Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
2.
J Hematol Oncol ; 16(1): 84, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37501059

RESUMO

Hyperhomocysteinemia (HHcy) is closely associated with thrombotic diseases such as myocardial infarction and stroke. Enhanced platelet activation was observed in animals and humans with HHcy. However, the influence of HHcy on thrombopoiesis remains largely unknown. Here, we reported increased platelet count (PLT) in mice and zebrafish with HHcy. In hypertensive patients (n = 11,189), higher serum level of total Hcy was observed in participants with PLT ≥ 291 × 109/L (full adjusted ß, 0.59; 95% CI 0.14, 1.04). We used single-cell RNA sequencing (scRNA-seq) to characterize the impact of Hcy on transcriptome, cellular heterogeneity, and developmental trajectories of megakaryopoiesis from human umbilical cord blood (hUCB) CD34+ cells. Together with in vitro and in vivo analysis, we demonstrated that Hcy promoted megakaryocytes (MKs) differentiation via growth hormone (GH)-PI3K-Akt axis. Moreover, the effect of Hcy on thrombopoiesis is independent of thrombopoietin (TPO) because administration of Hcy also led to a significant increase of PLT in homozygous TPO receptor (Mpl) mutant mice and zebrafish. Administration of melatonin effectively reversed Hcy-induced thrombopoiesis in mice. ScRNA-seq showed that melatonin abolished Hcy-facilitated MK differentiation and maturation, inhibited the activation of GH-PI3K-Akt signaling. Our work reveals a previously unrecognized role of HHcy in thrombopoiesis and provides new insight into the mechanisms by which HHcy confers an increased thrombotic risk.Trial Registration clinicaltrials.gov Identifier: NCT00794885.


Assuntos
Hiper-Homocisteinemia , Melatonina , Humanos , Camundongos , Animais , Trombopoese/genética , Megacariócitos , Plaquetas , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt/genética , Peixe-Zebra , Hormônio do Crescimento/farmacologia , Melatonina/farmacologia , Hiper-Homocisteinemia/complicações , Diferenciação Celular
3.
Cell Biol Toxicol ; 39(6): 3077-3100, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37495868

RESUMO

Hyperhomocysteinemia (HHcy) plays a salient role in male infertility. However, whether HHcy interferes with testosterone production remains inconclusive. Here, we reported a lower serum testosterone level in HHcy mice. Single-cell RNA sequencing revealed that genes related to testosterone biosynthesis, together with nuclear receptor subfamily 5 group A member 1 (Nr5a1), a key transcription factor for steroidogenic genes, were downregulated in the Leydig cells (LCs) of HHcy mice. Mechanistically, Hcy lowered trimethylation of histone H3 on lysine 4 (H3K4me3), which was bound on the promoter region of Nr5a1, resulting in downregulation of Nr5a1. Intriguingly, we identified an unknown cell cluster annotated as Macrophage-like Leydig cells (McLCs), expressing both LCs and macrophages markers. In HHcy mice, McLCs were shifted toward pro-inflammatory phenotype and thus promoted inflammatory response in LC. Betaine supplementation rescued the downregulation of NR5A1 and restored the serum testosterone level in HHcy mice. Overall, our study highlights an etiological role of HHcy in LCs dysfunction.


Assuntos
Hiper-Homocisteinemia , Células Intersticiais do Testículo , Camundongos , Masculino , Animais , Células Intersticiais do Testículo/metabolismo , Testosterona , Hiper-Homocisteinemia/metabolismo , Macrófagos/metabolismo , Fatores de Transcrição/genética
4.
Front Physiol ; 13: 996166, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36407000

RESUMO

Chronic kidney disease (CKD) is a global public health problem that shortens lifespan primarily by increasing the risk of cardiovascular diseases. Trimethylamine-N-oxide (TMAO), a gut microbiota-derived toxin produced by metabolizing high-choline or carnitine foods, is associated with cardiovascular events in patients with CKD. Although the deleterious effect of TMAO on CKD-induced cardiac injury has been confirmed by various researches, the mechanisms remain unclear. Here, we tested the hypothesis that TMAO aggravates CKD-induced cardiac injury and explores the potential mechanism. CD1 mice underwent 5/6 nephrectomy to induce CKD, and then fed with a diet supplemented with choline (1.2% total) for 8 weeks. Serum TMAO levels were elevated in CKD mice compared with SHAM group, and higher TMAO levels were found in choline-supplemented CKD mice compared with CKD group. Dietary choline aggravated CKD-induced cardiac dysfunction, and reducing TMAO levels via medicinal charcoal tablets improved cardiac dysfunction. RNA-seq analysis revealed that dietary choline affected cardiac angiogenesis in CKD mice. Reduced cardiac capillary density and expressions of angiogenesis-related genes were observed in choline-treated CKD mice. Furthermore, dietary choline inhibited cardiac Hif-1α protein level in CKD mice, and Hif-1α stabilizer FG-4592 could improve cardiac angiogenesis and dysfunction in CKD mice on a high-choline diet. In conclusion, these data indicate that dietary choline, via gut microbe-generated TMAO, inhibits cardiac angiogenesis by reducing Hif-1α protein level, ultimately aggravates cardiac dysfunction in CKD mice.

5.
EBioMedicine ; 86: 104312, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36335669

RESUMO

BACKGROUND: The role of the IL6 family members in organ fibrosis, including renal interstitial fibrosis (TIF), has been widely explored. However, few studies have ever simultaneously examined them in the same cohort of patients. Besides, the role of leukemia inhibitory factor (LIF) in TIF remains unclear. METHODS: RNA-seq data of kidney biopsies from chronic kidney disease (CKD) patients, in both public databases and our assays, were used to analyze transcript levels of IL6 family members. Two TIF mouse models, the unilateral ureteral obstruction (UUO) and the ischemia reperfusion injury (IRI), were employed to validate the finding. To assess the role of LIF in vivo, short hairpin RNA, lenti-GFP-LIF was used to knockdown LIF receptor (LIFR), overexpress LIF, respectively. LIF-neutralizing antibody was used in therapeutic studies. Whether urinary LIF could be used as a promising predictor for CKD progression was investigated in a prospective observation patient cohort. FINDINGS: Among IL6 family members, LIF is the most upregulated one in both human and mouse renal fibrotic lesions. The mRNA level of LIF negatively correlated with eGFR with the strongest correlation and the smallest P value. Baseline urinary concentrations of LIF in CKD patients predict the risk of CKD progression to end-stage kidney disease by Kaplan-Meier analysis. In mouse TIF models, knockdown of LIFR alleviated TIF; conversely, overexpressing LIF exacerbated TIF. Most encouragingly, visible efficacy against TIF was observed by administering LIF-neutralizing antibodies to mice. Mechanistically, LIF-LIFR-EGR1 axis and Sonic Hedgehog signaling formed a vicious cycle between fibroblasts and proximal tubular cells to augment LIF expression and promote the pro-fibrotic response via ERK and STAT3 activation. INTERPRETATION: This study discovered that LIF is a noninvasive biomarker for the progression of CKD and a potential therapeutic target of TIF. FUNDINGS: Stated in the Acknowledgements section of the manuscript.


Assuntos
Rim , Insuficiência Renal Crônica , Humanos , Camundongos , Animais , Fator Inibidor de Leucemia/genética , Rim/metabolismo , Interleucina-6/genética , Estudos Prospectivos , Proteínas Hedgehog , Fibrose , Insuficiência Renal Crônica/patologia
6.
Transl Res ; 246: 49-65, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35276386

RESUMO

ZAK (sterile alpha motif and leucine zipper-containing kinase) is a newly discovered member of the subfamily of mitogen-activated protein kinase kinase kinases (MAP3Ks). The role of ZAK in kidney disease remains largely unknown. In this study, we systematically investigated the expression and function of ZAK in the progression of tubulointerstitial fibrosis (TIF). ZAK was induced, predominantly in tubular epithelium, in both fibrotic kidneys of human and mouse models with TIF. ZAK expression level was correlated with the extent of renal fibrosis and the decline of eGFR of CKD patients. Depleting ZAK attenuated TIF and inflammation induced by unilateral ureteral occlusion (UUO) together with decreased activation of p38 MAPK and Smads signaling. Moreover, we demonstrated that overexpressed ZAK was in complex with Smad2/3 and TGF-ß receptor Ⅰ (TßRI). Whereas, silencing endogenous ZAK ameliorated the amount of Smad2/3 recruited to TßRI. Moreover, we discovered a novel small molecule inhibitor of ZAK, named 6p. In vitro, incubation with 6p inhibited TGF-ß1-induced fibrogenic response in NRK52E cells. In vivo, intragastric administration of 6p ameliorated TIF and inflammation in UUO and unilateral ischemia-reperfusion injury model. Delayed administration of 6p was also effective in retarding the progression of the established TIF. In conclusion, ZAK is a novel therapeutic target for TIF, and 6p might be a potential therapeutic agent for TIF.


Assuntos
Nefropatias , MAP Quinase Quinase Quinases , Obstrução Ureteral , Animais , Fibrose , Humanos , Inflamação/patologia , Rim/patologia , MAP Quinase Quinase Quinases/genética , Camundongos , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo
7.
Cell Death Differ ; 29(7): 1409-1422, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35017646

RESUMO

Given the critical role of SCF E3 ligases in autophagy by modulating the protein stability of various autophagic components, the activity of SCF should be tightly controlled to maintain the autophagic flux. We here showed that Numb, a multifunctional adaptor protein, increased the protein abundance of DEPTOR, which is an inhibitor of mTORC1, leading to increased autophagy flux. In vitro ubiquitination assay demonstrated that Numb inhibited SCFß-TrCP2 mediated ubiquitination of DEPTOR. Mechanistically, Numb interrupted the interaction between ß-TrCP2 and SKP1 by directly binding with SKP1. In the presence of wild type ß-TrCP2, Numb overexpression inhibited DEPTOR degradation. Whereas, in the presence of the mutant ß-TrCP2 which lacks the F-box domain, Numb overexpression did not affect the protein abundance of DEPTOR. In mouse model of renal fibrosis induced by unilateral ureteral obstruction, the expression of Numb was significantly increased. Consistently, the upregulation of Numb was observed in renal fibrotic lesions of chronic kidney disease patients. Specifically depleting Numb in proximal renal tubules decreased the protein abundance of DEPTOR, attenuated autophagy in fibrotic lesions and protected the kidney from development of renal fibrosis in vivo. Taken together, both in vitro and in vivo data indicated that Numb functions as a novel regulator to fine tuning the activity of SCFß-TrCP2 in modulating autophagy.


Assuntos
Autofagia , Proteínas de Membrana , Proteínas do Tecido Nervoso , Ubiquitina-Proteína Ligases , Animais , Fibrose , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
8.
Transl Res ; 240: 50-63, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34673277

RESUMO

Trimethylamine-N-oxide (TMAO), a gut microbiota-produced metabolite, is accumulated in chronic kidney disease (CKD) patients. It is well known to contribute to CKD-related cardiovascular complications. However, the effect of TMAO on peritoneal dialysis (PD)-related peritonitis remains largely unknown. Here, we demonstrate that serum concentrations of TMAO were positively correlated with C-reactive protein levels, and the appearance rate of dialysate IL-6 and PAI-1, in PD patients. During the follow-up period of 28.3 ± 8.0 months, patients with higher TMAO levels (≥50 µM) had a higher risk of new-onset peritonitis (HR, 3.60; 95%CI, 1.18-10.99; P=0.025) after adjusting for sex, age, diabetes, PD duration, BUN, rGFR, C-reactive protein, BMI and ß2-M. In CKD rat models, TMAO significantly promoted peritoneal dialysate-induced inflammatory cell infiltration, inflammatory cytokines production in the peritoneum. In vitro study revealed that TMAO directly induced primary peritoneal mesothelial cell necrosis, together with increased production of pro-inflammatory cytokines including CCL2, TNF-α, IL-6, and IL-1ß. In addition, TMAO significantly increased TNF-α-induced P-selectin production in mesothelial cells, as well as high glucose-induced TNF-α and CCL2 expression in endothelial cells. In conclusion, our data demonstrate that higher levels of TMAO exacerbate peritoneal inflammation and might be a risk factor of incidence of peritonitis in PD patients.


Assuntos
Microbioma Gastrointestinal , Inflamação/patologia , Metaboloma , Metilaminas/efeitos adversos , Diálise Peritoneal/efeitos adversos , Peritônio/patologia , Peritonite/epidemiologia , Peritonite/etiologia , Adulto , Animais , Morte Celular , Citocinas/metabolismo , Epitélio/patologia , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Glucose/toxicidade , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Metaboloma/efeitos dos fármacos , Pessoa de Meia-Idade , Selectina-P/metabolismo , Ratos Sprague-Dawley , Insuficiência Renal Crônica/complicações , Fatores de Risco , Regulação para Cima
9.
Kidney Blood Press Res ; 47(1): 61-71, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34788763

RESUMO

BACKGROUND: Trimethylamine-N-oxide (TMAO) is an intestinal metabolic toxin, which is produced by gut flora via metabolizing high-choline foods. TMAO is known to increase the risk of atherosclerosis and cardiovascular events in chronic kidney disease (CKD) patients. OBJECTIVES: The objective of this study was to explore the role and mechanism of TMAO aggravating kidney injury. METHOD: We used the five-sixths nephrectomy (5/6 Nx)-induced CKD rats to investigate whether TMAO could aggravate kidney damage and its possible mechanisms. Six weeks after the operation, the two groups of 5/6 Nx rats were subjected to intraperitoneal injection with 2.5% glucose peritoneal dialysis fluid (2.5% PDF) and 2.5% PDF plus TMAO 20 mg/kg/day. RESULTS: In this study, we provided evidence showing TMAO significantly aggravated renal failure as well as inflammatory cell infiltration and in five-sixths nephrectomy-induced CKD rats. We found that TMAO could upregulate inflammatory factors including MCP-1, TNF-α, IL-6, IL-1ß, and IL-18 by activating p38 phosphorylation and upregulation of human antigen R. TMAO could aggravate oxidative stress by upregulating NOX4 and downregulating SOD. The result also confirmed that TMAO promoted NLRP3 inflammasome formation as well as cleaved caspase-1 and IL-1ß activation in the kidney tissue. CONCLUSIONS: Taken together, the present study validates TMAO as a pro-inflammatory factor that causes renal inflammatory injury and renal function impairment. Inhibition of TMAO synthesis or promoting its clearance may be a potential therapeutic approach of CKD in the future.


Assuntos
Proteína Semelhante a ELAV 1/metabolismo , Metilaminas/metabolismo , Insuficiência Renal Crônica/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Inflamação/metabolismo , Inflamação/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Insuficiência Renal Crônica/patologia , Regulação para Cima
10.
Kidney Dis (Basel) ; 7(3): 186-199, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34179114

RESUMO

BACKGROUND: Progressive tubulointerstitial fibrosis (TIF) is the final common pathway leading to ESRD. There is an urgent need to develop effective approaches slowing the progression of TIF. Previous studies showed that systemic supplementation of nicotinamide (NAM) increases renal NAD+ and reverses ischemic-reperfusion induced acute renal injury. However, the role and mechanism of NAM in TIF has been unclear. METHODS: In vivo, we injected NAM (0.25 mg/g) 3 days before unilateral ureter obstruction (UUO) till day 7 post-operation. In vitro, mouse primary proximal tubular epithelial cells (PTCs), rat renal NRK-49F cells, and human renal proximal tubular epithelial cell (HK-2) were pretreated with the indicated concentration of NAM 1 h before incubation with transform growth factor-ß1 (TGF-ß1) or aristolochic acid (AA) for 24 or 48 h. To evaluate the role of sirtuins (SIRTs), PTCs were pretreated with EX527 or resveratrol 30 min before incubation with NAM and TGF-ß1. RESULTS: In the present study, we demonstrated that NAM supplementation prevented UUO-induced TIF, and AA-induced renal injury. NAM also decreased the expression of pro-fibrotic proteins and pro-inflammatory cytokines (IL-6 and TNF-α) and attenuated interstitial inflammation. In vitro experiment showed that, NAM inhibited AA-induced G2/M arrest of HK-2 cells by downregulating the expression of cyclin G1, a target gene of p53. In addition, NAM inhibited TGF-ß1-induced fibroblast proliferation and activation shown as downregulated expression of collagen I, fibronectin, PCNA, cyclin D1, IL-6, and TNF-α. NAM decreased the acetylation of Smad3 and p53. EX527, an inhibitor of SIRT1, reversed the effect of NAM on TGF-ß1-induced matrix protein production. However, resveratrol, a SIRT1 activator, did not further boost the protective effect of NAM on reducing matrix protein production. CONCLUSIONS: Taken together, these data indicate that NAM supplementation could inhibit TIF at least partially by boosting the activity of sirtuins.

11.
Biochim Biophys Acta Mol Basis Dis ; 1867(5): 166081, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33486098

RESUMO

Tubulointerstitial fibrosis is the ultimate common pathway of all manners of chronic kidney disease. We previously demonstrated that specific deletion of Numb in proximal tubular cells (PTCs) prevented G2/M arrest and attenuated renal fibrosis. However, how Numb modulates cell cycle arrest remains unclear. Here, we showed that Numb overexpression significantly increased the protein level of hypoxia-inducible factor-1α (HIF-1α). Numb overexpression-induced G2/M arrest was blocked by silencing endogenous HIF-1α, subsequently downregulated the expression of cyclin G1 which is an atypical cyclin to promote G2/M arrest of PTCs. Further analysis revealed that Numb-augmented HIF-1α protein was blocked by simultaneously overexpressing MDM2. Moreover, silencing Numb decreased TGF-ß1-induceded HIF-1α protein expression. While endogenous MDM2 was knocked down this reduction was reversed, indicating that Numb stabilized HIF-1α protein via interfering MDM2-mediated HIF-1α protein degradation. Interestingly, HIF-1α overexpression significantly upregulated the expression of Numb and silencing endogenous HIF-1α blocked CoCl2 or TGF-ß1-induced Numb expression. Chromatin immunoprecipitation (ChIP) assays demonstrated that HIF-1α binded to the promoter region of Numb. This binding was significantly increased by TGF-ß1. Collectively, these data indicate that Numb and HIF-1α cooperates to promote G2/M arrest of PTCs, and thus aggravates tubulointerstitial fibrosis. Numb might be a potential target for the therapy of tubulointerstitial fibrosis.


Assuntos
Fibrose/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/química , Nefropatias/patologia , Túbulos Renais/patologia , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Animais , Fibrose/etiologia , Fibrose/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Nefropatias/etiologia , Nefropatias/metabolismo , Túbulos Renais/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-mdm2/genética
12.
Clin Nutr ; 39(8): 2517-2524, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31806397

RESUMO

BACKGROUND & AIMS: Betaine (a micronutrient) has important biological functions (e.g., preventing premature apoptosis and serving as a methyl donor). We investigated the association between baseline serum betaine and the incident risk of first stroke in hypertensive patients. METHODS: We conducted a nested case-control study, including 622 patients with first stroke (including 502 ischemic stroke, 118 hemorrhagic stroke and 2 uncertain type of stroke) and 622 matched controls from the China Stroke Primary Prevention Trial (CSPPT). The study was conducted from May 2008 to August 2013. The study outcomes included first stroke and its subtypes: first ischemic and hemorrhagic stroke. RESULTS: There was a U-shaped association between baseline serum betaine and the risk of first ischemic stroke. The risk of first ischemic stroke decreased with the increment of betaine (per 10 µmol/L increase: OR, 0.87; 95%CI: 0.77-0.99) in patients with betaine <77.7 µmol/L, while the risk of first ischemic stroke increased with the betaine increment (OR, 1.17; 95%CI: 1.01-1.36) in patients with betaine ≥77.7 µmol/L. However, there was no significant association between serum betaine and risk of first hemorrhagic stroke (per 10 µmol/L increase: OR, 0.98; 95%CI: 0.82-1.17). CONCLUSIONS: There was a U-shaped association between baseline betaine levels and the risk of first ischemic stroke in hypertensive patients, with a turning point at about 77.7 µmol/L. CLINICAL TRIAL REGISTRATION-URL: http://www.clinicaltrials.gov. Unique identifier: NCT00794885.


Assuntos
Betaína/sangue , Acidente Vascular Cerebral Hemorrágico/etiologia , Hipertensão/sangue , AVC Isquêmico/etiologia , Medição de Risco/estatística & dados numéricos , Idoso , Estudos de Casos e Controles , China , Método Duplo-Cego , Feminino , Fatores de Risco de Doenças Cardíacas , Acidente Vascular Cerebral Hemorrágico/epidemiologia , Humanos , Hipertensão/complicações , Incidência , AVC Isquêmico/epidemiologia , Masculino , Pessoa de Meia-Idade , Ensaios Clínicos Controlados Aleatórios como Assunto
14.
Antioxid Redox Signal ; 30(13): 1635-1650, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30084650

RESUMO

AIMS: The risk factors promoting acute kidney injury (AKI) to chronic kidney disease (CKD) progression remain largely unknown. The aim of the present study was to investigate whether hyperhomocysteinemia (Hhcy) accelerates the development of renal fibrosis after AKI. RESULTS: Hhcy aggravated ischemia-reperfusion-induced AKI and the subsequent development of renal fibrotic lesions characterized by excessive extracellular matrix deposition. Mechanistically, the RNA binding protein human antigen R (HuR) bound to the 3'-untranslated region (3'-UTR) of heme oxygenase-1 (HO-1) messenger RNA (mRNA). Homocysteine (Hcy) downregulated HuR expression, reduced the binding of HuR to the 3'-UTR of HO-1, and thereafter decreased HO-1 expression. Administration of the HO-1 inducer cobalt protoporphyrin-IX significantly hindered Hhcy-augmented reactive oxygen species production and renal fibrotic lesions. Innovation and Conclusion: These data indicate that Hhcy might be a novel risk factor that promotes AKI to CKD progression. Lowering Hcy level or HO-1 induction might be a potential therapeutic strategy to improve the outcome of AKI.


Assuntos
Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Heme Oxigenase-1/genética , Hiper-Homocisteinemia/metabolismo , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Injúria Renal Aguda/etiologia , Biópsia , Progressão da Doença , Suscetibilidade a Doenças , Proteína Semelhante a ELAV 1/metabolismo , Expressão Gênica , Heme Oxigenase-1/metabolismo , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/complicações , Túbulos Renais/metabolismo , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Insuficiência Renal Crônica/etiologia
15.
Antioxid Redox Signal ; 30(15): 1797-1816, 2019 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-29890853

RESUMO

AIMS: Mitochondrial fragmentation is a crucial mechanism contributing to tubular cell apoptosis during acute kidney injury (AKI). However, the mechanism of modulating mitochondrial dynamics during AKI remains unclear. Numb is a multifunction adaptor protein that is expressed in renal tubules. The aim of the present study was to evaluate the role of Numb in mitochondrial dysfunction during AKI. RESULTS: The expression of Numb was upregulated in both ischemia-reperfusion- and cisplatin-induced AKI. Depletion of Numb from proximal tubules (PT-Nb-KO) exacerbated AKI shown as more severe renal tubular damage and higher serum creatinine than wild-type mice. Numb depletion alone significantly increased mitochondrial fragmentation without altering mitochondrial mass and function, including adenosine triphosphate production, mitochondrial membrane potential, oxygen consumption, and reactive oxygen species production. However, mitochondrial fragmentation and dysfunction were significantly aggravated after cisplatin exposure in PT-Nb-KO mice. Mechanistically, Numb depletion triggered dynamin-related protein 1 (Drp1) recruitment to mitochondria by increasing the phosphorylation of Drp1 at serine 656 residue (human Drp1 ser637). Inhibiting the activity of Rho-associated coiled-coil containing protein kinase (ROCK) by Y-27632 attenuated phosphorylation of Drp1 ser656 and mitochondrial fragmentation in Numb-deficient cells. Administration of mdivi-1, a pharmacological inhibitor of Drp1, restored mitochondrial morphology, attenuated cisplatin-induced tubular injury, and renal dysfunction in PT-Nb-KO mice. Innovation and Conclusion: Our data suggest that Numb depletion promotes mitochondrial fragmentation by promoting the phosphorylation of Drp1 Ser637 and thus exacerbates cisplatin-induced mitochondrial dysfunction and tubular cell apoptosis. These findings add a novel insight into modulating mechanism of mitochondrial dynamics during AKI.


Assuntos
Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Proteínas Quinases Associadas com Morte Celular/genética , Suscetibilidade a Doenças , Proteínas de Membrana/metabolismo , Dinâmica Mitocondrial/genética , Proteínas do Tecido Nervoso/metabolismo , Injúria Renal Aguda/patologia , Animais , Apoptose , Biomarcadores , Biópsia , Cisplatino/efeitos adversos , Proteínas Quinases Associadas com Morte Celular/metabolismo , Progressão da Doença , Técnicas de Silenciamento de Genes , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Proteínas de Membrana/genética , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/genética , Transporte Proteico , Radiação Ionizante , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
16.
Stroke ; 49(9): 2021-2028, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30354996

RESUMO

Background and Purpose- Trimethylamine N-oxide (TMAO)-a gut derived metabolite-has been shown to be atherogenic. It remains unknown whether TMAO is associated with the risk of first stroke. We aimed to determine the association between serum TMAO levels and first stroke in hypertensive patients without major cardiovascular diseases and examine any possible effect modifiers. Methods- We used a nested case-control design, using data from the CSPPT (China Stroke Primary Prevention Trial), including 622 patients with first stroke and 622 matched controls. The study was conducted from May 2008 to August 2013. The primary outcome was a first stroke. Results- After adjusting for choline, L-carnitine, and other important covariates, including baseline systolic blood pressure and time-averaged systolic blood pressure, during the treatment period, the risk of first stroke increased with each increment of TMAO level (per natural log [TMAO] increment: odds ratio, 1.22; 95% CI, 1.02-1.46). Consistently, compared with participants in the lowest tertile (<1.79 µmol/L) of serum TMAO levels, a significantly higher risk of first stroke was found in those in higher TMAO tertiles (≥1.79 µmol/L; odds ratio, 1.34; 95% CI, 1.00-1.81) or in TMAO tertile 3 (≥3.19 µmol/L; odds ratio, 1.43; 95% CI, 1.02-2.01). In the exploratory analysis, we observed an interaction between TMAO and folate levels (≥7.7 [median] versus <7.7 ng/mL) on first stroke ( P for interaction, 0.030). Conclusions- Higher TMAO levels were associated with increased risk of first stroke in hypertensive patients. Our finding, if further confirmed, calls for a carefully designed clinical trial to further evaluate the role of higher TMAO levels on outcomes in hypertensive patients. Clinical Trial Registration- URL: https://www.clinicaltrials.gov . Unique identifier: NCT00794885.


Assuntos
Hipertensão/sangue , Metilaminas/sangue , Acidente Vascular Cerebral/sangue , Idoso , Carnitina/sangue , Estudos de Casos e Controles , China/epidemiologia , Colina/sangue , Feminino , Ácido Fólico/sangue , Microbioma Gastrointestinal , Humanos , Hipertensão/epidemiologia , Masculino , Pessoa de Meia-Idade , Razão de Chances , Acidente Vascular Cerebral/epidemiologia
17.
Int J Biol Sci ; 13(2): 219-231, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28255274

RESUMO

Hyperhomocysteinemia (HHcy) has been linked to several clinical manifestations including chronic kidney disease. However, it is not known whether HHcy has a role in the development of acute kidney injury (AKI). In the present study, we reported that HHcy mice developed more severe renal injury after cisplatin injection and ischemia-reperfusion injury shown as more severe renal tubular damage and higher serum creatinine. In response to cisplatin, HHcy mice showed more prevalent tubular cell apoptosis and decreased tubular cell proliferation. Mechanistically, a heightened ER stress and a reduced Akt activity were observed in kidney tissues of HHcy mice after cisplatin injection. Stimulating cultured NRK-52E cells with Hcy significantly increased the fraction of cells in G2/M phase and cell apoptosis together with decreased Akt kinase activity. Akt agonist IGF-1 rescued HHcy-induced cell cycle arrest and cell apoptosis. In conclusion, the present study provides evidence that HHcy increases the sensitivity and severity of AKI.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/etiologia , Cisplatino/efeitos adversos , Hiper-Homocisteinemia/complicações , Injúria Renal Aguda/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Homocisteína/sangue , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos
18.
Kidney Int ; 92(3): 612-624, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28318631

RESUMO

Renal fibrosis is the final common pathway of all varieties of progressive chronic kidney disease. However, there are no effective therapies to prevent or slow the progression of renal fibrosis. Niclosamide is a US Food and Drug Administration-approved oral antihelminthic drug used for treating most tapeworm infections. Here, we demonstrated that phosphate niclosamide, the water-soluble form of niclosamide, significantly reduced proteinuria, glomerulosclrotic lesions, and interstitial fibrosis in a murine model of adriamycin nephropathy. In addition, phosphate niclosamide significantly ameliorated established renal interstitial fibrosis a murine model of unilateral ureteral obstruction. Mechanistically, phosphate niclosamide directly inhibited TGF-ß-induced expression of homeodomain-interacting protein kinase 2 (HIPK2) by interfering with the binding of Smad3 to the promoter of the HIPK2 gene, and subsequently mitigated the activation of its downstream signaling pathways including Smad, Notch, NF-κB and Wnt/ß-catenin pathway both in vitro and in vivo. Thus, phosphate niclosamide mitigates renal fibrosis at least partially by inhibiting HIPK2 expression. Hence, phosphate niclosamide might be a potential therapeutic agent for renal fibrosis.


Assuntos
Anti-Helmínticos/farmacologia , Proteínas de Transporte/metabolismo , Rim/patologia , Niclosamida/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Insuficiência Renal Crônica/tratamento farmacológico , Animais , Anti-Helmínticos/uso terapêutico , Células Cultivadas , Modelos Animais de Doenças , Doxorrubicina/toxicidade , Fibrose , Humanos , Rim/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Niclosamida/uso terapêutico , Proteinúria/tratamento farmacológico , Proteinúria/urina , Ratos , Receptores Notch/metabolismo , Insuficiência Renal Crônica/induzido quimicamente , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/urina , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos
19.
PLoS One ; 11(9): e0162873, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27636716

RESUMO

Chronic kidney disease (CKD) is becoming a worldwide problem. However, current treatment options are limited. In the current study we showed that QiShenYiQi (QSYQ), a water-ethanol extract from several Chinese medicines, is a potent inhibitor of renal interstitial fibrosis. QSYQ inhibited transforming growth factor-ß1 (TGF-ß1)-responsive α-smooth muscle actin (α-SMA), collagen I, and fibronectin up-regulation in obstructive nephropathy and cultured cells. Administration of QSYQ also inhibited the established renal interstitial fibrosis in obstructive nephropathy. Interestingly, QSYQ selectively inhibited TGF-ß1-induced ß-catenin up-regulation and downstream gene transcription. Taken together, our study suggests that QSYQ selectively inhibits TGF-ß1-induced ß-catenin up-regulation and might have significant therapeutic potential for the treatment of renal fibrosis.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , beta Catenina/metabolismo , Actinas/antagonistas & inibidores , Animais , Fibrose/tratamento farmacológico , Masculino , Ratos , Ratos Sprague-Dawley , Transcrição Gênica , Fator de Crescimento Transformador beta1/fisiologia
20.
Int J Mol Med ; 38(2): 513-20, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27314843

RESUMO

Hepatitis B virus (HBV)-associated glomerulo-nephritis is the most common extra-hepatic disorder occurring with hepatitis B virus infection. In the present study, we hypothesized that HBV X protein (HBx) may play a critical role in renal interstitial fibrosis, as HBx has been shown to induce epithelial-mesenchymal transition (EMT) in renal cells. For this purpose, we successfully transfected HBx plasmid into human renal proximal tubule epithelial cells (HK-2 cells). We found that transfection with HBx plasmid significantly downregulated E-cadherin expression and upregulated α-smooth muscle actin, collagen I and fibronectin expression in a time- and concentration-dependent manner (at the lower concentrations and earlier time points). HBx also increased nuclear factor-κB (NF-κB) phosphorylation in a time- and concentration-dependent manner (again at the lower concentrations and earlier time points); however, it did not alter the phosphorylation of Smad2, Smad3, p38, phosphoinositide 3-kinase (PI3K) or extracellular signal-regulated kinase (ERK). Thus, the findings of this study demonstrate that HBx promotes EMT in renal HK-2 cells, and the potential underlying mechanisms may involve the activation of the NF-κB signaling pathway.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal , Túbulos Renais Proximais/patologia , NF-kappa B/metabolismo , Transativadores/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Imunofluorescência , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Smad/metabolismo , Transfecção , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Virais Reguladoras e Acessórias , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...