Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.650
Filtrar
1.
Zool Res ; 45(5): 1161-1174, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39257378

RESUMO

Acute kidney injury (AKI) and chronic kidney disease (CKD) are significant public health issues associated with a long-term increase in mortality risk, resulting from various etiologies including renal ischemia, sepsis, drug toxicity, and diabetes mellitus. Numerous preclinical models have been developed to deepen our understanding of the pathophysiological mechanisms and therapeutic approaches for kidney diseases. Among these, rodent models have proven to be powerful tools in the discovery of novel therapeutics, while the development of kidney organoids has emerged as a promising advancement in the field. This review provides a comprehensive analysis of the construction methodologies, underlying biological mechanisms, and recent therapeutic developments across different AKI and CKD models. Additionally, this review summarizes the advantages, limitations, and challenges inherent in these preclinical models, thereby contributing robust evidence to support the development of effective therapeutic strategies.


Assuntos
Modelos Animais de Doenças , Animais , Nefropatias/etiologia , Nefropatias/patologia , Humanos , Injúria Renal Aguda/fisiopatologia , Injúria Renal Aguda/patologia , Insuficiência Renal Crônica/patologia
2.
Commun Biol ; 7(1): 1091, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39237614

RESUMO

Cisplatin is a common anticancer drug, but its frequent nephrotoxicity limits its clinical use. Small GTP-binding protein GDP dissociation stimulator (smgGDS), a small GTPase chaperone protein, was considerably downregulated during cisplatin-induced acute kidney injury (CDDP-AKI), especially in renal tubular epithelial cells. SmgGDS-knockdown mice was established and found that smgGDS knockdown promoted CDDP-AKI, as demonstrated by an increase in serum creatine, blood urea nitrogen levels and the appearance of tubular patterns. RNA sequencing suggested that protein kinase RNA-like ER kinase (PERK), which bridges mitochondria-associated ER membranes, was involved in smgGDS knockdown following CDDP-AKI, and then identified that smgGDS knockdown increased phosphorylated-PERK in vivo and in vitro. Furthermore, we confirmed that smgGDS deficiency aggravated apoptosis and ER stress in vivo and in vitro. And the ER stress inhibitor 4-Phenylbutyric acid and the inhibition of PERK phosphorylation mitigated smgGDS deficiency-induced ER stress related apoptosis following cisplatin treatment, while the eIF2α phosphorylation inhibitor could not reverse the smgGDS deficiency accelerated cell death. Furthermore, the over-expression of smgGDS could reverse the ER stress and apoptosis caused by CDDP. Overall, smgGDS regulated PERK-dependent ER stress and apoptosis, thereby influencing renal damage. This study identified a target for diagnosing and treating cisplatin-induced acute kidney injury.


Assuntos
Injúria Renal Aguda , Cisplatino , Estresse do Retículo Endoplasmático , eIF-2 Quinase , Cisplatino/efeitos adversos , Cisplatino/toxicidade , Animais , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/genética , eIF-2 Quinase/metabolismo , eIF-2 Quinase/genética , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Camundongos , Masculino , Apoptose/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Antineoplásicos/efeitos adversos , Antineoplásicos/toxicidade , Fosforilação
3.
Ren Fail ; 46(2): 2369342, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39230047

RESUMO

Sepsis represents an organ dysfunction resulting from the host's maladjusted response to infection, and can give rise to acute kidney injury (AKI), which significantly increase the morbidity and mortality of septic patients. This study strived for identifying a novel therapeutic strategy for patients with sepsis-induced AKI (SI-AKI). Rat tubular epithelial NRK-52E cells were subjected to lipopolysaccharide (LPS) exposure for induction of in-vitro SI-AKI. The expressions of E1A binding protein p300 (EP300) and methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) in NRK-52E cells were assessed by western blot and qRT-PCR, and their interaction was explored by chromatin immunoprecipitation performed with antibody for H3K27 acetylation (H3K27ac). The effect of them on SI-AKI-associated mitochondrial dysfunction of tubular epithelial cells was investigated using transfection, MTT assay, TUNEL staining, 2',7'-Dichlorodihydrofluorescein diacetate probe assay, Mitosox assay, and JC-1 staining. MTHFD2 and EP300 were upregulated by LPS exposure in NRK-52E cells. LPS increased the acetylation of H3 histone in the MTHFD2 promoter region, and EP300 suppressed the effect of LPS. EP300 ablation inhibited the expression of MTHFD2. MTHFD2 overexpression antagonized LPS-induced viability reduction, apoptosis promotion, reactive oxygen species overproduction, and mitochondrial membrane potential collapse of NRK-52E cells. By contrast, MTHFD2 knockdown and EP300 ablation brought about opposite consequences. Furthermore, MTHFD2 overexpress and EP300 ablation counteracted each other's effect in LPS-exposed NRK-52E cells. EP300-mediated H3 acetylation elevates MTHFD2 expression to reduce mitochondrial dysfunction of tubular epithelial cells in SI-AKI.


Assuntos
Injúria Renal Aguda , Proteína p300 Associada a E1A , Células Epiteliais , Lipopolissacarídeos , Metilenotetra-Hidrofolato Desidrogenase (NADP) , Mitocôndrias , Animais , Ratos , Acetilação , Metilenotetra-Hidrofolato Desidrogenase (NADP)/metabolismo , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Proteína p300 Associada a E1A/metabolismo , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Células Epiteliais/metabolismo , Mitocôndrias/metabolismo , Linhagem Celular , Histonas/metabolismo , Apoptose , Sepse/metabolismo , Túbulos Renais/patologia , Túbulos Renais/metabolismo , Regulação para Cima
4.
Int J Mol Sci ; 25(17)2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-39273595

RESUMO

Acute kidney injury (AKI) is widely recognized as a precursor to the onset or rapid progression of chronic kidney disease (CKD). However, there is currently no effective treatment available for AKI, underscoring the urgent need for the development of new strategies to improve kidney function. Human placental mesenchymal stromal cells (hpMSCs) were isolated from donor placentas, cultured, and characterized with regard to yield, viability, flow cytometry, and potency. To mimic AKI and its progression to CKD in a rat model, a dedicated sensitive non-clinical bilateral kidney ischemia-reperfusion injury (IRI) model was utilized. The experimental group received 3 × 105 hpMSCs into each kidney, while the control group received IRI and saline and the untreated group received IRI only. Urine, serum, and kidney tissue samples were collected over a period of 28 days. The hpMSCs exhibited consistent yields, viability, and expression of mesenchymal lineage markers, and were also shown to suppress T cell proliferation in a dose-dependent manner. To ensure optimal donor selection, manufacturing optimization, and rigorous quality control, the rigorous Good Manufacturing Practice (GMP) conditions were utilized. The results indicated that hpMSCs increased rat survival rates and improved kidney function by decreasing serum creatinine, urea, potassium, and fractionated potassium levels. Furthermore, the study demonstrated that hpMSCs can prevent the initial stages of kidney structural fibrosis and improve kidney function in the early stages by mitigating late interstitial fibrosis and tubular atrophy. Additionally, a robust manufacturing process with consistent technical parameters was established.


Assuntos
Injúria Renal Aguda , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Insuficiência Renal Crônica , Injúria Renal Aguda/terapia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/prevenção & controle , Animais , Insuficiência Renal Crônica/terapia , Insuficiência Renal Crônica/patologia , Humanos , Ratos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Feminino , Gravidez , Transplante de Células-Tronco Mesenquimais/métodos , Placenta/citologia , Traumatismo por Reperfusão/terapia , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/patologia , Modelos Animais de Doenças , Rim/patologia , Ratos Sprague-Dawley , Masculino , Progressão da Doença
5.
ACS Appl Mater Interfaces ; 16(37): 49913-49925, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39240782

RESUMO

Renal-specific nanoparticulate drug delivery systems have shown great potential in reducing systemic side effects and improving the safety and efficacy of treatments for renal diseases. Here, stearic acid-grafted chitosan oligosaccharide (COS-SA) was synthesized as a renal-targeted carrier due to the high affinity of the 2-glucosamine moiety on COS to the megalin receptor expressed on renal proximal tubular epithelial cells. Specifically, COS-SA/CLT micelles were prepared by encapsulating celastrol (CLT) with COS-SA, and different proportions of human serum albumin (HSA) were then adsorbed onto its surface to explore the interaction between the protein corona and cationic polymeric micelles. Our results showed that a multilayered protein corona, consisting of an inner "hard" corona and an outer "soft" corona, was formed on the surface of COS-SA/CLT@HSA8, which was beneficial in preventing its recognition and phagocytosis by macrophages. The formation of HSA protein corona on COS-SA/CLT micelles also increased its accumulation in the renal tubules. Furthermore, the electropositivity of COS-SA/CLT micelles affected the conformation of adsorbed proteins to various degrees. During the adsorption process, the protein corona on the surface of COS-SA/CLT@HSA1 was partially denatured. Overall, COS-SA/CLT and COS-SA/CLT@HSA micelles demonstrated sufficient safety with renal targeting potential, providing a viable strategy for the management of ischemia/reperfusion-induced acute kidney injury.


Assuntos
Injúria Renal Aguda , Quitosana , Micelas , Oligossacarídeos , Coroa de Proteína , Traumatismo por Reperfusão , Albumina Sérica Humana , Quitosana/química , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/patologia , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Animais , Oligossacarídeos/química , Oligossacarídeos/farmacologia , Humanos , Coroa de Proteína/química , Coroa de Proteína/metabolismo , Albumina Sérica Humana/química , Camundongos , Sistemas de Liberação de Medicamentos , Masculino , Portadores de Fármacos/química
6.
Ann Clin Lab Sci ; 54(4): 539-546, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39293843

RESUMO

OBJECTIVE: To study the effect of dexmedetomidine (Dex) on myocardial injury induced by acute kidney injury (AKI) in diabetes mellitus rats and explore the potential mechanisms. METHODS: The Type 2 diabetes mellitus (T2DM) model was prepared in 40 adult male Wistar rats. These rats were randomly divided into four groups (n=10/group), including the control (Con) group, AKI group, Dex preconditioning (DPreC) group, and resveratrol (Res) combined with Dex preconditioning (Res+DPreC) group. The AKI model was prepared in the AKI, DPreC, and Res+DPreC group. The DPreC group received Dex, while the Con and AKI group received normal saline. The Res+DPreC group received Res in addition to Dex preconditioning. Histopathologic, apoptotic, enzymatic, and inflammatory changes in myocardial tissue were observed or detected. RESULTS: Histopathologic, apoptotic, and enzymatic changes in myocardial tissue demonstrated that AKI induced myocardial injury in T2DM rats; Dex preconditioning could mitigate this injury; and RES enhanced this effect. Inflammatory changes suggested that Dex alleviated the inflammatory response induced by AKI in T2DM rats via regulating the expressions of SIRT1, TNF-α, IL-17A, and IL-10. CONCLUSIONS: Dex could alleviate myocardial injury induced by AKI in DM rats via regulating the inflammatory response associated with SIRT1, TNF-α, IL-17A, and IL-10, and Res could enhance this protective effect.


Assuntos
Injúria Renal Aguda , Dexmedetomidina , Diabetes Mellitus Experimental , Inflamação , Ratos Wistar , Animais , Dexmedetomidina/farmacologia , Dexmedetomidina/uso terapêutico , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/metabolismo , Masculino , Ratos , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/patologia , Inflamação/patologia , Inflamação/tratamento farmacológico , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Miocárdio/patologia , Miocárdio/metabolismo , Resveratrol/farmacologia , Sirtuína 1/metabolismo , Apoptose/efeitos dos fármacos
7.
Cell Commun Signal ; 22(1): 423, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39223553

RESUMO

Acute kidney injury (AKI) is a clinical condition characterized by a rapid decline in kidney function, which is associated with local inflammation and programmed cell death in the kidney. The G protein-coupled receptors (GPCRs) represent the largest family of signaling transduction proteins in the body, and approximately 40% of drugs on the market target GPCRs. The expressions of various GPCRs, prostaglandin receptors and purinergic receptors, to name a few, are significantly altered in AKI models. And the role of GPCRs in AKI is catching the eyes of researchers due to their distinctive biological functions, such as regulation of hemodynamics, metabolic reprogramming, and inflammation. Therefore, in this review, we aim to discuss the role of GPCRs in the pathogenesis of AKI and summarize the relevant clinical trials involving GPCRs to assess the potential of GPCRs and their ligands as therapeutic targets in AKI and the transition to AKI-CKD.


Assuntos
Injúria Renal Aguda , Receptores Acoplados a Proteínas G , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Animais , Transdução de Sinais
8.
Sci Rep ; 14(1): 20386, 2024 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-39223189

RESUMO

Sodium-glucose cotransporter 2 (SGLT2) inhibitors have been shown to be renoprotective in ischemia-reperfusion (I/R) injury, with several proposed mechanisms, though additional mechanisms likely exist. This study investigated the impact of luseogliflozin on kidney fibrosis at 48 h and 1 week post I/R injury in C57BL/6 mice. Luseogliflozin attenuated kidney dysfunction and the acute tubular necrosis score on day 2 post I/R injury, and subsequent fibrosis at 1 week, as determined by Sirius red staining. Metabolomics enrichment analysis of I/R-injured kidneys revealed suppression of the glycolytic system and activation of mitochondrial function under treatment with luseogliflozin. Western blotting showed increased nutrient deprivation signaling with elevated phosphorylated AMP-activated protein kinase and Sirtuin-3 in luseogliflozin-treated kidneys. Luseogliflozin-treated kidneys displayed increased protein levels of carnitine palmitoyl transferase 1α and decreased triglyceride deposition, as determined by oil red O staining, suggesting activated fatty acid oxidation. Luseogliflozin prevented the I/R injury-induced reduction in nuclear factor erythroid 2-related factor 2 activity. Western blotting revealed increased glutathione peroxidase 4 and decreased transferrin receptor protein 1 expression. Immunostaining showed reduced 4-hydroxynonenal and malondialdehyde levels, especially in renal tubules, indicating suppressed ferroptosis. Luseogliflozin may protect the kidney from I/R injury by inhibiting ferroptosis through oxidative stress reduction.


Assuntos
Injúria Renal Aguda , Ferroptose , Camundongos Endogâmicos C57BL , Insuficiência Renal Crônica , Inibidores do Transportador 2 de Sódio-Glicose , Animais , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/patologia , Injúria Renal Aguda/prevenção & controle , Ferroptose/efeitos dos fármacos , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Camundongos , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Masculino , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Sorbitol/análogos & derivados , Sorbitol/farmacologia , Rim/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Estresse Oxidativo/efeitos dos fármacos , Fibrose , Modelos Animais de Doenças , Transportador 2 de Glucose-Sódio
9.
Revista Digital de Postgrado ; 13(2): e396, ago.2024. ilus, graf
Artigo em Espanhol | LILACS, LIVECS | ID: biblio-1567349

RESUMO

Angostura trifoliata (Willd) T.S. Elías (Rutaceae) es una planta, cuya corteza es empleada en Venezuela para el tratamiento de la diabetes mellitus, la malaria y la disminución de peso. Sin embargo, se ha demostrado que altas dosis de su extracto administrados en forma aguda producen hiperglicemia y alteraciones neurológicas. El objetivo de este estudio fue correlacionar los efectos histológicos a nivel hepático y renal en ratones sanos con la hiperglicemia aguda producida por el extracto de la corteza de esta planta. Métodos: Se realizó un estudio experimental in vivo utilizando el extracto diluido en agua y administrado vía ip a dosis de 452 y 700 mg/kg; se determinó la glicemia utilizando un glucómetro comercial; los efectos histológicos con hematoxilina eosina previa fijación de los órganos con formaldehído al 10%. En todos los casos, se comparó con el grupo control. Resultados: el extracto produjo hiperglicemia significativamente P<0,05. En el tejido hepático causó: pérdida parcial de su arquitectura, binucleación, vasos congestivos con elementos inflamatorios, núcleos hipercromáticos, espacios de Disse dilatados con hematíes y áreas de necrosis. En el riñón originó congestión vascular en los tubos contorneados proximales y distales, concomitante con ruptura y necrosis de la membrana basal. Conclusión: el extracto produce toxicidad hepática y renal que se correlacionan con hiperglicemia, por lo que podría ser considerado como un agente hepatotóxico y nefrotóxico. (AU)


Angostura trifoliata (Willd) T.S. Elías (Rutaceae) is a plant, whose bark is used in Venezuela for the treatment of diabetes mellitus, malaria and weight loss. However, it has been shown that high doses of its extract administered acutely produce hyperglycemia and neurological alterations. The objective of this study was to correlate the histological effects at the liver and kidney level in healthy mice with the acute hyperglycemia produced by the bark extract of this plant. Methods: An in vivo experimental study was carried out using the extract diluted in water and administered ip at doses of 452 and 700 mg/kg; blood glucose was determined using a commercial glucometer; the histological effects with hematoxylin eosin after fixation of the organs with 10% formaldehyde. In all cases, it was compared with the control group. Results: the extract produced hyperglycemia significantly P<0.05. In the liver tissue it caused: partial loss of its architecture, binucleation, congested vessels with inflammatory elements, hyperchromatic nuclei, dilated spaces of Disse with red blood cells and areas of necrosis. In the kidney, it caused vascular congestion in the proximal and distal convoluted tubes, concomitant with rupture and necrosis of the basement membrane. Conclusion: the extract produces liver and kidney toxicity that correlates with hyperglycemia, so it could be considered a hepatotoxic and nephrotoxic agent. (AU)


Assuntos
Animais , Camundongos , Insuficiência Renal/sangue , Injúria Renal Aguda/patologia , Hiperglicemia/diagnóstico , Autopsia , Casca de Planta/toxicidade
10.
PLoS One ; 19(8): e0308977, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39159207

RESUMO

Perioperative acute kidney injury (AKI), which is mainly mediated by renal ischemia‒reperfusion (I/R) injury, is commonly observed in clinical practice. However, effective measures for preventing and treating this perioperative complication are still lacking in the clinic. Thus, we designed this study to examine whether remote liver ischemic preconditioning (RLIPC) has a protective effect on damage caused by renal I/R injury. In a rodent model, 30 mice were divided into five groups to assess the effects of RLIPC and ERK1/2 inhibition on AKI. The groups included the sham-operated (sham), kidney ischemia and reperfusion (CON), remote liver ischemic preconditioning (RLIPC), CON with the ERK1/2 inhibitor U0126 (CON+U0126), and RLIPC with U0126 (RLIPC+U0126). RLIPC consisted of 4 liver ischemia cycles before renal ischemia. Renal function and injury were assessed through biochemical assays, histology, cell apoptosis and protein phosphorylation analysis. RLIPC significantly mitigated renal dysfunction, tissue damage, inflammation, and apoptosis caused by I/R, which was associated with ERK1/2 phosphorylation. Furthermore, ERK1/2 inhibition with U0126 negated the protective effects of RLIPC and exacerbated renal injury. To summarize, we demonstrated that RLIPC has a strong renoprotective effect on kidneys post I/R injury and that this effect may be mediated by phosphorylation of ERK1/2.


Assuntos
Injúria Renal Aguda , Precondicionamento Isquêmico , Fígado , Proteína Quinase 3 Ativada por Mitógeno , Nitrilas , Traumatismo por Reperfusão , Animais , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/metabolismo , Precondicionamento Isquêmico/métodos , Fígado/metabolismo , Fígado/patologia , Fígado/irrigação sanguínea , Fosforilação , Camundongos , Masculino , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Nitrilas/farmacologia , Rim/patologia , Rim/irrigação sanguínea , Rim/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Butadienos/farmacologia , Apoptose/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos
11.
Ren Fail ; 46(2): 2379601, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39099238

RESUMO

Acute kidney injury (AKI) is a significant issue in public health, displaying a high occurrence rate and mortality rate. Ferroptosis, a form of programmed cell death (PCD), is characterized by iron accumulation and intensified lipid peroxidation. Recent studies have demonstrated the pivotal significance of ferroptosis in AKI caused by diverse stimuli, including ischemia-reperfusion injury (IRI), sepsis and toxins. Autophagy, a multistep process that targets damaged organelles and macromolecules for degradation and recycling, also plays an essential role in AKI. Previous research has demonstrated that autophagy deletion in proximal tubules could aggravate tubular injury and renal function loss, indicating the protective function of autophagy in AKI. Consequently, finding ways to stimulate autophagy has become a crucial therapeutic strategy. The recent discovery of the role of selective autophagy in influencing ferroptosis has identified new therapeutic targets for AKI and has highlighted the importance of understanding the cross-talk between autophagy and ferroptosis. This study aims to provide an overview of the signaling pathways involved in ferroptosis and autophagy, focusing on the mechanisms and functions of selective autophagy and autophagy-dependent ferroptosis. We hope to establish a foundation for future investigations into the interaction between autophagy and ferroptosis in AKI as well as other diseases.


Assuntos
Injúria Renal Aguda , Autofagia , Ferroptose , Transdução de Sinais , Humanos , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/etiologia , Animais , Traumatismo por Reperfusão/metabolismo , Peroxidação de Lipídeos
12.
J Med Chem ; 67(16): 14234-14255, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39137258

RESUMO

Cisplatin is a widely used drug for the clinical treatment of tumors. However, nephrotoxicity limits its widespread use. A series of compounds including eight analogs (G3-G10) and 40 simplifiers (G11-G50) were synthesized based on the total synthesis of Psiguamer A and B, which were novel meroterpenoids with unusual skeletons from the leaves of Psidium guajava. Among these compounds, (d)-G8 showed the strongest protective effect on cisplatin-induced acute kidney injury (AKI) in vitro and vivo, and slightly enhanced the antitumor efficacy of cisplatin. A mechanistic study showed that (d)-G8 promoted the efflux of cisplatin via upregulating the copper transporting efflux proteins ATP7A and ATP7B. It enhanced autophagy through the activation of the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway. (d)-G8 showed no acute toxicity or apparent pathological damage in the healthy mice at a single dose of 1 g/kg. This study provides a promising lead against cisplatin-induced AKI.


Assuntos
Injúria Renal Aguda , Antineoplásicos , Cisplatino , Psidium , Cisplatino/farmacologia , Animais , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/patologia , Camundongos , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/síntese química , Psidium/química , Terpenos/farmacologia , Terpenos/síntese química , Terpenos/química , Masculino , Relação Estrutura-Atividade
13.
J Transl Med ; 22(1): 789, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39192240

RESUMO

BACKGROUND: Kidneys are at risk from drug-induced toxicity, with a significant proportion of acute kidney injury (AKI) linked to medications, particularly cisplatin. Existing cytoprotective drugs for cisplatin-AKI carry side effects, prompting a search for better biological therapies. Mesenchymal Stem Cells (MSCs) are under consideration given their regenerative properties, yet their clinical application has not achieved their full potential, mainly due to variability in the source of MSC tested. In addition, translating treatments from rodent models to humans remains challenging due to a lack of standardized dosing and understanding potential differential responses to cisplatin between animal strains. METHOD: In the current study, we performed a time-course analysis of the effect of cisplatin across different mouse strains and evaluated gender related differences to create a robust preclinical model that could then be used to explore the therapeutic efficacy of different sources of MSCs for their ability to reverse AKI. RESULT: Our data indicated that different mouse strains produce differential responses to the same cisplatin dosing regimen. Despite this, we did not observe any gender-related bias towards cisplatin nephrotoxicity. Furthermore, our time-course analysis identified that cisplatin-induced inflammation was driven by a strong CXCL1 response, which was used as a putative biomarker to evaluate the comparative therapeutic efficacy of different MSC sources in reversing AKI. Our data indicates that UC-MSCs have a stronger anti-inflammatory effect compared to BM-MSCs and AD-MSCs, which helped to ameliorate cisplatin-AKI. CONCLUSION: Overall, our data underscores the importance of using an optimized preclinical model of cisplatin-AKI to test different therapies. We identified CXCL1 as a potential biomarker of cisplatin-AKI and identified the superior efficacy of UC-MSCs in mitigating cisplatin-AKI.


Assuntos
Injúria Renal Aguda , Cisplatino , Modelos Animais de Doenças , Células-Tronco Mesenquimais , Cisplatino/efeitos adversos , Animais , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Masculino , Feminino , Camundongos Endogâmicos C57BL , Fatores de Tempo , Quimiocina CXCL1/metabolismo , Camundongos , Rim/efeitos dos fármacos , Rim/patologia , Inflamação/patologia
14.
Cells ; 13(16)2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39195224

RESUMO

Under vasculogenic conditioning, certain pro-inflammatory subsets within peripheral blood mononuclear cells (PBMCs) undergo phenotypic transformation into pro-regenerative types, such as vasculogenic endothelial progenitor cells, M2 macrophages, and regulatory T cells. These transformed cells are collectively termed regeneration-associated cells (RACs). In this study, we aimed to investigate the therapeutic efficacy of RAC-derived extracellular vesicles (RACev) compared with a vehicle-treated group in the context of renal ischemia-reperfusion injury (R-IRI). Human PBMCs were cultured with defined growth factor cocktails for seven days to harvest RACs. EV quantity and size were characterized by nanoparticle tracking analysis. Notably, the systemic injection of RACev significantly decreased serum creatinine and blood urine nitrogen at day three compared to the control group. Histologically, the treatment group showed less fibrosis in the cortex and medullary areas (p < 0.04 and p < 0.01) compared to the control group. The CD31 staining confirmed enhanced capillary densities in the treatment group compared to the control group (p < 0.003). These beneficial effects were accompanied by angiogenesis, anti-fibrosis, anti-inflammation, and anti-apoptosis RACev miR delivery to ischemic injury to control inflammatory, endothelial mesenchymal transition, and hypoxia pathways. In vivo bioluminescence analysis demonstrated a preferential accumulation of RACev in the IR-injured kidney. The systemic transplantation of RACev beneficially restored kidney function by protecting from tissue fibrosis and through anti-inflammation, angiogenesis, and anti-apoptosis miR delivery to the ischemic tissue.


Assuntos
Injúria Renal Aguda , Vesículas Extracelulares , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/transplante , Injúria Renal Aguda/patologia , Injúria Renal Aguda/terapia , Humanos , Animais , Masculino , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/terapia , Leucócitos Mononucleares/metabolismo , Fibrose , Apoptose/efeitos dos fármacos , Neovascularização Fisiológica , MicroRNAs/metabolismo , MicroRNAs/genética , Camundongos
15.
Life Sci ; 354: 122955, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39122109

RESUMO

AIMS: Losartan potassium-laden pegylated nanocubic vesicles (LP-NCVs-PEG) have an intriguing kidney-targeted nanoplatform for acute renal injury via blocking apoptosis and activating wnt/ß-catenin pathway. MAIN METHODS: Utilizing a thin-film hydration methodology established on 42 full factorial design to produce LP loaded nanocubic formulations (LP-NCVs) which composed mainly from L-α-phosphatidylcholine and poloxamer. The optimization process was designed to select the formulation with maximum entrapment efficiency (EE %), maximum in-vitro drug release (Q8h), and minimum vesicle size (VS). The optimum formulation was then pegylated to obtain LP-NCVs-PEG formulation that shields NCVs from the harsh ecosystem of the stomach, improves their oral drug delivery performance and targets the proximal renal tubules with no systemic toxicity. Male albino rats were injected with Cisplatin (6 mg/kg, i.p.) alone or with LP-formulations (5 mg/kg/day). Kidney injury markers, inflammatory markers, apoptotic markers. Besides renal tissue expression of Wnt, ß-Catenin, GSK-3ß, renal RNA gene expression of TCF-4, LEF-1 and histopathology were also analyzed to display pharmacological study. KEY FINDINGS: The pharmacokinetics studies demonstrated that LP-NCVs-PEG boosted LP bioavailability approximately 3.61 times compared to LP oral solution. Besides LP-NCVs-PEG may have an intriguing kidney-targeted nanoplatform for acute renal injury via decreased renal toxicity markers, renal expression of LEF-1, GSK3-ß, caspase, TNF-α, NF-κB and TUNEL expression. Alternatively, increased renal tissue level of Bcl-2, wnt, ß-catenin and TCF-4. SIGNIFICANCE: LP-NCVs-PEG improved LP pharmacokinetics targeting the kidney and improved injury by activating wnt/ß-catenin/TCF-4 pathway, blocking apoptosis, inflammation and renal toxicity markers suggesting it might be successful nephroprotective adjuvant therapy.


Assuntos
Injúria Renal Aguda , Apoptose , Cisplatino , Losartan , Polietilenoglicóis , Via de Sinalização Wnt , Animais , Masculino , Ratos , Apoptose/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Polietilenoglicóis/química , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/patologia , Losartan/farmacologia , beta Catenina/metabolismo , Nanopartículas/química , Fator de Transcrição 4/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Antineoplásicos/farmacologia , Ratos Wistar , Liberação Controlada de Fármacos
16.
Theranostics ; 14(11): 4536-4553, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39113797

RESUMO

Rationale: Acute kidney injury (AKI) has substantial rates of mortality and morbidity, coupled with an absence of efficacious treatment options. AKI commonly transits into chronic kidney disease (CKD) and ultimately culminates in end-stage renal failure. The interferon-stimulated gene 15 (ISG15) level was upregulated in the kidneys of mice injured by ischemia-reperfusion injury (IRI), cisplatin, or unilateral ureteral obstruction (UUO), however, its role in AKI development and subsequent AKI-to-CKD transition remains unknown. Methods: Isg15 knockout (Isg15 KO) mice challenged with bilateral or unilateral IRI, cisplatin, or UUO were used to investigate its role in AKI. We established cellular models with overexpression or knockout of ISG15 and subjected them to hypoxia-reoxygenation, cisplatin, or transforming growth factor- ß1 (TGF-ß1) stimulation. Renal RNA-seq data obtained from AKI models sourced from public databases and our studies, were utilized to examine the expression profiles of ISG15 and its associated genes. Additionally, published single cell RNA-seq data from human kidney allograft biopsies and mouse IRI model were analyzed to investigate the expression patterns of ISG15 and the type I TGF-ß receptor (TGFßR1). Western blotting, qPCR, co-immunoprecipitation, and immunohistochemical staining assays were performed to validate our findings. Results: Alleviated pathological injury and renal function were observed in Isg15 KO mice with IRI-, cisplatin-, or UUO-induced AKI and the following AKI-to-CKD transition. In hypoxia-reoxygenation, cisplatin or TGF-ß1 treated HK-2 cells, knockout ISG15 reduced stimulus-induced cell fibrosis, while overexpression of ISG15 with modification capacity exacerbated cell fibrosis. Immunoprecipitation assays demonstrated that ISG15 promoted ISGylation of TGFßR1, and inhibited its ubiquitination. Moreover, knockout of TGFßR1 blocked ISG15's fibrosis-exacerbating effect in HK-2 cells, while overexpression of TGFßR1 abolished the renal protective effect of ISG15 knockout during IRI-induced kidney injury. Conclusions: ISG15 plays an important role in the development of AKI and subsequent AKI-to-CKD transition by promoting TGFßR1 ISGylation.


Assuntos
Injúria Renal Aguda , Cisplatino , Citocinas , Camundongos Knockout , Traumatismo por Reperfusão , Ubiquitinas , Animais , Humanos , Masculino , Camundongos , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Cisplatino/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Rim/metabolismo , Rim/patologia , Camundongos Endogâmicos C57BL , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/genética , Ubiquitinas/metabolismo , Ubiquitinas/genética , Obstrução Ureteral/metabolismo , Obstrução Ureteral/complicações , Obstrução Ureteral/genética
17.
Sci Adv ; 10(32): eado2849, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39110788

RESUMO

Acute kidney injury (AKI) causes epithelial damage followed by subsequent repair. While successful repair restores kidney function, this process is often incomplete and can lead to chronic kidney disease (CKD) in a process called failed repair. To better understand the epigenetic reprogramming driving this AKI-to-CKD transition, we generated a single-nucleus multiomic atlas for the full mouse AKI time course, consisting of ~280,000 single-nucleus transcriptomes and epigenomes. We reveal cell-specific dynamic alterations in gene regulatory landscapes reflecting, especially, activation of proinflammatory pathways. We further generated single-nucleus multiomic data from four human AKI samples including validation by genome-wide identification of nuclear factor κB binding sites. A regularized regression analysis identifies key regulators involved in both successful and failed repair cell fate, identifying the transcription factor CREB5 as a regulator of both successful and failed tubular repair that also drives proximal tubular cell proliferation after injury. Our interspecies multiomic approach provides a foundation to comprehensively understand cell states in AKI.


Assuntos
Injúria Renal Aguda , Epigênese Genética , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Camundongos , Humanos , Transcriptoma , NF-kappa B/metabolismo , NF-kappa B/genética , Modelos Animais de Doenças , Reprogramação Celular/genética , Proliferação de Células/genética , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/metabolismo
18.
Mol Med ; 30(1): 133, 2024 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-39217289

RESUMO

OBJECTIVE: Renal ischemia/reperfusion injury (IRI) is a major cause of acute kidney injury (AKI), which is associated with high incidence and mortality. AST-120 is an oral carbonaceous adsorbent that can alleviate kidney damage. This study aimed to explore the effects of AST-120 on renal IRI and the molecular mechanism. METHODS: A renal IRI mouse model was established and administrated AST-120, and differentially expressed genes were screened using RNA sequencing. Renal function and pathology were analyzed in mice. Hypoxia/reoxygenation (H/R) cell model was generated, and glycolysis was evaluated by detecting lactate levels and Seahorse analysis. Histone lactylation was analyzed by western blotting, and its relationship with hexokinase 2 (HK2) was assessed using chromatin immunoprecipitation. RESULTS: The results showed that HK2 expression was increased after IRI, and AST-120 decreased HK2 expression. Knockout of HK2 attenuated renal IRI and inhibits glycolysis. AST-120 inhibited renal IRI in the presence of HK2 rather than HK2 absence. In proximal tubular cells, knockdown of HK2 suppressed glycolysis and H3K18 lactylation caused by H/R. H3K18 lactylation was enriched in HK2 promoter and upregulated HK2 levels. Rescue experiments revealed that lactate reversed IRI that suppressed by HK2 knockdown. CONCLUSIONS: In conclusion, AST-120 alleviates renal IRI via suppressing HK2-mediated glycolysis, which suppresses H3K18 lactylation and further reduces HK2 levels. This study proposes a novel mechanism by which AST-120 alleviates IRI.


Assuntos
Carbono , Modelos Animais de Doenças , Glicólise , Hexoquinase , Óxidos , Traumatismo por Reperfusão , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Hexoquinase/metabolismo , Hexoquinase/genética , Glicólise/efeitos dos fármacos , Camundongos , Masculino , Óxidos/farmacologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Rim/metabolismo , Rim/patologia , Rim/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Histonas/metabolismo , Humanos , Linhagem Celular
19.
Toxicology ; 508: 153919, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39137829

RESUMO

Nephrotoxicity, including electrolytic disorders and acute kidney injury (AKI), limits the clinical dosage and utility of platinated antineoplastics such as cisplatin. Cisplatin nephrotoxicity embodies a tubulopathy involving the medullary S2 and S3 segments of the proximal and the distal tubules. Higher dosage extends damage over the cortical S1 segment and intensifies overall injury. However, the standard diagnosis based on plasma creatinine as well as novel injury biomarkers lacks enough pathophysiological specificity. Further granularity in the detection of renal injury would help understand the implications of individual damage patterns needed for personalized patient handling. In this article, we studied the association of urinary ganglioside GM2 activator protein (GM2AP) with the patterns of tubular damage produced by 5 and 10 mg/kg cisplatin in rats. Our results show that GM2AP appears in the urine only following damage to the cortical segment of the proximal tubule. The information provided by GM2AP is not redundant with but distinct and complementary to that provided by urinary neutrophil gelatinase-associated lipocalin (NGAL). Similarly, treatment with 150 mg/kg/day gentamicin damages the renal cortex and increases GM2AP urinary excretion; whereas renal ischemia, which does not affect the cortex, has no effect on GM2AP. Because of the key role of the cortical proximal tubule in renal function, we contend GM2AP as a potential diagnostic biomarker to stratify AKI patients according to the underlying damage and follow their evolution and prognosis. Prospectively, urinary GM2AP may help grade the severity of platinated antineoplastic nephrotoxicity by forming part of a non-invasive liquid biopsy.


Assuntos
Injúria Renal Aguda , Antineoplásicos , Biomarcadores , Cisplatino , Proteína Ativadora de G(M2) , Córtex Renal , Cisplatino/toxicidade , Animais , Masculino , Ratos , Biomarcadores/urina , Antineoplásicos/toxicidade , Córtex Renal/efeitos dos fármacos , Córtex Renal/patologia , Córtex Renal/metabolismo , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Injúria Renal Aguda/urina , Gentamicinas/toxicidade , Ratos Sprague-Dawley , Lipocalina-2/urina , Índice de Gravidade de Doença
20.
Life Sci ; 353: 122936, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39094904

RESUMO

Diclofenac (DF), a non-steroidal anti-inflammatory drug, is commonly used to relieve pain and inflammation. High doses of DF might induce acute kidney injury (AKI), particularly in elderly, a known vulnerable population. AIM: We aimed to assess the protective role of melatonin (Mel) on DF-induced AKI in aged rats and to highlight the underpinning mechanisms include, oxidative stress and inflammation focusing on microRNA-34a (miR-34a), nuclear factor erythroid-2-related factor-2/hemeoxygenase-1 (Nrf2/HO-1) and NLR family-pyrin domain containing-3 (NLRP3) inflammasome pathways, and to elucidate the possibility of epithelial sodium channel (ENaC) involvement. MATERIALS AND METHODS: Thirty old male Wistar rats were allocated randomly into 3 groups: Control, DF and Mel-DF groups. KEY FINDINGS: Melatonin provided nephroprotective effects against DF-induced AKI via attenuating the expression of renal miR-34a and subsequently promoting the signaling of Nrf2/HO-1 with elevation of the antioxidant defense capacity and suppressing NLRP3 inflammasomes. Melatonin alleviated DF-induced hypernatremia via decreasing the ENaC expression. Renal histopathological examination revealed significant reduction in vascular congestion, mononuclear infiltration, glomerulo-tubular damage, fibrosis and TNF-α optical density. SIGNIFICANCE: It can be assumed that melatonin is a promising safe therapeutic agent in controlling DF-induced AKI in elderly.


Assuntos
Injúria Renal Aguda , Anti-Inflamatórios não Esteroides , Diclofenaco , Melatonina , Estresse Oxidativo , Ratos Wistar , Animais , Melatonina/farmacologia , Melatonina/uso terapêutico , Masculino , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/patologia , Ratos , Anti-Inflamatórios não Esteroides/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Antioxidantes/farmacologia , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Substâncias Protetoras/farmacologia , Rim/efeitos dos fármacos , Rim/patologia , Rim/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...