Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.275
Filtrar
1.
Acta Pharmacol Sin ; 45(4): 777-789, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38200148

RESUMO

Renal fibrosis is the final pathological change in renal disease, and aging is closely related to renal fibrosis. Mitochondrial dysfunction has been reported to play an important role in aging, but the exact mechanism remains unclear. Disulfide-bond A oxidoreductase-like protein (DsbA-L) is mainly located in mitochondria and plays an important role in regulating mitochondrial function and endoplasmic reticulum (ER) stress. However, the role of DsbA-L in renal aging has not been reported. In this study, we showed a reduction in DsbA-L expression, the disruption of mitochondrial function and an increase in fibrosis in the kidneys of 12- and 24-month-old mice compared to young mice. Furthermore, the deterioration of mitochondrial dysfunction and fibrosis were observed in DsbA-L-/- mice with D-gal-induced accelerated aging. Transcriptome analysis revealed a decrease in Flt4 expression and inhibition of the PI3K-AKT signaling pathway in DsbA-L-/- mice compared to control mice. Accelerated renal aging could be alleviated by an AKT agonist (SC79) or a mitochondrial protector (MitoQ) in mice with D-gal-induced aging. In vitro, overexpression of DsbA-L in HK-2 cells restored the expression of Flt4, AKT pathway factors, SP1 and PGC-1α and alleviated mitochondrial damage and cell senescence. These beneficial effects were partially blocked by inhibiting Flt4. Finally, activating the AKT pathway or improving mitochondrial function with chemical reagents could alleviate cell senescence. Our results indicate that the DsbA-L/AKT/PGC-1α signaling pathway could be a therapeutic target for age-related renal fibrosis and is associated with mitochondrial dysfunction.


Assuntos
Glutationa Transferase , Nefropatias , Rim , Mitocôndrias , Animais , Camundongos , Envelhecimento , Fibrose , Homeostase , Rim/patologia , Nefropatias/enzimologia , Mitocôndrias/enzimologia , Doenças Mitocondriais/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Glutationa Transferase/metabolismo
2.
Molecules ; 27(2)2022 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-35056661

RESUMO

Cell cycle kinases represent an important component of the cell machinery that controls signal transduction involved in cell proliferation, growth, and differentiation. Nek2 is a mitotic Ser/Thr kinase that localizes predominantly to centrosomes and kinetochores and orchestrates centrosome disjunction and faithful chromosomal segregation. Its activity is tightly regulated during the cell cycle with the help of other kinases and phosphatases and via proteasomal degradation. Increased levels of Nek2 kinase can promote centrosome amplification (CA), mitotic defects, chromosome instability (CIN), tumor growth, and cancer metastasis. While it remains a highly attractive target for the development of anti-cancer therapeutics, several new roles of the Nek2 enzyme have recently emerged: these include drug resistance, bone, ciliopathies, immune and kidney diseases, and parasitic diseases such as malaria. Therefore, Nek2 is at the interface of multiple cellular processes and can influence numerous cellular signaling networks. Herein, we provide a critical overview of Nek2 kinase biology and discuss the signaling roles it plays in both normal and diseased human physiology. While the majority of research efforts over the last two decades have focused on the roles of Nek2 kinase in tumor development and cancer metastasis, the signaling mechanisms involving the key players associated with several other notable human diseases are highlighted here. We summarize the efforts made so far to develop Nek2 inhibitory small molecules, illustrate their action modalities, and provide our opinion on the future of Nek2-targeted therapeutics. It is anticipated that the functional inhibition of Nek2 kinase will be a key strategy going forward in drug development, with applications across multiple human diseases.


Assuntos
Doenças Ósseas/patologia , Inibidores Enzimáticos/farmacologia , Doenças do Sistema Imunitário/patologia , Nefropatias/patologia , Malária/patologia , Quinases Relacionadas a NIMA/antagonistas & inibidores , Neoplasias/patologia , Doenças Ósseas/tratamento farmacológico , Doenças Ósseas/enzimologia , Resistência a Medicamentos , Humanos , Doenças do Sistema Imunitário/tratamento farmacológico , Doenças do Sistema Imunitário/enzimologia , Nefropatias/tratamento farmacológico , Nefropatias/enzimologia , Malária/tratamento farmacológico , Malária/enzimologia , Metástase Neoplásica , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia
3.
J Mol Histol ; 53(1): 133-143, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34655350

RESUMO

Methotrexate is used for cure of many cancer types. It has many side effects. For this reason, obtaining a nephroprotective agent is obligatory. In the study, our aim is to determine probable effects of Vitamin B12 on MTX caused kidney damages in rats. Rats were randomly divided into 4 groups, including 8 animals in each group. Control group, VitB12 group (3 µg-kg-ip B12 throughout 15 days), MTX group (at the 8th day of experiment, a single dose of 20 mg-kg-ip MTX), Vit B12 + MTX group (3 µg-kg-ip B12 throughout 15 days and at the 8th day of experiment, a single dose of 20 mg-kg-ip MTX) Animals were anesthetized and kidney tissues were removed to evaluate biochemically, immunohistochemically and histopathologycally. There were histopathological deteriorations, rises of apoptotic cells, expressions of heat shock proteins, endoplasmic reticulum stress and inflammation markers in the MTX group. In the MTX group, Superoxide Dismutase (SOD), Total Antioxidant Status (TAS) and Catalase (CAT) levels decreased, but Total Oxidant Status TOS, Malondialdehyde (MDA) and interleukin-6 (IL6) levels increased. In addition, there was amelioration in kidney tissue in Vit B12 + MTX group compared to the MTX group. We suggest that Vit B12 can be used to reduce the toxic effects of MTX.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Nefropatias/prevenção & controle , Metotrexato/toxicidade , Vitamina B 12/uso terapêutico , Complexo Vitamínico B/uso terapêutico , Animais , Apoptose , Catalase/metabolismo , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Interleucina-6/metabolismo , Nefropatias/induzido quimicamente , Nefropatias/enzimologia , Masculino , Malondialdeído/metabolismo , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo
4.
FASEB J ; 36(1): e22122, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34958158

RESUMO

Lysine-specific histone demethylase 1 (LSD1) as the first identified histone/lysine demethylase regulates gene expression and protein functions in diverse diseases. In this study, we show that the expression of LSD1 is increased in mouse kidneys with unilateral ureteral obstruction (UUO) and in cultured NRK-52E cells undergoing TGF-ß1-induced epithelial-mesenchymal transition (EMT). Inhibition of LSD1 with its specific inhibitor ORY1001 attenuated renal EMT and fibrosis, which was associated with decreased the deposition of extracellular matrix proteins and the expression of fibrotic markers, including α-smooth muscle actin (α-SMA) and fibronectin, and the recovery of E-cadherin expression and decrease of N-cadherin expression in UUO kidneys and in NRK-52E cells induced with TGF-ß1. Targeting LSD1 also decreased the expression of Snail family transcriptional repressor 1 (Snail-1) and its interaction with LSD1 in UUO kidneys and in NRK-52E cells treated with TGF-ß1. In addition, we identified a novel LSD1-14-3-3ζ-PKCα axis in the regulation of the activation of AKT and Stat3 and then the activation of fibroblasts. This study suggests that LSD1 plays a critical role in regulation of renal EMT and fibrosis through activation of diverse signaling pathways and places an emphasis that LSD1 has potential as a therapeutic target for the treatment of renal fibrosis.


Assuntos
Sistemas de Liberação de Medicamentos , Inibidores Enzimáticos/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Histona Desmetilases , Rim/enzimologia , Animais , Linhagem Celular , Transição Epitelial-Mesenquimal/genética , Fibrose , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Nefropatias/tratamento farmacológico , Nefropatias/enzimologia , Nefropatias/genética , Masculino , Camundongos , Ratos
5.
Biochim Biophys Acta Mol Cell Res ; 1869(3): 119165, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34699872

RESUMO

Besides its involvement in blood and bone physiology, the kidney's main function is to filter substances and thereby regulate the electrolyte composition of body fluids, acid-base balance and toxin removal. Depending on underlying conditions, the nephron must undergo remodeling and cellular adaptations. The proteolytic removal of cell surface proteins via ectodomain shedding by A Disintegrin and Metalloproteases (ADAMs) is of importance for the regulation of cell-cell and cell-matrix adhesion of renal cells. ADAM10 controls glomerular and tubule development in a Notch1 signaling-dependent manner and regulates brush border composition. ADAM17 regulates the renin angiotensin system and is together with ADAM10 involved in calcium phosphate homeostasis. In kidney disease ADAMs, especially ADAM17 contribute to inflammation through their involvement in IL-6 trans-signaling, Notch-, epithelial growth factor receptor-, and tumor necrosis factor α signaling. ADAMs are interesting drug targets to reduce the inflammatory burden, defective cell adhesion and impaired signaling pathways in kidney diseases.


Assuntos
Proteínas ADAM/metabolismo , Nefropatias/patologia , Rim/fisiopatologia , Proteínas de Membrana/metabolismo , Proteólise , Proteínas ADAM/genética , Animais , Humanos , Rim/enzimologia , Nefropatias/enzimologia
6.
Am J Physiol Renal Physiol ; 321(6): F740-F756, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34747196

RESUMO

Renal fibrosis is the common pathological pathway in progressive renal diseases. In the present study, we analyzed the roles of semaphorin 3 A (SEMA3A) on renal fibrosis and the effect of SEMA3A inhibitor (SEMA3A-I) using a unilateral ureteral obstruction (UUO) mouse model. Expression of SEMA3A in the proximal tubulus and neuropilin-1, a recepor of SEMA3A, in fibloblast and tubular cells were increased in UUO kidneys. The expression of myofibroblast marker tenascin-C and fibronection as well as renal fibrosis were increased in UUO kidneys, all of which were ameliorated by SEMA3A-I. In addition, the JNK signaling pathway, known as the target of SEMA3A signaling, was activated in proximal tubular cells and fibroblast cells after UUO surgery, and SEMA3A-I significantly attenuated the activation. In vitro, treatments with SEMA3A as well as transforming growth factor-ß1 (TGF-ß1) in human proximal tubular cells lost epithelial cell characteristics, and SEMA3A-I significantly ameliorated this transformation. The JNK inhibitor SP600125 partially reversed SEMA3A and TGF-ß1-induced cell transformation, indicating that JNK signaling is involved in SEMA3A-induced renal fibrosis. In addition, treatment with SEMA3A in fibroblast cells activated expression of tenascin-C, collagen type I, and fibronection, indicating that SEMA3A may accelerate renal fibrosis through the activation of fibroblast cells. Analysis of human data revealed the positive correlation between urinary SEMA3A and urinary N-acetyl-ß-d-glucosaminidase, indicating the association between SEMA3A and tubular injury. In conclusion, SEMA3A signaling is involved in renal fibrosis through the JNK signaling pathway and SEMA3A-I might be a therapeutic option for protecting from renal fibrosis.NEW & NOTEWORTHY Renal fibrosis is the common pathological pathway in the progression of renal diseases. This study, using a unilateral ureteral obstruction (UUO) mouse model, indicated increased semaphorin3A (SEMA3A) signaling in renal tubular cells as well as fibroblast cells under UUO surgery, and SEMA3A inhibitor ameliorated UUO-induced renal fibrosis through the regulation of JNK signaling. The study proposes the potential therapeutic option of SEMA3A inhibitor to treat renal fibrosis.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Nefropatias/prevenção & controle , Rim/efeitos dos fármacos , Fármacos Renais/farmacologia , Semaforina-3A/antagonistas & inibidores , Adulto , Idoso , Animais , Modelos Animais de Doenças , Feminino , Fibrose , Humanos , Rim/enzimologia , Rim/metabolismo , Nefropatias/enzimologia , Nefropatias/etiologia , Nefropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Células NIH 3T3 , Semaforina-3A/metabolismo , Transdução de Sinais , Obstrução Ureteral/complicações
7.
Am J Physiol Renal Physiol ; 321(6): F675-F688, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34658261

RESUMO

Expansion of renal lymphatic networks, or lymphangiogenesis (LA), is well recognized during development and is now being implicated in kidney diseases. Although LA is associated with multiple pathological conditions, very little is known about its role in acute kidney injury. The purpose of this study was to evaluate the role of LA in a model of cisplatin-induced nephrotoxicity. LA is predominately regulated by vascular endothelial growth factor (VEGF)-C and VEGF-D, ligands that exert their function through their cognate receptor VEGF receptor 3 (VEGFR3). We demonstrated that use of MAZ51, a selective VEGFR3 inhibitor, caused significantly worse structural and functional kidney damage in cisplatin nephrotoxicity. Apoptotic cell death and inflammation were also increased in MAZ51-treated animals compared with vehicle-treated animals following cisplatin administration. Notably, MAZ51 caused significant upregulation of intrarenal phospho-NF-κB, phospho-JNK, and IL-6. Cisplatin nephrotoxicity is associated with vascular congestion due to endothelial dysfunction. Using three-dimensional tissue cytometry, a novel approach to explore lymphatics in the kidney, we detected significant vascular autofluorescence attributed to erythrocytes in cisplatin alone-treated animals. Interestingly, no such congestion was detected in MAZ51-treated animals. We found increased renal vascular damage in MAZ51-treated animals, whereby MAZ51 caused a modest decrease in the endothelial markers endomucin and von Willebrand factor, with a modest increase in VEGFR2. Our findings identify a protective role for de novo LA in cisplatin nephrotoxicity and provide a rationale for the development of therapeutic approaches targeting LA. Our study also suggests off-target effects of MAZ51 on the vasculature in the setting of cisplatin nephrotoxicity.NEW & NOTEWORTHY Little is known about injury-associated LA in the kidney and its role in the pathophysiology of acute kidney injury (AKI). Observed exacerbation of cisplatin-induced AKI after LA inhibition was accompanied by increased medullary damage and cell death in the kidney. LA inhibition also upregulated compensatory expression of LA regulatory proteins, including JNK and NF-κB. These data support the premise that LA is induced during AKI and lymphatic expansion is a protective mechanism in cisplatin nephrotoxicity.


Assuntos
Indóis/toxicidade , Nefropatias/induzido quimicamente , Rim/efeitos dos fármacos , Linfangiogênese/efeitos dos fármacos , Vasos Linfáticos/efeitos dos fármacos , Naftalenos/toxicidade , Inibidores de Proteínas Quinases/toxicidade , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Cisplatino , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Rim/enzimologia , Rim/patologia , Rim/fisiopatologia , Nefropatias/enzimologia , Nefropatias/patologia , Nefropatias/fisiopatologia , Vasos Linfáticos/enzimologia , Vasos Linfáticos/patologia , Vasos Linfáticos/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Fosforilação , Transdução de Sinais , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Oxid Med Cell Longev ; 2021: 2999296, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34712381

RESUMO

Calorie restriction (CR) extends lifespan and increases resistance to multiple forms of stress, including renal ischemia-reperfusion (I/R) injury. However, whether CR has protective effects on contrast-induced nephropathy (CIN) remains to be determined. In this study, we evaluated the therapeutic effects of CR on CIN and investigated the potential mechanisms. CIN was induced by the intravenous injection of iodinated contrast medium (CM) iopromide (1.8 g/kg) into Sprague Dawley rats with normal food intake or 40% reduced food intake, 4 weeks prior to iopromide administration. We found that CR was protective of CIN, assessed by renal structure and function. CM increased apoptosis, reactive oxygen species (ROS), and inflammation in the renal outer medulla, which were decreased by CR. The silent information regulator 1 (SIRT1) participated in the protective effect of CR on CIN, by upregulating glutathione peroxidase 4 (GPX4), a regulator of ferroptosis, because this protective effect was reversed by EX527, a specific SIRT1 antagonist. Our study showed that CR protected CIN via SIRT1/GPX4 activation. CR may be used to mitigate CIN.


Assuntos
Restrição Calórica , Nefropatias/prevenção & controle , Rim/enzimologia , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Sirtuína 1/metabolismo , Animais , Apoptose , Meios de Contraste , Citocinas/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Ferroptose , Mediadores da Inflamação/metabolismo , Iohexol/análogos & derivados , Rim/patologia , Nefropatias/induzido quimicamente , Nefropatias/enzimologia , Nefropatias/patologia , Masculino , Estresse Oxidativo , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
9.
Toxicol Appl Pharmacol ; 432: 115753, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34637808

RESUMO

T-2 toxin is a highly toxic trichothecene that can induce toxic effects in a variety of organs and tissues, but the pathogenesis of its nephrotoxicity has not been elucidated. In this study, we assessed the involvement of protein kinase RNA-like ER kinase (PERK)-mediated endoplasmic reticulum (ER) stress and apoptosis in PK-15 cells cultured at different concentrations of T-2 toxin. Cell viability, antioxidant capacity, intracellular calcium (Ca2+) content, apoptotic rate, levels of ER stress, and apoptosis-related proteins were studied. T-2 toxin inhibited cell proliferation; increased the apoptosis rate; and was accompanied by increased cleaved caspase-3 expression, altered intracellular oxidative stress marker levels, and intracellular Ca2+ overloading. The ER stress inhibitor 4-phenylbutyrate (4-PBA) and PERK selective inhibitor GSK2606414 prevented the decrease of cell activity and apoptosis caused by T-2 toxin. The altered expression of glucose regulatory protein 78 (GRP78), C/EBP homologous protein (CHOP), and caspase-12 proved that ER stress was involved in cell injury triggered by T-2 toxin. T-2 toxin activated the phosphorylation of PERK and the alpha subunit of eukaryotic initiation factor 2 (eIF2α) and upregulated the activating transcription factor 4 (ATF4), thereby triggering ER stress via the GRP78/PERK/CHOP signaling pathway. This study provides a new perspective for understanding the nephrotoxicity of T-2 toxin.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Fator de Iniciação 2 em Eucariotos/metabolismo , Nefropatias/induzido quimicamente , Rim/efeitos dos fármacos , Toxina T-2/toxicidade , eIF-2 Quinase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspase 12/metabolismo , Linhagem Celular , Chaperona BiP do Retículo Endoplasmático/metabolismo , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Rim/enzimologia , Rim/patologia , Nefropatias/enzimologia , Nefropatias/patologia , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais , Sus scrofa , Fator de Transcrição CHOP/metabolismo
10.
Cell Death Dis ; 12(8): 792, 2021 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-34392303

RESUMO

Impaired energy metabolism in proximal tubular epithelial cells (PTECs) is strongly associated with various kidney diseases. Here, we characterized proximal tubular phenotype alternations during kidney injury and repair in a mouse model of folic acid nephropathy, in parallel, identified carnitine palmitoyltransferase 1α (CPT1α) as an energy stress response accompanied by renal tubular dedifferentiation. Genetic ablation of Cpt1α aggravated the tubular injury and interstitial fibrosis and hampered kidney repair indicate that CPT1α is vital for the preservation and recovery of tubular phenotype. Our data showed that the lipid accumulation and mitochondrial mass reduction induced by folic acid were persistent and became progressively more severe in PTECs without CPT1α. Interference of CPT1α reduced capacities of mitochondrial respiration and ATP production in PTECs, and further sensitized cells to folic acid-induced phenotypic changes. On the contrary, overexpression of CPT1α protected mitochondrial respiration and prevented against folic acid-induced tubular cell damage. These findings link CPT1α to intrinsic mechanisms regulating the mitochondrial respiration and phenotype of kidney tubules that may contribute to renal pathology during injury and repair.


Assuntos
Carnitina O-Palmitoiltransferase/metabolismo , Túbulos Renais/enzimologia , Túbulos Renais/patologia , Mitocôndrias/metabolismo , Trifosfato de Adenosina/biossíntese , Animais , Nitrogênio da Ureia Sanguínea , Carnitina O-Palmitoiltransferase/deficiência , Respiração Celular , Células Cultivadas , Creatinina/metabolismo , Fibrose , Ácido Fólico , Nefropatias/enzimologia , Nefropatias/patologia , Túbulos Renais/lesões , Túbulos Renais/ultraestrutura , Metabolismo dos Lipídeos , Masculino , Camundongos Endogâmicos C57BL , Mitocôndrias/ultraestrutura , Fenótipo
11.
Life Sci ; 282: 119843, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34298037

RESUMO

AIMS: Ischemia/reperfusion (I/R) occurs in renal artery stenosis, partial nephrectomy and most commonly during kidney transplantation. It brings serious consequences such as DGF (Delayed Graft Function) or organ dysfunction leading to renal failure and ultimate death. There is no effective therapy to handle the consequences of Renal Ischemia/Reperfusion (I/R) injury. Cyclic nucleotides, cAMP and cGMP are the important second messengers that stimulate intracellular signal transduction for cell survival in response to growth factors and peptide hormones in normal tissues and in kidneys plays significant role that involves vascular tone regulation, inflammation and proliferation of parenchymal cells. Renal ischemia and subsequent reperfusion injury stimulate signal transduction pathways involved in oxidative stress, inflammation, alteration in renal blood flow leading to necrosis and apoptosis of renal cell. MATERIALS AND METHODS: An extensive literature review of various search engines like PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out. To understand the functioning of Phosphodiesterases (PDEs) and its pharmacological modulation in Renal Ischemia-Reperfusion Injury. KEY FINDINGS: Current therapeutic options may not be enough to treat renal I/R injury in group of patients and therefore, the current review has discussed the general characteristics and physiology of PDEs and preclinical-studies defining the relationship between PDEs expression in renal injury due to I/R and its outcome on renal function. SIGNIFICANCE: The role of PDE inhibitors in renal I/R injury and the clinical status of drugs for various renal diseases have been summarized in this review.


Assuntos
Nefropatias , Rim/enzimologia , Inibidores de Fosfodiesterase/uso terapêutico , Diester Fosfórico Hidrolases/metabolismo , Traumatismo por Reperfusão , Transdução de Sinais/efeitos dos fármacos , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Humanos , Nefropatias/tratamento farmacológico , Nefropatias/enzimologia , Nefropatias/patologia , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/enzimologia
13.
Front Immunol ; 12: 690697, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093596

RESUMO

Renal fibrosis is the final common pathway to chronic kidney diseases regardless of etiology. Parkinson disease protein 7 (PARK7) is a multifunctional protein involved in various cellular processes, but its pathophysiological role in kidneys remain largely unknown. Here, we have determined the role of PARK7 in renal fibrosis and have further elucidated the underlying mechanisms by using the in vivo mouse model of unilateral ureteric obstruction (UUO) and the in vitro model of transforming growth factor-b (TGFB1) treatment of cultured kidney proximal tubular cells. PARK7 decreased markedly in atrophic kidney tubules in UUO mice, and Park7 deficiency aggravated UUO-induced renal fibrosis, tubular cell apoptosis, ROS production and inflammation. In vitro, TGFB1 treatment induced fibrotic changes in renal tubular cells, which was accompanied by alterations of PARK7. Park7 knockdown exacerbated TGFB1-induced fibrotic changes, cell apoptosis and ROS production, whereas Park7 overexpression or treatment with ND-13 (a PARK7-derived peptide) attenuated these TGFB1-induced changes. Mechanistically, PARK7 translocated into the nucleus of renal tubular cells following TGFB1 treatment or UUO, where it induced the expression of SOD2, an antioxidant enzyme. Taken together, these results indicate that PARK7 protects against chronic kidney injury and renal fibrosis by inducing SOD2 to reduce oxidative stress in tubular cells.


Assuntos
Nefropatias/prevenção & controle , Túbulos Renais Proximais/enzimologia , Estresse Oxidativo , Proteína Desglicase DJ-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/biossíntese , Animais , Apoptose , Linhagem Celular , Modelos Animais de Doenças , Indução Enzimática , Fibrose , Nefropatias/enzimologia , Nefropatias/etiologia , Nefropatias/patologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Desglicase DJ-1/genética , Transdução de Sinais , Superóxido Dismutase/genética , Fator de Crescimento Transformador beta1/farmacologia , Obstrução Ureteral/complicações
15.
Mar Drugs ; 19(4)2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33810216

RESUMO

Mitochondrial dysfunction contributes to the pathogenesis of kidney injury related with cardiovascular disease. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) protects renal tubular cells by upregulating nuclear factor erythroid 2-related factor 2 (Nrf2). AMP-activated protein kinase (pAMPK)-mediated phosphorylation and sirtuin 1/3 (SIRT1/3)-mediated deacetylation are required for PGC-1α activation. In the present study, we aimed to investigate whether omega-3 fatty acids (FAs) regulate the expression of mediators of mitochondrial biogenesis in 5/6 nephrectomy (Nx) rats. Male Sprague-Dawley rats were assigned to the following groups: sham control, Nx, and Nx treated with omega-3 FA. The expression of PGC-1α, phosphorylated PGC-1α (pPGC-1α), acetylated PGC-1α, and factors related to mitochondrial biogenesis was examined through Western blot analysis. Compared to the control group, the expression of PGC-1α, pAMPK, SIRT1/3, Nrf1, mTOR, and Nrf2 was significantly downregulated, and that of Keap 1, acetylated PGC-1α, and FoxO1/3, was significantly upregulated in the Nx group. These changes in protein expression were rescued in the omega-3 FA group. However, the expression of pPGC-1α was similar among the three groups. Omega-3 FAs may involve mitochondrial biogenesis by upregulating Nrf1 and Nrf2. This protective mechanism might be attributed to the increased expression and deacetylation of PGC-1α, which was triggered by SIRT1/3.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Nefropatias/tratamento farmacológico , Rim/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Fator 1 Nuclear Respiratório/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Sirtuína 1/metabolismo , Sirtuínas/metabolismo , Acetilação , Animais , Modelos Animais de Doenças , Rim/enzimologia , Rim/patologia , Nefropatias/enzimologia , Nefropatias/etiologia , Nefropatias/patologia , Masculino , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Nefrectomia , Biogênese de Organelas , Processamento de Proteína Pós-Traducional , Ratos Sprague-Dawley , Transdução de Sinais
16.
Toxicol Lett ; 345: 12-23, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-33857584

RESUMO

We previously determined that specific microRNAs (miRNAs) are involved in renal pathophysiological occurrences induced by cadmium (Cd) in rats. This study expands our studies on miRNAs, determining their role in Cd-induced nephrotoxicity in occupational workers. We performed miRNA microarray analyses of blood and urine samples from patients diagnosed as occupational chronic Cd poisoning (OCCP) with abnormally elevated concentrations of urinary beta-2-microglobulin (U-ß2-MG), an indicator of tubular proteinuria. We also performed in vitro bioinformatics-based investigations of apoptosis-related genes targeted by miRNAs involved in the biological response to Cd exposure. We tested five differentially expressed miRNAs and determined a significant increase of sera miR-363-3p. Also, we determined that miR-363-3p increase is associated with phosphoinositide 3-kinase (PI3K) down-regulation and the suppressed proliferation and enhanced apoptosis of renal tubule epithelial cells. The obtained results suggest miR-363-3p involvement in the pathophysiology of Cd-induced renal injury in humans and maybe considered for possible interventional therapeutic strategies for Cd-associated kidney damage.


Assuntos
Apoptose/efeitos dos fármacos , Cádmio/efeitos adversos , Nefropatias/induzido quimicamente , Túbulos Renais Proximais/efeitos dos fármacos , MicroRNAs/metabolismo , Exposição Ocupacional/efeitos adversos , Fosfatidilinositol 3-Quinase/metabolismo , Adulto , Animais , Estudos de Casos e Controles , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo , Feminino , Regulação Enzimológica da Expressão Gênica , Humanos , Nefropatias/enzimologia , Nefropatias/genética , Nefropatias/patologia , Túbulos Renais Proximais/enzimologia , Túbulos Renais Proximais/patologia , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Saúde Ocupacional , Fosfatidilinositol 3-Quinase/genética , Ratos , Transdução de Sinais
17.
Pancreas ; 50(3): 393-398, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33835971

RESUMO

OBJECTIVE: The clinical significance of increased serum pancreatic enzymes (PEs) in coronavirus disease 2019 (COVID-19) patients has not yet been fully understood. We aimed to investigate the frequency and the impact on clinical outcome of PE elevation and acute pancreatitis in such patients. METHODS: Clinical data, laboratory tests, and cross-sectional images were analyzed from COVID-19 patients admitted to the Tor Vergata Hospital in Rome. Variables associated with PE abnormalities, intensive care unit (ICU) admission, or death were investigated through univariate and multivariate analyses and Cox proportional hazard model. RESULTS: Pancreatic enzymes were available in 254 of 282 COVID-19 patients. Among these, 66 patients (26%) showed mild elevation of PE, and 11 patients (4.3%) had severe elevation (>3 times of the upper limit of normal). Overall, 2 patients met the diagnostic criteria for acute pancreatitis. Hepatic and renal involvements were associated with PE elevation. Multivariate analysis showed that mild and severe PE elevations were significantly associated with ICU admission (odds ratios, 5.51 [95% confidence interval, 2.36-12.89; P < 0.0001] and 26.2 [95% confidence interval, 4.82-142.39; P < 0.0001]). CONCLUSIONS: Increase in serum PE, but not acute pancreatitis, is frequent in hospitalized COVID-19 patients and associates with ICU admission.


Assuntos
COVID-19/epidemiologia , Hospitalização/estatística & dados numéricos , Unidades de Terapia Intensiva , Pâncreas/enzimologia , Pancreatite/epidemiologia , Idoso , Idoso de 80 Anos ou mais , COVID-19/sangue , COVID-19/enzimologia , COVID-19/mortalidade , Feminino , Humanos , Nefropatias/sangue , Nefropatias/enzimologia , Nefropatias/epidemiologia , Hepatopatias/sangue , Hepatopatias/enzimologia , Hepatopatias/epidemiologia , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Razão de Chances , Pancreatite/sangue , Pancreatite/enzimologia , Prognóstico , Modelos de Riscos Proporcionais
18.
Clin Exp Nephrol ; 25(6): 585-597, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33754203

RESUMO

The kidneys are vital organs that play an important role in removing waste materials from the blood, electrolyte balance, blood pressure regulation, and red blood cell genesis. Kidney disease can be caused by various factors, including diabetes, ischemia/reperfusion injury, and nephrotoxic agents. Inflammation and oxidative stress play a key role in the progression and pathogenesis of kidney diseases. Acute kidney injury (AKI) and chronic kidney disease (CKD) are important health problems worldwide, as they are associated with a long-term hospital stay, and increased morbidity and mortality in high-risk patients. Current standard therapeutic options are not sufficient to delay or stop the loss of kidney function. Therefore, it is necessary to develop new therapeutic options. Phosphodiesterase 5 inhibitors (PDE5Is) are a currently available class of drugs that are used to treat erectile dysfunction and pulmonary hypertension in humans. However, recent evidence suggests that PDE5Is have beneficial renoprotective effects via a variety of mechanisms. In this review, the benefits of PDE5 inhibitors in clinical conditions associated with kidney disease, such as diabetic nephropathy, ischemia-reperfusion injury, and acute and chronic kidney injury, are summarized.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Nefropatias/tratamento farmacológico , Rim/efeitos dos fármacos , Inibidores da Fosfodiesterase 5/uso terapêutico , Animais , Humanos , Rim/enzimologia , Rim/patologia , Rim/fisiopatologia , Nefropatias/enzimologia , Nefropatias/patologia , Nefropatias/fisiopatologia , Inibidores da Fosfodiesterase 5/efeitos adversos , Transdução de Sinais , Resultado do Tratamento
19.
Int J Mol Sci ; 22(5)2021 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-33669059

RESUMO

The renin-angiotensin-aldosterone system (RAAS) is implicated in hypertension and kidney disease. The developing kidney can be programmed by various early-life insults by so-called renal programming, resulting in hypertension and kidney disease in adulthood. This theory is known as developmental origins of health and disease (DOHaD). Conversely, early RAAS-based interventions could reverse program processes to prevent a disease from occurring by so-called reprogramming. In the current review, we mainly summarize (1) the current knowledge on the RAAS implicated in renal programming; (2) current evidence supporting the connections between the aberrant RAAS and other mechanisms behind renal programming, such as oxidative stress, nitric oxide deficiency, epigenetic regulation, and gut microbiota dysbiosis; and (3) an overview of how RAAS-based reprogramming interventions may prevent hypertension and kidney disease of developmental origins. To accelerate the transition of RAAS-based interventions for prevention of hypertension and kidney disease, an extended comprehension of the RAAS implicated in renal programming is needed, as well as a greater focus on further clinical translation.


Assuntos
Hipertensão/metabolismo , Nefropatias/metabolismo , Rim/crescimento & desenvolvimento , Néfrons/crescimento & desenvolvimento , Sistema Renina-Angiotensina , Renina/metabolismo , Adulto , Animais , Modelos Animais de Doenças , Disbiose/metabolismo , Epigênese Genética , Humanos , Hipertensão/genética , Rim/metabolismo , Nefropatias/enzimologia , Nefropatias/genética , Néfrons/citologia , Néfrons/metabolismo , Óxido Nítrico/deficiência , Óxido Nítrico/metabolismo , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia , Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/fisiologia
20.
Int J Mol Sci ; 22(4)2021 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-33670516

RESUMO

The incidence of kidney disease is rising, constituting a significant burden on the healthcare system and making identification of new therapeutic targets increasingly urgent. The heme oxygenase (HO) system performs an important function in the regulation of oxidative stress and inflammation and, via these mechanisms, is thought to play a role in the prevention of non-specific injuries following acute renal failure or resulting from chronic kidney disease. The expression of HO-1 is strongly inducible by a wide range of stimuli in the kidney, consequent to the kidney's filtration role which means HO-1 is exposed to a wide range of endogenous and exogenous molecules, and it has been shown to be protective in a variety of nephropathological animal models. Interestingly, the positive effect of HO-1 occurs in both hemolysis- and rhabdomyolysis-dominated diseases, where the kidney is extensively exposed to heme (a major HO-1 inducer), as well as in non-heme-dependent diseases such as hypertension, diabetic nephropathy or progression to end-stage renal disease. This highlights the complexity of HO-1's functions, which is also illustrated by the fact that, despite the abundance of preclinical data, no drug targeting HO-1 has so far been translated into clinical use. The objective of this review is to assess current knowledge relating HO-1's role in the kidney and its potential interest as a nephroprotection agent. The potential therapeutic openings will be presented, in particular through the identification of clinical trials targeting this enzyme or its products.


Assuntos
Injúria Renal Aguda/metabolismo , Heme Oxigenase-1/metabolismo , Heme/metabolismo , Nefropatias/metabolismo , Estresse Oxidativo , Injúria Renal Aguda/enzimologia , Injúria Renal Aguda/patologia , Animais , Humanos , Rim/citologia , Rim/enzimologia , Rim/metabolismo , Nefropatias/enzimologia , Nefropatias/patologia , Túbulos Renais/citologia , Túbulos Renais/enzimologia , Túbulos Renais/metabolismo , Substâncias Protetoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...