1
|
Biswas S, Mondal M, Pakhira S, Ghosh R, Samanta P, Basu J, Bhowmik A, Hajra S, Saha P. Attenuation of paclitaxel-induced toxicities by polyphenolic natural compound rutin through inhibition of apoptosis and activation of NRF2/ARE signaling pathways. Food Chem Toxicol 2025; 200:115408. [PMID: 40154830 DOI: 10.1016/j.fct.2025.115408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Paclitaxel is the first microtubule-stabilizing drug widely used as an antineoplastic agent. Hepatotoxicity, nephrotoxicity and myeloid suppression may lead to secondary malignancy which is an important adverse effect of paclitaxel-therapy. In this study, we have evaluated the potential protective role of natural flavonoid rutin against paclitaxel-induced toxicities in BALB/cmice. Paclitaxel was administered intraperitoneally (in alternate days at a dose of 8.5 mg/kg b. w.) and rutin was given every day by oral gavages (20 mg/kg b. w.) in BALB/c mice. Results showed that administration of paclitaxel significantly (P < 0.05) increased the generation of ROS and NO in bone marrow, liver and kidney tissues. In contrast, co-administration of rutin and paclitaxel significantly (p < 0.05) reduced the intracellular ROS and NO levels, reversed the toxic effects of paclitaxel through NRF2-mediated activation of antioxidant response element (ARE) pathway and upregulated activity of several phase-II antioxidant enzymes. Furthermore, rutin treatment inhibited apoptosis by downregulated expression of Bax, caspase-3 and cPARP in bone marrow, liver and kidney tissues. Additionally, the chemoprotective potential of rutin was confirmed by histopathological analysis. Thus, our results suggest that co-administration of rutin may serve as a promising preventive strategy against paclitaxel induced toxicities and indicate its future use as an adjuvant in chemotherapy.
Collapse
Affiliation(s)
- Souradeep Biswas
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, West Bengal, India.
| | - Mrinmoyee Mondal
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Shampa Pakhira
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Rituparna Ghosh
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Priya Samanta
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Jhinuk Basu
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Arijit Bhowmik
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Subhadip Hajra
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, 700 026, West Bengal, India.
| |
Collapse
|
2
|
Gao Y, Han C, Chen Z, Huang J, Peng T, Ding X, Zhong H, Liao L, He C, Huang J. Theabrownins improve burn-induced kidney injury by increasing the levels of guanidinoacetic acid and fumaric acid. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156609. [PMID: 40090045 DOI: 10.1016/j.phymed.2025.156609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Burns are a common and serious health issue, with severe burn-induced acute kidney injury (AKI) being a major factor contributing to poor recovery and increased mortality in patients. Theabrownins (TBs), bioactive compounds formed during tea leaf fermentation, have shown promising effects on reducing inflammation, combating oxidative stress, and enhancing metabolic function. However, the roles and mechanisms of TBs in burn-induced kidney injury are still not fully understood. METHODS The dorsal skin of 3-month-old mice was exposed to hot water for 10 s to induce burn-related renal injury. The mice were then orally administered TBs (40 mg/kg and 400 mg/kg). After 24 h of treatment, the mice were sacrificed for tissue collection. Transcriptomic and metabolomic analyses were performed to identify the pathways modulated by TBs. Metabolomics revealed TB-associated renal metabolites, such as guanidinoacetic acid (GAA) and fumaric acid (FA). Renal tubular epithelial (HK2) cells pretreated with GAA and FA were exposed to hydrogen peroxide (H2O2), cisplatin (CDDP) and erastin to establish a cell injury model. Changes in the levels of relevant molecules were assessed using quantitative RT-PCR, Western blotting, and fluorescence staining. RESULTS TB treatment significantly increased the survival rate and reduced kidney injury in mice with burn injury. Multiomics analyses and molecular experimental validation revealed that TB treatment downregulated the inflammation, apoptosis, and ferroptosis pathways in the kidneys of mice with burn injury and increased the levels of the renal metabolites GAA and FA. Cellular experiments confirmed that GAA and FA alleviated H2O2-, CDDP- and erastin-induced renal tubular epithelial cell injury by inhibiting apoptosis and ferroptosis. CONCLUSIONS Burns induce inflammation and kidney damage by upregulating the apoptosis and ferroptosis pathways in renal tissue. TBs alleviate burn-induced renal apoptosis and ferroptosis by increasing the levels of GAA and FA in the kidneys, thereby ameliorating kidney damage. This study innovatively and systematically evaluated the ability of TBs to ameliorate burn-induced kidney injury and, for the first time, identified the potential mechanism by which TBs ameliorate burn-induced kidney damage by increasing the levels of the metabolites GAA and FA in the kidneys.
Collapse
Affiliation(s)
- You Gao
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China; Department of Plastic and Aesthetic Burn, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Changshun Han
- Department of Anesthesiology, School of Medicine, Xiang'an Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhiyuan Chen
- Department of Anesthesiology, School of Medicine, Xiang'an Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jiancheng Huang
- Department of Anesthesiology, School of Medicine, Xiang'an Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Tianyun Peng
- Department of Anesthesiology, School of Medicine, Xiang'an Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiaoyan Ding
- Department of Anesthesiology, School of Medicine, Xiang'an Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Hongbin Zhong
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The Fifth Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Lixin Liao
- Department of Plastic and Aesthetic Burn, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Chengyong He
- Department of Anesthesiology, School of Medicine, Xiang'an Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| | - Jiyi Huang
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China; Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The Fifth Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
3
|
Abdelhady R, Arab HH, Fakhr Eldeen RR, Shalaby HN, Nawwar DA, Elhemely MA, Sayed RH. Unveiling the Therapeutic Potential of Dulaglutide in Mitigating Tacrolimus-Induced Nephrotoxicity Through Targeting the miR-22/HMGB-1/TLR4/MyD88/NF-κB Trajectory. Arch Pharm (Weinheim) 2025; 358:e3127. [PMID: 40205909 PMCID: PMC11983086 DOI: 10.1002/ardp.202500023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/23/2025] [Accepted: 03/22/2025] [Indexed: 04/11/2025]
Abstract
Tacrolimus (Tac) is an immunosuppressive drug used to reduce the risk of allograft rejection; however, it can induce renal injury. High mobility group box 1 (HMGB-1) protein, which induces inflammation through the aberrant stimulation of the Toll-like receptor 4 (TLR4)/myeloid differentiation primary response protein (MyD88)/nuclear factor kappa B (NF-κB) trajectory, could represent a molecular target for alleviating Tac-induced renal damage. The present study aimed to investigate the potential protective role of the GLP-1 agonist, dulaglutide (Dula), against Tac-induced nephrotoxicity in rats. Rats were administered Tac (5 mg/kg/day) and vehicle or Dula (0.2 mg/kg once a week) for 14 days. Treatment with Dula reduced serum creatinine plus blood urea nitrogen and attenuated Tac-induced renal histopathological changes. Dula treatment also hampered renal inflammation and restored redox homeostasis, as indicated by remarkably reduced tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), malondialdehyde (MDA), and NADPH oxidase 1 levels alongside marked replenishment in reduced glutathione (GSH) content. These effects were mediated through the upregulation of miR-22 expression and the consequent inhibition of the HMGB-1/TLR4/MyD88/NF-κB trajectory. Collectively, Dula has been demonstrated to protect rats against Tac-induced nephrotoxicity by reducing inflammation, restoring redox homeostasis, and modulation of the miR-22/HMGB-1/TLR4/MyD88/NF-κB trajectory. Dula may be beneficial clinically in preventing Tac-induced renal injury.
Collapse
Affiliation(s)
- Rasha Abdelhady
- Pharmacology and Toxicology Department, Faculty of PharmacyFayoum UniversityFayoumEgypt
- Pharmacology and Toxicology Department, Faculty of PharmacyEgyptian Chinese UniversityCairoEgypt
| | - Hany H. Arab
- Department of Pharmacology and Toxicology, College of PharmacyTaif UniversityTaifSaudi Arabia
| | - Rasha R. Fakhr Eldeen
- Biochemistry Department, College of Pharmaceutical Sciences and Drug ManufacturingMisr University for Science and Technology6th of October CityEgypt
| | - Heba Nasr Shalaby
- Pharmacology and Toxicology Department, Faculty of PharmacyCairo UniversityCairoEgypt
| | - Dalia A. Nawwar
- Pharmacology and Toxicology Department, Faculty of PharmacyCairo UniversityCairoEgypt
| | - Mai Abdallah Elhemely
- School of Medical Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Department of Pharmacology and Toxicology, Faculty of PharmacyBeni‐Suef UniversityBeni‐SuefEgypt
| | - Rabab H. Sayed
- Pharmacology and Toxicology Department, Faculty of PharmacyCairo UniversityCairoEgypt
- School of PharmacyNewgiza UniversityGizaEgypt
| |
Collapse
|
4
|
Geng M, Cheng Y, Duan L, Dong G, Yang S, Jiao X, Jia W, Huang H, Dou M, Fu X, Yu F, Du Y, Zhou G, Ba Y. Inflammation May Mediate the Effects of Fluoride on Liver and Kidney Function of Adults: Cross-Sectional Studies in China. Biol Trace Elem Res 2025:10.1007/s12011-025-04583-4. [PMID: 40117029 DOI: 10.1007/s12011-025-04583-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/13/2025] [Indexed: 03/23/2025]
Abstract
To assess associations among fluoride exposure, the inflammation, and liver and kidney functions, a total of 1646 adults aged ≥ 18 years were recruited in cross-sectional studies conducted in 2017 and 2022 in fluorosis areas. Questionnaire surveys were administered to obtain the demographic information. Urine and blood samples were collected for determinations of urinary fluoride (UF), aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (TBIL), blood urea nitrogen (BUN), serum creatinine (SCr), serum uric acid (SUA), and white blood cell (WBC) counts. Linear regressions, generalized linear models, and mediation analyses were employed here. We found that AST, BUN, and SUA increased by 0.589, 0.087, and 4.226 with a 1 mg/L increment of UF, and the SCr showed a negative linear association with UF (β = - 0.790) in all. Similar results were also observed in female. However, we only found the positive linear association between UF and AST/ALT in male. We also detected a significant modification by gender on associations between UF and values of AST, ALT, BUN, and SCr. Besides, participants with higher UF levels had higher counts of monocytes (β = 0.014) and neutrophils (β = 0.147) both in all and in stratified analyses of gender. Associations between AST/ALT, TBIL, SCr, SUA, and counts of WBC were revealed. In addition, there were mediator effects of monocytes and neutrophils in associations between UF and BUN (or SUA). We observed similar results in the age group of 46-64 years with those in all participants. This study observed a statistically significant association between UF and adult levels of AST, SCr, BUN, SUA, as well as monocyte and neutrophil counts, particularly in females. Furthermore, alterations in monocyte and neutrophil counts partially mediate the association between UF and BUN (or SUA). Our findings reveal the effects of fluoride exposure on liver and kidney function and provide clues for analyzing the relevant mechanism from an inflammatory perspective.
Collapse
Affiliation(s)
- Minghao Geng
- Department of Preventive Medicine, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yi Cheng
- Department of Environmental Health & Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Leizhen Duan
- Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Guangshen Dong
- Department of Basic Medicine, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Shuo Yang
- Department of Environmental Health & Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xuecheng Jiao
- Department of Endemic Diseases, Puyang Center for Disease Control and Prevention, Puyang, 457005, Henan, China
| | - Weihua Jia
- Department of Endemic Diseases, Zhengzhou Center for Disease Control and Prevention, Zhengzhou, 450006, Henan, China
| | - Hui Huang
- Department of Environmental Health & Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Ming Dou
- School of Water Conservancy and Transportation, School of Ecology and Environment, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xiaoli Fu
- Department of Preventive Health Care, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangfang Yu
- Department of Environmental Health & Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yuhui Du
- National Experimental Teaching Demonstration Center of Public Health and Preventive Medicine, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Guoyu Zhou
- Department of Environmental Health & Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Yue Ba
- Department of Environmental Health & Environment and Health Innovation Team, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| |
Collapse
|
5
|
Ye S, Song S, Liu X, Luo Y, Cai S. A small-molecule screen identifies novel aging modulators by targeting 5-HT/DA signaling pathway. Aging Cell 2025; 24:e14411. [PMID: 39552540 PMCID: PMC11896485 DOI: 10.1111/acel.14411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/15/2024] [Accepted: 10/29/2024] [Indexed: 11/19/2024] Open
Abstract
The risk of many human diseases including cardiovascular diseases, cancer, neurodegenerative diseases, and musculoskeletal disorders rises significantly in the elderly. With the increase in the aging population, it is becoming increasingly important to understand the biology of healthy aging and develop interventions that slow down the aging process or prevent age-related diseases. In this study, by a high-throughput screen in Caenorhabditis elegans (C. elegans), we identified 11 small molecules that promote healthy aging. Among them, Carbamazepine (a voltage-gated channels inhibitor) and Calmagite (a calcium and magnesium indicator) enhanced serotonin (5-HT) and dopamine (DA) levels, extended lifespan, and preserved several important behaviors in aging C. elegans. These behaviors include slowing responses to food, pharyngeal pumping, locomotion, and male mating. Interestingly, we further found that administration of Carbamazepine or Calmagite alleviated hyperexcitability of aging male diagonal muscles and improved behavioral performance by ameliorating Ca2+ homeostasis. Mechanistically, administration of Carbamazepine or Calmagite induced nuclear translocation of the transcription factor DAF-16 and thus up-regulated its downstream genes numr-1/-2, which are known to promote resistance to metal-induced stresses and longevity. Taken together, our study offers a way for the discovery of drugs that promote healthy aging, and provides potential interventions for preventing behavioral deterioration in the elderly.
Collapse
Affiliation(s)
- Shi‐Wei Ye
- Institute of Neuroscience and State key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Shuang‐Di Song
- Institute of Neuroscience and State key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xi‐Juan Liu
- Institute of Neuroscience and State key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
| | - Yun Luo
- Institute of Neuroscience and State key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
| | - Shi‐Qing Cai
- Institute of Neuroscience and State key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
| |
Collapse
|
6
|
Faheem H, Alawadhi R, Basha EH, Ismail R, Ibrahim HA, Elshamy AM, Motawea SM, Seleem MA, Elkordy A, Homouda AA, Khaled HE, Aboeida RA, Abdel Ghafar MT, Rizk FH, El-Harty YM. Ameliorating immune-dependent inflammation and apoptosis by targeting TLR4/MYD88/NF-κB pathway by celastrol mitigates the diabetic reproductive dysfunction. Physiol Genomics 2025; 57:103-114. [PMID: 39510137 DOI: 10.1152/physiolgenomics.00072.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
This study aimed to examine the protective effect of celastrol on testicular dysfunction in diabetic rats and the potential underlying mechanisms. All rats included in the study were divided into four groups: a control group treated with sodium citrate buffer and vehicle), a celastrol-treated control group, a streptozotocin (STZ)-induced diabetic group following insulin resistance, and a celastrol-treated diabetic group. Serum glucose, triglyceride, total cholesterol, high-density lipoprotein cholesterol, interleukin (IL)-1β, tumor necrosis factor-α, and testosterone levels were measured. In addition, the levels of testicular homogenate superoxide dismutase and malondialdehyde were assessed. Furthermore, testicular tissue relative toll-like receptor 4 (TLR4), nuclear factor kappa B (NF-κB), and myeloid differentiation factor 88 (MYD88) expressions were quantitatively measured using polymerase chain reaction. Histopathological and immunohistochemical studies were also conducted. The results revealed that treatment with celastrol significantly reduced TLR4, MyD88, and NF-κB expressions, and the levels of inflammatory mediators such as tumor necrosis factor-α and IL-1β in the testicular tissue of treated rats. These findings suggest that celastrol has the potential to be effective in the treatment of diabetes-induced testicular injury by inhibiting testicular inflammation, apoptosis, and oxidative stress.NEW & NOTEWORTHY Celastrol inhibits the production of proinflammatory cytokines in the testicular tissue by specifically targeting the TLR4/MyD88/NF-κB signaling cascade pathways. This indicates that celastrol may serve as a promising new therapeutic target for treating diabetic reproductive dysfunction.
Collapse
Affiliation(s)
- Heba Faheem
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rana Alawadhi
- Science Department, College of Basic Education, PAAET, Ardhiya, Kuwait
| | - Eman H Basha
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
- Department of Basic Medical Sciences-Physiology, Faculty of Medicine, Ibn Sina University for Medical Sciences, Amman, Jordan
| | - Radwa Ismail
- Department of Anatomy and Embryology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hoda A Ibrahim
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Amira M Elshamy
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Shaimaa M Motawea
- Department of Anatomy and Embryology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Monira A Seleem
- Department of Medical Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Alaa Elkordy
- Department of Neuropsychiatry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Abdallah A Homouda
- Department of Urology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Howayda E Khaled
- Department of Zoology, Faculty of Science, Suez University, Suez, Egypt
| | - Reham A Aboeida
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Fatma H Rizk
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Yasmeen M El-Harty
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
7
|
Tao S, Qi Y, Gao J, Yuan H, Wang R, Shen X, Wei G, Peng Z. Ameliorative Effect of Sipunculus nudus Hydrolysate on Cisplatin-Induced Nephrotoxicity by Mitigating Oxidative Stress, Inflammation and Apoptosis. Mar Drugs 2025; 23:100. [PMID: 40137286 PMCID: PMC11943811 DOI: 10.3390/md23030100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 03/27/2025] Open
Abstract
The present study investigated the protective effects and possible mechanisms of an ultrafiltration fraction of Sipunculus nudus hydrolysate (UFSH) on cisplatin-induced kidney damage in a mouse model. The results showed that UFSH significantly attenuated cisplatin-induced nephrotoxicity by inhibiting increases in blood urea nitrogen (BUN) and serum creatinine (SCr). Additionally, UFSH treatment significantly alleviated cisplatin-induced renal histopathological changes, such as significant dilation of renal tubules, cast formation, and tubular cell necrosis, as well as tubulointerstitial fibrosis. Moreover, UFSH decreased cisplatin-induced oxidative stress by increasing the activities of antioxidant enzymes SOD and GSH-Px, while reducing the malondialdehyde (MDA) level in the kidney. Furthermore, UFSH significantly inhibited cisplatin-induced increases in inflammatory cytokines, including Interleukin 1-beta (IL-1β), Interleukin-6 (IL-6), and Tumor necrosis factor-alpha (TNF-α). Western blotting revealed that UFSH inhibited the phosphorylation of the inflammation-associated MAPK/NF-κB signaling pathway, lowered the expression of the apoptosis-related protein Bax, and reversed the reduction in the anti-apoptotic Bcl-2 protein. This investigation demonstrated that UFSH can ameliorate cisplatin-induced nephrotoxicity by mitigating oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Susu Tao
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
| | - Yi Qi
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
| | - Jialong Gao
- College of Food Science & Technology, Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Zhanjiang 524088, China;
| | - Huafang Yuan
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Ruimin Wang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Xiaoqin Shen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Gang Wei
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Zhilan Peng
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
- College of Food Science & Technology, Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Zhanjiang 524088, China;
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| |
Collapse
|
8
|
Wang L, Tang Y, Hou T, Gao Y, Ci X, Peng L. Hesperetin alleviates PM2.5-induced lung injury by inhibiting ferroptosis in an Nrf2-dependent manner. Int Immunopharmacol 2025; 148:114123. [PMID: 39884081 DOI: 10.1016/j.intimp.2025.114123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Fine particulate matter (PM2.5) is a global environmental problem that threatens public health because it can induce ferroptosis and cause lung injury. Hesperetin (Hes), a natural compound widely present in fruits and vegetables, can activate nuclear factor erythroid 2-related factor 2 (Nrf2), thereby exerting powerful antioxidant effects. PURPOSE We explored the antioxidant effects of Hes on lung injury caused by PM2.5 exposure. METHODS In vivo, a mouse model of PM2.5-induced lung injury was used to evaluate the protective effect of Hes. In vitro, the effects of Hes on PM2.5-induced ferroptosis were examined in BEAS-2B cells. RESULTS In vivo, Hes activated the Nrf2 signaling pathway and protected lung tissues from damage induced by PM2.5. In vitro, Hes activated Nrf2 by promoting PI3K/AKT phosphorylation and inhibiting PM2.5-induced ferroptosis. However, in siNrf2-treated BEAS-2B cells, the protective effects of Hes were eliminated. In addition, Nrf2-KO mice exhibited more severe lung injury than did wild-type (WT) mice after PM2.5 exposure. Besides, the protective effects of Hes on PM2.5-exposed Nrf2-KO mice were strongly compromised. CONCLUSION Hes activates Nrf2 by promoting PI3K/AKT phosphorylation to exert a protective effect on PM2.5-induced ferroptosis and lung injury.
Collapse
Affiliation(s)
- Lu Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021 China
| | - Ying Tang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021 China
| | - Tianhua Hou
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021 China
| | - Yun Gao
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021 China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130001 China.
| | - Liping Peng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021 China.
| |
Collapse
|
9
|
Zhou Y, Wan Z, Xiong D, Gong Z, Liu F. Nur77 Promotes Inflammation in Cisplatin-Induced Acute Kidney Injury Through Transactivation of SERPINA3 Mediating Wnt/β-Catenin Pathway. Nephrology (Carlton) 2025; 30:e70006. [PMID: 39957271 DOI: 10.1111/nep.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/16/2025] [Accepted: 02/01/2025] [Indexed: 02/18/2025]
Abstract
AIM Acute kidney injury (AKI) is the most common complication in the treatment of cisplatin, which is a clinically effective and classical anticancer drug. Orphan Nuclear Receptor Nur77 has been found to promote renal ischaemia-reperfusion injury. In this study, we aim to explore the effects of Nur77 on cisplatin-induced AKI (CI-AKI) and its underlying mechanism. METHODS HK-2 cells treated with cisplatin were used to construct the CI-AKI model in vitro. Cell viability and cell proliferation were analysed using CCK-8 and EdU assays, respectively. Cell apoptosis was analysed by flow cytometry. The inflammation release level was detected using ELISA. Molecular abundance was evaluated using qPCR, Western blot and immunofluorescence. The interaction between Nur77 and SERPINA3 was clarified using ChIP and dual-luciferase reporter gene assays. RESULTS Our works demonstrated that Nur77 and SERPINA3 expression were considerably ascended in cisplatin-induced HK-2 cells. The silence of SERPINA3 alleviated cisplatin-stimulated HK-2 cell injury, which was characterised by increased cell viability and proliferation, and decreased apoptosis and inflammatory cytokine release. In addition, Nur77 promotes SERPINA3 transcription by binding to the SERPINA3 promoter region (-182 to -175), thereby upregulating SERPINA3 expression and activating the Wnt/β-catenin pathway. Moreover, HK-2 cell injury induced by cisplatin was notably inhibited by the knockdown of Nur77. Furthermore, the efficacy of Nur77 downregulation on the cell injury in cisplatin-stimulated HK-2 cells was antagonised by SERPINA3 overexpression. CONCLUSION Taken together, our findings revealed that Nur77 knockdown resisted cisplatin-induced HK-2 cells injury through lessening the expression of SERPINA3 mediated by transcriptional regulation and inactivating the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Ying Zhou
- Hemodialysis Room, The First Hospital of Nanchang, Nanchang, China
| | - Zhen Wan
- Hemodialysis Room, The First Hospital of Nanchang, Nanchang, China
| | - Di Xiong
- Hemodialysis Room, The First Hospital of Nanchang, Nanchang, China
| | - Zhijun Gong
- Hemodialysis Room, The First Hospital of Nanchang, Nanchang, China
| | - Feiyan Liu
- Hemodialysis Room, The First Hospital of Nanchang, Nanchang, China
| |
Collapse
|
10
|
Karunakar KK, Edwin ER, Gopalakrishnan M, Cheriyan BV, Ramaiyan V, Karthikha VS, Justin JP. Advances in nephroprotection: the therapeutic role of selenium, silver, and gold nanoparticles in renal health. Int Urol Nephrol 2025; 57:479-510. [PMID: 39312019 DOI: 10.1007/s11255-024-04212-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 01/29/2025]
Abstract
Renal toxicity is a disorder that causes considerable issues in healthcare systems world, highlighting the critical importance of creating alternative treatments. Metallic nanoparticles have recently emerged as promising therapeutic agents for nephroprotection because of their remarkable properties. Numerous disciplines, including medicine, biotechnology, and the food industry, are currently investigating and exploring metallic nanoparticles, such as selenium, silver, and gold, with promising outcomes. In this overview, we provide the most current findings on cutting-edge nephroprotection through metallic nanoparticles, especially selenium, silver, and gold nanoparticles. While outlining the benefits, we outline possible methods for developing metallic nanoparticles, characterization techniques, and nephroprotection therapies. Selenium nanoparticles (SeNPs) minimize oxidative stress, a primary cause of nephrotoxicity through cell regeneration which protects kidneys. Silver nanoparticles (AgNPs) have anti-inflammatory capabilities that help alleviate kidney damage and nephrotoxicity. Gold nanoparticles (AuNPs), which are biocompatible and immune-modifying, reduce inflammation and promote renal cell regeneration, indicating nephroprotective advantages. Renal protection via the use of metallic nanoparticles represents a promising new frontier in the fight against kidney disease and other renal disorders. Metallic nanoparticles of selenium, silver, and gold can protect the kidneys by lowering oxidative stress, reducing inflammation, and improving cell repair. Through their mechanisms, these nanoparticles effectively safeguard and repair kidney function, making them suitable for treating renal diseases. The potential applications of selenium, silver, and gold nanoparticles, as well as their complex modes of action and renal penetration, provide fresh hope for improving renal health and quality of life in patients with kidney disease. The current study highlights therapeutic ability, stability, nephroprotection, and toxicity profiles, as well as the importance of continuous research in this dynamic and evolving field.
Collapse
Affiliation(s)
- Karthik K Karunakar
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Elizabeth Rani Edwin
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Meenaloshini Gopalakrishnan
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Binoy Varghese Cheriyan
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India.
| | - Velmurugan Ramaiyan
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - V S Karthikha
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Jerry Peliks Justin
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| |
Collapse
|
11
|
Bian Y, Dong J, Zhou Z, Zhou H, Xu Y, Zhang Q, Chen C, Pi J. The spatiotemporal and paradoxical roles of NRF2 in renal toxicity and kidney diseases. Redox Biol 2025; 79:103476. [PMID: 39724848 PMCID: PMC11732127 DOI: 10.1016/j.redox.2024.103476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Over 10% of the global population is at risk to kidney disorders. Nuclear factor erythroid-derived 2-related factor 2 (NRF2), a pivotal regulator of redox homeostasis, orchestrates antioxidant response that effectively counters oxidative stress and inflammatory response in a variety of acute pathophysiological conditions, including acute kidney injury (AKI) and early stage of renal toxicity. However, if persistently activated, NRF2-induced transcriptional cascade may disrupt normal cell signaling and contribute to numerous chronic pathogenic processes such as fibrosis. In this concise review, we assembled experimental evidence to reveal the cell- and pathophysiological condition-specific roles of NRF2 in renal chemical toxicity, AKI, and chronic kidney disease (CKD), all of which are closely associated with oxidative stress and inflammation. By incorporating pertinent research findings on NRF2 activators, we dissected the spatiotemporal roles of NRF2 in distinct nephrotoxic settings and kidney diseases. Herein, NRF2 exhibits diverse expression patterns and downstream gene profiles across distinct kidney regions and cell types, and during specific phases of nephropathic progression. These changes are directly or indirectly connected to altered antioxidant defense, damage repair, inflammatory response, regulated cell death and fibrogenesis, culminating ultimately in either protective or deleterious outcomes. The spatiotemporal and paradoxical characteristics of NRF2 in mitigating nephrotoxicity suggest that translational application of NRF2 activation strategy for prevention and interventions of kidney injury are unlikely to be straightforward - right timing and spatial precision must be taken into consideration.
Collapse
Affiliation(s)
- Yiying Bian
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China.
| | - Jize Dong
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Zhengsheng Zhou
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Hua Zhou
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), China; Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, China
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, GA, 30322, USA
| | - Chengjie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China.
| | - Jingbo Pi
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
12
|
Hashimoto K, Yoshida Y, Kamesawa M, Yazawa N, Tominaga H, Aisyah R, Chen S, Bumrungkit C, Kawamoto S, Kumrungsee T, Yanaka N. Glucosyl Hesperidin Supplementation Prevents Tubulointerstitial Fibrosis and Immune Activation in Diabetic Nephropathy in Mice. Nutrients 2025; 17:383. [PMID: 39940240 PMCID: PMC11820413 DOI: 10.3390/nu17030383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a serious condition that can result in end-stage renal failure. Recent evidence has focused on the dietary effects of polyphenols on blood glucose levels and the complications of diabetes. OBJECTIVES In this study, we investigated the protective effect of glucosyl hesperidin (G-Hes), composed of glucose and hesperidin, against streptozotocin (STZ)-induced nephropathy in mice. METHODS We used an STZ-induced diabetic mouse model to investigate the preventive effect of G-Hes on renal pathology. After G-Hes supplementation for 4 weeks, we investigated the renal gene expression profiles using DNA microarray analysis and renal histology to examine the underlying molecular mechanism. RESULTS G-Hes suppressed the increase in kidney weight without any change in the blood glucose levels. This study identified 511 genes whose expression levels were substantially increased during DN development but were downregulated by G-Hes supplementation. G-Hes prevented mRNA expression associated with renal tubule injury, fibrosis, and immune responses. Notably, G-Hes supplementation considerably decreased the complement component C3 at the mRNA and protein levels in the glomeruli and ameliorated glomerular and mesangial matrix expansion in diabetic nephropathy. CONCLUSIONS G-Hes supplementation is useful in preventing tubulointerstitial fibrosis and inflammation in a mouse model of DN, without exhibiting a hypoglycemic effect.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Noriyuki Yanaka
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8528, Japan (S.K.); (T.K.)
| |
Collapse
|
13
|
Xia L, Yuan H, Gao Z, Lv Y, Xu L, Hu F. The role of mitochondrial reactive oxygen species in initiating mitochondrial damage and inflammation in wasp-venom-induced acute kidney injury. J Toxicol Pathol 2025; 38:17-26. [PMID: 39839726 PMCID: PMC11745504 DOI: 10.1293/tox.2024-0046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 08/02/2024] [Indexed: 01/23/2025] Open
Abstract
Acute kidney injury induced by stings from multiple wasps is a medical emergency and is a driving factor of acute renal dysfunction. Numerous studies have shown that mitochondrial reactive oxygen species (mtROS) play a key role in ischemia-reperfusion injury-, cisplatin-, and sepsis-induced acute kidney injury. However, the role of mtROS and its underlying mechanisms in wasp-venom-induced acute kidney injury remain inconclusive. In this study, we investigated the role and mechanisms of mtROS in mitochondrial damage and inflammation in a mouse model of acute kidney injury induced using wasp venom. Changes in mitochondrial function, transcription factor A (TFAM) expression, and DNA maintenance levels, renal function, stimulator of interferon gene (STING) expression, and inflammatory mediator levels in model mice with or without the mtROS scavenger Mito-Tempo were analyzed in vivo. Downregulation of mtROS levels reversed renal damage and mitochondrial dysfunction, and reduced STING expression and inflammation in the kidneys of model mice. The suppression of mtROS levels also improved the decrease in TFAM levels and mitochondrial DNA copy numbers in the kidneys of the model mice. In summary, the existing evidence in this study shows that mtROS contribute significantly to mitochondrial damage and inflammation in acute kidney injury induced by wasp venom.
Collapse
Affiliation(s)
- Lingya Xia
- School of Medicine, Wuhan University of Science and
Technology, Wuhan 430065, China
- Department of Nephrology, Xiangyang Central Hospital,
Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China
| | - Hai Yuan
- Department of Nephrology, Xiangyang Central Hospital,
Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China
| | - Zhao Gao
- Department of Nephrology, Xiangyang Central Hospital,
Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China
| | - Ying Lv
- Department of Nephrology, Xiangyang Central Hospital,
Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China
| | - Liang Xu
- Department of Nephrology, Xiangyang Central Hospital,
Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China
| | - Fengqi Hu
- Department of Nephrology, Xiangyang Central Hospital,
Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China
| |
Collapse
|
14
|
Wang Y, Xue P, Gao L, Wang X, Zhou S, Wu X, Guo C. Improved bioavailability of polydatin and its protective effect against cisplatin induced nephrotoxicity through self-assembled fucoidan and carboxymethyl chitosan delivery system. Int J Biol Macromol 2025; 287:138577. [PMID: 39657878 DOI: 10.1016/j.ijbiomac.2024.138577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/26/2024] [Accepted: 12/07/2024] [Indexed: 12/12/2024]
Abstract
Cisplatin induced acute kidney injury (AKI) is clinically prevalent, with a complex pathogenesis and a lack of effective therapeutic drugs. Polydatin (Po) has excellent biological activity, but its low solubility and bioavailability limit its application. In this study, fucoidan (Fu) and carboxymethyl chitosan (Cs) self-assembled into nanoparticles through electrostatic interactions/hydrogen bonding and loaded Po (Fu/Cs Po NPs). In vitro studies found that Fu/Cs Po NPs protected human renal tubular epithelial (HK-2) cells from cisplatin induced damage and accumulation of reactive oxygen species (ROS). Mechanistic studies showed that Fu/Cs Po NPs inhibited cisplatin induced DNA damage and activation of cyclic guanosine monophosphate synthase (cGAS) and intron gene stimulator (STING) pathways. In vivo studies showed that Fu/Cs Po NPs treatment alleviated cisplatin induced AKI symptoms, including elevated blood urea nitrogen (BUN) and serum creatinine (SCr), as well as pathological damage to kidney tissues. In vivo mechanism studies also showed that Fu/Cs Po NPs treatment inhibited cisplatin induced DNA damage and activation of the cGAS-STING pathway. The pharmacokinetic and tissue distribution results demonstrated that the Fu/Cs delivery system enhanced the bioavailability and kidney accumulation of Po in vivo. In summary, our study provided potential drugs for the treatment of cisplatin induced AKI.
Collapse
Affiliation(s)
- Yinghan Wang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Pengyu Xue
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Liang Gao
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xuefei Wang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Shilin Zhou
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiaochen Wu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Chuanlong Guo
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| |
Collapse
|
15
|
Pan X, Zhu R, Peng J, Liu H, Pan W, Jin Y, Pei J, Zhang L. Molecular mechanisms and potential targets of lycopene for alleviating renal ischemia-reperfusion injury revealed by network pharmacology and animal experiments. Int Immunopharmacol 2024; 143:113421. [PMID: 39442187 DOI: 10.1016/j.intimp.2024.113421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE Renal IRI is one of the leading causes of AKI. How to effectively mitigate renal IRI is important for the recovery of renal function. The regulatory mechanism of lycopene, a natural antioxidant, in renal IRI is currently unknown. Therefore, we utilized network pharmacology and animal experiments to explore the possible mechanisms and potential targets of lycopene for alleviating renal IRI. METHODS We obtained lycopene-regulated genes and renal IRI-related genes from the CTD database and GeneCards database, respectively. Subsequently, the two were intersected and the intersecting genes we defined as lycopene-regulated genes in renal IRI. Next, we explored their potential biological functions and mechanisms through enrichment analysis. Meanwhile, we constructed a rat renal IRI model and validated the protective effects of lycopene and related mechanisms. To further explore the Hub genes regulated by lycopene, we constructed a PPI protein interactions network and characterized the Hub genes using Cytoscape software. We also verified the expression of Hub genes using animal experiments and molecular docking techniques. Finally, we constructed TF-Hub gene and miRNA-Hub gene regulatory networks. RESULTS We obtained a total of 255 lycopene-regulated genes and 327 renal IRI-related genes. The enrichment analysis revealed that they were closely related to the regulation of oxidative stress as well as the regulation of inflammatory factors. At the same time, the MAPK signaling pathway was significantly enriched. Next, we found in animal experiments that lycopene significantly alleviated the level of oxidative stress and inflammation during renal IRI, and had a protective effect on kidney damage. Also, we found that this protective effect may be achieved by inhibiting the MAPK signaling pathway. Next, we identified a total of five Hub genes using Cytoscape software: TNF, AKT1, MAPK3, IL6 and CASP3. Both animal experiments and molecular docking techniques demonstrated that lycopene can effectively regulate the expression of Hub genes. Finally, our constructed TF-Hub gene and miRNA-Hub gene regulatory network provide a theoretical basis for further regulation of Hub genes in follow-up. CONCLUSIONS This study suggests that lycopene is a promising option in mitigating renal IRI. Lycopene may exert protective effects by inhibiting the MAPK signaling pathway.
Collapse
Affiliation(s)
- Xingyu Pan
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Rong Zhu
- Department of Pediatric Surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Jinpu Peng
- Department of Pediatric Surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Hongli Liu
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Wenqing Pan
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Yuhan Jin
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Jun Pei
- Department of Pediatric Surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| | - Li Zhang
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China.
| |
Collapse
|
16
|
Xu N, Mu R, Deng S, Han Y, Shi Y, Fu X, Li H, Yao Q. Reserpine alleviates cisplatin-induced acute kidney injury via anti-ferroptosis and cGAS/STING pathway. Ren Fail 2024; 46:2406395. [PMID: 39377110 PMCID: PMC11463010 DOI: 10.1080/0886022x.2024.2406395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/03/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024] Open
Abstract
Cisplatin plays a pivotal role in the chemotherapy treatment of various cancers, but its use is often limited due to its nephrotoxic side effects. Identifying compounds that can mitigate cisplatin-induced nephrotoxicity is therefore of great importance. This study focused on evaluating the protective effects of reserpine against cisplatin-induced acute kidney injury. Reserpine was found to significantly safeguard against kidney damage caused by cisplatin, as indicated by the decreased levels of serum creatinine, blood urea nitrogen, and lactate dehydrogenase induced by cisplatin. Moreover, reserpine improved kidney histology damage caused by cisplatin treatment, with hematoxylin-eosin and periodic acid-Schiff staining revealing notable recovery from renal injury. Mechanistically, reserpine mitigated oxidative stress triggered by cisplatin and exhibits the ability to inhibit ferroptosis both in vivo and in vitro. Additionally, reserpine blocked the activation of the cGAS/STING signaling pathway and the subsequent expression of inflammatory genes, thus reducing inflammation-driven kidney damage. In summary, the findings suggest that reserpine offers a promising new strategy for preventing nephrotoxicity induced by cisplatin.
Collapse
Affiliation(s)
- Nahua Xu
- Zunyi Medical University, Zunyi, China
| | - Rong Mu
- Zunyi Medical University, Zunyi, China
| | - Siyuan Deng
- School of Medicine, Chongqing University, Chongqing, China
| | - Ye Han
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children’s Hospital of Chongqing Medical University), Chongqing, China
| | - Yanyun Shi
- Medical College of Guizhou University, Guiyang, China
| | - Xuemei Fu
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children’s Hospital of Chongqing Medical University), Chongqing, China
| | - Hui Li
- Zunyi Medical University, Zunyi, China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children (Women and Children’s Hospital of Chongqing Medical University), Chongqing, China
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qi Yao
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
17
|
Song B, Hao M, Zhang S, Niu W, Li Y, Chen Q, Li S, Tong C. Comprehensive review of Hesperetin: Advancements in pharmacokinetics, pharmacological effects, and novel formulations. Fitoterapia 2024; 179:106206. [PMID: 39255908 DOI: 10.1016/j.fitote.2024.106206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/01/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024]
Abstract
Hesperetin is a flavonoid compound naturally occurring in the peel of Citrus fruits from the Rutaceae family. Previous studies have demonstrated that hesperetin exhibits various pharmacological effects, such as anti-inflammatory, anti-tumor, antioxidative, anti-aging, and neuroprotective properties. In recent years, with the increasing prevalence of diseases and the rising awareness of traditional Chinese medicine, hesperetin has garnered growing attention for its wide-ranging pharmacological effects. To substantiate its health benefits and elucidate potential mechanisms, knowledge of pharmacokinetics is crucial. However, the limited solubility of hesperetin restricts its bioavailability, thereby diminishing its efficacy as a beneficial health agent. To enhance the bioavailability of hesperetin, various novel formulations have been developed, including nanoparticles, liposomes, and cyclodextrin inclusion complexes. This article reviews recent advances in the pharmacokinetics of hesperetin and methods to improve its bioavailability, as well as its pharmacological effects and mechanisms, aiming to provide a theoretical basis for clinical applications.
Collapse
Affiliation(s)
- Bocui Song
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, China.
| | - Meihan Hao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Shuang Zhang
- Administration Committee of Jilin Yongji Economic Development Zone, Jilin, Jilin, China
| | - Wenqi Niu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Yuqi Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Qian Chen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Shuang Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Chunyu Tong
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China.
| |
Collapse
|
18
|
Lou J, Wu F, He W, Hu R, Cai Z, Chen G, Zhao W, Zhang Z, Si Y. Hesperidin activates Nrf2 to protect cochlear hair cells from cisplatin-induced damage. Redox Rep 2024; 29:2341470. [PMID: 38629504 PMCID: PMC11025410 DOI: 10.1080/13510002.2024.2341470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024] Open
Abstract
Cisplatin is widely employed in clinical oncology as an anticancer chemotherapy drug in clinical practice and is known for its severe ototoxic side effects. Prior research indicates that the accumulation of reactive oxygen species (ROS) plays a pivotal role in cisplatin's inner ear toxicity. Hesperidin is a flavanone glycoside extracted from citrus fruits that has anti-inflammatory and antioxidant effects. Nonetheless, the specific pharmacological actions of hesperidin in alleviating cisplatin-induced ototoxicity remain elusive. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a critical mediator of the cellular oxidative stress response, is influenced by hesperidin. Activation of Nrf2 was shown to have a protective effect against cisplatin-induced ototoxicity. The potential of hesperidin to stimulate Nrf2 in attenuating cisplatin's adverse effects on the inner ear warrants further investigation. This study employs both in vivo and in vitro models of cisplatin ototoxicity to explore this possibility. Our results reveal that hesperidin mitigates cisplatin-induced ototoxicity by activating the Nrf2/NQO1 pathway in sensory hair cells, thereby reducing ROS accumulation, preventing hair cell apoptosis, and alleviating hearing loss.
Collapse
Affiliation(s)
- Jintao Lou
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Fan Wu
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Wuhui He
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Rui Hu
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Ziyi Cai
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Guisheng Chen
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Wenji Zhao
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Zhigang Zhang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yu Si
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
19
|
Zheng J, Wang Q, Xu K, Ma M, Wang Z, Sun Z, Yang S, Wang X, Yan N, Duan X. Fluoride induces immune-inflammatory disorder in the kidneys via histone lysine crotonylation in vivo. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117385. [PMID: 39581112 DOI: 10.1016/j.ecoenv.2024.117385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Fluoride is an essential trace element for human. Adequate levels of fluoride are crucial for maintaining skeletal growth, but excessive fluoride exposure entering the body can cause renal damage, including damaged renal tubules and impaired renal function. However, the mechanism on fluoride-induced kidney injury remains unclear. This study aimed to explore the immune-inflammatory imbalance induced by fluoride and its possible mechanism in the kidneys. Mice were exposed to sodium fluoride (NaF) (0, 25, 50 and 100 mg/L) for five months. The results showed that NaF increased the renal weight and renal index. The NaF-treated groups exhibited higher serum creatinine (Cre), blood urea nitrogen (BUN), albumin (ALB) total protein (TP) levels. Further, NaF increased reactive oxygen species (ROS) levels, lipid peroxidation (LPO) levels and malondialdehyde (MDA) level. Superoxide dismutase (SOD) activity was reduced and glutathione (GSH) activities were reduced in fluoride-treated group. NaF treatment also downregulated the nuclear factor E2-related factor (Nrf2) protein and its downstream enzymes heme oxygenase-1 (HO-1) and NAD(P)H: Quinone Oxidoreductase 1(NQO1) in the kidneys. Further, NaF shifted Th1/Th2 balance toward Th1 bias. Similarly, NaF exhibited increased macrophages and augmented M1 differentiation but suppressed M2 differentiation. The renal inflammatory response was also induced by fluoride via activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome and increase of the pro-inflammatory factors tumour necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β), interleukin-6 (IL-6) and interleukin-18 (IL-18). In addition, NaF treatment reduced the expression of the histone 2B lysine 12 crotonylation (H2BK12cr) and H4K8cr proteins as well as decreased the histone acetyltransferase P300 protein. NaF incresed the protein expression of histone decrotonylation enzyme sirtuin1 (sirt1) and histone deacetylase 3 (HDAC3) and upregulated HDAC2 protein. These findings demonstrate that fluoride exposure induces renal dysfunction and oxidative injury, affects M1/M2 polarization and Th1/Th2 differentiation, and promotes the inflammatory response via histone lysine crotonylation, ultimately resulting in nephrotoxicity.
Collapse
Affiliation(s)
- Jingwen Zheng
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang 110034, PR China
| | - Qian Wang
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang 110034, PR China
| | - Kangjie Xu
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang 110034, PR China
| | - Mingyue Ma
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang 110034, PR China
| | - Zhengdong Wang
- Department of Human Anatomy, Shenyang Medical College, Shenyang 110034, PR China
| | - Zhenxiang Sun
- Department of Human Anatomy, Shenyang Medical College, Shenyang 110034, PR China
| | - Shuang Yang
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang 110034, PR China
| | - Xinyue Wang
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang 110034, PR China
| | - Nan Yan
- Department of Medical Applied Technology, Shenyang Medical College, Shenyang 110034, PR China.
| | - Xiaoxu Duan
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang 110034, PR China.
| |
Collapse
|
20
|
Giri D, Dey SK, Manna S, Das Chaudhuri A, Mahata R, Pradhan A, Roy T, Jana K, Das S, Roy S, Maiti Choudhury S. Nanoconjugate Carrying pH-Responsive Transferrin Receptor-Targeted Hesperetin Triggers Triple-Negative Breast Cancer Cell Death through Oxidative Attack and Assemblage of Pro-Apoptotic Proteins. ACS APPLIED BIO MATERIALS 2024; 7:7556-7573. [PMID: 39504304 DOI: 10.1021/acsabm.4c01131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Triple-negative breast cancer (TNBC) is recognized as a major aggressive subtype of breast cancer due to its expeditious worsening growth, extensive metastatic capability, and recalcitrance to standard current treatments. Hesperetin (HSP), a natural bioflavonoid from citrus fruits, demonstrates pronounced anticancer efficacy, but its hydrophobicity limits its clinical development. The present study reports the fabrication of a biocompatible and pH-responsive transferrin (TF) receptor-targeted HSP-loaded poly(lactic-co-glycolic acid) (PLGA) nanobioconjugate (PLGA-HSP-TF NPs) and the exploration of its in vitro and in vivo antineoplastic potential. PLGA nanoparticles (NPs), PLGA-HSP NPs, and PLGA-HSP-TF NPs were synthesized and characterized by DLS, FTIR, FE-SEM, and 1H NMR spectroscopy. The stability and in vitro release profile of nanoparticles were inspected, and anticancer efficacy was scrutinized in terms of in vitro cytotoxicity, oxidative stress and apoptosis biomarkers, and cell cycle arrest. In vivo tumor regression and host survival studies were executed in Ehrlich ascites carcinoma (EAC) cell-bearing Swiss albino mice. The drug uptake of highly stable PLGA-HSP-TF NPs was accomplished effectively in MDA-MB-231 cells and showed the pH-dependent intracellular release of HSP, which generated excessive intracellular reactive oxygen species (ROS) that led to oxidative assault to the TNBC cells. This elevated ROS dropped the mitochondrial membrane potential and triggered apoptosis-mediated cell death by arresting the cell cycle at the G0/G1 phase. Furthermore, PLGA-HSP-TF NPs unveiled significant in vivo Ehrlich ascites carcinoma regression and host survival compared to free HSP with minimum toxicity at a minimum dose of 20 mg/kg body weight. The study divulges that PLGA-HSP-TF NPs may be an astounding anticancer nanocandidate for aggressive triple-negative breast cancer therapy.
Collapse
Affiliation(s)
- Dibyendu Giri
- Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India, 721102
- Department of Physiology, Ghatal Rabindra Satabarsiki Mahavidyalaya, Ghatal, Paschim Medinipur, West Bengal, India, 721212
| | - Surya Kanta Dey
- Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India, 721102
| | - Sounik Manna
- Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India, 721102
| | - Angsuman Das Chaudhuri
- Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India, 721102
| | - Rumi Mahata
- Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India, 721102
| | - Ananya Pradhan
- Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India, 721102
| | - Tamanna Roy
- Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India, 721102
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, P1/12 CIT scheme VIIM, Kolkata, West Bengal, India, 700054
| | - Subhasis Das
- Department of Surgery, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Sumita Roy
- Department of Chemistry and Chemical Technology, Vidyasagar University, Midnapore, West Bengal, India, 721102
| | - Sujata Maiti Choudhury
- Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India, 721102
| |
Collapse
|
21
|
Baig SI, Naseer M, Munir AR, Ali Y, Razzaq MA. Immunoinformatic-based drug design utilizing hesperetin to target CISD2 activation for liver aging in humans. Biogerontology 2024; 25:1189-1213. [PMID: 39196437 DOI: 10.1007/s10522-024-10130-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024]
Abstract
The CISD protein family, consisting of CISD1, CISD2, and CISD3, encodes proteins that feature CDGSH iron-sulfur domains crucial for cellular functions and share a common 2Fe-2S domain. CISD2, which is pivotal in cells, regulates intracellular calcium levels, maintains the endoplasmic reticulum and mitochondrial function, and is associated with longevity and overall health, with exercise stimulating CISD2 production. However, CISD2 expression decreases with age, impacting age-related processes. According to in silico docking, HST is a CISD2 activator that affects metabolic dysfunction and age-related illnesses by affecting metabolic pathways. This study investigated the ability of CISD2 and HST to reduce age-related ailments, with a particular emphasis on liver aging. CISD2 deficiency has a major effect on the function of cells, as it undermines the integrity of the ER, mitochondria, and calcium homeostasis. It also increases susceptibility to oxidative stress and metabolic dysregulation, which is linked to Wolfram syndrome and exacerbates age-related illnesses and metabolic disorders. By shielding cells from stress, CISD2 extends the life of cells and maintains liver health as people age. Its protective effecfts on the liver during aging are further enhanced by its control of translation factors such as Nrf2 and IL-6. This work paves the way for future investigations and clinical applications by examining the structural and functional properties of CISD2 and the interaction between CISD2 and HST. This highlights the therapeutic potential of these findings in promoting healthy livers in humans and battling age-related illnesses.
Collapse
Affiliation(s)
- Saad Ilyas Baig
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan.
| | - Maria Naseer
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan
| | - Abdur-Rehman Munir
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan
| | - Yasir Ali
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan
| | | |
Collapse
|
22
|
Zhao Y, Song JY, Feng R, Hu JC, Xu H, Ye ML, Jiang JD, Chen LM, Wang Y. Renal Health Through Medicine-Food Homology: A Comprehensive Review of Botanical Micronutrients and Their Mechanisms. Nutrients 2024; 16:3530. [PMID: 39458524 PMCID: PMC11510533 DOI: 10.3390/nu16203530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/18/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND As an ancient concept and practice, "food as medicine" or "medicine-food homology" is receiving more and more attention these days. It is a tradition in many regions to intake medicinal herbal food for potential health benefits to various organs and systems including the kidney. Kidney diseases usually lack targeted therapy and face irreversible loss of function, leading to dialysis dependence. As the most important organ for endogenous metabolite and exogenous nutrient excretion, the status of the kidney could be closely related to daily diet. Therefore, medicinal herbal food rich in antioxidative, anti-inflammation micronutrients are ideal supplements for kidney protection. Recent studies have also discovered its impact on the "gut-kidney" axis. METHODS Here, we review and highlight the kidney-protective effects of botanicals with medicine-food homology including the most frequently used Astragalus membranaceus and Angelica sinensis (Oliv.) Diels, concerning their micronutrients and mechanism, offering a basis and perspective for utilizing and exploring the key substances in medicinal herbal food to protect the kidney. RESULTS The index for medicine-food homology in China contains mostly botanicals while many of them are also consumed by people in other regions. Micronutrients including flavonoids, polysaccharides and others present powerful activities towards renal diseases. CONCLUSIONS Botanicals with medicine-food homology are widely speeded over multiple regions and incorporating these natural compounds into dietary habits or as supplements shows promising future for renal health.
Collapse
Affiliation(s)
- Yi Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jian-Ye Song
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ru Feng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jia-Chun Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hui Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Meng-Liang Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jian-Dong Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Li-Meng Chen
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
23
|
Xiao L, Zhao M, Linghu KG, Wu G, Zhang T, Chen C, Guan J, Cao Z, Hu Y, Yu H. Ganweikang extract protects hepatocytes from oxidative injury by activating Nrf2/HO-1 and MAPKs pathways. Fitoterapia 2024; 178:106146. [PMID: 39089591 DOI: 10.1016/j.fitote.2024.106146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Ganweikang tablet (GWK) is a traditional Chinese prescription and has been clinically used in treating liver diseases for decades. Although GWK has been shown to exert potential therapeutic effect for hepatotoxicity protection, the underlying biological mechanisms are still not well clarified. In the present study, the compositional analysis of GWK was performed by HPLC analysis, and the hepato-protective effects of GWK were assessed in H2O2-stimulated acute oxidative injured HL-7702 hepatocytes in vitro. As a result, 7 components in GWK were quantified to be 0.06 ± 0.01% (calycosin), 0.46 ± 0.02% (calycosin-7-glucoside), 0.13 ± 0.01% (liquiritin), 0.17 ± 0.02% (glycyrrhizic acid), 0.45 ± 0.02% (forsythoside A), 0.07 ± 0.01% (5-O-methylvisammioside) and 0.45 ± 0.02% (forsythin), respectively. Furthermore, GWK (100, 200 and 400 μg/mL, 24 h) dose-dependently alleviated HL-7702 hepatocytes from H2O2 (200 μM, 2 h)-induced cell apoptosis by decreasing the intracellular reactive oxygen species (ROS) generation and malondialdehyde (MDA) level, as well as the cellular aminotransferases (ALT and AST) activities. GWK increased the expressions of HO-1, NQO1 and Nrf2, while suppressing the expression of KEAP1 in H2O2-stimulated HL-7702 cells. A specific Nrf2 inhibitor, ML385, was further employed to investigate the regulation of Nrf2 in HL-7702 cells stimulated by H2O2. In addition, the activation of MAPKs (JUN, ERK and p38) was simultaneously detected in H2O2-stimulated HL-7702 cells. In conclusion, GWK exerted potential therapeutic effect to protect hepatocytes from acute oxidative injury through activating the Nrf2/HO-1 and MAPKs pathways.
Collapse
Affiliation(s)
- Linxuan Xiao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Mingming Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Ke-Gang Linghu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Guoping Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Tian Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Chengyu Chen
- Jiaheng Pharmaceutical Technology Co., Ltd, Zhuhai 519000, China
| | - Jianli Guan
- Henan Fusen Pharmaceutical Co., Ltd, Nanyang 473000, China
| | - Zhiming Cao
- Jiaheng Pharmaceutical Technology Co., Ltd, Zhuhai 519000, China; Henan Fusen Pharmaceutical Co., Ltd, Nanyang 473000, China
| | - Yuanjia Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Hua Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China.
| |
Collapse
|
24
|
Queirós V, Azeiteiro UM, Santos JL, Alonso E, Soares AMVM, Barata C, Freitas R. Unravelling biochemical responses in the species Mytilus galloprovincialis exposed to the antineoplastics ifosfamide and cisplatin under different temperature scenarios. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 943:173668. [PMID: 38839013 DOI: 10.1016/j.scitotenv.2024.173668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
This study investigates the chronic impact of two of the most widely consumed antineoplastic drugs, Ifosfamide (IF) and Cisplatin (CDDP), on the bivalve species Mytilus galloprovincialis under current (17 °C) and predicted warming conditions (21 °C). Accompanying the expected increase in worldwide cancer incidence, antineoplastics detection in the aquatic environment is also expected to rise. Mussels were exposed to varying concentrations of IF (10, 100, 500 ng/L) and CDDP (10, 100, 1000 ng/L) for 28 days. Biochemical analyses focused on metabolic, antioxidant and biotransformation capacities, cellular damage, and neurotoxicity. Results showed temperature-dependent variations in biochemical responses. Metabolic capacity remained stable in mussels exposed to IF, while CDDP exposure increased it at 1000 ng/L for both temperatures. Antioxidant enzyme activities were unaffected by IF, but CDDP activated them, particularly at 21 °C. Biotransformation capacity was unchanged by IF but enhanced by CDDP. Nevertheless, cellular damage occurred at CDDP concentrations above 100 ng/L, regardless of temperature. Integrated biomarker responses highlighted CDDP's greater impact, emphasizing the critical role of temperature in shaping organismal responses and underscoring the complexity of environmental stressor interactions.
Collapse
Affiliation(s)
- Vanessa Queirós
- Centre for Environmental and Marine Studies (CESAM) and Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Ulisses M Azeiteiro
- Centre for Environmental and Marine Studies (CESAM) and Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Juan Luis Santos
- Departamento de Química Analítica, Escuela Politécnica Superior, Universidad de Sevilla, C/ Virgen de África 7, 41011 Sevilla, Spain
| | - Esteban Alonso
- Departamento de Química Analítica, Escuela Politécnica Superior, Universidad de Sevilla, C/ Virgen de África 7, 41011 Sevilla, Spain
| | - Amadeu M V M Soares
- Centre for Environmental and Marine Studies (CESAM) and Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Carlos Barata
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research, IDAEA-CSIC, C/ Jordi Girona 18, 08034 Barcelona, Spain
| | - Rosa Freitas
- Centre for Environmental and Marine Studies (CESAM) and Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
25
|
Lu C, Yang W, Chu F, Wang S, Ji Y, Liu Z, Yu H, Qin S, Sun D, Jiao Z, Sun H. Hesperetin Attenuates T-2 Toxin-Induced Chondrocyte Injury by Inhibiting the p38 MAPK Signaling Pathway. Nutrients 2024; 16:3107. [PMID: 39339707 PMCID: PMC11434908 DOI: 10.3390/nu16183107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/01/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Hesperetin, a flavonoid derived from citrus fruits, exhibits potent antioxidant and anti-inflammatory activities and has been implicated in cartilage protection. However, its effectiveness against T-2 toxin-induced knee cartilage damage remains unclear. METHODS In this study, high-throughput sequencing analysis was employed to identify the key signaling pathways involved in T-2 toxin-induced articular cartilage damage in rats. Animal models were divided into the following groups: control, low-dose T-2 toxin, high-dose T-2 toxin, T-2 toxin + hesperetin, hesperetin, and vehicle. Pathological staining and immunohistochemistry were used to assess pathological changes, as well as the expression levels of the cartilage matrix-related proteins MMP13 and collagen II, along with the activation of the p38 MAPK signaling pathway. Additionally, primary rat chondrocytes were cultured to establish an in vitro model for investigating the underlying mechanism. RESULTS High-throughput sequencing analysis revealed the involvement of the MAPK signaling pathway in T-2 toxin-induced articular cartilage damage in rats. Hesperetin intervention in T-2 toxin-exposed rats attenuated pathological cartilage damage. Immunohistochemistry results demonstrated a significant reduction in collagen II protein expression in the high-dose T-2 toxin group (p < 0.01), accompanied by a significant increase in MMP13 protein expression (p < 0.01). In both the articular cartilage and the epiphyseal plate, the T-2 toxin + hesperetin group exhibited significantly higher collagen II protein expression than the high-dose T-2 toxin group (p < 0.05), along with significantly lower MMP13 protein expression (p < 0.05). Hesperetin inhibited the over-activation of the p38/MEF2C signaling axis induced by T-2 toxin in primary rat chondrocytes. Compared to the T-2 toxin group, the T-2 toxin + hesperetin group showed significantly reduced phosphorylation levels of p38 and protein expression levels of MEF2C (p < 0.001 or p < 0.05). Moreover, the T-2 toxin + hesperetin group exhibited a significant decrease in MMP13 protein expression (p < 0.05) and a significant increase in collagen II protein expression (p < 0.01) compared to the T-2 toxin group. CONCLUSIONS T-2 toxin activates the p38 MAPK signaling pathway, causing knee cartilage damage in rats. Treatment with hesperetin inhibits the p38/MEF2C signaling axis, regulates collagen II and MMP13 protein expression, and reduces cartilage injury significantly.
Collapse
Affiliation(s)
- Chunqing Lu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Wenjing Yang
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Fang Chu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Sheng Wang
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Yi Ji
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
- Institute of Keshan Disease, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China
| | - Zhipeng Liu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Hao Yu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Shaoxiao Qin
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Dianjun Sun
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Zhe Jiao
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
- Institute for Kashin Beck Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China
| | - Hongna Sun
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
26
|
Mokhtar HI, Abd El-Fadeal NM, El-Sayed RM, Hegazy A, El-Kherbetawy MK, Hamad AG, ElSayed MH, Zaitone SA. Computational Analysis and Experimental Data Exploring the Role of Hesperetin in Ameliorating ADHD and SIRT1/Nrf2/Keap1/OH-1 Signaling. Int J Mol Sci 2024; 25:9284. [PMID: 39273230 PMCID: PMC11395673 DOI: 10.3390/ijms25179284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 09/15/2024] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) manifests as poor attention, hyperactivity, as well as impulsive behaviors. Hesperetin (HSP) is a citrus flavanone with strong antioxidant and anti-inflammatory activities. The present study aimed to test hesperetin efficacy in alleviating experimental ADHD in mice and its influence on hippocampal neuron integrity and sirtuin 1 (SIRT1) signaling. An in silico study was performed to test the related proteins. Groups of mice were assigned as control, ADHD model, ADHD/HSP (25 mg/kg), and ADHD/HSP (50 mg/kg). ADHD was induced by feeding with monosodium glutamate (0.4 g/kg, for 8 weeks) and assessed by measuring the motor and attentive behaviors (open filed test, Y-maze test, and marble burying test), histopathological examination of the whole brain tissues, and estimation of inflammatory markers. The in-silico results indicated the putative effects of hesperetin on ADHD by allowing the integration and analysis of large-scale genomic, transcriptomic, and proteomic data. The in vivo results showed that ADHD model mice displayed motor hyperactivity and poor attention in the behavioral tasks and shrank neurons at various hippocampal regions. Further, there was a decline in the mRNA expression and protein levels for SIRT1, the erythroid 2-related factor-2 (Nrf2), kelch like ECH associated protein 1 (Keap1) and hemeoxygenase-1 (OH-1) proteins. Treatment with HSP normalized the motor and attentive behaviors, prevented hippocampal neuron shrinkage, and upregulated SIRT1/Nrf2/Keap1/OH-1 proteins. Taken together, HSP mainly acts by its antioxidant potential. However, therapeutic interventions with hesperetin or a hesperetin-rich diet can be suggested as a complementary treatment in ADHD patients but cannot be suggested as an ADHD treatment per se as it is a heterogeneous and complex disease.
Collapse
Affiliation(s)
- Hatem I. Mokhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University-Kantara Branch, Ismailia 41636, Egypt
| | - Noha M. Abd El-Fadeal
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Rehab M. El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sinai University—Arish Branch, Arish, 45511, Egypt
| | - Ann Hegazy
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | | | - Ahmed G. Hamad
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed H. ElSayed
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo 11757, Egypt
| | - Sawsan A. Zaitone
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| |
Collapse
|
27
|
Zhao X, Meng Y, Dang C, Xue L, Zhang J, Ma S, Li H. VALD-2 mitigates cisplatin-induced acute kidney injury: Mechanistic insights into oxidative stress modulation and inflammation suppression. J Biochem Mol Toxicol 2024; 38:e23786. [PMID: 39072927 DOI: 10.1002/jbt.23786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
This study explores the compelling antitumor properties of VALD-2, a synthetic Schiff base ligand known for its low toxicity. The focus is on investigating VALD-2's protective role against cisplatin-induced acute kidney injury (AKI) in mice, with a specific emphasis on mitigating oxidative stress and inflammation. The study involves daily intraperitoneal injections of amifostine or VALD-2 over 7 days to establish an AKI model. Subsequently, mice were assigned to normal control, cisplatin group, cisplatin + amifostine group, and cisplatin + VALD-2 10 mg/kg group, cisplatin + VALD-2 20 mg/kg, and cisplatin + VALD-2 40 mg/kg. Kidney injury is assessed through serum blood urea nitrogen (BUN) and creatinine (Cr) activity assays. Levels of inflammatory factors, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), in kidney tissue of mice were assessed through enzyme-linked immunosorbent assay (ELISA). The protective effect of VALD-2 is further examined through HE staining to observe pathological changes in kidney injury. The ultrastructural changes of renal cells and tubular epithelial cells were observed by electron microscopy under experimental conditions, indicating the effect of VALD-2 on reversing cisplatin-induced renal injury. The study delves into VALD-2's protective mechanisms against cisplatin-induced kidney injury by using western blot analysis to assess the expression levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and malondialdehyde (MDA) in kidney tissues. VALD-2 demonstrates significant improvement in cisplatin-induced AKI, as evidenced by increased BUN and Cr levels. It effectively protects kidney tissue from oxidative damage, enhancing SOD and GSH-Px activities while reducing MDA levels. The study also reveals a decrease in TNF-α and IL-6 levels, supported by ELISA results, and histological findings confirm anti-nephrotoxic effects. Western blot analysis shows an upregulation of antioxidant enzymes (SOD, GSH-Px) and a reduction in MDA production. VALD-2 emerges as a promising mitigator of cisplatin-induced AKI, showcasing its ability to enhance oxidative stress-related protein expression. The findings suggest VALD-2 as a potential therapeutic agent for protecting against cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Xuhui Zhao
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Yuna Meng
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Chunyan Dang
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Li Xue
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Jing Zhang
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Shuping Ma
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Hongling Li
- Department of Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
28
|
Wang L, Xie Z, Wu M, Chen Y, Wang X, Li X, Liu F. The role of taurine through endoplasmic reticulum in physiology and pathology. Biochem Pharmacol 2024; 226:116386. [PMID: 38909788 DOI: 10.1016/j.bcp.2024.116386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Taurine is a sulfur-containing amino acid found in many cell organelles that plays a wide range of biological roles, including bile salt production, osmoregulation, oxidative stress reduction, and neuromodulation. Taurine treatments have also been shown to ameliorate the onset and development of many diseases, including hypertension, fatty liver, neurodegenerative diseases and ischemia-reperfusion injury, by exerting antioxidant, anti-inflammatory, and antiapoptotic effects. The endoplasmic reticulum (ER) is a dynamic organelle involved in a wide range of cellular functions, including lipid metabolism, calcium storage and protein stabilization. Under stress, the disruption of the ER environment leads to the accumulation of misfolded proteins and a characteristic stress response called the unfolded protein response (UPR). The UPR protects cells from stress and helps to restore cellular homeostasis, but its activation promotes cell death under prolonged ER stress. Recent studies have shown that ER stress is closely related to the onset and development of many diseases. This article reviews the beneficial effects and related mechanisms of taurine by regulating the ER in different physiological and pathological states, with the aim of providing a reference for further research and clinical applications.
Collapse
Affiliation(s)
- Linfeng Wang
- Institute of Microbial Engineering, School of Life Sciences, Henan University, Kaifeng 475004, China; Engineering Research Center for Applied Microbiology of Henan Province, Kaifeng, 475004, China
| | - Zhenxing Xie
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Mengxian Wu
- Institute of Microbial Engineering, School of Life Sciences, Henan University, Kaifeng 475004, China; Engineering Research Center for Applied Microbiology of Henan Province, Kaifeng, 475004, China
| | - Yunayuan Chen
- Institute of Microbial Engineering, School of Life Sciences, Henan University, Kaifeng 475004, China; Engineering Research Center for Applied Microbiology of Henan Province, Kaifeng, 475004, China
| | - Xin Wang
- Institute of Microbial Engineering, School of Life Sciences, Henan University, Kaifeng 475004, China; Engineering Research Center for Applied Microbiology of Henan Province, Kaifeng, 475004, China
| | - Xingke Li
- Institute of Microbial Engineering, School of Life Sciences, Henan University, Kaifeng 475004, China; Engineering Research Center for Applied Microbiology of Henan Province, Kaifeng, 475004, China.
| | - Fangli Liu
- College of Nursing and Health, Henan University, Kaifeng 475004, China.
| |
Collapse
|
29
|
Qiao O, Zhang L, Han L, Wang X, Li Z, Bao F, Hao H, Hou Y, Duan X, Li N, Gong Y. Rosmarinic acid plus deferasirox inhibits ferroptosis to alleviate crush syndrome-related AKI via Nrf2/Keap1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155700. [PMID: 38704914 DOI: 10.1016/j.phymed.2024.155700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/03/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND Myoglobin (Mb) induced death of renal tubular epithelial cells (RTECs) is a major pathological factor in crush syndrome-related acute kidney injury (CS-AKI). It is unclear whether ferroptosis is involved and could be a target for treatment. PURPOSE This study aimed to evaluate the potential therapeutic effects of combining the natural small molecule rosemarinic acid (RA) and the iron chelator deferasirox (Dfe) on CS-AKI through inhibition of ferroptosis. METHODS Sequencing data were downloaded from the GEO database, and differential expression analysis was performed using the R software limma package. The CS-AKI mouse model was constructed by squeezing the bilateral thighs of mice for 16 h with 1.5 kg weight. TCMK1 and NRK-52E cells were induced with 200 μM Mb and then treated with RA combined with Dfe (Dfe + RA, both were 10 μM). Functional and pathological changes in mouse kidney were evaluated by glomerular filtration rate (GFR) and HE pathology. Immunofluorescence assay was used to detect Mb levels in kidney tissues. The expression levels of ACSL4, GPX4, Keap1, and Nrf2 were analyzed by WB. RESULTS We found that AKI mice in the GSE44925 cohort highly expressed the ferroptosis markers ACSL4 and PTGS2. CS-AKI mice showed a rapid decrease in GFR, up-regulation of ACSL4 expression in kidney tissue, and down-regulation of GPX4 expression, indicating activation of the ferroptosis pathway. Mb was found to deposit in renal tubules, and it has been proven to cause ferroptosis in TCMK1 and NRK-52E cells in vitro. We found that Dfe had a strong iron ion scavenging effect and inhibited ACSL4 expression. RA could disrupt the interaction between Keap1 andNrf2, stabilize Nrf2, and promote its nuclear translocation, thereby exerting antioxidant effects. The combination of Dfe and RA effectively reversed Mb induced ferroptosis in RTECs. CONCLUSION In conclusion, we found that RA combined with Dfe attenuated CS-AKI by inhibiting Mb-induced ferroptosis in RTECs via activating the Nrf2/Keap1 pathway.
Collapse
Affiliation(s)
- Ou Qiao
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China
| | - Li Zhang
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China
| | - Lu Han
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China
| | - Xinyue Wang
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China
| | - Zizheng Li
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China
| | - Fengjiao Bao
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China
| | - Herui Hao
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China
| | - Yingjie Hou
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China
| | - Xiaohong Duan
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China
| | - Ning Li
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China; Key Laboratory for Disaster Medicine Technology, Tianjin, China.
| | - Yanhua Gong
- Medical School, Faculty of Medicine, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Institute of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University, China; Key Laboratory for Disaster Medicine Technology, Tianjin, China.
| |
Collapse
|
30
|
He Y, Hu C, Zhang X. GW1929 (an agonist of PPARγ) inhibits excessive production of reactive oxygen species in cisplatin-stimulated renal tubular epithelial cells, hampers cell apoptosis, and ameliorates renal injury. J Histotechnol 2024; 47:68-79. [PMID: 38018414 DOI: 10.1080/01478885.2023.2286692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023]
Abstract
Cisplatin-induced nephrotoxicity has long been explored for development of preventative and therapeutic drugs. The current investigation focused on the renal protective effect of GW1929, an agonist for peroxisome proliferator-activated receptors gamma (PPARγ), on cisplatin-induced kidney injury. HK2 cells treated with 20 μM cisplatin and C57BL/6 mice injected with 20 mg/kg cisplatin were used as the cell model and animal model for acute kidney injury. HK2 cell viability after cisplatin or GW1929 (0-80 μM) treatment was tested using methyl thiazolyl tetrazolium assays. Flow cytometry analysis and TUNEL assays were used to measure cell apoptosis. Intracellular reactive oxygen species (ROS) level was measured through fluorescence intensities. Levels of blood urea nitrogen (BUN) and serum creatinine (SCr) were measured to evaluate the renal function of mice. For renal morphology observation and cell apoptosis assessment in vivo, hematoxylin-eosin staining and TUNEL assays were conducted. The concentrations of oxidative stress markers in renal samples were measured using colorimetric tests. It was found that GW1929 dose-dependently enhanced protein levels of PPARγ, PGC-1α and TFEB in HK2 cells. Meanwhile, intracellular ROS overproduction, the decrease in cell viability and excessive cell apoptosis mediated by cisplatin were reversed by GW1929. For in vivo experiments, GW1929 notably attenuated cisplatin-stimulated nephrotoxicity and oxidative stress while reducing BUN and Scr levels in cisplatin-challenged model mice. Moreover, GW1929 significantly dampened renal cell apoptosis in vivo. GW1929 mitigates renal tubular epithelial cell injury and renal damage by inhibiting oxidative stress and renal cell apoptosis.
Collapse
Affiliation(s)
- Yong He
- Department of Nephrology, The Fifth Hospital of Wuhan, Wuhan, China
| | - Caihong Hu
- Department of Clinical Internal Medicine, Wuhan Hospital of China University of Geoscience, Wuhan, China
| | - Xin Zhang
- Department of Nephrology, The Fifth Hospital of Wuhan, Wuhan, China
| |
Collapse
|
31
|
Zhang Y, Chen X, Wang X, Xu Y, Li J, Wu Y, Wang Z, Zhang S, Hu J, Qi Q. Hesperetin ameliorates spinal cord injury in rats through suppressing apoptosis, oxidative stress and inflammatory response. Eur J Pharmacol 2024; 971:176541. [PMID: 38556120 DOI: 10.1016/j.ejphar.2024.176541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
Spinal cord injury (SCI), a fatal condition, is characterized by progressive tissue degradation and extreme functional deficits with limited treatment options. Hesperetin, a natural flavonoid with potent antioxidant, antiapoptotic and anti-inflammatory properties, has yet to be systematically investigated for its therapeutic effects on neurological damage in rat models of SCI. In this study, rats were given oral hesperetin once daily for 28 days, and their locomotion and histopathological changes were assessed. The findings demonstrated that hesperetin alleviates neurological damage caused by SCI. The observed behavioral improvement could be due to an increase in the survival rate of neurons and oligodendrocytes. This improvement further boosted the ability to repair tissue and form myelin after SCI, ultimately resulting in better neurological outcomes. Furthermore, the present study revealed that hesperetin possesses potent antioxidant capabilities in the context of SCI, reducing the levels of harmful oxygen free radicals and increasing the activity of antioxidant enzymes. Additionally, hesperetin markedly inhibited injury-induced apoptosis, as assessed by caspase-3 immunofluorescence staining and the expression level of caspase-3, indicating the ability of hesperetin to prevent cell death after SCI. Finally, after SCI, hesperetin treatment effectively reduced the expression of inflammatory factors, including IL-1β, TNFα, and NF-kB, demonstrating the anti-inflammatory effect of hesperetin. Together, our results suggest that hesperetin should be considered a valuable therapeutic aid following SCI, as its positive effects on the nervous system, including antioxidant, anti-inflammatory and antiapoptotic effects, may be crucial mechanisms through which hesperetin exerts neuroprotective effects against SCI.
Collapse
Affiliation(s)
- Yuxin Zhang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Xiaojie Chen
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Xiaoxuan Wang
- School of Laboratory Medicine, Bengbu Medical University, Bengbu, China; Clinical Laboratory, Bengbu Municipal Second People Hospital, Bengbu, China
| | - Yibo Xu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Jiaxin Li
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Yimin Wu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Ziyao Wang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Suhui Zhang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China
| | - Jianguo Hu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China.
| | - Qi Qi
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China; School of Basic Medicine, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
32
|
Zhang W, Chan C, Zhang K, Qin H, Yu BY, Xue Z, Zheng X, Tian J. Discovering a New Drug Against Acute Kidney Injury by Using a Tailored Photoacoustic Imaging Probe. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311397. [PMID: 38221651 DOI: 10.1002/adma.202311397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/29/2023] [Indexed: 01/16/2024]
Abstract
Acute kidney injury (AKI) has become an increasing concern for patients due to the widespread clinical use of nephrotoxic drugs. Currently, the early diagnosis of AKI is still challenging and the available therapeutic drugs cannot meet the clinical demand. Herein, this work has investigated the key redox couple involved in AKI and develops a tailored photoacoustic (PA) imaging probe (AB-DiOH) which can reversibly respond to hypochlorite (ClO-)/glutathione (GSH) with high specificity and sensitivity. This probe enables the real-time monitoring of AKI by noninvasive PA imaging, with better detection sensitivity than the blood test. Furthermore, this probe is utilized for screening nephroprotective drugs among natural products. For the first time, astragalin is discovered to be a potential new drug for the treatment of AKI. After oral administration, astragalin can be efficiently absorbed by the animal body, alleviate kidney injury, and meanwhile induce no damage to other normal tissues. The treatment mechanism of astragalin has also been revealed to be the simultaneous inhibition of oxidative stress, ferroptosis, and cuproposis. The developed PA imaging probe and the discovered drug candidate provide a promising new tool and strategy for the early diagnosis and effective treatment of AKI.
Collapse
Affiliation(s)
- Wangning Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Chenming Chan
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Kaiyu Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Haifeng Qin
- Guangdong Provincial Key Laboratory of Nanophotonic Manipulation, Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Bo-Yang Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhaoli Xue
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Xianchuang Zheng
- Guangdong Provincial Key Laboratory of Nanophotonic Manipulation, Institute of Nanophotonics, Jinan University, Guangzhou, 511443, China
| | - Jiangwei Tian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
33
|
Gökçek İ. Cardioprotective effect of oleuropein in a cisplatin-induced cardiotoxicity model in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3403-3410. [PMID: 37955692 DOI: 10.1007/s00210-023-02828-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 10/29/2023] [Indexed: 11/14/2023]
Abstract
This study investigated the cardioprotective effect of oleuropein against cisplatin-induced cardiac damage in terms of inflammatory, oxidative stress and cardiac parameters. In this study, 40 female Wistar albino rats were divided into four groups: control, cisplatin, oleuropein and cisplatin+oleuropein. To establish the experimental model, oleuropein (200 mg/kg) was administered for 14 days and cisplatin (7 mg/kg) was administered as a single dose on the seventh day. Cisplatin increased MDA cardiac parameters (CK, CK-MB and cTnI) and inflammatory cytokines (TNF-α, IL-1β and IL-6) in cardiac tissue and decreased GSH, GSH-Px and catalase levels. On the other hand, oleuropein improved cardiac parameters and decreased inflammatory cytokine and oxidative stress levels in cardiac tissue.
Collapse
Affiliation(s)
- İshak Gökçek
- Veterinary Faculty, Veterinary Physiology Department, Hatay Mustafa Kemal University, Hatay, 31000, Turkey.
| |
Collapse
|
34
|
Tang J, Li L, Chen Z, Liao C, Hu K, Yang Y, Huang J, Tang L, Zhang L, Li L. Agrimol B alleviates cisplatin-induced acute kidney injury by activating the Sirt1/Nrf2 signaling pathway in mice. Acta Biochim Biophys Sin (Shanghai) 2024; 56:551-563. [PMID: 38404180 DOI: 10.3724/abbs.2023285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024] Open
Abstract
Cisplatin (CDDP) is a widely used chemotherapeutic agent that has remarkable antineoplastic effects. However, CDDP can cause severe acute kidney injury (AKI), which limits its clinical application. Agrimol B is the main active ingredient found in Agrimonia pilosa Ledeb and has a variety of pharmacological activities. The effect of agrimol B on CDDP-induced renal toxicity has not been determined. To investigate whether agrimol B has a protective effect against CDDP-induced AKI, we first identify Sirtuin 1 (Sirt1) as a critical target protein of agrimol B in regulating AKI through network pharmacology analysis. Subsequently, the AKI mouse model is induced by administering a single dose of CDDP via intraperitoneal injection. By detecting the serum urea nitrogen and creatinine levels, as well as the histopathological changes, we confirm that agrimol B effectively reduces CDDP-induced AKI. In addition, treatment with agrimol B counteracts the increase in renal malondialdehyde level and the decrease in superoxide dismutase (SOD), catalase and glutathione levels induced by CDDP. Moreover, western blot results reveal that agrimol B upregulates the expressions of Sirt1, SOD2, nuclear factor erythroid2-related factor 2, and downstream molecules, including heme oxygenase 1 and NAD(P)H quinone dehydrogenase 1. However, administration of the Sirt1 inhibitor EX527 abolishes the effects of agrimol B. Finally, we establish a tumor-bearing mouse model and find that agrimol B has a synergistic antitumor effect with CDDP. Overall, agrimol B attenuates CDDP-induced AKI by activating the Sirt1/Nrf2 signaling pathway to counteract oxidative stress, suggesting that this compound is a potential therapeutic agent for the treatment of CDDP-induced AKI.
Collapse
Affiliation(s)
- Jiarui Tang
- Department of Pathophysiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Longhui Li
- Department of Health Management Centre, Chongqing General Hospital, Chongqing 401147, China
| | - Zhijian Chen
- Department of Pathophysiology, Shihezi University School of Medicine, Shihezi 832000, China
| | - Cuiting Liao
- Department of Pathophysiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Kai Hu
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yongqiang Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Jiayi Huang
- Department of Pathophysiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Li Tang
- Department of Pathophysiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Li Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Longjiang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
35
|
Guo H, Wang M, Shang Y, Zhang B, Zhang S, Liu X, Cao P, Fan Y, Tan K. Apoptosis-related prognostic biomarkers and potential targets for acute kidney injury based on machine learning algorithm and in vivo experiments. Apoptosis 2024; 29:303-320. [PMID: 37789227 DOI: 10.1007/s10495-023-01896-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 10/05/2023]
Abstract
Acute kidney injury (AKI) is a common critical illness in hospitalized patients, characterized by a rapid decline in kidney function over a short period, which can seriously endanger the patient's life. Currently, there is a lack of precise and universal AKI diagnostic biomarkers in clinical practice. In this study, weighted gene coexpression network analysis (WGCNA), differential expression analysis, univariate and multivariate logistic regression analyses, receiver operating characteristic (ROC) curves, and immune cell infiltration were performed to identify apoptosis-related biomarkers that can be used for AKI diagnosis. Three core apoptosis-related genes (ARGs), CBFB, EGF and COL1A1, were identified as AKI biomarkers. More importantly, an apoptosis-related signature containing three hub ARGs was validated as a diagnostic model. The hub genes exhibited good correlations with glomerular filtration rate (GFR) and serum creatinine (SCr) in the Nephroseq kidney disease database. Additionally, CIBERSORT immune infiltration analysis indicated that these core ARGs may affect immune cell recruitment and infiltration in AKI patients. Subsequently, we investigated the alteration of the expression levels of three core ARGs in AKI samples using single-cell RNA sequencing analysis and analyzed the cell types that mainly expressed these ARGs. More importantly, the expression of core ARGs was validated in folic acid- and cisplatin-induced AKI mouse models. In summary, our study identified three diagnostic biomarkers for AKI, explored the roles of ARGs in AKI progression and provided new ideas for the clinical diagnosis and treatment of AKI.
Collapse
Affiliation(s)
- Hanyao Guo
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Meixia Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Yanan Shang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Bo Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Sidi Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Xiaoyu Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Pengxiu Cao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Yumei Fan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Ke Tan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, Hebei Province Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China.
| |
Collapse
|
36
|
Gao Y, Lu X, Zhang G, Liu C, Sun S, Mao W, Jiang G, Zhou Y, Zhang N, Tao S, Chen M, Chen S, Zhang L. DRD4 alleviates acute kidney injury by suppressing ISG15/NOX4 axis-associated oxidative stress. Redox Biol 2024; 70:103078. [PMID: 38354631 PMCID: PMC10876914 DOI: 10.1016/j.redox.2024.103078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 02/16/2024] Open
Abstract
Acute kidney injury (AKI) is a life-threatening health condition associated with increasing morbidity and mortality. Despite extensive research on the mechanisms underlying AKI, effective clinical tools for prediction and treatment remain scarce. Oxidative stress and mitochondrial damage play a critical role in AKI and dopamine D4 receptor (DRD4) has been confirmed to be associated with oxidative stress. In this study, we hypothesized that DRD4 could attenuate AKI through its antioxidative and antiapoptotic effects. In vivo, DRD4 was remarkably decreased in the kidneys of mice subjected to ischemia/reperfusion injury (IRI) or cisplatin treatment. Notably, DRD4 significantly attenuated nephrotoxicity by suppressing oxidative stress and enhancing mitochondrial bioenergetics through the downregulation of reactive oxygen species (ROS) generation and NADPH oxidase 4 (NOX4) expression. In vitro, DRD4 demonstrated the ability to ameliorate oxidative stress-induced apoptosis in HK-2 cells subjected to hypoxia/reoxygenation- or cisplatin treatment. Transcriptome sequencing revealed that, mechanistically, DRD4 reduced the expression of its downstream target, interferon-stimulated gene 15 (ISG15), suppressing NOX4 ISGylation, enhancing the ubiquitination of NOX4, leading to its degradation, and ultimately counteracting oxidative stress-induced AKI. Altogether, these findings underscore the significance of DRD4 in AKI and elucidate DRD4 as a potential protectant against IRI or cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Yue Gao
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Xun Lu
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Guangyuan Zhang
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Chunhui Liu
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Si Sun
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Weipu Mao
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Guiya Jiang
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Yu Zhou
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China
| | - Nieke Zhang
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Shuchun Tao
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Ming Chen
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.
| | - Shuqiu Chen
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.
| | - Lei Zhang
- Surgical Research Center, Institute of Urology, Medical School of Southeast University, Nanjing, China; Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.
| |
Collapse
|
37
|
Wang X, Zhang L. The systemic oxidative stress score has a prognostic value on gastric cancer patients undergoing surgery. Front Oncol 2024; 14:1307662. [PMID: 38525419 PMCID: PMC10957578 DOI: 10.3389/fonc.2024.1307662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/26/2024] [Indexed: 03/26/2024] Open
Abstract
Background Oxidative stress is strongly associated with the development, recurrence metastasis, and treatment of gastric cancer. It is yet unknown, though, how systemic oxidative stress levels relate to the surgically treated gastric cancer patients' clinical results. This research aims to investigate the prognostic effect of systemic oxidative stress score, also known as systematic oxidative stress score (SOS), on gastric cancer patients undergoing surgical treatment. Methods Development of the SOS Formula through Least Absolute Shrinkage and Selection Operator LASSO Cox Regression. By using optimal cut-off values, the 466 patients included in the study had been split into high SOS and low SOS groups. Utilizing Chi-square test and the Wilcoxon rank sum test, this research examined the relationship between SOS and clinical traits. With the aid of Kaplan-Meier and COX regression analysis, the prognosis of patients with gastric cancer was examined. Results SOS consisted of four oxidative stress-related laboratory indices. Univariate and multivariate COX regression analyses revealed that SOS, Age, CA724, Radical resection and TNM stage were crucial prognostic factors for OS, and the independent prognostic factors for PFS included Age, CA724, TNM stage and SOS. They could have their prognosis correctly predicted using a nomogram built around SOS and independent prognostic variables. Conclusion SOS is a practical and reasonably priced tool for determining a patient's prognosis for gastric cancer. More notably, SOS is an accurate prognostic factor for patients with advanced gastric cancer who has undergone radical surgery.
Collapse
Affiliation(s)
| | - Limin Zhang
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
38
|
Wang Q, Xu J, Li M, Chen Y, Xu Y, Li L, Gong Y, Yang Y. Nrf2 knockout attenuates the astragaloside IV therapeutic effect on kidney fibrosis from liver cancer by regulating pSmad3C/3L pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1687-1700. [PMID: 37712971 DOI: 10.1007/s00210-023-02711-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023]
Abstract
Fibrotic kidney injury from hepatocarcinogenesis seriously impacts treatment effect. Astragaloside IV (AS-IV), an extract of Astragalus membranaceus, has several pharmacological activities, which are useful in the treatment of edema and fibrosis. Nrf2/HO-1 is a key antioxidant stress pathway and help treatment of kidney injury. Smad3 phosphorylation is implicated in hepatocarcinogenesis. Our previous study clarified that Smad3 is differentially regulated by different phosphorylated forms of Smad3 on hepatocarcinogenesis. Therefore, we investigated the contribution of AS-IV on the therapy of kidney fibrosis from hepatocarcinogenesis. And the focus was on whether the phosphorylation of Smad3 and the regulation of Nrf2/HO-1 pathway were involved during AS-IV therapy and whether there is an effect of Nrf2 knockout on the phosphorylation of Smad3. We performed TGF-β1 stimulation on HK-2 cells and intervened with AS-IV. Furtherly, we investigated renal injury of AS-IV on Nrf2 knockout mice during hepatocarcinogenesis and its mechanism of action. On the one hand, in vitro results showed that AS-IV reduced the ROS and α-SMA expression of HK-2 by promoting the expression pSmad3C/p21 of and Nrf2/HO-1 and suppressed the expression of pSmad3L/PAI-1. On the other hand, the in vivo results of histopathological features, serological biomarkers, and oxidative damage indicators showed that Nrf2 knockout aggravated renal injury. Besides, Nrf2 deletion decreased the nephroprotective effect of AS-IV by suppressing the pSmad3C/p21 pathway and promoting the pSmad3L/PAI-1 pathway. The experimental results were as we suspected. And we identify for the first time that Nrf2 deficiency increases renal fibrosis from hepatocarcinogenesis and attenuates the therapeutic effects of AS-IV via regulating pSmad3C/3L signal pathway.
Collapse
Affiliation(s)
- Qin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China
| | - Jiacheng Xu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China
| | - Miaomiao Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China
| | - Yuqing Chen
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China
| | - Yingying Xu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China
| | - Lili Li
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China
| | - Yongfang Gong
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China
| | - Yan Yang
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-Inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, 81 Meishan Road, Hefei, 230032, China.
| |
Collapse
|
39
|
Yu X, Liu Z, Yu Y, Qian C, Lin Y, Jin S, Wu L, Li S. Hesperetin promotes diabetic wound healing by inhibiting ferroptosis through the activation of SIRT3. Phytother Res 2024; 38:1478-1493. [PMID: 38234096 DOI: 10.1002/ptr.8121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/04/2023] [Accepted: 12/27/2023] [Indexed: 01/19/2024]
Abstract
Hesperetin (HST) is a flavonoid compound naturally occurring in citrus fruits and is widespread in various traditional medicinal herbs such as grapefruit peel, orange peel, and tangerine peel. These plant materials are commonly used in traditional Chinese medicine to prepare herbal remedies. The study aimed to investigate the potential molecular mechanisms through which HST reduces ferroptosis in human umbilical vein endothelial cells (HUVECs) and promotes angiogenesis and wound healing. We employed network pharmacology to predict the downstream targets affected by HST. The expression of markers related to ferroptosis was assessed through Western blot (WB) and polymerase chain reaction. Intracellular levels of ferroptosis-related metabolism were examined using glutathione/oxidized glutathione (GSH/GSSG) and malondialdehyde (MDA) assay kits. Mitochondrial status and iron levels within the cells were investigated through staining with Mitosox, FerroOrange, and JC1 staining. Potential downstream direct targets of HST were identified using molecular docking. Additionally, wound healing and neovascularization within the wound site were analyzed using various methods including HE staining, Masson's staining, immunohistochemistry, and Doppler hemodynamics assessment. HST effectively inhibits the elevated levels of intracellular ferroptosis stimulated by ERASTIN. Furthermore, we observed that HST achieves this inhibition of ferroptosis by activating SIRT3. In a diabetic rat wound model, HST significantly promotes wound healing, reducing levels of tissue ferroptosis, consistent with our in vitro findings. This study demonstrates that HST can inhibit the progression of ferroptosis and protect the physiological function of HUVECs by activating SIRT3. HST holds promise as a natural compound for promoting diabetic wound healing.
Collapse
Affiliation(s)
- Xianbin Yu
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zhixuan Liu
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- Alberta Institute, Wenzhou Medical University, Wenzhou, China
| | - Yitian Yu
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Chengjie Qian
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yuzhe Lin
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shuqing Jin
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Long Wu
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shi Li
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
40
|
Li Z, He R, Liu J, Jin X, Jiang B, Lao Y, Yang S. JianPiYiShen formula prevents cisplatin-induced acute kidney injury in mice by improving necroptosis through MAPK pathway. BMC Complement Med Ther 2024; 24:101. [PMID: 38402163 PMCID: PMC10893720 DOI: 10.1186/s12906-024-04366-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 01/19/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Acute kidney injury (AKI), characterized by necroptosis and activation of MAPK pathway, causes sudden declines in renal function. To date, efficacious treatments are lacking. JianPiYiShen Formula (JPYSF) has a protective effect on the kidneys. The aim of this study is to explore the mechanism of JPYSF in cisplatin-induced AKI. METHODS Male C57/BL6J mice were divided into control group, cisplatin group and cisplatin + JPYSF group. Before establishing the model, the cisplatin + JPYSF group was administered JPYSF (18.35 g/kg/day) by gavage for 5 consecutive days. A single intraperitoneal injection of cisplatin (20 mg/kg) was used to establish AKI model. Measurement of renal function and H&E staining were performed to assess renal damage. WB, PCR, TUNEL staining and immunohistochemistry were used to detect related indicators of mitochondrial function, oxidative stress, necroptosis, inflammation and MAPK pathway. And one-way analysis of variance was used to compare group differences. RESULTS Compared with the cisplatin group, JPYSF can attenuate AKI, reflected by the decrease in Scr and BUN levels, the improvement of renal tubular injury, and the downregulation of NGAL and KIM1. Cisplatin can induce mitochondrial dysfunction and oxidative stress, triggering necroptosis. In this study, JPYSF improved mitochondrial dysfunction to enhance oxidative stress, as manifested by upregulation of OPA1, PGC-1α, SOD and CAT, and downregulation of DRP1 and MFF. Then JPYSF showed a significant protective effect in necroptosis, as embodied by reduced number of TUNEL-positive cells, decreased the gene expression of RIPK3 and MLKL, as well as downregulation the proteins expression of P-RIPK1, P-RIPK3, and P-MLKL. Moreover, necroptosis can aggravate inflammation. JPYSF ameliorated inflammation by improving inflammatory and anti-inflammatory indexes, including downregulation of TNF-α, IL-6, MCP-1 and LY6G, and upregulation of IL-10. In addition, JPYSF also inhibited MAPK pathway to improve necroptosis by decreasing the expression of P-JNK and P-ERK. CONCLUSION Our data showed that JPYSF prevents cisplatin-induced AKI by improving necroptosis through MAPK pathway, which is related to the improvement of mitochondrial dysfunction, oxidative stress, and inflammation.
Collapse
Affiliation(s)
- Zhongtang Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1, Fuhua Road, Futian District, Shenzhen, Guangdong, 518033, China
| | - Riming He
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1, Fuhua Road, Futian District, Shenzhen, Guangdong, 518033, China
| | - Jiahui Liu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1, Fuhua Road, Futian District, Shenzhen, Guangdong, 518033, China
| | - Xiaoming Jin
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1, Fuhua Road, Futian District, Shenzhen, Guangdong, 518033, China
| | - Beibei Jiang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1, Fuhua Road, Futian District, Shenzhen, Guangdong, 518033, China
| | - Yunlan Lao
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1, Fuhua Road, Futian District, Shenzhen, Guangdong, 518033, China
| | - Shudong Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1, Fuhua Road, Futian District, Shenzhen, Guangdong, 518033, China.
| |
Collapse
|
41
|
Liu J, Xie L, Zhai H, Wang D, Li X, Wang Y, Song M, Xu C. Exploration of the protective mechanisms of Icariin against cisplatin-induced renal cell damage in canines. Front Vet Sci 2024; 11:1331409. [PMID: 38455257 PMCID: PMC10919057 DOI: 10.3389/fvets.2024.1331409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
This study delves into the protective mechanisms of Icariin (ICA) against cisplatin-induced damage in Madin-Darby canine kidney (MDCK) cells. Comprising two distinct phases, the investigation initially employed a single-factor randomized design to ascertain the minimal cisplatin concentration eliciting MDCK cell damage, spanning concentrations from 0 to 16 mmol/L. Concurrently, various concentrations of ICA (ranging from 5 to 50 mmol/L) were combined with 1 mmol/L cisplatin to determine the most efficacious treatment concentration. Subsequent investigations utilized four treatment groups: control, 1 mmol/L cisplatin, 1 mmol/L cisplatin + 20 mmol/L ICA, and 1 mmol/L cisplatin + 25 mmol/L ICA, aimed at elucidating ICA's protective mechanisms. Findings from the initial phase underscored a significant reduction in MDCK cell viability with 1 mmol/L cisplatin in comparison to the control (P < 0.01). Notably, the inclusion of 20 and 25 mmol/L ICA substantively ameliorated MDCK cell viability under 1 mmol/L cisplatin (P < 0.01). Moreover, cisplatin administration induced an elevation in inflammatory factors, malondialdehyde (MDA), reactive oxygen species (ROS), and Bax protein levels, while concurrently suppressing superoxide dismutase (SOD), catalase (CAT), and Bcl-2 expression (P < 0.01). Conversely, supplementation of 20 and 25 mmol/L ICA demonstrated a marked increase in mitochondrial membrane potential and levels of SOD, CAT, and Bcl-2 (P < 0.01). These interventions effectively attenuated inflammatory responses and suppressed Bax protein expression (P < 0.05), consequently mitigating cisplatin-induced apoptosis in MDCK cells (P < 0.01). In summary, these findings elucidate the role of ICA in impeding apoptosis in cisplatin-induced MDCK cells by regulating inflammatory responses, oxidative stress, and autophagic protein expression.
Collapse
Affiliation(s)
- Jiayi Liu
- College of Police Dog Technology of Criminal Investigation Police University of China, Shenyang, China
| | - Liuwei Xie
- College of Police Dog Technology of Criminal Investigation Police University of China, Shenyang, China
| | - He Zhai
- College of Police Dog Technology of Criminal Investigation Police University of China, Shenyang, China
| | - Dongwei Wang
- College of Police Dog Technology of Criminal Investigation Police University of China, Shenyang, China
| | - Xiao Li
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yao Wang
- College of Police Dog Technology of Criminal Investigation Police University of China, Shenyang, China
| | - Mingqiang Song
- College of Police Dog Technology of Criminal Investigation Police University of China, Shenyang, China
| | - Chao Xu
- Institute of Special Wild Economic Animals and Plants, Chinese Academy of Agricultural Sciences, Changchun, China
| |
Collapse
|
42
|
Wang L, Xie Y, Xiao B, He X, Ying G, Zha H, Yang C, Jin X, Li G, Ping L, Wang J, Weng Q. Isorhamnetin alleviates cisplatin-induced acute kidney injury via enhancing fatty acid oxidation. Free Radic Biol Med 2024; 212:22-33. [PMID: 38101584 DOI: 10.1016/j.freeradbiomed.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/03/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023]
Abstract
Cisplatin is an effective chemotherapy drug widely used in the treatment of various solid tumors. However, the clinical usage of cisplatin is limited by its nephrotoxicity. Isorhamnetin, a natural flavanol compound, displays remarkable pharmacological effects, including anti-inflammatory and anti-oxidation. In this study, we aimed to investigate the potential of isorhamnetin in alleviating acute kidney injury induced by cisplatin. In vitro study showed that isorhamnetin significantly suppressed the cytotoxic effects of cisplatin on human tubular epithelial cells. Furthermore, isorhamnetin exerted significantly inhibitory effects on cisplatin-induced apoptosis and inflammatory response. In acute kidney injury mice induced by a single intraperitoneal injection with 20 mg/kg cisplatin, oral administration of isorhamnetin two days before or 2 h after cisplatin injection effectively ameliorated renal function and renal tubule injury. Transcriptomics RNA-seq analysis of the mice kidney tissues suggested that isorhamnetin treatment may protect against cisplatin-induced nephrotoxicity via PGC-1α mediated fatty acid oxidation. Isorhamnetin achieved significant enhancements in the lipid clearance, ATP level, as well as the expression of PGC-1α and its downstream target genes PPARα and CPT1A, which were otherwise impaired by cisplatin. In addition, the protection effects of isorhamnetin against cisplatin-induced nephrotoxicity were abolished by a PGC-1α inhibitor, SR-18292. In conclusion, our findings indicate that isorhamnetin could protect against cisplatin-induced acute kidney injury by inducing PGC-1α-dependent reprogramming of fatty acid oxidation, which highlights the clinical potential of isorhamnetin as a therapeutic approach for the management of cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Lingkun Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310007, China
| | - Yaochen Xie
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310007, China
| | - Boneng Xiao
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310007, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xuelin He
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Department of Nephrology, Beilun People's Hospital, Ningbo, 315826, China
| | - Guanghui Ying
- Department of Nephrology, Beilun People's Hospital, Ningbo, 315826, China
| | - Huiyan Zha
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310007, China
| | - Chen Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310007, China
| | - Xuejin Jin
- Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310053, China
| | - Guilin Li
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310007, China
| | - Li Ping
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310007, China
| | - Jincheng Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310007, China; Research Institute of Zhejiang University-Taizhou, Taizhou, 318000, China; Beijing Life Science Academy, Beijing, 102200, China.
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310007, China; Research Institute of Zhejiang University-Taizhou, Taizhou, 318000, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
43
|
Pisoschi AM, Iordache F, Stanca L, Cimpeanu C, Furnaris F, Geicu OI, Bilteanu L, Serban AI. Comprehensive and critical view on the anti-inflammatory and immunomodulatory role of natural phenolic antioxidants. Eur J Med Chem 2024; 265:116075. [PMID: 38150963 DOI: 10.1016/j.ejmech.2023.116075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023]
Abstract
The immune response encompasses innate and adaptive immunity, each with distinct and specific activities. The innate immune system is constituted by phagocytic cells, macrophages, monocytes and neutrophils, the cascade system, and different classes of receptors such as toll-like receptors that are exploited by the innate immune cells. The adaptive immune system is antigen-specific, encompassing memory lymphocytes and the corresponding specific receptors. Inflammation is understood as an activation of different signaling pathways such as toll-like receptors or nuclear factor kappa-light-chain-enhancer of activated B cells, with an increase in nitric oxide, inflammatory cytokines and chemokines. Increased oxidative stress has been identified as main source of chronic inflammation. Phenolic antioxidants modulate the activities of lymphocytes and macrophages by impacting cytokines and nitric oxide release, exerting anti-inflammatory effect. The nuclear-factor kappa-light-chain-enhancer of activated B cells signaling pathway and the mitogen-activated protein kinase pathway are targeted, alongside an increase in nuclear factor erythroid 2-related factor mediated antioxidant response, triggering the activity of antioxidant enzymes. The inhibitive potential on phospholipase A2, cyclooxygenase and lipoxygenase in the arachidonic acid pathway, and the subsequent reduction in prostaglandin and leukotriene generation, reveals the potential of phenolics as inflammation antagonists. The immunomodulative potential encompasses the capacity to interfere with proinflammatory cytokine synthesis and with the expression of the corresponding genes. A diet rich in antioxidants can result in prevention of inflammation-related pathologies. More investigations are necessary to establish the role of these antioxidants in therapy. The appropriate delivery system and the prooxidant effects exhibited at large doses, or in the presence of heavy metal cations should be regarded.
Collapse
Affiliation(s)
- Aurelia Magdalena Pisoschi
- University of Agronomic Sciences and Veterinary Medicine of Bucharest, Faculty of Veterinary Medicine, Department Preclinical Sciences, 105 Splaiul Independentei, 050097, Bucharest, Romania.
| | - Florin Iordache
- University of Agronomic Sciences and Veterinary Medicine of Bucharest, Faculty of Veterinary Medicine, Department Preclinical Sciences, 105 Splaiul Independentei, 050097, Bucharest, Romania
| | - Loredana Stanca
- University of Agronomic Sciences and Veterinary Medicine of Bucharest, Faculty of Veterinary Medicine, Department Preclinical Sciences, 105 Splaiul Independentei, 050097, Bucharest, Romania
| | - Carmen Cimpeanu
- University of Agronomic Sciences and Veterinary Medicine of Bucharest, Faculty of Land Reclamation and Environmental Engineering, 59 Marasti Blvd, 011464, Bucharest, Romania
| | - Florin Furnaris
- University of Agronomic Sciences and Veterinary Medicine of Bucharest, Faculty of Veterinary Medicine, Department Preclinical Sciences, 105 Splaiul Independentei, 050097, Bucharest, Romania
| | - Ovidiu Ionut Geicu
- University of Agronomic Sciences and Veterinary Medicine of Bucharest, Faculty of Veterinary Medicine, Department Preclinical Sciences, 105 Splaiul Independentei, 050097, Bucharest, Romania; University of Bucharest, Faculty of Biology, Department Biochemistry and Molecular Biology, 91-95 Splaiul Independentei, 050095, Bucharest, Romania
| | - Liviu Bilteanu
- Molecular Nanotechnology Laboratory, National Institute for Research and Development in Microtechnologies, 126A, Erou Iancu Nicolae Street, 077190, Bucharest, Romania
| | - Andreea Iren Serban
- University of Agronomic Sciences and Veterinary Medicine of Bucharest, Faculty of Veterinary Medicine, Department Preclinical Sciences, 105 Splaiul Independentei, 050097, Bucharest, Romania; University of Bucharest, Faculty of Biology, Department Biochemistry and Molecular Biology, 91-95 Splaiul Independentei, 050095, Bucharest, Romania
| |
Collapse
|
44
|
Ullah S, Burki S, Munir AB, Yousaf G, Shafique M. Nanocarrier-based localized and effective treatment of renal disorders: currently employed targeting strategies. Nanomedicine (Lond) 2024; 19:345-361. [PMID: 38293889 DOI: 10.2217/nnm-2023-0251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024] Open
Abstract
Renal disorders pose a global health threat, with targeted drug-delivery systems emerging as a promising strategy to enhance therapy safety and efficacy. Recent efforts have harnessed targeted nanomaterials for kidney disease treatment. While some systems remain in the early stages, they show immense potential in delivering cargo to specific sites. Through animal model experimentations, it has been demonstrated to reduce systemic side effects and enhance treatment effectiveness. This review presents current strategies for kidney disorder treatment, emphasizing site-specific targeting critical to renal disease pathophysiology. Recent advancements in nano-drug delivery systems for kidney targeting are explored. Finally, toxicological aspects and prospects of the most promising kidney-targeting delivery systems are discussed in this review article.
Collapse
Affiliation(s)
- Shafi Ullah
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Superior University, Lahore, Punjab, 54000, Pakistan
| | - Samiullah Burki
- Department of Pharmacology, Jinnah Sindh Medical University, Karachi, 75510, Pakistan
| | - Abu Bakar Munir
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Superior University, Lahore, Punjab, 54000, Pakistan
| | - Ghulam Yousaf
- PAF Ruth Pfau Medical College and Hospital Faisal Base Karachi, Karachi, 75350, Pakistan
| | - Muhammad Shafique
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia
| |
Collapse
|
45
|
Kaya K, Ciftci O, Basak Turkmen N, Taşlıdere A, Gül CC. β-Glucan ameliorates cisplatin-induced oxidative and histological damage in kidney and liver of rats. Biotech Histochem 2024; 99:92-100. [PMID: 38444353 DOI: 10.1080/10520295.2024.2320626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
We investigated the effects of β-glucan (βg) on kidney and liver damage caused by cisplatin (CP), an antineoplastic agent widely used to treat many types of cancer, in a rat model. The side effects of CP in many tissues and organs limit its usage. βg is a natural polysaccharide that is an effective free radical scavenger. A total of 28 rats were randomly divided into four groups. Group 1 was a non-intervention control, only feed and water were given. Group 2 was administered 7 mg/kg CP in a single dose. Group 3 was administered 50 mg/kg βg orally for 14 days. Group 4 was administered βg for 14 days, following a single dose of CP. At the end of the experiment, kidney and liver tissues were evaluated biochemically and histopathologically. Increased thiobarbituric acid-reactive substances (TBARS) levels, as well as decreased catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx) activities, and reduced glutathione (GSH) levels, as well as histological damage, were noted in both the kidney and liver tissues of the CP group. However, βg treatment prevented the oxidative and histopathological effects of CP. The study demonstrates the protective efficacy of βg against CP-induced kidney and liver damage through the effect of its antioxidant properties.
Collapse
Affiliation(s)
- Kürşat Kaya
- Department of Medicinal Biochemistry, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| | - O Ciftci
- Department of Pharmacology, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| | - N Basak Turkmen
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, İnönü University, Malatya, Türkiye
| | - A Taşlıdere
- Department of Histology and Embryology, Faculty of Medicine, İnönü University, Malatya, Türkiye
| | - C C Gül
- Department of Histology and Embryology, Faculty of Medicine, İnönü University, Malatya, Türkiye
| |
Collapse
|
46
|
Pushpan CK, Kresock DF, Ingersoll MA, Lutze RD, Keirns DL, Hunter WJ, Bashir K, Teitz T. Repurposing AZD5438 and Dabrafenib for Cisplatin-Induced AKI. J Am Soc Nephrol 2024; 35:22-40. [PMID: 37962623 PMCID: PMC10786615 DOI: 10.1681/asn.0000000000000261] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/09/2023] [Indexed: 11/15/2023] Open
Abstract
SIGNIFICANCE STATEMENT To combat both untoward effects of nephrotoxicity and ototoxicity in cisplatin-treated patients, two potential therapeutic oral anticancer drugs AZD5438 and dabrafenib, a phase-2 clinical trial protein kinase CDK2 inhibitor and an US Food and Drug Administration-approved drug BRAF inhibitor, respectively, were tested in an established mouse AKI model. Both drugs have previously been shown to protect significantly against cisplatin-induced hearing loss in mice. Each drug ameliorated cisplatin-induced increases in the serum biomarkers BUN, creatinine, and neutrophil gelatinase-associated lipocalin. Drugs also improved renal histopathology and inflammation, mitigated cell death by pyroptosis and necroptosis, and significantly enhanced overall survival of cisplatin-treated mice. BACKGROUND Cisplatin is an effective chemotherapy agent for a wide variety of solid tumors, but its use is dose-limited by serious side effects, including AKI and hearing loss. There are no US Food and Drug Administration-approved drugs to treat both side effects. Recently, two anticancer oral drugs, AZD5438 and dabrafenib, were identified as protective against cisplatin-induced hearing loss in mice. We hypothesize that similar cell stress and death pathways are activated in kidney and inner ear cells when exposed to cisplatin and tested whether these drugs alleviate cisplatin-induced AKI. METHODS The HK-2 cell line and adult FVB mice were used to measure the protection from cisplatin-induced cell death and AKI by these drugs. Serum markers of kidney injury, BUN, creatinine, and neutrophil gelatinase-associated lipocalin as well as histology of kidneys were analyzed. The levels of markers of kidney cell death, including necroptosis and pyroptosis, pERK, and proliferating cell nuclear antigen, were also examined by Western blotting and immunofluorescence. In addition, CDK2 knockout (KO) mice were used to confirm AZD5438 protective effect is through CDK2 inhibition. RESULTS The drugs reduced cisplatin-induced cell death in the HK-2 cell line and attenuated cisplatin-induced AKI in mice. The drugs reduced serum kidney injury markers, inhibited cell death, and reduced the levels of pERK and proliferating cell nuclear antigen, all of which correlated with prolonged animal survival. CDK2 KO mice were resistant to cisplatin-induced AKI, and AZD5438 conferred no additional protection in the KO mice. CONCLUSIONS Cisplatin-induced damage to the inner ear and kidneys shares similar cellular beneficial responses to AZD5438 and dabrafenib, highlighting the potential therapeutic use of these agents to treat both cisplatin-mediated kidney damage and hearing loss.
Collapse
Affiliation(s)
- Chithra K. Pushpan
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska
| | - Daniel F. Kresock
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska
| | - Matthew A. Ingersoll
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska
| | - Richard D. Lutze
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska
| | - Darby L. Keirns
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska
| | - William J. Hunter
- Department of Pathology, Creighton University School of Medicine, Omaha, Nebraska
| | - Khalid Bashir
- Renal Division, Department of Medicine, CHI Nephrology and Creighton University Medical Center, Omaha, Nebraska
| | - Tal Teitz
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska
| |
Collapse
|
47
|
Al-Bataineh WM, Alzoubi KH, Khabour OF, Mahasneh A, Al Momany EM. Vitamin B12 Protects against Genotoxicity Induced by Cisplatin. Curr Cancer Drug Targets 2024; 24:1169-1176. [PMID: 38299397 DOI: 10.2174/0115680096284684240110044954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/18/2023] [Accepted: 11/29/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Cisplatin is an effective synthetic chemotherapeutic drug used for cancer treatment. Vitamin B12 has been shown to possess anti-genotoxic activity. This study aimed to investigate the effect of vitamin B12 on chromosomal damage induced by cisplatin. METHODS The level of sister chromatid exchanges (SCEs) and chromosomal aberrations (CAs) were measured in cultured human blood lymphocytes treated with cisplatin and/or vitamin B12. RESULTS The results showed a significantly elevated frequency of CAs and SCEs of cisplatin-treated cultures compared to the control (P < 0.05). The CAs and SCEs induced by cisplatin were significantly lowered by pretreatment of cell cultures with vitamin B12. In addition, cisplatin caused a slight reduction in the mitotic index (MI), while vitamin B12 did not modulate the effect of cisplatin on MI. CONCLUSION Vitamin B12 can protect human lymphocytes against genotoxicity associated with cisplatin.
Collapse
Affiliation(s)
- Wejdan M Al-Bataineh
- Department of Applied Biological Sciences, Faculty of Science and Art, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, UAE
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Amjad Mahasneh
- Department of Applied Biological Sciences, Faculty of Science and Art, Jordan University of Science and Technology, Irbid, Jordan
| | - Enaam M Al Momany
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, P.O. box 330127, Zarqa 13133, Jordan
| |
Collapse
|
48
|
Li Y, Shi L, Zhao F, Luo Y, Zhang M, Wu X, Zhu J. PIM1 attenuates cisplatin-induced AKI by inhibiting Drp1 activation. Cell Signal 2024; 113:110969. [PMID: 37967691 DOI: 10.1016/j.cellsig.2023.110969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/23/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023]
Abstract
Cisplatin, an effective anti-cancer drug, always causes acute kidney injury (AKI) by inducing mitochondrial damage. PIM1 is a serine/threonine kinase, which has been shown to regulate mitochondrial function. However, the role and mechanisms of PIM1 in cisplatin-induced AKI remain unexplored. This study aimed to investigate the effects of PIM1 in cisplatin-induced AKI and its underlying mechanisms. To established Cisplatin-induced AKI model, mice were given a single intraperitoneal injection(20 mg/kg) and BUMPT cells were treated with cisplatin(20 μM). PIM1 inhibitor AZD1208 was used to inhibit PIM1 and PIM1-experssing adenovirus was used to overexpress PIM1. Drp1 inhibitor P110 and pcDNA3-Drp1K38A were used to inhibit the activation of Drp1 and mitochondrial fission. The indicators of renal function, renal morphology, apoptosis and mitochondrial dysfunction were assessed to evaluate cisplatin-induced nephrotoxicity. We observed that PIM1 was activated in cisplatin-induced AKI in vivo and cisplatin-induced tubular cells injury in vitro. PIM1 inhibition aggravated cisplatin-induced AKI in vivo, while PIM1 overexpression attenuated cisplatin-induced kidney injury in vivo and in vitro. Moreover, inhibiting PIM1 exacerbated mitochondrial damage in mice, but overexpressing PIM1 relieved mitochondrial damage in mice and BUMPT cells. In mice and BUMPT cells, inhibiting PIM1 deregulated the expression of p-Drp1S637, overexpressing PIM1 upregulated the ex-pression of p-Drp1S637. And inhibiting Drp1 activity alleviated cell damage in BUMPT cells with PIM1 knockdown or inhibition. This study demonstrated the protective effect of PIM1 in cisplatin-induced AKI, and regulation of Drp1 activation might be the underlying mechanism. Altogether, PIM1 may be a potential therapeutic target for cisplatin-induced AKI.
Collapse
Affiliation(s)
- Yuzhen Li
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Lang Shi
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Fan Zhao
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yanwen Luo
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Mingjiao Zhang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xiongfei Wu
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China.
| | - Jiefu Zhu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China.
| |
Collapse
|
49
|
Rabienezhad Ganji N, Urzì O, Tinnirello V, Costanzo E, Polito G, Palumbo Piccionello A, Manno M, Raccosta S, Gallo A, Lo Pinto M, Calligaris M, Scilabra SD, Di Bella MA, Conigliaro A, Fontana S, Raimondo S, Alessandro R. Proof-of-Concept Study on the Use of Tangerine-Derived Nanovesicles as siRNA Delivery Vehicles toward Colorectal Cancer Cell Line SW480. Int J Mol Sci 2023; 25:546. [PMID: 38203716 PMCID: PMC10779162 DOI: 10.3390/ijms25010546] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/23/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
In the last years, the field of nanomedicine and drug delivery has grown exponentially, providing new platforms to carry therapeutic agents into the target sites. Extracellular vesicles (EVs) are ready-to-use, biocompatible, and non-toxic nanoparticles that are revolutionizing the field of drug delivery. EVs are involved in cell-cell communication and mediate many physiological and pathological processes by transferring their bioactive cargo to target cells. Recently, nanovesicles from plants (PDNVs) are raising the interest of the scientific community due to their high yield and biocompatibility. This study aims to evaluate whether PDNVs may be used as drug delivery systems. We isolated and characterized nanovesicles from tangerine juice (TNVs) that were comparable to mammalian EVs in size and morphology. TNVs carry the traditional EV marker HSP70 and, as demonstrated by metabolomic analysis, contain flavonoids, organic acids, and limonoids. TNVs were loaded with DDHD1-siRNA through electroporation, obtaining a loading efficiency of 13%. We found that the DDHD1-siRNA complex TNVs were able to deliver DDHD1-siRNA to human colorectal cancer cells, inhibiting the target expression by about 60%. This study represents a proof of concept for the use of PDNVs as vehicles of RNA interference (RNAi) toward mammalian cells.
Collapse
Affiliation(s)
- Nima Rabienezhad Ganji
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, 90133 Palermo, Italy; (N.R.G.); (O.U.); (V.T.); (E.C.); (M.A.D.B.); (A.C.); (S.F.); (R.A.)
| | - Ornella Urzì
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, 90133 Palermo, Italy; (N.R.G.); (O.U.); (V.T.); (E.C.); (M.A.D.B.); (A.C.); (S.F.); (R.A.)
| | - Vincenza Tinnirello
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, 90133 Palermo, Italy; (N.R.G.); (O.U.); (V.T.); (E.C.); (M.A.D.B.); (A.C.); (S.F.); (R.A.)
| | - Elisa Costanzo
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, 90133 Palermo, Italy; (N.R.G.); (O.U.); (V.T.); (E.C.); (M.A.D.B.); (A.C.); (S.F.); (R.A.)
| | - Giulia Polito
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy; (G.P.); (A.P.P.)
| | - Antonio Palumbo Piccionello
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy; (G.P.); (A.P.P.)
| | - Mauro Manno
- Institute of Biophysics, National Research Council of Italy, 90146 Palermo, Italy; (M.M.); (S.R.)
| | - Samuele Raccosta
- Institute of Biophysics, National Research Council of Italy, 90146 Palermo, Italy; (M.M.); (S.R.)
| | - Alessia Gallo
- Research Department, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy;
| | - Margot Lo Pinto
- Proteomics Group of Fondazione Ri.MED, Department of Research IRCCS-ISMETT, via Ernesto Tricomi 5, 90145 Palermo, Italy (M.C.)
| | - Matteo Calligaris
- Proteomics Group of Fondazione Ri.MED, Department of Research IRCCS-ISMETT, via Ernesto Tricomi 5, 90145 Palermo, Italy (M.C.)
| | - Simone Dario Scilabra
- Proteomics Group of Fondazione Ri.MED, Department of Research IRCCS-ISMETT, via Ernesto Tricomi 5, 90145 Palermo, Italy (M.C.)
| | - Maria Antonietta Di Bella
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, 90133 Palermo, Italy; (N.R.G.); (O.U.); (V.T.); (E.C.); (M.A.D.B.); (A.C.); (S.F.); (R.A.)
| | - Alice Conigliaro
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, 90133 Palermo, Italy; (N.R.G.); (O.U.); (V.T.); (E.C.); (M.A.D.B.); (A.C.); (S.F.); (R.A.)
| | - Simona Fontana
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, 90133 Palermo, Italy; (N.R.G.); (O.U.); (V.T.); (E.C.); (M.A.D.B.); (A.C.); (S.F.); (R.A.)
| | - Stefania Raimondo
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, 90133 Palermo, Italy; (N.R.G.); (O.U.); (V.T.); (E.C.); (M.A.D.B.); (A.C.); (S.F.); (R.A.)
| | - Riccardo Alessandro
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università degli Studi di Palermo, 90133 Palermo, Italy; (N.R.G.); (O.U.); (V.T.); (E.C.); (M.A.D.B.); (A.C.); (S.F.); (R.A.)
| |
Collapse
|
50
|
Yuan F, Xia GQ, Cai JN, Lv X, Dai M. Hesperitin attenuates alcoholic steatohepatitis by regulating TLR4/NF-κB signaling in mice. Anal Biochem 2023; 682:115339. [PMID: 37805041 DOI: 10.1016/j.ab.2023.115339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/22/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
In the peel of citrus (Rutaceae) fruit, hesperitin (Hesp), a flavanone glycoside chemical, is found naturally. Hesp has been found to have a wide range of pharmacological actions, including anti-inflammatory, antioxidant, antiviral, and anticancer properties, according to earlier research. However, nothing is known regarding its function in alcoholic liver steatosis and inflammation. In this study, we employed a network pharmacology approach to identify the TLR4 signaling pathway as a primary target of Hesp for the treatment of alcoholic steatohepatitis (ASH). Molecular docking results showed that Hesp bound to the representative target TLR4 and exhibited good affinity. In addition, Hesp inhibits the TLR4 target and consequently the NF-κB signaling pathway, which in turn slows the evolution of alcoholic steatohepatitis, according to further in vitro and in vivo tests. The results of this study preliminarily indicate that Hesp is an ideal drug candidate for the treatment of ASH.
Collapse
Affiliation(s)
- Fei Yuan
- Department of Pharmacy, Anhui Provincial Cancer Hospital, West Branch of The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Guo-Qing Xia
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Jun-Nan Cai
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Xiongwen Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China.
| | - Meng Dai
- Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| |
Collapse
|