determined mitochondrial-associated proteins are MA-induced AKI affinity proteins. We hypothesized that mitochondrial dysfunction in tubular epithelial cells plays a critical role in AKI. In vivo and in vitro systems have been used to test this hypothesis. For the in vivo model, C57BL/6 mice were intraperitoneally injected with 400 mg/kg body weight MA. For the in vitro model, HK-2 human proximal tubular epithelial cells were treated with 2 mM or 5 mM MA for 24 h. AKI can be induced by administration of MA. In the mice injected with MA, the levels of blood urea nitrogen (BUN) and creatinine in the sera were significantly increased (p < 0.005). From the pathological analysis, MA-induced AKI aggravated renal tubular injuries, increased kidney injury molecule-1 (KIM-1) expression and caused renal tubular cell apoptosis. At the cellular level, mitochondrial dysfunction was found with increasing mitochondrial reactive oxygen species (ROS) (p < 0.001), uncoupled mitochondrial respiration with decreasing electron transfer system activity (p < 0.001), and decreasing ATP production (p < 0.05). Under transmission electron microscope (TEM) examination, the cristae formation of mitochondria was defective in MA-induced AKI. To unveil the potential target in mitochondria, gene expression analysis revealed a significantly lower level of ATPase6 (p < 0.001). Renal mitochondrial protein levels of ATP subunits 5A1 and 5C1 (p < 0.05) were significantly decreased, as confirmed by protein analysis. Our study demonstrated that dysfunction of mitochondria resulting from altered expression of ATP synthase in renal tubular cells is associated with MA-induced AKI. This finding provides a potential novel target to develop new strategies for better prevention and treatment of MA-induced AKI.


Asunto(s)
Lesión Renal Aguda , Apoptosis , Maleatos , Ratones Endogámicos C57BL , Mitocondrias , ATPasas de Translocación de Protón Mitocondriales , Animales , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/genética , Humanos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Ratones , ATPasas de Translocación de Protón Mitocondriales/metabolismo , ATPasas de Translocación de Protón Mitocondriales/genética , Maleatos/farmacología , Apoptosis/efectos de los fármacos , Masculino , Especies Reactivas de Oxígeno/metabolismo , Túbulos Renales/patología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Línea Celular , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología
19.
Sci Rep ; 14(1): 439, 2024 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172172

RESUMEN

Examining kidney fibrosis is crucial for mechanistic understanding and developing targeted strategies against chronic kidney disease (CKD). Persistent fibroblast activation and tubular epithelial cell (TEC) injury are key CKD contributors. However, cellular and transcriptional landscapes of CKD and specific activated kidney fibroblast clusters remain elusive. Here, we analyzed single cell transcriptomic profiles of two clinically relevant kidney fibrosis models which induced robust kidney parenchymal remodeling. We dissected the molecular and cellular landscapes of kidney stroma and newly identified three distinctive fibroblast clusters with "secretory", "contractile" and "vascular" transcriptional enrichments. Also, both injuries generated failed repair TECs (frTECs) characterized by decline of mature epithelial markers and elevation of stromal and injury markers. Notably, frTECs shared transcriptional identity with distal nephron segments of the embryonic kidney. Moreover, we identified that both models exhibited robust and previously unrecognized distal spatial pattern of TEC injury, outlined by persistent elevation of renal TEC injury markers including Krt8 and Vcam1, while the surviving proximal tubules (PTs) showed restored transcriptional signature. We also found that long-term kidney injuries activated a prominent nephrogenic signature, including Sox4 and Hox gene elevation, which prevailed in the distal tubular segments. Our findings might advance understanding of and targeted intervention in fibrotic kidney disease.


Asunto(s)
Túbulos Renales , Insuficiencia Renal Crónica , Humanos , Túbulos Renales/patología , Riñón/patología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Fibroblastos/fisiología , Fibrosis
20.
Annu Rev Physiol ; 86: 379-403, 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38012047

RESUMEN

Mitochondria play a key role in kidney physiology and pathology. They produce ATP to fuel energy-demanding water and solute reabsorption processes along the nephron. Moreover, mitochondria contribute to cellular health by the regulation of autophagy, (oxidative) stress responses, and apoptosis. Mitochondrial abundance is particularly high in cortical segments, including proximal and distal convoluted tubules. Dysfunction of the mitochondria has been described for tubulopathies such as Fanconi, Gitelman, and Bartter-like syndromes and renal tubular acidosis. In addition, mitochondrial cytopathies often affect renal (tubular) tissues, such as in Kearns-Sayre and Leigh syndromes. Nevertheless, the mechanisms by which mitochondrial dysfunction results in renal tubular diseases are only scarcely being explored. This review provides an overview of mitochondrial dysfunction in the development and progression of kidney tubulopathies. Furthermore, it emphasizes the need for further mechanistic investigations to identify links between mitochondrial function and renal electrolyte reabsorption.


Asunto(s)
Síndrome de Bartter , Síndrome de Kearns-Sayre , Enfermedades Renales , Humanos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Síndrome de Bartter/metabolismo , Síndrome de Bartter/patología , Síndrome de Kearns-Sayre/metabolismo , Síndrome de Kearns-Sayre/patología , Enfermedades Renales/patología , Mitocondrias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...