51
|
Krajka-Kuźniak V, Paluszczak J, Kleszcz R, Baer-Dubowska W. (+)-Usnic acid modulates the Nrf2-ARE pathway in FaDu hypopharyngeal carcinoma cells. Mol Cell Biochem 2021; 476:2539-2549. [PMID: 33635505 PMCID: PMC8119403 DOI: 10.1007/s11010-021-04092-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/29/2021] [Indexed: 11/24/2022]
Abstract
Naturally occurring phytochemicals of different origin and structure, arctigenin, bergenin, usnic acid and xanthohumol, were shown to affect Nrf2 pathway in the context of various diseases, but their effect on this pathway in cancer cells was not extensively investigated. This study aimed to evaluate the effect of these compounds on Nrf2 expression and activation in hypopharyngeal FaDu squamous cell carcinoma cells. FaDu cells were treated with 2 or 10 μM arctigenin, bergenin, (+)-usnic acid or xanthohumol for 24 h. While arctigenin, bergenin, and xanthohumol did not affect either Nrf2 expression or activation, (+)-usnic acid treatment increased its transcript level and increased the nuclear/cytosol Nrf2 protein ratio—the measure of Nrf2 pathway activation. Consequently, (+)-usnic acid enhanced the transcription and translation of Nrf2 target genes: NQO1, SOD, and to a lesser extent, GSTP. The treatment of FaDu cells with (+)-usnic acid decreased both GSK-3β transcript and protein level, indicating its possible involvement in Nrf2 activation. All the tested compounds decreased Bax mRNA but did not change the level of Bax protein. (+)-Usnic acid tended to increase the percentage of early apoptotic cells and LC3 protein, autophagy marker. Significant induction of p53 also was observed after treatment with (+)-usnic acid. In summary, the results of this study indicate that low concentrations of (+)-usnic acid activate Nrf2 transcription factor, most probably as a result of ROS accumulation, but do not lead to FaDu hypopharyngeal carcinoma cells death.
Collapse
Affiliation(s)
- Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznan, Poland.
| | - Jarosław Paluszczak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznan, Poland
| | - Robert Kleszcz
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznan, Poland
| | - Wanda Baer-Dubowska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
52
|
Hiebert P. The Nrf2 transcription factor: A multifaceted regulator of the extracellular matrix. Matrix Biol Plus 2021; 10:100057. [PMID: 34195594 PMCID: PMC8233472 DOI: 10.1016/j.mbplus.2021.100057] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/13/2021] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
The transcription factor nuclear factor-erythroid 2-related factor 2 (Nrf2) is widely recognized as a master regulator of the cellular stress response by facilitating the transcription of cytoprotective genes. As such, the Nrf2 pathway is critical in guarding the cell from the harmful effects of excessive reactive oxygen species/reactive nitrogen species (ROS/RNS) and in maintaining cellular redox balance. While excessive ROS/RNS are harmful to the cell, physiological levels of ROS/RNS play important roles in regulating numerous signaling pathways important for normal cellular function, including the synthesis of extracellular matrix (ECM). Recent advances have underscored the importance of ROS/RNS, and by extension, factors that influence redox-balance such as Nrf2, in regulating ECM production and deposition. In addition to reducing the oxidative burden in the cell, the discovery that Nrf2 can also directly target genes that regulate and form the ECM has cemented it as a multifaceted player in the regulation of ECM proteins, and provides new insight into its potential usefulness as a target for treating ECM-related pathologies. Reactive oxygen/nitrogen species regulate extracellular matrix. Nrf2 can directly target extracellular matrix gene transcription. Regulation of extracellular matrix by Nrf2 potentially impacts tissue repair/cancer.
Collapse
Affiliation(s)
- Paul Hiebert
- Institute for Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich 8093, Switzerland
| |
Collapse
|
53
|
Fisetin protects against high fat diet-induced nephropathy by inhibiting inflammation and oxidative stress via the blockage of iRhom2/NF-κB signaling. Int Immunopharmacol 2021; 92:107353. [PMID: 33429334 DOI: 10.1016/j.intimp.2020.107353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023]
Abstract
Promoted inflammation enhances the development of nephropathy in obesity. Fisetin (3,3',4',7-tetrahydroxyflavone, FIS) is a naturally occurring dietary flavonoid, and exhibits anti-inflammatory and anti-oxidative properties. Inactive rhomboid protein 2 (iRhom2), an inactive member of the rhomboid intramembrane proteinase family, is an essential inflammation-associated regulator. Here, we attempted to investigate the protective mechanisms of FIS against high fat diet (HFD)-induced nephropathy, with particular focus on iRhom2. We found that HFD induced systematic and renal pro-inflammatory cytokine production. Furthermore, iRhom2 expression was markedly elevated in kidney of HFD-fed mice, and in PAL-incubated macrophages, accompanied with high phosphorylation of NF-κB. Significant oxidative stress was observed in kidney of HFD-fed mice through suppressing Nrf-2/HO-1 signaling. Moreover, activation of iRhom2/NF-κB signaling and oxidative stress by PAL was detected in macrophages, which were effectively reversed by FIS. Importantly, we showed that iRhom2 knockdown significantly abrogated the ability of FIS to restrain inflammation and oxidative stress induced by PAL in macrophages, indicating that iRhom2 might be a potential therapeutic target for FIS during nephropathy treatment. Together, these results revealed that FIS could mitigate HFD-induced renal injury by regulating iRhom2/NF-κB and Nrf-2/HO-1 signaling pathways.
Collapse
|
54
|
Liu J, Zhang Y, Yu C, Zhang P, Gu S, Wang G, Xiao H, Li S. Bergenin inhibits bladder cancer progression via activating the PPARγ/PTEN/Akt signal pathway. Drug Dev Res 2020; 82:278-286. [PMID: 33112006 DOI: 10.1002/ddr.21751] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/21/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022]
Abstract
Bladder cancer is one of the most common malignant tumors in the urinary system with high mortality and morbidity. Evidence revealed that bergenin could affect the development of cancer. Here, we aimed to investigate the effect of bergenin on bladder cancer progression and its mechanism. The effect of bergenin on cell function was first detected, followed by assessing the changes of the epithelial-mesenchymal transition (EMT) in bergenin-treated cells. The effect of bergenin on peroxisome proliferator-activated receptor γ (PPARγ)/phosphatase and tensin homolog (PTEN)/Akt signal pathway was measured by Western blotting, followed by the rescue experiments. The results showed that bergenin treatment significantly decreased cell viability and increased G1 phase arrest, accompanied by reduced expression of Ki67, cycling D1, and cycling B1 in bladder cancer cells. Apoptosis was induced by bergenin in bladder cancer cells, as evidenced by increased Bax and cleaved caspase 3 protein levels and decreased Bcl-2 level in bergenin-treated cells. Meanwhile, the inhibition of the invasion, migration, and EMT was also observed in bergenin-treated cells. Mechanism studies showed that bergenin treatment could activate PPARγ/PTEN/Akt signal pathway, as evidence by the increased nucleus PPARγ and phosphatase and tensin homolog (PTEN) expression and decreased Akt expression. Moreover, PPARγ inhibitor administration inverted the effects of bergenin on bladder cancer cell function, including the proliferation, apoptosis, invasion, and migration in bladder cancer cells. Our findings revealed that bergenin could inhibit bladder cancer progression via activating the PPARγ/PTEN/Akt signal pathway, indicating that bergenin may be a potential therapeutic medicine for bladder cancer treatment.
Collapse
Affiliation(s)
- Junjiang Liu
- Department of Urology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Yunxia Zhang
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Chunhong Yu
- Physical Examination Center, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Panying Zhang
- Department of Urology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Shouyi Gu
- Department of Urology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Gang Wang
- Department of Urology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Helong Xiao
- Department of Urology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| | - Shoubin Li
- Department of Urology, Hebei General Hospital, Shijiazhuang, Hebei, P.R. China
| |
Collapse
|
55
|
Liu R, Chen L, Wang Z, Zheng X, Hou Z, Zhao D, Long J, Liu J. Omega-3 polyunsaturated fatty acids prevent obesity by improving tricarboxylic acid cycle homeostasis. J Nutr Biochem 2020; 88:108503. [PMID: 32956825 DOI: 10.1016/j.jnutbio.2020.108503] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/29/2020] [Accepted: 09/03/2020] [Indexed: 02/08/2023]
Abstract
The beneficial effects of omega-3 polyunsaturated fatty acids (n-3 PUFAs) on preventing obesity are well known; however, the underlying mechanism by which n-3 PUFAs influence tricarboxylic acid (TCA) cycle under obesity remains unclear. We randomly divided male C57BL/6 mice into 5 groups (n=10) and fed for 12 weeks as follows: mice fed a normal diet (Con, 10% kcal); mice fed a high-fat diet (HFD, lard, 60% kcal); and mice fed a high-fat diet (60% kcal) substituting half the lard with safflower oil (SO), safflower oil and fish oil (SF) and fish oil (FO), respectively. Then we treated HepG2 cells with palmitic acid and DHA for 24 h. We found that body weight in FO group was significantly lower than it in HFD and SO groups. N-3 PUFAs reduced the transcription and translation of TCA cycle enzymes, including IDH1, IDH2, SDHA, FH and MDH2, to enhance mitochondrial function in vivo and vitro. DHA significantly inhibited protein expression of the mTORC1 signaling pathway, increased p-AKT protein expression to alleviate insulin resistance and improved mitochondrial oxygen consumption rate and glycolysis ability in HepG2 cells. In addition, the expressions of IDH2 and SDHB were reduced by rapamycin. N-3 PUFAs could prevent obesity by improving TCA cycle homeostasis and mTORC1 signaling pathway may be upstream.
Collapse
Affiliation(s)
- Run Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Lei Chen
- Institute of Nutrition & Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China; Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhen Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xuewei Zheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhanwu Hou
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Daina Zhao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
56
|
Wang G, Wu B, Zhang B, Wang K, Wang H. LncRNA CTBP1-AS2 alleviates high glucose-induced oxidative stress, ECM accumulation, and inflammation in diabetic nephropathy via miR-155-5p/FOXO1 axis. Biochem Biophys Res Commun 2020; 532:308-314. [PMID: 32868076 DOI: 10.1016/j.bbrc.2020.08.073] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND This study aimed to investigate the involvement of lncRNA CTBP1-AS2 in the progression of diabetic nephropathy (DN) by affecting high glucose (HG)-induced human glomerular mesangial cells (HGMCs). METHODS HGMCs were selected for the establishment of cell injury induced by HG. The expression of CTBP1-AS2, miR-155-5p and FOXO1 was detected by real-time PCR and western blotting. The target association between miR-155-5p and CTBP1-AS2 or FOXO1 was confirmed by dual-luciferase reporter assays. Cell proliferation and oxidative stress were revealed by CCK-8 colorimetry, and the measurement of reactive oxygen species (ROS) and the activities of antioxidant enzymes. Extracellular matrix (ECM) protein accumulation and the production of inflammatory cytokines were investigated by western blotting and ELISA. RESULTS The expression of CTBP1-AS2 was downregulated, and miR-155-5p was highly expressed in peripheral blood of DN patients and HG-treated HGMCs. Further investigation revealed that CTBP1-AS2 overexpression inhibited proliferation, oxidative stress, ECM accumulation and inflammatory response in HG-induced HGMCs. Mechanical analysis revealed that CTBP1-AS2 regulated FOXO1 expression via sponging miR-155-5p. Rescue experiments demonstrated that miR-155-5p overexpression or FOXO1 inhibition reversed the effects of CTBP1-AS2 in HG-stimulated HGMCs. CONCLUSION Taken together, this study revealed CTBP1-AS2 attenuated HG-induced HGMC proliferation, oxidative stress, ECM accumulation, and inflammation through miR-155-5p/FOXO1 signaling.
Collapse
Affiliation(s)
- Guang Wang
- Department of Intensive Care Unit, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Bing Wu
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, 130033, China
| | - Bo Zhang
- Department of Pediatric Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130020, China
| | - Kun Wang
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of JiLin University, 126Xiantai Street, Changchun, Jilin Province, 130021, China
| | - Heyuan Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
57
|
Villarreal CF, Santos DS, Lauria PSS, Gama KB, Espírito-Santo RF, Juiz PJL, Alves CQ, David JM, Soares MBP. Bergenin Reduces Experimental Painful Diabetic Neuropathy by Restoring Redox and Immune Homeostasis in the Nervous System. Int J Mol Sci 2020; 21:ijms21144850. [PMID: 32659952 PMCID: PMC7420298 DOI: 10.3390/ijms21144850] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetic neuropathy is a frequent complication of diabetes. Symptoms include neuropathic pain and sensory alterations—no effective treatments are currently available. This work characterized the therapeutic effect of bergenin in a mouse (C57/BL6) model of streptozotocin-induced painful diabetic neuropathy. Nociceptive thresholds were assessed by the von Frey test. Cytokines, antioxidant genes, and oxidative stress markers were measured in nervous tissues by ELISA, RT-qPCR, and biochemical analyses. Single (3.125–25 mg/kg) or multiple (25 mg/kg; twice a day for 14 days) treatments with bergenin reduced the behavioral signs of diabetic neuropathy in mice. Bergenin reduced both nitric oxide (NO) production in vitro and malondialdehyde (MDA)/nitrite amounts in vivo. These antioxidant properties can be attributed to the modulation of gene expression by the downregulation of inducible nitric oxide synthase (iNOS) and upregulation of glutathione peroxidase and Nrf2 in the nervous system. Bergenin also modulated the pro- and anti-inflammatory cytokines production in neuropathic mice. The long-lasting antinociceptive effect induced by bergenin in neuropathic mice, was associated with a shift of the cytokine balance toward anti-inflammatory predominance and upregulation of antioxidant pathways, favoring the reestablishment of redox and immune homeostasis in the nervous system. These results point to the therapeutic potential of bergenin in the treatment of painful diabetic neuropathy.
Collapse
Affiliation(s)
- Cristiane F. Villarreal
- Faculdade de Farmácia, Universidade Federal da Bahia, CEP 40.170-115 Salvador, Brazil; (D.S.S.); (P.S.S.L.); (R.F.E.-S.)
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, CEP 40.296-710 Salvador, Brazil; (K.B.G.); (M.B.P.S.)
- Correspondence: ; Tel.: +55-(71)3283-6933
| | - Dourivaldo S. Santos
- Faculdade de Farmácia, Universidade Federal da Bahia, CEP 40.170-115 Salvador, Brazil; (D.S.S.); (P.S.S.L.); (R.F.E.-S.)
| | - Pedro S. S. Lauria
- Faculdade de Farmácia, Universidade Federal da Bahia, CEP 40.170-115 Salvador, Brazil; (D.S.S.); (P.S.S.L.); (R.F.E.-S.)
| | - Kelly B. Gama
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, CEP 40.296-710 Salvador, Brazil; (K.B.G.); (M.B.P.S.)
| | - Renan F. Espírito-Santo
- Faculdade de Farmácia, Universidade Federal da Bahia, CEP 40.170-115 Salvador, Brazil; (D.S.S.); (P.S.S.L.); (R.F.E.-S.)
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, CEP 40.296-710 Salvador, Brazil; (K.B.G.); (M.B.P.S.)
| | - Paulo J. L. Juiz
- Universidade Federal do Recôncavo da Bahia, CEP 44.042-280 Feira de Santana, Brazil;
| | - Clayton Q. Alves
- Departamento de Ciências Exatas, Universidade Estadual de Feira de Santana, CEP 44.036-336 Feira de Santana, Brazil;
| | - Jorge M. David
- Instituto de Química, Universidade Federal da Bahia, CEP 40.170-280 Salvador, Brazil;
| | - Milena B. P. Soares
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, CEP 40.296-710 Salvador, Brazil; (K.B.G.); (M.B.P.S.)
| |
Collapse
|
58
|
Vasileva LV, Savova MS, Amirova KM, Dinkova-Kostova AT, Georgiev MI. Obesity and NRF2-mediated cytoprotection: Where is the missing link? Pharmacol Res 2020; 156:104760. [DOI: 10.1016/j.phrs.2020.104760] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/15/2020] [Accepted: 03/17/2020] [Indexed: 12/29/2022]
|
59
|
Punicalagin Protects Diabetic Nephropathy by Inhibiting Pyroptosis Based on TXNIP/NLRP3 Pathway. Nutrients 2020; 12:nu12051516. [PMID: 32456088 PMCID: PMC7284711 DOI: 10.3390/nu12051516] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/14/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic nephropathy is a diabetic complication caused by chronic inflammation. As the primary polyphenol in pomegranate, punicalagin is believed to have significant anti-inflammatory properties. In this study, we established a mice model for diabetes induced by high-fat diet (HFD)/ streptozotocin (STZ) to verify the protective effect of punicalagin in vivo. The results show that the blood urea nitrogen (BUN), serum creatinine (CREA), and the urine albumin to creatinine ratio (UACR) were significantly decreased in diabetic mice after punicalagin intervention, and the symptoms of glomerular interstitial hyperplasia and glomerular hypertrophy were alleviated. Pyroptosis is an essential manner of programmed cell death in the inflammatory response; the expression of pyroptosis-related proteins such as interleukin-1 (IL-1β), cysteinyl aspartate-specific protease-1 (caspase-1), gasdermin D (GSDMD), and nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain containing protein 3 (NLRP3) was decreased in our study, which proved that the administration of punicalagin for eight weeks can significantly inhibit pyroptosis in mice. In addition, punicalagin reduced high glucose-mediated protein expressions of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) and alleviated mitochondria damage. Low expression of NOX4 inhibits the dissociation of thioredoxin (Trx) and thioredoxin-interacting protein (TXNIP) and the suppression of NLRP3 inflammasome activation. To summarize, our study provided evidence that punicalagin can alleviate diabetic nephropathy, and the effect is associated with downregulating the expression of NOX4, inhibiting TXNIP/NLRP3 pathway-mediated pyroptosis, suggesting its therapeutic implications for complications of diabetes.
Collapse
|
60
|
Shen X, Dong X, Han Y, Li Y, Ding S, Zhang H, Sun Z, Yin Y, Li W, Li W. Ginsenoside Rg1 ameliorates glomerular fibrosis during kidney aging by inhibiting NOX4 and NLRP3 inflammasome activation in SAMP8 mice. Int Immunopharmacol 2020; 82:106339. [PMID: 32114413 DOI: 10.1016/j.intimp.2020.106339] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 01/08/2023]
Abstract
Aging is closely related to the progress of renal fibrosis, which eventually results in renal dysfunction. Ginsenoside Rg1 (Rg1) has been reported to have an extensive anti-aging effect. However, the role and mechanism of Rg1 in aging-related renal fibrosis remain unclear. The present study aimed to evaluate the protective effect and mechanism of Rg1 in renal fibrosis during kidney aging in a model of SAMP8 mice. Taking SAMR1 mice as the control group, SAMP8 mice were administered Apocynin (50 mg/kg), Tempol (50 mg/kg), or Rg1 (5, 10 mg/kg) intragastrically for 9 weeks as treatment groups. The results showed that the elevated levels of blood urea nitrogen, serum creatinine and senescence-associated β-galactosidase (β-Gal) were markedly decreased, the glomerular mesangial proliferation was significantly alleviated and the increased levels of collagen IV and TGF-β1 were significantly downregulated by Rg1 in SAMP8 mice. In addition, the generation of ROS and the expression of NADHP oxidase 4 (NOX4) in the renal cortex were significantly reduced by Rg1 treatment. The expression levels of NLRP3 inflammasome-related proteins and the inflammation-related cytokine IL-1β were also inhibited by Rg1 treatment in the SAMP8 mice. These results suggested that Rg1 could delay kidney aging and inhibit aging-related glomerular fibrosis by reducing NOX4-derived ROS generation and downregulating NLRP3 inflammasome expression.
Collapse
Affiliation(s)
- Xiaoyan Shen
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Xianan Dong
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Yuli Han
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Yan Li
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Shixin Ding
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Han Zhang
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Zhenghao Sun
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Yanyan Yin
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China
| | - Weiping Li
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China; Anqing Medical and Pharmaceutical College, Anqing 246052, Anhui, China.
| | - Weizu Li
- Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
61
|
Xu Z, Dai XX, Zhang QY, Su SL, Yan H, Zhu Y, Shang EX, Qian DW, Duan JA. Protective effects and mechanisms of Rehmannia glutinosa leaves total glycoside on early kidney injury in db/db mice. Biomed Pharmacother 2020; 125:109926. [PMID: 32028239 DOI: 10.1016/j.biopha.2020.109926] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 12/10/2019] [Accepted: 12/15/2019] [Indexed: 02/07/2023] Open
Abstract
The spontaneous db/db mice were used to elucidate the biological effects and mechanisms of Rehmannia glutinosa leaves total glycoside (DHY) on kidney injury through biochemical indicators, kidney pathological section analysis, metabolic profiling, intestinal flora analysis and in vitro Human renal tubular epithelial (HK-2) cell model induced by high glucose. It was found that DHY can decrease the blood sugar level (insulin, INS; fasting blood glucose, FBG), blood lipid level (Total Cholesterol, T-CHO; Triglyceride, TG) significantly and improve kidney injury level (blood urea nitrogen, BUN; urine microalbumin, mALB; serum creatinine, Scr). It can also alleviate kidney tubular epithelial cell oedema and reduce interstitial connective tissue hyperplasia of the injury kidney induced by high glucose. 13 endogenous metabolites were identified in serum, which involved of ether lipid metabolism, sphingolipid metabolism, glyoxylic acid and dicarboxylic acid metabolism and arachidonic acid metabolism. High glucose can also lead to the disorder of intestinal flora, especially Firmicutes and Bacteroides. Meanwhile, DHY also inhibited the expression of α-SMA, TGF- β1, Smad3 and Smad4 in the kidney tissues of db/db mice and HK-2 cells. To sum up, DHY may restore the dysfunctional intestinal flora to normal and regulate glycolipid level of db/db mice as well as TGF-β/Smad signalling pathway regulation to improve early kidney damage caused by diabetes.
Collapse
Affiliation(s)
- Zhuo Xu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, State Administration of Traditional Chinese Medicine, Traditional Chinese Medicine Resource Recycling, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Xin-Xin Dai
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, State Administration of Traditional Chinese Medicine, Traditional Chinese Medicine Resource Recycling, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Qing-Yang Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, State Administration of Traditional Chinese Medicine, Traditional Chinese Medicine Resource Recycling, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Shu-Lan Su
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, State Administration of Traditional Chinese Medicine, Traditional Chinese Medicine Resource Recycling, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Hui Yan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, State Administration of Traditional Chinese Medicine, Traditional Chinese Medicine Resource Recycling, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yue Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, State Administration of Traditional Chinese Medicine, Traditional Chinese Medicine Resource Recycling, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Er-Xin Shang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, State Administration of Traditional Chinese Medicine, Traditional Chinese Medicine Resource Recycling, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Da-Wei Qian
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, State Administration of Traditional Chinese Medicine, Traditional Chinese Medicine Resource Recycling, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, State Administration of Traditional Chinese Medicine, Traditional Chinese Medicine Resource Recycling, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
62
|
Ashrafizadeh M, Ahmadi Z, Samarghandian S, Mohammadinejad R, Yaribeygi H, Sathyapalan T, Sahebkar A. MicroRNA-mediated regulation of Nrf2 signaling pathway: Implications in disease therapy and protection against oxidative stress. Life Sci 2020; 244:117329. [PMID: 31954747 DOI: 10.1016/j.lfs.2020.117329] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/12/2020] [Accepted: 01/15/2020] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRs) are small non-coding pieces of RNA that are involved in a variety of physiologic processes such as apoptosis, cell proliferation, cell differentiation, cell cycle and cell survival. These multifunctional nucleotides are also capable of preventing oxidative damages by modulating antioxidant defense systems in a variety of milieu, such as in diabetes. Although the exact molecular mechanisms by which miRs modulate the antioxidant defense elements are unclear, some evidence suggests that they may exert these effects via nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. This intracellular mechanism is crucial in the maintenance of the physiologic redox balance by regulating the expression and activity of various cellular antioxidative defense elements and thereby plays a pivotal role in the development of oxidative stress. Any impairment in the Nrf2 signaling pathway may result in oxidative damage-dependent complications such as various diabetic complications, neurological disorders and cancer. In the current review, we discuss the modulatory effects of miRs on the Nrf2 signaling pathway, which can potentially be novel therapeutic targets.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Zahra Ahmadi
- Department of Basic Science, Shoushtar Branch, Islamic Azad University, Shoushtar, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
63
|
Dong Z, Iqbal S, Zhao Z. Preparation of Ergosterol-Loaded Nanostructured Lipid Carriers for Enhancing Oral Bioavailability and Antidiabetic Nephropathy Effects. AAPS PharmSciTech 2020; 21:64. [PMID: 31932990 DOI: 10.1208/s12249-019-1597-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023] Open
Abstract
In our previously studies, we confirmed that ergosterol could ameliorate diabetic nephropathy by suppressing the proliferation of mesangial cells and the accumulation of extracellular matrix (ECM). However, the therapeutic application of ergosterol may be confined due to poor aqueous solubility and low oral bioavailability. We aim to prepare ergosterol-loaded nanostructured lipid carriers (ERG-NLCs) to enhance the solubility and oral bioavailability of ergosterol. ERG-NLCs were prepared using glyceryl monostearate and decanoyl/octanoyl-glycerides by hot emulsification-ultrasonication method and characterized by dynamic light scattering (DLS), transmission electron microscopy (TEM), differential scanning calorimetry (DSC), powder X-ray diffraction (PXRD) analysis, entrapment efficiency (EE), and drug loading (DL) capacity studies. The prepared ERG-NLCs were spherical, with particle size of 81.39 nm and negative zeta potential of 30.77 mV. Ergosterol was successfully encapsulated in NLCs with a high EE of 92.95% and a DL capacity of 6.51%. In pharmacokinetic study, Cmax and AUC0-∞ of ergosterol in ERG-NLCs were obviously enhanced, and the relative oral bioavailability of ERG-NLCs was 277.56% higher than that of raw ergosterol. Moreover, the in vitro pharmacodynamic study indicated that ERG-NLCs inhibited high-glucose-stimulated mesangial cells over proliferation and ECM accumulation more effectively compared to raw ergosterol. In conclusion, the validated ERG-NLCs showed that NLCs mediated delivery could be used as potential vehicle to enhance solubility, oral bioavailability and therapeutic efficacy of ergosterol.
Collapse
|