Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Publication year range
1.
Environ Toxicol ; 39(6): 3481-3499, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38456329

ABSTRACT

CONTEXT: Qi-dan-dihuang decoction (QDD) has been used to treat diabetic kidney disease (DKD), but the underlying mechanisms are poorly understood. OBJECTIVE: This study reveals the mechanism by which QDD ameliorates DKD. MATERIALS AND METHODS: The compounds in QDD were identified by high-performance liquid chromatography and quadrupole-time-of-flight tandem mass spectrometry (HPLC-Q-TOF-MS). Key targets and signaling pathways were screened through bioinformatics. Nondiabetic Lepr db/m mice were used as control group, while Lepr db/db mice were divided into model group, dapagliflozin group, 1% QDD-low (QDD-L), and 2% QDD-high (QDD-H) group. After 12 weeks of administration, 24 h urinary protein, serum creatinine, and blood urea nitrogen levels were detected. Kidney tissues damage and fibrosis were evaluated by pathological staining. In addition, 30 mmol/L glucose-treated HK-2 and NRK-52E cells to induce DKD model. Cell activity and migration capacity as well as protein expression levels were evaluated. RESULTS: A total of 46 key target genes were identified. Functional enrichment analyses showed that key target genes were significantly enriched in the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) and mitogen-activated protein kinase (MAPK) signaling pathways. In addition, in vivo and in vitro experiments confirmed that QDD ameliorated renal fibrosis in diabetic mice by resolving inflammation and inhibiting the epithelial-mesenchymal transition (EMT) via the p38MAPK and AKT-mammalian target of rapamycin (mTOR) pathways. DISCUSSION AND CONCLUSION: QDD inhibits EMT and the inflammatory response through the p38MAPK and AKT/mTOR signaling pathways, thereby playing a protective role in renal fibrosis in DKD.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Drugs, Chinese Herbal , Signal Transduction , Animals , Humans , Male , Mice , Rats , Cell Line , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Drugs, Chinese Herbal/pharmacology , Fibrosis , Kidney/drug effects , Kidney/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
2.
Transl Androl Urol ; 9(4): 1712-1724, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32944532

ABSTRACT

BACKGROUND: To study the protective effect of berberine (BBR) on cisplatin-induced acute kidney injury (AKI) and its effect on mitophagy. METHODS: (I) Male C57BL/6 mice aged 6-8 weeks were randomly divided into control group (saline), cisplatin group (cisplatin), and cisplatin + BBR (5, 10 mg/kg) groups. In the cisplatin group and BBR groups, mice were injected intraperitoneally with 15 mg/kg of cisplatin. Mice in BBR groups were given BBR at 72, 48, 24, 0.5 h before and 24, 48 h after cisplatin injection. Mice were sacrificed 72 h after cisplatin injection, and blood were collected for detecting serum creatinine (SCr) and blood urea nitrogen (BUN) levels. Kidneys were collected for detecting protein expression levels of Kidney injury molecule 1 (KIM-1), LC3 II/LC3 I, p62, PINK 1, Parkin in the renal tissue by Western blotting. The pathological changes in renal tissues were observed using periodic acid-Schiff (PAS) staining. (II) Renal tubular epithelial cells (RTECs) were pretreated with different concentrations (1, 2, and 4 µM) of BBR, and then incubated with cisplatin. Changes in autophagy proteins LC3 II/LC3 I, p62, PINK 1, and Parkin were detected by Western blotting, and changes in cellular reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were detected by flow cytometry. RESULTS: (I) Mice treated with BBR at dosage of 5 and 10 mg/kg for 6 days showed significant reduction in SCr and BUN compared to that in mice treated with cisplatin. PAS staining and immunohistochemistry showed that BBR ameliorated cisplatin-induced nephrotoxicity and reduced cisplatin-induced increase in protein expression levels of KIM-1. Compared to cisplatin-treated mice, the mice treated with BBR showed increased LC3 II/LC3 I, PINK 1, and Parkin, and decreased p62 protein expression. (II) Compared to cisplatin-incubated RTECs, cells pretreated with BBR for 24 h exhibited increased protein expressions of LC3 II/LC3 I, PINK1, and Parkin and decreased protein expression of p62. BBR reversed cellular ROS and cell MMP level induced by cisplatin. CONCLUSIONS: BBR plays a protective role in cisplatin-induced AKI by up-regulating mitophagy in RTECs.

3.
Article in English | MEDLINE | ID: mdl-30425748

ABSTRACT

Qi-deficiency (QX) is thought to promote the body's susceptibility to disease, but the underlying mechanism through which this occurs is not clear. We surveyed the traditional Chinese medicine constitution (TCMC) of healthy college students to identify those that were PH (balanced TCMC constitution) and QX (unbalanced TCMC constitution). We then used high-throughput sequencing of the 16SrRNA V3-4 region in fecal microbiota samples to identify differences between those obtained from PH and QX individuals. Our results demonstrated that the alpha diversity of QX samples was significantly lower than that of PH samples (p < 0.05) and that beta diversity was remarkably different in QX and PH samples. Four and 122 bacterial taxa were significantly overrepresented in QX and PH groups, respectively. The genera Sphingobium, Clostridium, and Comamonas were enriched in the QX group and had a certain pathogenic role. The QX group also showed a statistically significant lack of probiotics and anti-inflammatory bacteria such as Bifidobacterium and Bdellovibrio. The functional potential of QX bacterial taxa was reduced in fatty acid metabolism and butanoate metabolism. We contend that the imbalanced intestinal microbiota in QX and the following functional changes in metabolism influence immunity and energy metabolism, which could increase susceptibility to disease.

4.
Nan Fang Yi Ke Da Xue Xue Bao ; 37(2): 184-191, 2016 Feb 20.
Article in Chinese | MEDLINE | ID: mdl-28219861

ABSTRACT

OBJECTIVE: To determine evaluate the effect of health-promoting lifestyle on the outcomes of suboptimal health status (SHS). METHODS: A prospective population cohort was conducted by consecutively enrolling 5676 college students who took routine health examination from March to May 2013. The participants were assessed for baseline health status and lifestyle and 2972 participants with SHS were followed up for 1.5 years. Exposure was defined as an unhealthy lifestyle. The health-promoting lifestyle was assessed via the Health-promoting Lifestyle Profile (HPLP-II). SHS was evaluated using the medical examination report and Sub-health Measurement Scale V1.0 (SHMS V1.0). RESULTS: Among the 2972 students with SHS, 422 showed recovery of the healthy status at 1.5 year follow-up, 579 showed progression into disease conditions, and 1971 remained in SHS. The participants with recovered health status presented with significant increase of SHMS V1.0 scores by 8.75∓6.95 points compared to the baseline assessment (t=-2.14, P=0.000) in physiological, psychological and social dimensions; they also showed a marked improvement of HPLP-II scores by 14.73 points in 6 dimensions (t=-15.34, P=0.000). Multivariable regression analyses with adjusted demographic variables revealed a significant association between health status and health-promoting lifestyle (P<0.05). Compared with a healthy lifestyle (minimal exposure), a 'poor' lifestyle (the highest level of exposure) was associated with a 30 times higher risk of developing SHS (OR: 30.598, 95% CI: 3.928-238.331), while a 'moderate' lifestyle (a relatively high-level exposure) had a 24 times higher risk of SHS (OR: 23.988, 95%CI: 14.695-39.158), and a suboptimal lifestyle had a nearly 4 times higher risk of SHS (OR: 4.306, 95%CI: 2.767-6.702). CONCLUSION: s SHS may evolve into either a healthy or a disease condition. A unhealthy lifestyle is the important risk factor contributing to the progression of SHS into a disease condition, suggesting the importance of intervention of unhealthy lifestyles in promoting good health.


Subject(s)
Health Behavior , Health Status , Healthy Lifestyle , Humans , Prospective Studies , Regression Analysis , Risk Factors , Students
SELECTION OF CITATIONS
SEARCH DETAIL
...