1
|
Abdulla MH, Murphy E, Mulcahy L, Johns EJ. TRPV1 blockade restores the baroreflex control of renal sympathetic nerve activity in cisplatin-induced renal injury in rats. Exp Physiol 2025. [PMID: 40349310 DOI: 10.1113/ep092618] [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: 02/07/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025]
Abstract
Renal injury is associated with inflammatory responses within the kidney which could involve activation of transient receptor potential vanilloid 1 (TRPV1) channels. This study investigated whether TRPV1 channels modulate baroreflex regulation of renal sympathetic nerve activity (RSNA) in a rat model of cisplatin-mediated renal injury. Rats were anaesthetised and prepared for measurement of mean arterial pressure (MAP), heart rate (HR) and RSNA 4 days after a single i.p. dose of cisplatin (5 mg kg-1). RSNA and HR baroreflex gain curves (BRC) were generated and the decrease in RSNA to volume expansion was determined during intrarenal capsazepine (CPZ, 15 µg kg-1 h-1) infusion. In the cisplatin group (MAP: 85 ± 13 mmHg; HR: 328 ± 17 bpm; RSNA: 0.83 ± 0.41 µV s), the slope and maximum gain of the BRC were approximately 50% lower (P = 0.015-0.033) than the control group (MAP: 78 ± 12 mmHg; HR: 352 ± 27 bpm; RSNA: 0.57 ± 0.36 µV s). Intrarenal CPZ infusion in the cisplatin group restored the slope (0.15 ± 0.04 vs. 0.09 ± 0.02, P = 0.014) of the RSNA BRC to near normal values. The RSNA response to volume expansion in the cisplatin group was enhanced following CPZ compared to vehicle infusion (-24 ± 14% vs. 1.7 ± 39%, P = 0.015). Intrarenal tumour necrosis factor α (TNF-α) infusion ( 2 µg kg-1 h-1) in normal rats decreased the slope of the BRC by 40% (P = 0.035) compared to vehicle infusion, which was slightly enhanced following intrarenal CPZ infusion. These findings demonstrate that TRPV1 channels contribute to the depressed baroreceptor control of RSNA in renal injury. Furthermore, the action of TNF-α in disrupting the baroreflex control mechanism partially involves TRPV1 channels.
Collapse
Affiliation(s)
| | - Ella Murphy
- Department of Physiology, University College Cork, Cork, Ireland
| | - Lauren Mulcahy
- Department of Physiology, University College Cork, Cork, Ireland
| | - Edward J Johns
- Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
2
|
Miyatsu M, Sukhbaatar A, Mishra R, Dorai A, Mori S, Kodama T. Optimization of lymphatic drug delivery system with carboplatin for metastatic lymph nodes. Sci Rep 2025; 15:16037. [PMID: 40341825 PMCID: PMC12062337 DOI: 10.1038/s41598-025-99602-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
Systemic chemotherapy is a common method for treatment of metastatic lymph nodes (LNs), but it has low tissue selectivity and high toxicity. Lymphatic drug delivery system (LDDS) is a novel approach to treat and prevent LN metastases. In a previous study, it was found that the increase of osmotic pressure with varied viscosity of the drug reagent enhances drug retention in the LNs. Here, we optimized the administration conditions to achieve a long-term therapeutic response by varying the dosages and injection rate, using the optimized osmotic pressure and varied viscosity of drug reagent for LDDS. A metastatic LN mouse model was created with MXH10/Mo/lpr mice. Luciferase labelled FM3A mouse mammary carcinoma cells were inoculated in subiliac LN (SiLN) to induce metastasis to the proper axillary LN (PALN). 4 days post tumor cell inoculation, carboplatin (CBDCA) was injected into the tumor-bearing SiLN under different administration conditions. Superior drug retention was observed in the group that received two-doses of CBDCA solution adjusted to an osmotic pressure and viscosity of 1897 kPa and 12 mPa·s, at an injection rate of 10 µL/min. Furthermore, this effect persisted for 42 days. This effect was accompanied by an upregulated expression of CD8, IL-12a, and IFN-γ in the spleen. These results suggest that dual-dose administration at 10 µL/min with hyper-osmotic and high viscosity formulation is optimal and can improve the long-term therapeutic efficacy of LN metastasis.
Collapse
Affiliation(s)
- Miriu Miyatsu
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
| | - Ariunbuyan Sukhbaatar
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
- Division of Oral and Maxillofacial Oncology and Surgical Sciences, Graduate School of Dentistry, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
| | - Radhika Mishra
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
| | - Arunkumar Dorai
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2‑1-1 Katahira, Aoba, Sendai, Miyagi, 980‑8577, Japan
| | - Shiro Mori
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
- Division of Oral and Maxillofacial Oncology and Surgical Sciences, Graduate School of Dentistry, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
| | - Tetsuya Kodama
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan.
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan.
| |
Collapse
|
3
|
Taliento C, Restaino S, Arcieri M, Scutiero G, Greco P, Scambia G, Vizzielli G. Cytoreductive Surgery Plus HIPEC in Recurrent or Newly Diagnosed Advanced Epithelial Ovarian Cancer: a Meta-analysis. Ann Surg Oncol 2025; 32:3648-3659. [PMID: 39904852 PMCID: PMC11976761 DOI: 10.1245/s10434-025-16979-6] [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: 12/20/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND In 2024, two randomized controlled trials (RCTs) were published, providing new high-quality evidence on HIPEC in epithelial ovarian cancer (EOC). Updating data on progression-free survival (PFS) and adverse events could offer a clearer understanding of the benefits and risks of HIPEC combined with cytoreductive surgery (CRS), with or without prior neoadjuvant chemotherapy (NACT). PATIENTS AND METHODS An electronic search was conducted using PubMed, Web of Science, EBSCO, and CENTRAL up to 23 November 2024. We only included RCTs reporting PFS and adverse events of interval or secondary CRS, with or without HIPEC, in newly diagnosed or recurrent EOC. RESULTS The meta-analysis included six RCTs. The addition of HIPEC to surgery significantly improved PFS in patients with newly diagnosed advanced-stage EOC who received NACT (HR 0.59; 95% CI 0.39-0.88; p = 0.01). No significant difference in PFS was observed between secondary CRS plus HIPEC and CRS alone in recurrent ovarian cancer without prior NACT (HR 1.22; 95% CI 0.82-1.83; p = 0.32). Regarding adverse events, a decrease in platelet count of any grade was more frequent in the HIPEC group (p = 0.03). The overall risk of acute kidney failure (AKF) was 10.6%, with a significantly higher incidence compared with CRS alone (p = 0.003). CONCLUSIONS The addition of HIPEC to CRS significantly improved PFS compared with surgery alone in patients with advanced EOC who received NACT. However, the treatment was associated with a higher incidence of AKF, which occurred in 10.6% of patients who underwent HIPEC.
Collapse
Affiliation(s)
- C Taliento
- Department of Medical Sciences, Institute of Obstetrics and Gynecology, University of Ferrara, Ferrara, Italy
| | - S Restaino
- Clinic of Obstetrics and Gynecology, "Santa Maria della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
- PhD School in Biomedical Sciences, Gender Medicine, Child and Women Health, University of Sassari, Sassari, Italy
| | - M Arcieri
- Clinic of Obstetrics and Gynecology, "Santa Maria della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - G Scutiero
- Department of Medical Sciences, Institute of Obstetrics and Gynecology, University of Ferrara, Ferrara, Italy
| | - P Greco
- Department of Medical Sciences, Institute of Obstetrics and Gynecology, University of Ferrara, Ferrara, Italy
| | - G Scambia
- Dipartimento per la salute della Donna e del Bambino e della Salute Pubblica, UOC Ginecologia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - G Vizzielli
- Clinic of Obstetrics and Gynecology, "Santa Maria della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy.
- Department of Medicine (DMED), University of Udine, Udine, Italy.
| |
Collapse
|
4
|
Niu Q, Zhang T. Synergistic mechanism of olaparib and cisplatin on breast cancer elucidated by network pharmacology. Sci Rep 2025; 15:14800. [PMID: 40295796 PMCID: PMC12038030 DOI: 10.1038/s41598-025-99741-y] [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: 12/02/2024] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
Cisplatin is an important chemotherapeutic agent is widely used to treat breast cancer and olaparib is the most studied PARP inhibitor to date. To explore the combinational anti-cancer potential and synergistic mechanism of Olaparib and cisplatin in breast cancer using network pharmacology. Drugs targets were drawn from PharmMapper, DrugBank, BATMAN-TCM, DrugCentral, STITCH, Swiss Institude of Bioinformatics and Comparative Toxigenomics Database (CTD). Breast cancer targets were extracted from OMIM, KEGG, GeneCards and DrugBank. The protein-protein interaction (PPI) network was created using the STRING database. Core targets were selected by incorporating PPI networks using Cytoscape 3.9.1. GO and KEGG analyses were performed to investigate common targets of Olaparib and cisplatin in breast cancer. The drug-disease-target network contained 82 nodes and 901 edges. The common targets obtained from Olaparib, cisplatin and breast cancer were identified, including ATK, p53, caspase-3, HSP90AA1, IL-6, IL-1β, ANXA5, SIRT1, caspase-9 and PARP. Core targets were primarily related to response to reactive oxygen species, regulation of apoptotic signaling pathway, regulation of DNA metabolic process, and regulation of cell activation. The KEGG pathway analysis revealed that Olaparib and cisplatin may affect breast cancer through platinum drug resistance and longevity regulating pathway. Furthermore, Olaparib combined with cisplatin downregulated the expression of caspase-3 and caspase-9 proteins and upregulated p53, PARP, and SIRT1 protein levels in MCF-7 cells. Functionally, the cooperative effect of Olaparib and cisplatin reduced the applied concentration of cisplatin and enhanced the anticancer effect, emphasizing the importance of combination therapy to overcome side effects and significantly improve the anticancer efficacy of cisplatin.
Collapse
Affiliation(s)
- Qiang Niu
- Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Tao Zhang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, Shandong Province, China.
| |
Collapse
|
5
|
Saad KM, Elmasry K, Baban B, Livingston MJ, Dong Z, Abdelmageed ME, Abdelaziz RR, Suddek GM, Elmarakby AA. Protocatechuic Acid Ameliorates Cisplatin-Induced Inflammation and Apoptosis in Mouse Proximal Tubular Cells. Int J Mol Sci 2025; 26:4115. [PMID: 40362355 PMCID: PMC12071929 DOI: 10.3390/ijms26094115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Cisplatin is a highly cytotoxic drug used for the treatment of head, neck, and soft tissue cancers; however, it has nephrotoxic effects that can lead to acute kidney injury. Protocatechuic acid (PCA) is a natural widely available antioxidant found in many fruits such as kiwi, mango, and berries. We have recently shown that PCA reduced renal injury in a mouse model of unilateral ureteral obstruction. The current study aims to investigate the protective effects of PCA in Cisplatin-induced inflammation in vitro in Boston University Mouse Proximal Tubular (BUMPT) cells. BUMPT cells were cultured in complete DMEM. Confluent BUMPT cells were then treated with 20 μM Cisplatin ± PCA 50 or 100 μM for 24 h. PCA treatment showed a dose-depending increase in % cell viability in Cisplatin-treated BUMPT cells. PCA treatment also dose-dependently decreased Cisplatin-induced increases in oxidative stress (ROS and TBARS), inflammation (p-NF-κB and IL-6), and apoptosis (cleaved caspase-3 and % of TUNEL+ cells) compared to Cisplatin-only treatment. The reduction in oxidative stress, inflammation, and apoptosis with PCA treatment in Cisplatin-treated BUMPT cells was associated with decreases in tubular physical barrier resistance and the expression of the tight junction protein zonula occludens-1 (ZO-1) when compared to BUMPT cells treated with Cisplatin alone. The current findings suggest that PCA treatment improves tubular barrier function in Cisplatin-treated BUMPT cells via reductions in oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Karim M. Saad
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Khaled Elmasry
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
- Department of Cellular Biology and Anatomy, The Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.J.L.); (Z.D.)
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
| | - Man J. Livingston
- Department of Cellular Biology and Anatomy, The Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.J.L.); (Z.D.)
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, The Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.J.L.); (Z.D.)
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Marwa E. Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Rania R. Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Ghada M. Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Ahmed A. Elmarakby
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| |
Collapse
|
6
|
Latif S, Sohaib M, Iqbal S, Mushtaq MH, Sultan MT. Nephroprotective Potential of Lyophilized Grewia asiatica Powder Against Renal Biomarkers and Inflammation In Vivo. J Nutr Metab 2025; 2025:3726752. [PMID: 40291032 PMCID: PMC12033067 DOI: 10.1155/jnme/3726752] [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: 05/20/2024] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction: Phalsa (Grewia asiatica) fruit is known for its rich nutritional profile and diverse pharmacological properties such as antioxidants, anti-inflammatory, and anti-cancer, making it a promising contender for preventive measures against cisplatin-induced acute kidney injury (AKI) in living organisms. Material and Methods: In the present study, rats were provided with different levels of lyophilized Grewia asiatica, i.e., 200, 300, and 400 mg/kg body weight along with control, fed on the basal diet. After trial completion, blood serum samples of rats subjected to renal biomarkers, hematology, and liver function tests, interleukin-6 (IL-6), whereas enzymes (alanine aminotransferase (ALT); sodium oxide dismutase, and glutathione) for kidney tissues along with photomicrographs for kidney tissue damage were measured. Results: The findings revealed that lyophilized Grewia asiatica provision effectively reduced renal biomarkers, blood urea nitrogen, and creatinine with AKI in the rats as well as treatments demonstrated significant improvements in antioxidant activity by reducing malonaldehyde levels and increasing the activity of glutathione, catalase, and superoxide dismutase in groups treated with dosages of 300 and 400 mg/kg powder. Conclusion: Grewia asiatica exhibited remarkable hepatoprotective properties by decreasing ALT and displayed anti-inflammatory properties, as evidenced by a substantial decrease in interleukin-6 serum levels. The study findings also added valuable insight into the multiform nephroprotective reverberation of lyophilized phalsa powder, emphasizing its plausible protective use in reducing cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Saima Latif
- Department of Food Science & Human Nutrition, University of Veterinary & Animal Sciences, Lahore 54000, Punjab, Pakistan
| | - Muhammad Sohaib
- Department of Food Science & Human Nutrition, University of Veterinary & Animal Sciences, Lahore 54000, Punjab, Pakistan
| | - Sanaullah Iqbal
- Department of Food Science & Human Nutrition, University of Veterinary & Animal Sciences, Lahore 54000, Punjab, Pakistan
| | - Muhammad Hassan Mushtaq
- Department of Epidemiology & Public Health, University of Veterinary and Animal Sciences, Lahore 54000, Punjab, Pakistan
| | | |
Collapse
|
7
|
Chen G. Investigation of polyvinylpyrrolidone-catechol-derived chitosan nanoconjugates allowed for kidney-targeted treatment of cisplatin-induced acute kidney injury and nursing care management. J Biomater Appl 2025; 39:1084-1096. [PMID: 39772864 DOI: 10.1177/08853282241304396] [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: 01/11/2025]
Abstract
Acute kidney injury (AKI) resulting from cisplatin (Cs) chemotherapy presents a significant challenge in clinical management. The study aimed to fabricate a novel compound Polyvinylpyrrolidone-catechol-derived chitosan nanoconjugates (PCChi-NC) for targeting Cs-induced AKI. Characterization studies utilizing UV-visible spectrophotometry, FT-IR, XRD, and TEM revealed a spherical morphology with diameters ranging from 20 to 60 nm. In vitro assessments utilizing HEK 293 cell lines demonstrated the biocompatibility of PCChi-NC without eliciting toxic effects. Furthermore, PCChi-NC exhibited a notable reduction in Cs-induced cell death in kidney cells, as evidenced by biomarker analysis. Anti-inflammatory analysis of mouse kidney homogenates revealed a decrease in TNF-α and IL-1β levels, indicative of the therapeutic efficacy of PCChi-NC in mitigating Cs-induced kidney inflammation. Moreover, In vivo, experimental analysis was evidenced by stable body weight and histopathological changes in mice. Our findings highlight the potential of PCChi-NC as a promising candidate for targeted therapy in Cs-induced AKI, owing to its unique renal targeting capacity.
Collapse
Affiliation(s)
- Guixian Chen
- Blood Purification Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, PR China
| |
Collapse
|
8
|
Hazem SH, Saad KM, Samaha MM. Protective effects of BTK inhibition by acalabrutinib on cisplatin-induced renal and testicular injury in mice: Modulation of mTOR/AMPK, NLRP3/GSDMD-N, and apoptotic pathways. Int Immunopharmacol 2025; 149:114256. [PMID: 39938312 DOI: 10.1016/j.intimp.2025.114256] [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: 10/17/2024] [Revised: 01/07/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Cisplatin-induced nephrotoxicity and testicular injury pose significant challenges during chemotherapy. AIM The current study evaluates the efficacy of acalabrutinib (ACB), a Bruton's tyrosine kinase inhibitor, in mitigating cisplatin-induced damage in renal and testicular tissues in mice. METHODS Testicular and renal toxicity was induced by a single I.P. injection of cisplatin (25 mg/kg). Mice were randomized into four groups: Normal (treated with vehicle), Cis (cisplatin + vehicle), Cis + ACB (6 mg/kg), and Cis + ACB (12 mg/kg). ACB was administered orally for three consecutive days, starting at Day 0 (1 h before single I.P. injection of cisplatin) and continued for Day 1 and Day 2. RESULTS ACB treatment (6 mg/kg and 12 mg/kg) significantly improved renal function by reducing serum creatinine, BUN, and KIM-1 levels, while also attenuating inflammation and apoptosis, as evidenced by decreased NLRP3, CD68, and caspase-3 expression. Additionally, it mitigated molecular damage by downregulating mTOR, AMPK, and GSDMD-N. In testicular tissues, ACB preserved structure, restored spermatogenesis, and improved sperm viability and testosterone levels. The protective effects were associated with reduced inflammation, apoptosis, and pyroptosis, indicated by lower levels of cathepsin L, NLRP3, and GSDMD-N. CONCLUSIONS These findings suggest that ACB offers a promising therapeutic approach to reduce the adverse effects of cisplatin, potentially enhancing the overall efficacy and safety of chemotherapy regimens.
Collapse
Affiliation(s)
- Sara H Hazem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516 Egypt.
| | - Karim M Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516 Egypt.
| | - Mahmoud M Samaha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516 Egypt.
| |
Collapse
|
9
|
Makled MN, Makled NN, Abdel-Rahman AM, Sharawy MH. Inhibition of p75 NTR/p53 axis by ambrisentan suppresses apoptosis and oxidative stress-mediated renal damage in a cisplatin AKI model. Chem Biol Interact 2025; 408:111408. [PMID: 39892498 DOI: 10.1016/j.cbi.2025.111408] [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: 11/21/2024] [Revised: 01/25/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
Cisplatin (CP) is a potent antineoplastic agent that triggers nephrotoxicity as a major adverse effect which can cause treatment interruptions and limitations to its clinical use. Nephrotoxicity associated with CP involves inflammation, oxidative stress, and apoptosis in kidney tubules. The objective of this work was to assess the effect of the blockade of endothelin-1 (ET-1) receptor with ambrisentan on altered renal function induced by CP. Swiss albino mice were assigned into control, CP, CP/Amb-5, and CP/Amb-10 groups. Ambrisentan improved kidney function (serum creatinine and BUN) and histopathological changes in comparison to CP-treated group. Ambrisentan significantly reduced protein expression of p75NTR and protein level of JNK influencing renal apoptosis as evidenced by reducing p53, caspase-3, and Bax levels and elevating Bcl-2 level (p < 0.05 vs CP group). Moreover, vasodilatory effect of ambrisentan was indicated by significant increase in level of vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase (eNOS) (p < 0.05 vs CP group). Ambrisentan also significantly restored oxidative balance in renal tissues as evidenced by reduced malondialdehyde and increased total antioxidant capacity and superoxide dismutase activity, in addition to decreasing nitric oxide levels (p < 0.05 vs CP group). This protective effect of ambrisentan might be mediated through the downregulation of death receptor, P75NTR that in turn restores renal blood flow and oxidative balance and regulates p53, VEGF/eNOS, NF-κB, and Bcl-2/Bax/caspase-3 signaling.
Collapse
Affiliation(s)
- Mirhan N Makled
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Noran N Makled
- Department of Urology, Faculty of Medicine, Damietta University, Damietta, Egypt; Department of Urology, Urology and Nephrology Center, Mansoura University, Mansoura, 35516, Egypt
| | - Ahmed M Abdel-Rahman
- Department of Nephrology, Urology and Nephrology Center, Mansoura University, Mansoura, 35516, Egypt
| | - Maha H Sharawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
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
|
Salah TM, Rabie MA, El Sayed NS. Renoprotective effect of berberine in cisplatin-induced acute kidney injury: Role of Klotho and the AMPK/mtor/ULK1/Beclin-1 pathway. Food Chem Toxicol 2025; 196:115179. [PMID: 39645019 DOI: 10.1016/j.fct.2024.115179] [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: 10/13/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Cisplatin (Cisp) is a potent cancer drug, but its use is limited by acute kidney injury (AKI). Autophagy, a process that removes damaged proteins and maintains cellular homeostasis, has been shown to alleviate Cisp-induced AKI. The balance between autophagy and apoptosis is crucial to kidney protection. Treatment with Berberine, known for its antioxidant and anti-inflammatory effects in nephrotoxicity models, was studied for its potential to enhance autophagy in Cisp-induced AKI. Treatment with Berberine (Berb) upregulated Klotho gene expression, enhancing autophagy as indicated by elevated protein levels of pS486-AMPK, pS638-ULK1, and Beclin-1, accompanied by a decrease in pS248-mTOR protein expression. Also, Berb mitigated oxidative stress by reducing elevated MDA levels and boosting SOD activity, which in turn suppressed inflammation by down-regulating HMGB1 and RAGE gene expression, as well as reducing pS536-NF-κB and IL-6 protein contents. Additionally, Berb reduced apoptosis by increasing Bcl-2 and decreasing Bax. This coordinated action preserved kidney function, evidenced by reductions in early injury markers (cystatin C, KIM-1, NGAL) and late markers (creatinine, BUN), along with attenuation of histopathological alterations. The use 3-MA, autophagy inhibitor, nullified these protective effects, highlighting Berb's role in promoting autophagy, reducing oxidative stress, inflammation, and apoptosis, and preserving renal health in Cisp-induced AKI.
Collapse
Affiliation(s)
- Tasneem M Salah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| |
Collapse
|
12
|
Zheng F, Lei JZ, Wang JX, Xu XY, Zhou B, Ge R, Dai M, Dong HK, Wu N, Li YH, Zhu GQ, Zhou YB. Crucial roles of asprosin in cisplatin-induced ferroptosis and acute kidney injury. Free Radic Biol Med 2025; 227:296-311. [PMID: 39653130 DOI: 10.1016/j.freeradbiomed.2024.12.024] [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: 06/09/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
Ferroptosis is a type of non-apoptotic regulated cell death characterized by iron accumulation and lipid peroxidation. Cisplatin is an effective chemotherapy drug with several serious side effects including acute kidney injury (AKI). Asprosin is a peptide contributing to metabolism regulation and metabolic disorders. This study aimed to determine the role and mechanism of asprosin in AKI. Cisplatin was used to induce cell damage in mouse renal tubular epithelial (TCMK-1) cells and AKI in C57BL/6 mice. Cisplatin caused asprosin upregulation in cisplatin-treated TCMK-1 cells and mice. In TCMK-1 cells, asprosin overexpression led to iron overload and lipid peroxidation, while asprosin knockdown attenuated cisplatin-induced iron overload, lipid peroxidation and ferroptosis. Exogenous asprosin promoted cell damage and ferroptosis, which were attenuated by ferroptosis inhibitors. Asprosin-induced iron overload, lipid peroxidation, cell damage and SMAD1/5/8 phosphorylation were prevented by bone morphogenetic protein (BMP) type I receptor inhibitor. Integrin antagonist prevented asprosin-induced SMAD1/5/8 phosphorylation, and asprosin can specifically bind to integrin β3. Inhibition of integrin β3 reduced the asprosin-induced increases in Fe2+ and MDA levels. Asprosin knockdown relieved cisplatin-induced hepcidin upregulation, while hepcidin knockdown attenuated asprosin-induced iron overload, lipid peroxidation and ferroptosis. In cisplatin-induced AKI mice, specific knockdown of asprosin in the kidney not only attenuated renal dysfunction and damage, but also alleviated iron overload, lipid peroxidation and ferroptosis. These results indicated that excessively increased asprosin promotes TCMK-1 cells ferroptosis and damage via integrin β3/BMP/hepcidin-mediated iron overload and lipid peroxidation. Silencing of asprosin attenuates renal injury and dysfunction in cisplatin-induced AKI by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Fen Zheng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jian-Zhen Lei
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jing-Xiao Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiao-Yu Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Bing Zhou
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, 241001, China
| | - Rui Ge
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Min Dai
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hong-Ke Dong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Nan Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Ye-Bo Zhou
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
13
|
Tekes E, Ickin Gulen M, Silan C, Guven Bagla A. Humic acid attenuates cisplatin-induced nephrotoxicity in rats. Drug Chem Toxicol 2025:1-9. [PMID: 39871462 DOI: 10.1080/01480545.2025.2453590] [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: 09/03/2024] [Revised: 11/24/2024] [Accepted: 01/09/2025] [Indexed: 01/29/2025]
Abstract
Cisplatin-induced nephrotoxicity, a major limitation of this chemotherapeutic agent, involves oxidative stress, inflammation, and apoptosis. This study investigated the potential renoprotective effects of humic acid in a rat model of cisplatin-induced nephrotoxicity. Forty-two male Wistar rats were assigned to six groups: control, humic acid, cisplatin, cisplatin + humic acid 10 mg/kg, cisplatin + humic acid 20 mg/kg, and cisplatin + humic acid 40 mg/kg. On day 7, the rats were sacrificed, and cardiac blood and kidneys were collected for biochemical and histopathological examinations. Humic acid administration significantly attenuated the cisplatin-induced increases in renal TNF-α and NF-κB levels, indicating a reduction in inflammation. Humic acid also ameliorated histopathological damage, including Bowman's capsule dilatation, tubular cell degeneration, and hemorrhage. However, humic acid did not significantly alter oxidative stress parameters or caspase-3 levels. Humic acid demonstrates a protective effect against cisplatin-induced nephrotoxicity in rats, primarily by mitigating the inflammatory response. While HA's beneficial effects on oxidative stress and apoptosis were limited in this study, its ability to reduce inflammation highlights its potential as a therapeutic strategy to mitigate cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Ender Tekes
- Department of Pharmacology, Faculty of Medicine, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Meltem Ickin Gulen
- Department of Histology & Embryology, Faculty of Medicine, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Coskun Silan
- Department of Pharmacology, Faculty of Medicine, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Aysel Guven Bagla
- Department of Histology & Embryology, Faculty of Medicine, Canakkale Onsekiz Mart University, Canakkale, Turkey
| |
Collapse
|
14
|
Khalil A, Al Toufaily S, Shebaby W, Hage ME, Mroue D, Faour W, Mroueh M. Lebanese Cannabis sativa L. extract protects from cisplatin-induced nephrotoxicity in mice by inhibiting podocytes apoptosis. J Cannabis Res 2025; 7:3. [PMID: 39819647 PMCID: PMC11737194 DOI: 10.1186/s42238-025-00260-4] [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: 11/05/2023] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Cisplatin is an anti-cancer drug used to treat a plethora of solid tumors. However, it is associated with dose dependent nephrotoxicity limiting its use as anticancer agent. OBJECTIVE The current study aimed to investigate the nephroprotective effect of native Lebanese Cannabis sativa in both in vitro and in vivo mice model of cisplatin-induced nephrotoxicity. METHODS Podocytes cell viability was assessed using MTS assay with cisplatin (30µM) in presence or absence of Cannabis oil extract (COE) at 0.5, 1 and 2µg/ml for 24h. Acute renal injury was established in adult female C57BL/6 mice with 20mg/kg, i.p. single dose cisplatin. Mice were divided into control group (vehicle), COE group, cisplatin group and cisplatin plus COE (2.5, 5 and 20mg/kg, i.p.). Animal body weight, serum creatinine, blood urea nitrogen (BUN), and proteinuria were measured. RESULTS Cell viability assay and western blot analysis revealed that COE prevented apoptosis induced by cisplatin in cultured immortalized rat podocytes. In addition, in vitro scratch assay demonstrated the ability of COE to promote and restore the migratory capacity of podocytes in cisplatin-treated cells. Interestingly, COE treatment improved urinary and serum parameters characterized by a significant decrease in serum creatinine, urea, and proteinuria at various COE doses. Western blot analysis showed that COE inhibited COX-2 protein induction as well as apoptosis marker production (Bax/Bcl2 ratio) in cisplatin-treated mice when compared to mice treated with cisplatin alone. CONCLUSION Collectively, the aforementioned findings indicate that COE could be a promising approach to protect against cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Alia Khalil
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box36, Byblos, Lebanon
| | - Sahar Al Toufaily
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
| | - Wassim Shebaby
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
| | - Marissa El Hage
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
- Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Dima Mroue
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos, Lebanon
| | - Wissam Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box36, Byblos, Lebanon.
| | - Mohamad Mroueh
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
| |
Collapse
|
15
|
Jana S, Mitra P, Panchali T, Khatun A, Das TK, Ghosh K, Pradhan S, Chakrabarti S, Roy S. Evaluating anti-inflammatory and anti-oxidative potentialities of the chloroform fraction of Asparagus racemosus roots against cisplatin induced acute kidney injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119084. [PMID: 39566865 DOI: 10.1016/j.jep.2024.119084] [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: 07/04/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute kidney injury (AKI), a global public health concern that increases the risk of death, end-stage renal disease, and prolonged hospital admissions. As of this point, supportive measures like fluid resuscitation and replacement therapy for renal failure are the only treatments available for treating AKI. Asparagus racemosus (AR) also known as Shatavari, belongs to family Liliaceae and is considered exceptional in Ayurvedic medicine due to its versatility in treating and preventing a variety of illnesses. AIM OF THE STUDY The purpose of this study is to determine the effectiveness of chloroform fraction of Asparagus racemosus (CFAR) against cisplatin (CP) induced AKI. MATERIALS AND METHODS HPLC was used to analyze the presence of bioactive phytocompounds in CFAR using standard quercetin. Further LC-MS study indicated the existence of different bioactive compounds. Normal Rat Kidney (NRK-52E) cells were used to study the nephroprotective effect of CFAR. Cells were untreated, treated or cotreated with CP (20 μM) and CFAR (5, 25, 50, 100, 200 and 400μg/mL) for 24 h. After 24 h of treatment, cell viability assay and assay of apoptosis parameters were performed. The CFAR at the dose of 50 mg, 100 mg and 200 mg/kg/day was administered orally for 15 days and acute kidney injury was induced in rats by intraperitoneal injection of CP (10 mg/kg body weight) at the 10th day of experimentation. Biochemical studies were performed to evaluate kidney function; protein expression by Western blot and mRNA expression of related gene were studied from the kidney tissues to evaluate the effects of CFAR. Histopathological analysis was done to investigate the structural abnormalities and fibrosis of renal tissues. RESULT Our result reported that CFAR contain many bioactive phytomolecules having many pharmacological properties. Cell viability assay and assay of apoptosis reported that different doses of CFAR could reduced CP-induced cell death and cell apoptosis. The levels of kidney injury markers (BUN, sCr and eGFR), inflammatory markers (Interleukin-18, KIM-1, Cys-C, NF-kB and NGAL), and antioxidant markers (SOD, GSH, CAT, Nrf2 and Bcl2) and lipid peroxidation (MDA) were settled to a normal level by the oral administration of high doses (100 and 200 mg/kg body weight) of CFAR after intraperitoneal injection of CP as suggested by biochemical, histopathological, protein and gene expression studies. CONCLUSION In conclusion, CFAR at the high doses (100 and 200 mg/kg body weight) could able to protect the kidneys from CP induced oxidative stress and inflammation due to presence of bioactive phytomolecules that prevent the activation of oxidative stress induced signalling cascades leading to kidney damage.
Collapse
Affiliation(s)
- Sahadeb Jana
- Biodiversity and Environmental Studies Research Centre affiliated to Vidyasagar University, Midnapore City College, Bhadutala, Paschim Medinipur, 721129, West Bengal, India; Department of Paramedical and Allied Health Sciences, Midnapore City College, Kuturiya, Bhadutala, Midnapore, 721129, India.
| | - Palash Mitra
- Biodiversity and Environmental Studies Research Centre affiliated to Vidyasagar University, Midnapore City College, Bhadutala, Paschim Medinipur, 721129, West Bengal, India; Department of Paramedical and Allied Health Sciences, Midnapore City College, Kuturiya, Bhadutala, Midnapore, 721129, India.
| | - Titli Panchali
- Biodiversity and Environmental Studies Research Centre affiliated to Vidyasagar University, Midnapore City College, Bhadutala, Paschim Medinipur, 721129, West Bengal, India; Department of Paramedical and Allied Health Sciences, Midnapore City College, Kuturiya, Bhadutala, Midnapore, 721129, India.
| | - Amina Khatun
- Biodiversity and Environmental Studies Research Centre affiliated to Vidyasagar University, Midnapore City College, Bhadutala, Paschim Medinipur, 721129, West Bengal, India; Department of Paramedical and Allied Health Sciences, Midnapore City College, Kuturiya, Bhadutala, Midnapore, 721129, India.
| | - Tridip Kumar Das
- Biodiversity and Environmental Studies Research Centre affiliated to Vidyasagar University, Midnapore City College, Bhadutala, Paschim Medinipur, 721129, West Bengal, India; Department of Paramedical and Allied Health Sciences, Midnapore City College, Kuturiya, Bhadutala, Midnapore, 721129, India.
| | - Kuntal Ghosh
- Department of Paramedical and Allied Health Sciences, Midnapore City College, Kuturiya, Bhadutala, Midnapore, 721129, India.
| | - Shrabani Pradhan
- Department of Paramedical and Allied Health Sciences, Midnapore City College, Kuturiya, Bhadutala, Midnapore, 721129, India.
| | - Sudipta Chakrabarti
- Department of Paramedical and Allied Health Sciences, Midnapore City College, Kuturiya, Bhadutala, Midnapore, 721129, India.
| | - Suchismita Roy
- Department of Paramedical and Allied Health Sciences, Midnapore City College, Kuturiya, Bhadutala, Midnapore, 721129, India.
| |
Collapse
|
16
|
Li C, Yang H, Wu Y, Zhou M, Luo H, Yuan P, Shen F. Carnosol alleviates cisplatin-induced acute kidney injury by regulating apoptosis and pyroptosis. Cell Biol Int 2025; 49:101-117. [PMID: 39523493 DOI: 10.1002/cbin.12258] [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: 04/22/2024] [Revised: 09/10/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
The use of the common anticancer drug cisplatin (CP) in clinical practice often leads to acute kidney injury (AKI); however, no protective therapy is available. Therefore, new drugs that reduce the nephrotoxicity induced by CP are urgently needed. Carnosol (CA) is an antioxidant found. We investigated the renoprotective effects of CA on CP-induced AKI in male C57BL/6 mice and HK2 cells. CA mitigated renal dysfunction, histopathological changes and tubular injury in vivo, as indicated by the expression of NGAL, KIM1 and HMGB1. Moreover, the numbers of apoptotic cells and the expression of apoptotic proteins were dramatically reduced after CA treatment in mouse kidneys and HK2 cells. CA significantly ameliorated CP-induced inflammation and decreased TNF-α and IL-1β levels in vivo and in vitro and macrophage infiltration in the mouse kidney. CA decreased the expression levels of p-p65/p65, NLRP3 and ASC, which indicates that CA suppressed the activation of the NF-κB/NLRP3 signaling axis induced by CP in vivo and in vitro. In addition, CA decreased the levels of certain protein in pyroptotic cells, as indicated by the expression of cleaved caspase-1, GSDMD, and mature IL-1β and IL-18 in vivo and in vitro. Finally, CA reduced the level of cleaved caspase-1, but those of GSDMD and NLRP3 protein were not significantly different after treatment with the NLRP3 inhibitor MCC950 and were elevated by the NLRP3 activator nigericin. In conclusion, this study revealed that CA protects against CP-induced AKI by decreasing apoptosis and NF-κB/NLRP3/GSDMD-mediated pyroptosis, which provides new insight into the prevention of AKI.
Collapse
Affiliation(s)
- Chunjie Li
- Xinxiang Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Hongyan Yang
- Xinxiang Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yuan Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Mingke Zhou
- Xinxiang Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Hengbiao Luo
- Xinxiang Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Peng Yuan
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Fengge Shen
- Xinxiang Key Laboratory of Molecular Neurology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
17
|
Taguchi H, Sumi D, Uemura A, Matsumoto K, Fujishiro H. Cisplatin caused highly delayed cytotoxicity in the immortalized cells derived from S3 segment of mouse kidney proximal tubules. Toxicol Appl Pharmacol 2025; 494:117171. [PMID: 39592085 DOI: 10.1016/j.taap.2024.117171] [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: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
Anti-cancer drug cisplatin (CDDP) causes severe acute kidney injury (AKI). CDDP-induced AKI does not occur immediately after administration, but rather 6 to 10 days after administration. However, the mechanism underling the delayed renal injury by CDDP is not well understood. In a previous investigation using immortalized cells derived from the S1, S2, and S3 segments of the proximal tubules, we found that S3 cells were more sensitive to CDDP than S1 and S2 cells. In this study, we examined whether S1, S2, and S3 cells would be useful in elucidating the mechanism of CDDP-induced delayed renal injury and whether the high sensitivity of S3 cells contributes to CDDP-induced delayed renal injury. Measurement of platinum (Pt) content by ICP-MS showed that Pt accumulation peaked at 15 min after CDDP exposure in each cell type. Even when the medium was replaced with CDDP-free medium after the 15-min CDDP exposure and the cells were further incubated, delayed cytotoxicity was still observed. The S3 cells exhibited greater sensitivity to CCDP than the S1 and S2 cells at all time points after the medium change. To investigate the mechanism of the CDDP-induced delayed cytotoxicity, we examined the cell cycle distribution of cells after CDDP exposure. The results showed that CDDP-induced perturbation of cell cycle was greater in S3 than in S1 and S2 cells. These results suggest that perturbation of the cell cycle in S3 cells due to enhanced CDDP-DNA adduct formation contributes to the high susceptibility of S3 cells to CDDP-induced delayed cytotoxicity.
Collapse
Affiliation(s)
- Hiroki Taguchi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Daigo Sumi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Ayumi Uemura
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Kanako Matsumoto
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Hitomi Fujishiro
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| |
Collapse
|
18
|
Wang H, Wu X, Li Z, Rong K, Gao S, Tang W, Zhang J. Novel Glycyrrhetin Ureas Possessing 2-Hydroxy-3-enone A Ring: Modification, Anti-inflammatory Activity, and Targeted STING for the Remedy of Acute Kidney Injury. ACS OMEGA 2024; 9:48821-48834. [PMID: 39676967 PMCID: PMC11635493 DOI: 10.1021/acsomega.4c09003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024]
Abstract
Glycyrrhetin urea has emerged as a privileged scaffold with anti-inflammatory activity for the treatment and prevention of acute kidney injury (AKI). In this study, structural modifications of the A ring of glycyrrhetinic acid yielded a series of urea derivatives, among which compound 7o exhibited the most promising anti-inflammatory activity. 7o was confirmed to interact with STING through a cellular heat shift assay and to inhibit the STING/NF-κB pathway in RAW264.7 cells. It acted on the STING pathway, inhibited NF-κB phosphorylation, and subsequently reduced the level of release of inflammatory factors. Additionally, 7o significantly increased the survival rate of renal tubular epithelial cells, demonstrating a protective effect against cisplatin-induced cell death and mitigating inflammation activation. The in vivo AKI mouse model showed that 7o significantly downregulated serum creatinine (Scr), blood urea nitrogen (BUN), and levels of inflammatory factors (IL-1β, IL-6, and TNF-α), thereby improving renal function. Morphological analysis revealed that 7o attenuated the cisplatin-induced renal tubular injury. Therefore, 7o represents a promising lead for the prevention and treatment of AKI.
Collapse
Affiliation(s)
- Hongbo Wang
- Department
of Pharmacy, Shandong Medical College, Linyi 276000, People’s Republic of China
- School
of Pharmacy, Anhui Medical University, Hefei 230032, People’s Republic of China
| | - Xiaoming Wu
- School
of Pharmacy, Anhui Medical University, Hefei 230032, People’s Republic of China
| | - Ziyun Li
- School
of Pharmacy, Anhui Medical University, Hefei 230032, People’s Republic of China
| | - Kuanrong Rong
- School
of Pharmacy, Anhui Medical University, Hefei 230032, People’s Republic of China
| | - Shan Gao
- School
of Pharmacy, Anhui Medical University, Hefei 230032, People’s Republic of China
| | - Wenjian Tang
- School
of Pharmacy, Anhui Medical University, Hefei 230032, People’s Republic of China
| | - Jing Zhang
- Anhui
Province Key Laboratory of Occupational Health, Anhui No. 2 Provincial People’s Hospital, Hefei 230041, People’s Republic of China
| |
Collapse
|
19
|
Manavi MA, Fathian Nasab MH, Mohammad Jafari R, Dehpour AR. Mechanisms underlying dose-limiting toxicities of conventional chemotherapeutic agents. J Chemother 2024; 36:623-653. [PMID: 38179685 DOI: 10.1080/1120009x.2023.2300217] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Dose-limiting toxicities (DLTs) are severe adverse effects that define the maximum tolerated dose of a cancer drug. In addition to the specific mechanisms of each drug, common contributing factors include inflammation, apoptosis, ion imbalances, and tissue-specific enzyme deficiencies. Among various DLTs are bleomycin-induced pulmonary fibrosis, doxorubicin-induced cardiomyopathy, cisplatin-induced nephrotoxicity, methotrexate-induced hepatotoxicity, vincristine-induced neurotoxicity, paclitaxel-induced peripheral neuropathy, and irinotecan, which elicits severe diarrhea. Currently, specific treatments beyond dose reduction are lacking for most toxicities. Further research on cellular and molecular pathways is imperative to improve their management. This review synthesizes preclinical and clinical data on the pharmacological mechanisms underlying DLTs and explores possible treatment approaches. A comprehensive perspective reveals knowledge gaps and emphasizes the need for future studies to develop more targeted strategies for mitigating these dose-dependent adverse effects. This could allow the safer administration of fully efficacious doses to maximize patient survival.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Thompson LE, Joy MS. Understanding Cisplatin Pharmacokinetics and Toxicodynamics to Predict and Prevent Kidney Injury. J Pharmacol Exp Ther 2024; 391:399-414. [PMID: 39322416 PMCID: PMC11585315 DOI: 10.1124/jpet.124.002287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
Cisplatin is a common platinum-based chemotherapeutic that induces acute kidney injury (AKI) in about 30% of patients. Pharmacokinetic/toxicodynamic (PKTD) models of cisplatin-induced AKI have been used to understand risk factors and evaluate potential mitigation strategies. While both traditional clinical biomarkers of kidney function [e.g., serum creatinine (SCr), blood urea nitrogen (BUN), estimated glomerular filtration rate (eGFR), and creatinine clearance (CrCl)] and newer subclinical biomarkers of kidney injury [e.g., urinary kidney injury molecule 1 (KIM-1), beta-2 microglobulin (B2M), neutrophil gelatinase-associated lipocalin (NGAL), calbindin, etc.] can be used to detect cisplatin-induced AKI, published PKTD models are limited to using only traditional clinical biomarkers. Previously identified risk factors for cisplatin nephrotoxicity have included dose, age, sex, race, body surface area, genetics, concomitant medications, and comorbid conditions. However, the relationships between concentrations and the pharmacokinetics (PK) of platinum and biomarkers of kidney injury have not been well elucidated. This review discusses the evaluation of cisplatin-induced nephrotoxicity in clinical studies, mouse models, and in vitro models, and examines the available human PK and toxicodynamic (TD) data. Improved understanding of the relationships between platinum PK and TD, in the presence of identified risk factors, will enable the prediction and prevention of cisplatin kidney injury. SIGNIFICANCE STATEMENT: As cisplatin treatment continues to cause AKI in a third of patients, it is critical to improve the understanding of the relationships between platinum PK and nephrotoxicity as assessed by traditional clinical and contemporary subclinical TD markers of kidney injury. Prediction and prevention of cisplatin-induced nephrotoxicity will be advanced by the evolving development of PKTD models that incorporate kidney injury biomarkers with enhanced sensitivity and include covariates that can impact risk of developing cisplatin-induced AKI.
Collapse
Affiliation(s)
- Lauren E Thompson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (L.E.T., M.S.J.), University of Colorado Cancer Center (M.S.J.), and Division of Renal Diseases and Hypertension (M.S.J.), University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Melanie S Joy
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (L.E.T., M.S.J.), University of Colorado Cancer Center (M.S.J.), and Division of Renal Diseases and Hypertension (M.S.J.), University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
21
|
Zhang D, Jin R, Li G, Zhang C, Zhou Y. Ameliorative effect of rutecarpine supplementation against cisplatin-induced nephrotoxicity in rats via inhibition of monocyte chemoattractant protein-1, intercellular adhesion molecule-1, high-mobility group box 1, and nuclear factor kappa B. Biotechnol Appl Biochem 2024. [PMID: 39506243 DOI: 10.1002/bab.2692] [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: 08/06/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/08/2024]
Abstract
Cisplatin, the pioneering heavy metal compound, stands out as a potent drug for the treatment of various solid tumors. However, its clinical utility is hampered by notable toxicity and adverse effects, particularly nephrotoxicity. The potency of rutecarpine, a phytochemical, in mitigating cisplatin-induced nephrotoxicity was assessed in the present study. In this experimental setup, healthy male Wistar rats were grouped into four and Group I rats served as the control group, receiving only vehicle control. Group II rats were subjected to cisplatin treatment alone, administered intraperitoneally at a dosage of 7 mg/kg body weight on the 19th, 20th, and 21st days. Group III and IV rats were orally administered with rutecarpine at doses of 10 and 20 mg/kg body weight, respectively, starting from Day 1 and continuing daily for 21 days. Additionally, they were injected intraperitoneally with cisplatin at the same dosage and schedule as Group II. Relative kidney weight and renal biochemical markers blood urea nitrogen, lactate dehydrogenase, serum urea, and creatinine were measured to assess rutecarpine inhibitory potency against cisplatin toxicity. Markers of oxidative damage and antioxidants levels were quantified in the ruteacarpine- and cisplatin-treated rats. The study investigated the anti-inflammatory property of rutecarpine in cisplatin-induced nephrotoxicity by analyzing inflammatory cytokines. Renal tissue levels of monocyte chemoattractant protein-1, intercellular adhesion molecule-1, high-mobility group box 1, and nuclear factor kappa B, key markers of nephrotoxicity, were quantified to assess rutecarpine's potential to mitigate cisplatin-triggered damage. Histopathological examinations were performed to confirm the impact of rutecarpine against cisplatin-induced nephrotoxicity. Treatment with rutecarpine notably reduced renal biochemical markers, prevented renal edema, and attenuated oxidative stress-induced damage in cisplatin-treated rats. Both inflammatory and nephrotoxicity markers showed significant decreases in rats treated with rutecarpine along with cisplatin. Histological analysis affirmed that rutecarpine pretreatment effectively prevented cisplatin-induced nephrotoxicity. The study findings demonstrate that rutecarpine ameliorates cisplatin-triggered nephrotoxicity through its antioxidant and anti-inflammatory properties, suggesting that rutecarpine supplementation alongside cisplatin treatment could potentially reduce nephrotoxicity in cancer patients.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Emergency, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Rui Jin
- Department of Emergency, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu, China
| | - Guoxing Li
- Outpatient Department, General Hospital of Western Theater of Chinese People's Liberation Army, Chengdu, Sichuan, China
| | - CaiFeng Zhang
- Department of Encephalopathy, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Yanhong Zhou
- Department of Emergency, Banan Hospital Affiliated to Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Alqahtani MM. Cisplatin-Induced Renal Failure Measured by Glomerular Filtration Rate (GFR) with 99mTc-DTPA Scans in Cancer Patients: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2024; 14:2468. [PMID: 39594134 PMCID: PMC11593308 DOI: 10.3390/diagnostics14222468] [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: 09/03/2024] [Revised: 10/27/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Cisplatin is a potent agent commonly used to treat cancer, but its effects pose a significant risk to renal function. Therefore, the present study aimed to evaluate the impact of cisplatin on renal function as measured by glomerular filtration rate (GFR) using diethyltriamine-penta-acetic acid (DTPA) renal scintigraphy. Methods: Extensive literature searches were performed using PRISMA guidelines that investigated cisplatin-induced renal failure by measuring GFR with DTPA. Eligible studies were included based on predefined criteria. Data on GFR, serum creatinine levels, and acute kidney injury (AKI) before and after cisplatin therapy were extracted and analyzed. A meta-analysis was performed utilizing RevMan 5.4 to determine the overall effect of cisplatin on GFR before and after treatment. For non-randomized controlled trials (RCTs), quality assessment was performed using the Newcastle-Ottawa Scale, while for RCT, the Cochrane risk of bias tool was utilized. Results: Initially, 1003 studies were searched from different databases, including ScienceDirect, PubMed, Scopus, Google Scholar, and The Cochrane Library, and after screening, 8 studies (PubMed, Scopus, and GoogleS cholar) with 489 patients were found eligible for inclusion in the present study. Cisplatin was administrated with varying doses ranging from 20 mg/m2 to 114.02 mg/m2. The findings underscore the nephrotoxic effects of cisplatin, a widely used chemotherapeutic agent, as demonstrated by the significant decline in GFR observed across multiple treatment cycles, and these findings were also supported by the findings of a meta-analysis that showed a significant (p < 0.01) difference between peri- and post-treatment GFR level with 37.06 (95% CI, 10.90-63.23) effect size and 96% heterogeneity. In addition, the included studies were found to be of high quality. Conclusions: Cisplatin significantly affects renal function, as evidenced by a decrease in GFR measured with DTPA. The findings underscore the importance of the routine monitoring of GFR to detect early renal injury and guide treatment modification. Future research should focus on strategies to reduce cisplatin-induced toxicity and explore alternative therapies with reduced renal risk.
Collapse
Affiliation(s)
- Mansour M Alqahtani
- Department of Radiological Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| |
Collapse
|
23
|
Ahmed HA, Shaaban AA, Ibrahim TM, Makled MN. G protein-coupled estrogen receptor activation attenuates cisplatin-induced CKD in C57BL/6 mice: An insight into sex-related differences. Food Chem Toxicol 2024; 194:115079. [PMID: 39491767 DOI: 10.1016/j.fct.2024.115079] [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: 08/25/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Gender contributes to differences in incidence and progression of chronic kidney disease (CKD) post-cisplatin therapy. This study aims at investigating the potential effect of G1 compound, a GPER agonist, on attenuating cisplatin-induced CKD. To induce CKD in male, intact female, and ovariectomized (OVX) mice, CKD was induced by injecting two cycles of 2.5 mg/kg cisplatin with a 16-day recovery period between cycles). G1 (50 or 100 μg/kg was administered daily for 6 weeks. Severity of renal damage was more pronounced in males than females. Interestingly, OVX resulted in renal damage that is non-significant compared to males and significantly higher than females. G1 improved renal function and blood flow as evidenced by reduction of serum creatinine and elevation of creatinine clearance, NO production, and reduction of ET1. This renoprotective effect could be attributed to its immunomodulatory effect regulated by TGF-β that shifted the balance to favor anti-inflammatory cytokine production (increased IL-10) rather than pro-inflammatory cytokines (decreased Th17 expression). Reduction of TGF-β activation also inhibited epithelial-to-mesenchymal transition that eventually ameliorated CKD development. Antioxidant potential of G1 has been demonstrated by upregulation of Nrf2 and subsequent antioxidant enzymes. These data suggest that G1 could be a promising therapeutic tool to attenuate CP-induced CKD.
Collapse
Affiliation(s)
- Hala A Ahmed
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, Delta University for Science and Technology, Egypt; Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Egypt
| | - Ahmed A Shaaban
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Egypt
| | - Tarek M Ibrahim
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Egypt
| | - Mirhan N Makled
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Egypt.
| |
Collapse
|
24
|
Bakr AF, El-Shiekh RA, Mahmoud MY, Khalil HMA, Alyami MH, Alyami HS, Galal O, Mansour DF. Efficacy of Quercetin and Quercetin Loaded Chitosan Nanoparticles Against Cisplatin-Induced Renal and Testicular Toxicity via Attenuation of Oxidative Stress, Inflammation, and Apoptosis. Pharmaceuticals (Basel) 2024; 17:1384. [PMID: 39459023 PMCID: PMC11510010 DOI: 10.3390/ph17101384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/16/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Flavonoids, including quercetin, have attracted much attention due to their potential health-promoting effects. METHODS The current experiment aims to see whether quercetin (QUE) in nanoparticle form could mitigate testicular and renal toxicity caused by cisplatin (CIS) more effectively than normally formulated QUE. Rats were randomly treated with CIS alone or in combination with QUE or QUE.NPs (Quercetin-loaded chitosan nanoparticles) for 4 weeks. QUE and QUE.NPs were given orally (10 mg/kg, three times a week), while CIS was given intraperitoneally (2 mg/kg, twice a week). RESULTS Compared to QUE- and CIS + QUE.NP-treated rats, CIS exposure induced anxiety and emotional stress as well as promoted oxidative stress in both testicular and renal tissues. Moreover, CIS reduced serum testosterone levels and diminished testicular IL-10, as well as CIS-induced renal failure, as indicated by hypokalemia, and increased levels of creatinine, urea, sodium, IL-18, and KIM-1. Further, severe histological changes were observed in the testis and kidney of CIS-intoxicated rats. Regarding immunohistochemical staining, CIS significantly upregulated Bax, downregulated Bcl-2, and moderately enhanced PCNA expression. CONCLUSIONS Our findings suggest that both QUE and QUE.NPs modulated emotional disturbance and improved testicular and renal functions via modulation of oxidation, inflammation, and apoptosis. However, QUE.NPs performed better than QUE-treated rats.
Collapse
Affiliation(s)
- Alaa F. Bakr
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Riham A. El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt;
| | - Mohamed Y. Mahmoud
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Heba M. A. Khalil
- Department of Veterinary Hygiene and Management, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
- Faculty of Veterinary Medicine, King Salman International University, South Sinai, Ras Sudr 43312, Egypt
| | - Mohammad H. Alyami
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 66462, Saudi Arabia;
| | - Hamad S. Alyami
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 66462, Saudi Arabia;
| | - Omneya Galal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza 12581, Egypt;
| | - Dina F. Mansour
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Cairo 12622, Egypt;
- Department of Pharmacy, Faculty of Pharmacy, Galala University, Attaka, Suez 43511, Egypt
| |
Collapse
|
25
|
Kimura H, Kamiyama K, Imamoto T, Takeda I, Kobayashi M, Takahashi N, Kasuno K, Sugaya T, Iwano M. Dichloroacetate reduces cisplatin-induced apoptosis by inhibiting the JNK/14-3-3/Bax/caspase-9 pathway and suppressing caspase-8 activation via cFLIP in murine tubular cells. Sci Rep 2024; 14:24307. [PMID: 39414949 PMCID: PMC11484893 DOI: 10.1038/s41598-024-75229-z] [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: 04/27/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024] Open
Abstract
Cisplatin-induced injury to renal proximal tubular cells stems from mitochondrial damage-induced apoptosis and inflammation. Dichloroacetate (DCA), a pyruvate dehydrogenase kinase (PDK) inhibitor, a potential generator of ROS and ATP, protects against cisplatin-induced nephrotoxicity by promoting the TCA cycle. However, its effects on apoptotic pathways and ROS production in renal tubular cells remain unclear. Here, we investigated the detailed molecular mechanisms of the DCA's effects by immunoblot, RT-PCR, RNA-sequencing, and RNA-silencing in a murine renal proximal tubular (mProx) cell line and mouse kidneys. In mProx cells, DCA suppressed cisplatin-induced apoptosis by attenuating the JNK/14-3-3/Bax/caspase-9 and death receptor/ligand/caspase-8 pathways without impeding inflammatory signaling. RNA-sequencing demonstrated that DCA increased the cisplatin-reduced expression of cFLIP, a caspase-8 inactivator, and decreased the expression of almost all oxidative phosphorylation (OXPHOS) genes. DCA also increased NF-kB activation and ROS production, probably enhancing the cFLIP induction and OXPHOS gene reduction, respectively. Furthermore, cFLIP silencing weakened the DCA's anti-apoptotic effects. Finally, in mouse kidneys, DCA attenuated cisplatin-caused injuries such as functional and histological damages, caspase activation, JNK/14-3-3 activation, and cFLIP reduction. Conclusively, DCA mitigates cisplatin-induced nephrotoxicity by attenuating the JNK/14-3-3/Bax/caspase-9 pathway and inhibiting the caspase-8 pathways via cFLIP induction, probably outweighing the cisplatin plus DCA-derived cytotoxic effects including ROS.
Collapse
Affiliation(s)
- Hideki Kimura
- Department of Clinical Laboratory, University of Fukui Hospital, 23-3 Matsuoka-shimoaizuki, Eiheiji-Cho, Yoshida, Fukui, 910-1193, Japan.
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan.
| | - Kazuko Kamiyama
- Department of Clinical Laboratory, University of Fukui Hospital, 23-3 Matsuoka-shimoaizuki, Eiheiji-Cho, Yoshida, Fukui, 910-1193, Japan
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Toru Imamoto
- Department of Clinical Laboratory, University of Fukui Hospital, 23-3 Matsuoka-shimoaizuki, Eiheiji-Cho, Yoshida, Fukui, 910-1193, Japan
| | - Izumi Takeda
- Department of Clinical Laboratory, University of Fukui Hospital, 23-3 Matsuoka-shimoaizuki, Eiheiji-Cho, Yoshida, Fukui, 910-1193, Japan
| | - Mamiko Kobayashi
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Naoki Takahashi
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Kenji Kasuno
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | | | - Masayuki Iwano
- Division of Nephrology, Department of General Medicine, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
26
|
Botrous S, Elmaghraby A, Achy SE, Mustafa Y, Abdel-Rahman S. Artemisinin pre-treatment fore cisplatin dosage enhances high grade urothelial carcinoma treatment in male albino mice via reverse gene expression modulation of FGFR3, HRAS, P53 and KDM6A. BMC Cancer 2024; 24:971. [PMID: 39118085 PMCID: PMC11308388 DOI: 10.1186/s12885-024-12683-y] [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: 04/21/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Urinary bladder cancer, is the 10th most common global cancer, diagnosed in over 600,000 people causing 200,000 deaths annually. Artemisinin and its derivatives are safe compounds that have recently been proven to possess potent anti-tumor effects in vivo, through inhibition of cancer cell growth. The aim of this study is to assess the efficiency of artemisinin as a cancer treatment alone and as a pre-treatment fore cisplatin therapy for high grade urothelial carcinoma. METHODS Sixty male albino mice were divided into six groups, and BBN was used to induce urinary bladder cancer. Blood samples were tested for renal functions and complete blood counts, kidney and urinary bladder tissues were harvested for histopathological examination. Total RNAs from urinary bladder tissues was collected, and gene expression of FGFR3, HRAS, P53, and KDM6A was quantified using qRT-PCR. RESULTS Compared to the induced cancer group, the results revealed that FGFR3 expression levels were down-regulated in the induced cancer group treated by artemisinin only and the induced cancer group pre-treated with artemisinin prior to cisplatin by ~ 0.86-fold and 0.4-folds, respectively, aligning with HRAS down-regulation by ~ 9.54-fold and 9.05-fold, respectively. Whereas, P53 expression levels were up-regulated by ~ 0.68-fold and 0.84-fold, respectively, in parallel with KDM6A expression, which is up-regulated by ~ 0.95-folds and 5.27-folds, respectively. Also, serum creatinine and urea levels decreased significantly in the induced cancer group treated by artemisinin alone and the induced cancer group pre-treated with artemisinin prior to cisplatin, whereas the induced cancer group treated by cisplatin their levels increased significantly. Moreover, Hb, PLT, RBC, and WBC counts improved in both cancer groups treated by artemisinin alone and pre-treated with artemisinin prior to cisplatin. Histologically, in kidney tissues, artemisinin pre-treatment significantly reduced renal injury caused by cisplatin. While Artemisinin treatment for cancer in bladder tissues reverted invasive urothelial carcinoma to moderate urothelial dysplasia. CONCLUSIONS This study indicates that artemisinin demonstrated a significant effect in reversal of the multi-step carcinogenesis process of high grade urothelial carcinoma and could enhance the effect of cisplatin therapy using artemisinin pre-treatment.
Collapse
Affiliation(s)
- Silvia Botrous
- Department of Genetics, Faculty of Agriculture, Alexandria University, Alexandria, Egypt.
| | - Ayaat Elmaghraby
- Department of Nucleic Acid Research, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, Alexandria, Egypt
| | - Samar El Achy
- Department of Surgical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Yehia Mustafa
- Department of Genetics, Faculty of Agriculture, Alexandria University, Alexandria, Egypt
| | - Salah Abdel-Rahman
- Department of Nucleic Acid Research, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, Alexandria, Egypt.
| |
Collapse
|
27
|
Ejtema M, Chegeni N, Zarei-Ahmady A, Salehnia Z, Shamsi M, Razmjoo S. Exploring the combined impact of cisplatin and copper-cysteamine nanoparticles through Chemoradiation: An in-vitro study. Toxicol In Vitro 2024; 99:105878. [PMID: 38906201 DOI: 10.1016/j.tiv.2024.105878] [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: 03/17/2024] [Revised: 05/28/2024] [Accepted: 06/14/2024] [Indexed: 06/23/2024]
Abstract
Copper-Cysteamine nanoparticles (Cu-Cy NPs) have emerged as promising radiosensitizers in cancer treatment. This study aims to investigate the combined therapeutic effect of these nanoparticles and cisplatin using a clinical linear accelerator to enhance the efficacy of chemoradiation therapy for cervical cancer. Following successful synthesis and characterization of Cu-Cy NPs, the cytotoxicity effect of these nanoparticles and cisplatin in various concentrations was evaluated on HeLa cancer cells, individually and in combination. Additionally, the radiobiological effects of these agents were investigated under a 6MV linear accelerator. At a concentration of 25 mg/L, Cu-Cy NPs displayed no significant cytotoxicity toward HeLa cancer cells. However, when combined with 2Gy X-ray irradiation at this concentration, the nanoparticles demonstrated a potent radiosensitizing effect. Notably, cell viability and migration rate in the combination group (Cu-Cy NPs + cisplatin + radiation) were significantly reduced compared to the radiation-alone group. Additionally, the combination treatment induced a significantly higher rate of apoptosis compared to the radiation-alone group. Overall, Cu-Cy NPs exhibited a significant dose-dependent synergistic enhancement of radiation efficacy when combined with cisplatin under X-ray exposure, and may provide a promising approach to improve the therapeutic effect of conventional radiation therapy.
Collapse
Affiliation(s)
- Mahsa Ejtema
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Medical Physics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nahid Chegeni
- Department of Medical Physics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Amanollah Zarei-Ahmady
- Marine Pharmaceutical Science Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zeinab Salehnia
- Department of Radiology, School of Paramedicine, Behbahan University of Medical Sciences, Behbahan, Iran
| | - Masoumeh Shamsi
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sasan Razmjoo
- Department of Clinical Oncology, Golestan Hospital, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
28
|
Zheng D, Ruan X, Wu Q, Qiu Y, Ruan S. Yishen Jiangzhuo decoction attenuates cisplatin‑induced acute kidney injury by inhibiting inflammation, oxidative stress and apoptosis through the TNF signal pathway. Exp Ther Med 2024; 28:331. [PMID: 38979022 PMCID: PMC11228562 DOI: 10.3892/etm.2024.12620] [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: 11/14/2023] [Accepted: 04/22/2024] [Indexed: 07/10/2024] Open
Abstract
The present study aimed to investigate the therapeutic effects and mechanisms of Yishen Jiangzhuo decoction (YSJZD) in a mouse model of cisplatin-induced acute kidney injury (AKI). The mice were divided into the NC, cisplatin and cisplatin + YSJZD groups. A concentration-dependent effect of YSJZD on cisplatin-induced AKI was observed and the optimal concentration for intervention was calculated. Changes in blood urea nitrogen and serum creatinine levels combined with hematoxylin and eosin and periodic acid-Schiff staining and transmission electron microscopy observations indicated that YSJZD enhanced renal function, reduced pathological injury and protected renal tubular epithelial cells in cisplatin-induced AKI mice. The results of the transcriptomic and enrichment analyses showed that the mechanisms of YSJZD may be associated with inflammation, oxidation, apoptosis and the TNF signal pathway. Immunofluorescence, oxidative stress index, terminal deoxynucleotidyl transferase dUTP nick end labeling assay and western blotting revealed that YSJZD downregulated apoptosis in the renal tissues of AKI mice and further decreased the expression levels of p-p65, p-p38 MAPK, TNF-α, cleaved-caspase-3 and malondialdehyde, while increasing the levels of NAD-dependent protein deacetylase sirtuin-3, glutathione and superoxide dismutase. Overall, the results showed that YSJZD could effectively abrogate cisplatin-induced AKI in mice through mechanisms primarily related to its anti-inflammatory, antioxidative and antiapoptotic effects by inhibited the TNF signal pathway. YSJZD warrants further investigation as a clinical empirical prescription.
Collapse
Affiliation(s)
- Dengyong Zheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Department of Nephrology, The Second Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Xinglin Ruan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Qiang Wu
- Department of Nephrology, The Second Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Yuliang Qiu
- Department of Nephrology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350004, P.R. China
| | - Shiwei Ruan
- Department of Nephrology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350004, P.R. China
| |
Collapse
|
29
|
Oliveira CA, Mercês ÉAB, Portela FS, Malheiro LFL, Silva HBL, De Benedictis LM, De Benedictis JM, Silva CCDE, Santos ACL, Rosa DP, Velozo HS, de Jesus Soares T, de Brito Amaral LS. An integrated view of cisplatin-induced nephrotoxicity, hepatotoxicity, and cardiotoxicity: characteristics, common molecular mechanisms, and current clinical management. Clin Exp Nephrol 2024; 28:711-727. [PMID: 38678166 DOI: 10.1007/s10157-024-02490-x] [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: 10/11/2023] [Accepted: 03/18/2024] [Indexed: 04/29/2024]
Abstract
Cisplatin (CP) is a chemotherapy drug widely prescribed to treat various neoplasms. Although fundamental for the therapeutic action of the drug, its cytotoxic mechanisms trigger adverse effects in several tissues, such as the kidney, liver, and heart, which limit its clinical use. In this sense, studies point to an essential role of damage to nuclear and mitochondrial DNA associated with oxidative stress, inflammation, and apoptosis in the pathophysiology of tissue injuries. Due to the limitation of effective preventive and therapeutic measures against CP-induced toxicity, new strategies with potential cytoprotective effects have been studied. Therefore, this article is timely in reviewing the characteristics and main molecular mechanisms common to renal, hepatic, and cardiac toxicity previously described, in addition to addressing the main validated strategies for the current management of these adverse events in clinical practice. We also handle the main promising antioxidant substances recently presented in the literature to encourage the development of new research that consolidates their potential preventive and therapeutic effects against CP-induced cytotoxicity.
Collapse
Affiliation(s)
- Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Érika Azenathe Barros Mercês
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Fernanda Santos Portela
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Lara Fabiana Luz Malheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | | | | | | | | | | | | | - Helloisa Souza Velozo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
| |
Collapse
|
30
|
Kazak F, Coskun P, Yarim GF, Baspinar N, Ozdemir O, Ates MB, Altug ME, Deveci MZY. Protective and therapeutic effects of nobiletin against cisplatin-induced nephrotoxicity in rats. Biotech Histochem 2024; 99:278-285. [PMID: 39092624 DOI: 10.1080/10520295.2024.2380663] [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: 08/04/2024] Open
Abstract
Possible protective and therapeutic effects of nobiletin on kidney in a cisplatin-induced nephrotoxicity rat model were investigated. Forty male albino rats were divided into four groups: control, cisplatin (CIS), cisplatin+nobiletin (CIS+NOB), and nobiletin+cisplatin (NOB+CIS). At the end of the study, the rats were subjected to biochemical, histological and immunohistochemical analyzes. Compared to the control group, tGSH (p < 0.05) levels, and G6PD (p < 0.05) and GPx (p < 0.001) activities, were increased in the CIS group; while significant (p < 0.05) decreases occurred in the MDA and TOC levels. Histopathologically, the kidneys of the groups administered nobiletin (CIS+NOB, NOB+CIS) were significantly different from the CIS group, being closer to control group in terms of degeneration and hyaline cylinder formation in the tubules (p < 0.05). While dilatation in the tubules, protein-rich fluid and hyaline cylinder formation in the lumen were most common in the CIS group, a significant decrease (p < 0.05) of these parameters was seen in the nobiletin groups (CIS+NOB, NOB+CIS). This study suggests that nobiletin can be effective in preventing and ameliorating toxic effects of cisplatin on the kidney.
Collapse
Affiliation(s)
- Filiz Kazak
- Department of Biochemistry, Veterinary Faculty, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Pinar Coskun
- Department of Biochemistry, Veterinary Faculty, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Gul Fatma Yarim
- Department of Biochemistry, Veterinary Faculty, Ondokuz Mayis University, Samsun, Turkey
| | - Nuri Baspinar
- Department of Biochemistry, Veterinary Faculty, Selcuk University, Konya, Turkey
| | - Ozgur Ozdemir
- Department of Pathology, Veterinary Faculty, Selcuk University, Konya, Turkey
| | - Mehmet Burak Ates
- Department of Pathology, Veterinary Faculty, Selcuk University, Konya, Turkey
| | - Muhammed Enes Altug
- Department of Surgery, Veterinary Faculty, Hatay Mustafa Kemal University, Hatay, Turkey
| | | |
Collapse
|
31
|
Zhang Z, Yu G, Eresen A, Hou Q, Yaghmai V, Zhang Z. MRI monitoring of combined therapy with transcatheter arterial delivery of NK cells and systemic administration of sorafenib for the treatment of HCC. Am J Cancer Res 2024; 14:2216-2227. [PMID: 38859849 PMCID: PMC11162671 DOI: 10.62347/iaro1564] [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: 03/19/2024] [Accepted: 04/27/2024] [Indexed: 06/12/2024] Open
Abstract
This preclinical study explored the synergistic potential of sorafenib and NK cell chemoimmunotherapy to combat hepatocellular carcinoma (HCC) in a rat model. We aimed to enhance NK cell cytotoxicity through IL-12/18 cytokines supplementation and elucidate the underlying molecular mechanisms driving this collaborative antitumor action. Twenty-four Sprague-Dawley rats were divided into distinct treatment groups, receiving sorafenib via gavage and NK cells via catheterization of the proper hepatic artery. Tumor growth and treatment response were monitored through weekly MRI scans, including T1w, T2w, DCE, and DWI sequences. Histological examinations assessed tumor cell viability, apoptosis fraction, and microvessel density. The combined therapy demonstrated significant inhibition of tumor growth, angiogenesis, and induction of durable antitumor immunity compared to either modality alone. DCE-MRI and DWI revealed distinct alterations in tumor microvasculature, highlighting the effectiveness of the combination. Our findings highlight the promise of sorafenib-augmented NK cell chemoimmunotherapy as a potential therapeutic strategy for HCC management. The targeted delivery of IL-12/18 cytokines supplemented NK cells effectively enhanced cytotoxicity within the tumor microenvironment, leading to improved antitumor responses. Further investigation in clinical trials is warranted to validate these findings in human patients and explore the translational potential of this approach.
Collapse
Affiliation(s)
- Zigeng Zhang
- Department of Radiological Sciences, University of California IrvineIrvine, CA, USA
| | - Guangbo Yu
- Department of Biomedical Engineering, University of California IrvineIrvine, CA, USA
| | - Aydin Eresen
- Department of Radiological Sciences, University of California IrvineIrvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California IrvineIrvine, CA, USA
| | - Qiaoming Hou
- Department of Radiological Sciences, University of California IrvineIrvine, CA, USA
| | - Vahid Yaghmai
- Department of Radiological Sciences, University of California IrvineIrvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California IrvineIrvine, CA, USA
| | - Zhuoli Zhang
- Department of Radiological Sciences, University of California IrvineIrvine, CA, USA
- Department of Biomedical Engineering, University of California IrvineIrvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California IrvineIrvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California IrvineIrvine, CA, USA
| |
Collapse
|
32
|
Zhai Y, Chavez JA, D'Aquino KE, Meng R, Nawrocki AR, Pocai A, Wang L, Ma LJ. Kynurenine 3-monooxygenase limits de novo NAD + synthesis through dietary tryptophan in renal proximal tubule epithelial cell models. Am J Physiol Cell Physiol 2024; 326:C1423-C1436. [PMID: 38497113 DOI: 10.1152/ajpcell.00445.2023] [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: 09/13/2023] [Revised: 02/15/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a pivotal coenzyme, essential for cellular reactions, metabolism, and mitochondrial function. Depletion of kidney NAD+ levels and reduced de novo NAD+ synthesis through the tryptophan-kynurenine pathway are linked to acute kidney injury (AKI), whereas augmenting NAD+ shows promise in reducing AKI. We investigated de novo NAD+ biosynthesis using in vitro, ex vivo, and in vivo models to understand its role in AKI. Two-dimensional (2-D) cultures of human primary renal proximal tubule epithelial cells (RPTECs) and HK-2 cells showed limited de novo NAD+ synthesis, likely due to low pathway enzyme gene expression. Using three-dimensional (3-D) spheroid culture model improved the expression of tubular-specific markers and enzymes involved in de novo NAD+ synthesis. However, de novo NAD+ synthesis remained elusive in the 3-D spheroid culture, regardless of injury conditions. Further investigation revealed that 3-D cultured cells could not metabolize tryptophan (Trp) beyond kynurenine (KYN). Intriguingly, supplementation of 3-hydroxyanthranilic acid into RPTEC spheroids was readily incorporated into NAD+. In a human precision-cut kidney slice (PCKS) ex vivo model, de novo NAD+ synthesis was limited due to substantially downregulated kynurenine 3-monooxygenase (KMO), which is responsible for KYN to 3-hydroxykynurenine conversion. KMO overexpression in RPTEC 3-D spheroids successfully reinstated de novo NAD+ synthesis from Trp. In addition, in vivo study demonstrated that de novo NAD+ synthesis is intact in the kidney of the healthy adult mice. Our findings highlight disrupted tryptophan-kynurenine NAD+ synthesis in in vitro cellular models and an ex vivo kidney model, primarily attributed to KMO downregulation.NEW & NOTEWORTHY Nicotinamide adenine dinucleotide (NAD+) is essential in regulating mitochondrial function. Reduced NAD+ synthesis through the de novo pathway is associated with acute kidney injury (AKI). Our study reveals a disruption in de novo NAD+ synthesis in proximal tubular models, but not in vivo, attributed to downregulation of enzyme kynurenine 3-monooxygenase (KMO). These findings highlight a crucial role of KMO in governing de novo NAD+ biosynthesis within the kidney, shedding light on potential AKI interventions.
Collapse
Affiliation(s)
- Yougang Zhai
- CVMR-PH Discovery, Johnson & Johnson Innovative Medicine Research & Development, Spring House, Pennsylvania, United States
| | - Jose A Chavez
- CVMR-PH Discovery, Johnson & Johnson Innovative Medicine Research & Development, Spring House, Pennsylvania, United States
| | - Katharine E D'Aquino
- CVMR-PH Discovery, Johnson & Johnson Innovative Medicine Research & Development, Spring House, Pennsylvania, United States
| | - Rong Meng
- CVMR-PH Discovery, Johnson & Johnson Innovative Medicine Research & Development, Spring House, Pennsylvania, United States
| | - Andrea R Nawrocki
- CVMR-PH Discovery, Johnson & Johnson Innovative Medicine Research & Development, Spring House, Pennsylvania, United States
| | - Alessandro Pocai
- CVMR-PH Discovery, Johnson & Johnson Innovative Medicine Research & Development, Spring House, Pennsylvania, United States
| | - Lifeng Wang
- CVMR-PH Discovery, Johnson & Johnson Innovative Medicine Research & Development, Spring House, Pennsylvania, United States
| | - Li-Jun Ma
- CVMR-PH Discovery, Johnson & Johnson Innovative Medicine Research & Development, Spring House, Pennsylvania, United States
| |
Collapse
|
33
|
Khurshid F, Iqbal J, Ahmad FUD, Lodhi AH, Malik A, Akhtar S, Khan AA, Bux MI, Younis M. A combination of generated hydrogen sulfide and nitric oxide activity has a potentiated protectant effect against cisplatin induced nephrotoxicity. Heliyon 2024; 10:e29513. [PMID: 38655296 PMCID: PMC11036060 DOI: 10.1016/j.heliyon.2024.e29513] [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: 11/14/2023] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Aim Hydrogen sulfide and nitric oxide possess cytoprotective activity and in vivo, they are generated from exogenous sodium hydrosulfide and L-arginine respectively. Cisplatin is a major chemotherapeutic agent used to treat cancer and has a high incidence of nephrotoxicity as a side effect. The study aim was to explore the effects of NaHS and L-arginine or their combination on cisplatin induced nephrotoxicity in rats. Methods Wistar Kyoto rats were given a single intraperitoneal dose of cisplatin (5 mg/kg) followed either by NaHS (56 μmol/kg, i. p.), L-arginine (1.25 g/L in drinking water) or their combination daily for 28-days. Post-mortem plasma, urine and kidney samples were collected for biochemical assays and histopathological analysis. Results Cisplatin decreased body weights and increased urinary output, while plasma creatinine and urea levels were elevated, but sodium and potassium concentrations were diminished. The renal function parameters, blood urea nitrogen and creatinine clearance, were raised and decreased respectively. Regarding markers of reactive oxygen species, plasma total superoxide dismutase was reduced, whereas malondiadehyde was augmented.Cisplatin also diminished plasma and urinary H2S as well as plasma NO, while NaHS and L-arginine counteracted this activity on both redox-active molecules. Cisplatin cotreatment with NaHS, and/or L-arginine exhibited a reversal of all other measured parameters. Conclusion In current study, NaHS and L-arginine as monotherapy protected the rats from cisplatin-induced nephrotoxicity but the combination of both worked more effectively suggesting the augmented anti-inflammatory and antioxidative potential of test treatments when administered together.
Collapse
Affiliation(s)
- Faria Khurshid
- Department of Pharmacology, Faculty of Pharmacy, University of Balochistan, Pakistan
| | - Javeid Iqbal
- Department of Pharmacology, Faculty of Pharmacy, University of Balochistan, Pakistan
| | - Fiaz-Ud-Din Ahmad
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Pakistan
| | - Arslan Hussain Lodhi
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Pakistan
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Suhail Akhtar
- Department of Biochemistry, A.T. Still University of Health Sciences, Kirksville, MO, USA
| | - Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Marvi Imam Bux
- Department of Pharmacology, Faculty of Pharmacy, University of Balochistan, Pakistan
| | - Mohammed Younis
- Department of Pharmacology, Faculty of Pharmacy, University of Balochistan, Pakistan
| |
Collapse
|
34
|
Yuan Z, Yang X, Hu Z, Gao Y, Wang M, Xie L, Zhu H, Chen C, Lu H, Bai Y. Fraxetin pretreatment alleviates cisplatin-induced kidney injury by antagonizing autophagy and apoptosis via mTORC1 activation. Phytother Res 2024; 38:2077-2093. [PMID: 38558449 DOI: 10.1002/ptr.8073] [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: 05/18/2023] [Revised: 10/24/2023] [Accepted: 11/05/2023] [Indexed: 04/04/2024]
Abstract
Cisplatin-induced kidney injury (CKI) is a common complication of chemotherapy. Fraxetin, derived from Fraxinus bungeana A. DC. bark, has antioxidant, anti-inflammatory, and anti-fibrotic effects. This study aims to investigate fraxetin's effects on CKI and its underlying mechanism in vivo and in vitro. Tubular epithelial cells (TECs) and mice were exposed to cisplatin with and without fraxetin preconditioning assess fraxetin's role in CKI. TECs autophagy was observed using transmission electron microscopy. Apoptosis levels in animal tissues were measured using TUNEL staining. The protective mechanism of fraxetin was explored through pharmacological and genetic regulation of mTORC1. Molecular docking was used to identify potential binding sites between fraxetin and mTORC1. The results indicated that fraxetin pretreatment reduced cisplatin-induced kidney injury in a time- and concentration-dependent way. Fraxetin also decreased autophagy in TECs, as observed through electron microscopy. Tissue staining confirmed that fraxetin pretreatment significantly reduced cisplatin-induced apoptosis. Inhibition of mTORC1 using rapamycin or siRNA reversed the protective effects of fraxetin on apoptosis and autophagy in cisplatin-treated TECs, while activation of mTORC1 enhanced fraxetin's protective effect. Molecular docking analysis revealed that fraxetin can bind to HEAT-repeats binding site on mTORC1 protein. In summary, fraxetin pretreatment alleviates CKI by antagonizing autophagy and apoptosis via mTORC1 activation. This provides evidence for the potential therapeutic application of fraxetin in CKI.
Collapse
Affiliation(s)
- Ziwei Yuan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuejia Yang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zujian Hu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuanyuan Gao
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengsi Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lili Xie
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hengyue Zhu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chaosheng Chen
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Chronic Nephropathy, Wenzhou Medical University, Wenzhou, China
| | - Hong Lu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Chronic Nephropathy, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
35
|
Kazak F, Deveci MZY, Akçakavak G. Eucalyptol alleviates cisplatin-induced kidney damage in rats. Drug Chem Toxicol 2024; 47:172-179. [PMID: 36514998 DOI: 10.1080/01480545.2022.2156530] [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: 09/28/2022] [Revised: 11/15/2022] [Accepted: 12/03/2022] [Indexed: 12/15/2022]
Abstract
This study was aimed to explore the therapeutic effect of eucalyptol on cisplatin induced kidney damage in Wistar albino rats. The animals were divided into four groups: sham (S), eucalyptol (E), cisplatin (C), and cisplatin + eucalyptol (CE) randomly, six animals in each group. Groups C and CE were received cisplatin (12 mg/kg, a single dose, intraperitoneally (i.p.)). Groups E and CE were treated with eucalyptol (100 mg/kg, for seven days, orally). The blood samples and kidney tissues were collected following sacrification and analyzed histopathologically and biochemically. Histopathological results revealed tubular degeneration and necrosis, inflammatory cell infiltration, tubular lumen dilatation, enlargement of bowman's space and hyaline cast were significantly irregular in the group C than group S. However, eucalyptol treatment (CE) modulated the alterations in the group C. Serum levels of blood urea nitrogen (BUN) and creatinine (CRE) were considerably higher in the group C compared to the other groups. There was no significant difference among the other groups statistically (except group C) in terms of BUN and CRE values. Eucalyptol treatment (at 100 mg/kg, for seven days) decreased the cisplatin induced increase in serum BUN and CRE levels and restored the reduced Vit C level and CAT activity of kidneys caused by cisplatin. Thus, eucalyptol's antioxidative, nephroprotective, and curative effects indicated the potential for future drug development.
Collapse
Affiliation(s)
- Filiz Kazak
- Department of Biochemistry, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Antakya, Turkey
| | - Mehmet Zeki Yılmaz Deveci
- Department of Surgery, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Antakya, Turkey
- Laboratory Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Gökhan Akçakavak
- Department of Pathology, Faculty of Veterinary Medicine, Bozok University, Yozgat, Turkey
| |
Collapse
|
36
|
Alqahtani NF, Alfaifi MY, Shati AA, Elbehairi SEI, Elshaarawy RFM, Serag WM, Hassan YA, El-Sayed WN. Exploring the chondroitin sulfate nanogel's potential in combating nephrotoxicity induced by cisplatin and doxorubicin-An in-vivo study on rats. Int J Biol Macromol 2024; 258:128839. [PMID: 38134998 DOI: 10.1016/j.ijbiomac.2023.128839] [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: 07/21/2023] [Revised: 12/02/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
In this study, we aim to unveil the potential of itaconyl chondroitin sulfate nanogel (ICSNG) in tackling chronic kidney diseases triggered by the administration of CDDP and doxorubicin (Adriamycin, ADR). To that end, the new drug delivery system (ICSNG) was initially prepared, characterized, and loaded with the target drugs. Thereafter, the in-vivo studies were performed using five equally divided groups of 100 male Sprague-Dawley (SD) rats. Biochemical evaluation and immunohistochemistry studies have revealed the renal toxicity and the ameliorative effects of ICSNG on renal function. When ICSNG-based treatments were contrasted with the CDDP and ADR infected groups, they significantly increased paraoxonase-1 (PON-1), superoxide dismutase (SOD), catalase (CAT) and albumin activity and significantly decreased nitric oxide (NO), tumor necrosis factor alpha (TNF-α), creatinine, urea, and cyclooxygenase-2 (COX-2) activity (p < 0.001). The findings of the current study imply that ICSNG may be able to lessen renal inflammation and damage in chronic kidney disorders brought on by the administration of CDDP and ADR. Interestingly, according to the estimated selectivity indices, the ICSNG-encapsulated drugs have demonstrated superior selectivity for cancer MCF-7 cells, over healthy HSF cells, in comparison to the bare drugs.
Collapse
Affiliation(s)
- Norah F Alqahtani
- Department of Chemistry, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Ali A Shati
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | | | - Reda F M Elshaarawy
- Department of Chemistry, Faculty of Science, Suez University, 43533 Suez, Egypt; Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany.
| | - Waleed M Serag
- Department of Chemistry, Faculty of Science, Suez University, 43533 Suez, Egypt
| | - Yasser A Hassan
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Al-Kitab University, Kirkuk, Iraq; Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Al-Qalam University College, Kirkuk, Iraq; Department of pharmaceutics and Pharmaceutical Technology, Faculty of pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - W N El-Sayed
- Department of Chemistry, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| |
Collapse
|
37
|
Rendra E, Uhlig S, Moskal I, Thielemann C, Klüter H, Bieback K. Adipose Stromal Cell-Derived Secretome Attenuates Cisplatin-Induced Injury In Vitro Surpassing the Intricate Interplay between Proximal Tubular Epithelial Cells and Macrophages. Cells 2024; 13:121. [PMID: 38247813 PMCID: PMC10814170 DOI: 10.3390/cells13020121] [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: 12/06/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
(1) Background: The chemotherapeutic drug cisplatin exerts toxic side effects causing acute kidney injury. Mesenchymal stromal cells can ameliorate cisplatin-induced kidney injury. We hypothesize that the MSC secretome orchestrates the vicious cycle of injury and inflammation by acting on proximal tubule epithelial cells (PTECs) and macrophages individually, but further by counteracting their cellular crosstalk. (2) Methods: Conditioned medium (CM) from adipose stromal cells was used, first assessing its effect on cisplatin injury in PTECs. Second, the effects of cisplatin and the CM on macrophages were measured. Lastly, in an indirect co-culture system, the interplay between the two cell types was assessed. (3) Results: First, the CM rescued PTECs from cisplatin-induced apoptosis by reducing oxidative stress and expression of nephrotoxicity genes. Second, while cisplatin exerted only minor effects on macrophages, the CM skewed macrophage phenotypes to the anti-inflammatory M2-like phenotype and increased phagocytosis. Finally, in the co-culture system, the CM suppressed PTEC death by inhibiting apoptosis and nuclei fragmentation. The CM lowered TNF-α release, while cisplatin inhibited macrophage phagocytosis, PTECs, and the CM to a greater extent, thus enhancing it. The CM strongly dampened the inflammatory macrophage cytokine secretion triggered by PTECs. (4) Conclusions: ASC-CM surpasses the PTEC-macrophage crosstalk in cisplatin injury. The positive effects on reducing cisplatin cytotoxicity, on polarizing macrophages, and on fine-tuning cytokine secretion underscore MSCs' CM benefit to prevent kidney injury progression.
Collapse
Affiliation(s)
- Erika Rendra
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
| | - Stefanie Uhlig
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
- Flow Core Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Isabell Moskal
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
| | - Corinna Thielemann
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
| | - Harald Klüter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
- Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany; (E.R.); (H.K.)
- Flow Core Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
38
|
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
|
39
|
Mahmod II, Ismail IS, Normi YM, Chong SG. Protective effect of Clinacanthus nutans in cisplatin-induced nephrotoxicity on human kidney cell (PCS-400-010) elucidated by an LCMS-based metabolomics approach. Biomed Chromatogr 2023; 37:e5750. [PMID: 37778127 DOI: 10.1002/bmc.5750] [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: 03/23/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 10/03/2023]
Abstract
Cisplatin-induced nephrotoxicity has been widely reported in numerous studies. The objective of this study is to assess the potential nephroprotective effects of Clinacanthus nutans (Burm. f.) Lindau (Acanthaceae) leaf extracts on human kidney cells (PCS-400-010) in vitro using an LCMS-based metabolomics approach. Orthogonal partial least square-discriminant analysis identified 16 significantly altered metabolites when comparing the control and pre-treated C. nutans cisplatin-induced groups. These metabolites were found to be associated with glycerophospholipid, purine, and amino acid metabolism, as well as the glycolysis pathway. Pre-treatment with C. nutans aqueous extract (125 μg/mL) for 24 h, followed by 48 h of cisplatin induction in PCS-400-010 cells, demonstrated a nephroprotective effect, particularly involving the regulation of amino acid metabolism.
Collapse
Affiliation(s)
- Ilya Iryani Mahmod
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Intan Safinar Ismail
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Yahaya M Normi
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Siok-Geok Chong
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
40
|
Abd Alhusen SK, Hasan AF. Evaluating the renoprotective effects of omega-3-6-9 against cisplatin-induced nephrotoxicity in mice. J Med Life 2023; 16:1756-1759. [PMID: 38585532 PMCID: PMC10994620 DOI: 10.25122/jml-2023-0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/13/2023] [Indexed: 04/09/2024] Open
Abstract
Fatty acids, particularly omega-3, omega-6, and omega-9, play a vital role in various biological processes. As the body cannot synthesize omega-3 and omega-6, dietary sources of these fatty acids are essential. Each omega fatty acid has a distinct chemical structure, source, and function. Cisplatin (CP) treatment is known to cause acute kidney injury (AKI) due to its inflammatory effects. This study explored the renoprotective potential of omega-3-6-9 when co-administered with cisplatin in a mice model. We divided adult mice into five groups: a control group received 0.5 ml of liquid paraffin; a cisplatin-only group; two groups were treated with low (50 mg/kg) and high (100 mg/kg) doses of omega-3-6-9 plus cisplatin; and a final group received vitamin E before cisplatin administration. The administration of omega-3-6-9 significantly decreased pro-inflammatory modulators and kidney function markers such as TNF-α, IL-1β, blood urea nitrogen, and creatinine, indicating potential renoprotective effects. Our research concluded that omega-3- 6- 9 had anti-inflammatory properties and was effective against the harmful effects of cisplatin.
Collapse
Affiliation(s)
- Saja Kareem Abd Alhusen
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Baghdad, Baghdad, Iraq
| | - Ali Faris Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Baghdad, Baghdad, Iraq
| |
Collapse
|
41
|
Zhang D, Luo G, Jin K, Bao X, Huang L, Ke J. The underlying mechanisms of cisplatin-induced nephrotoxicity and its therapeutic intervention using natural compounds. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2925-2941. [PMID: 37289283 DOI: 10.1007/s00210-023-02559-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 05/28/2023] [Indexed: 06/09/2023]
Abstract
Cisplatin is an effective chemotherapeutic drug widely used for the treatment of various solid tumors; however, its clinical use and efficacy are limited by its inherent nephrotoxicity. The pathogenesis of cisplatin-induced nephrotoxicity is complex and has not been fully elucidated. Cellular uptake and transport, DNA damage, apoptosis, oxidative stress, inflammatory response, and autophagy are involved in the development of cisplatin-induced nephrotoxicity. Currently, despite some deficiencies, hydration regimens remain the major protective measures against cisplatin-induced nephrotoxicity. Therefore, effective drugs must be explored and developed to prevent and treat cisplatin-induced kidney injury. In recent years, many natural compounds with high efficiency and low toxicity have been identified for the treatment of cisplatin-induced nephrotoxicity, including quercetin, saikosaponin D, berberine, resveratrol, and curcumin. These natural agents have multiple targets, multiple effects, and low drug resistance; therefore, they can be safely used as a supplementary regimen or combination therapy for cisplatin-induced nephrotoxicity. This review aimed to comprehensively describe the molecular mechanisms underlying cisplatin-induced nephrotoxicity and summarize natural kidney-protecting compounds to provide new ideas for the development of better therapeutic agents.
Collapse
Affiliation(s)
- Doudou Zhang
- Jinhua Municipal Central Hospital, Jinhua, 321000, China
| | - Guangwen Luo
- Jinhua Municipal Central Hospital, Jinhua, 321000, China.
| | - Kaixiang Jin
- Jinhua Municipal Central Hospital, Jinhua, 321000, China
| | - Xiaodong Bao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lili Huang
- Ningbo Medical Center Lihuili Hospital, Ningbo, 315040, China
| | - Jianghuan Ke
- Jinhua Municipal Central Hospital, Jinhua, 321000, China
| |
Collapse
|
42
|
Mapuskar KA, Pulliam CF, Zepeda-Orozco D, Griffin BR, Furqan M, Spitz DR, Allen BG. Redox Regulation of Nrf2 in Cisplatin-Induced Kidney Injury. Antioxidants (Basel) 2023; 12:1728. [PMID: 37760031 PMCID: PMC10525889 DOI: 10.3390/antiox12091728] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Cisplatin, a potent chemotherapeutic agent, is marred by severe nephrotoxicity that is governed by mechanisms involving oxidative stress, inflammation, and apoptosis pathways. The transcription factor Nrf2, pivotal in cellular defense against oxidative stress and inflammation, is the master regulator of the antioxidant response, upregulating antioxidants and cytoprotective genes under oxidative stress. This review discusses the mechanisms underlying chemotherapy-induced kidney injury, focusing on the role of Nrf2 in cancer therapy and its redox regulation in cisplatin-induced kidney injury. We also explore Nrf2's signaling pathways, post-translational modifications, and its involvement in autophagy, as well as examine redox-based strategies for modulating Nrf2 in cisplatin-induced kidney injury while considering the limitations and potential off-target effects of Nrf2 modulation. Understanding the redox regulation of Nrf2 in cisplatin-induced kidney injury holds significant promise for developing novel therapeutic interventions. This knowledge could provide valuable insights into potential strategies for mitigating the nephrotoxicity associated with cisplatin, ultimately enhancing the safety and efficacy of cancer treatment.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Casey F. Pulliam
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Diana Zepeda-Orozco
- Pediatric Nephrology and Hypertension at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Benjamin R. Griffin
- Division of Nephrology, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Muhammad Furqan
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
43
|
Nagasaki T, Maeda H, Yanagisawa H, Nishida K, Kobayashi K, Wada N, Noguchi I, Iwakiri R, Taguchi K, Sakai H, Saruwatari J, Watanabe H, Otagiri M, Maruyama T. Carbon Monoxide-Loaded Red Blood Cell Prevents the Onset of Cisplatin-Induced Acute Kidney Injury. Antioxidants (Basel) 2023; 12:1705. [PMID: 37760008 PMCID: PMC10526101 DOI: 10.3390/antiox12091705] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/26/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Cisplatin-induced acute kidney injury (AKI) is an important factor that limits the clinical use of this drug for the treatment of malignancies. Oxidative stress and inflammation are considered to be the main causes of not only cisplatin-induced death of cancer cells but also cisplatin-induced AKI. Therefore, developing agents that exert antioxidant and anti-inflammatory effects without weakening the anti-tumor effects of cisplatin is highly desirable. Carbon monoxide (CO) has recently attracted interest due to its antioxidant, anti-inflammatory, and anti-tumor properties. Herein, we report that CO-loaded red blood cell (CO-RBC) exerts renoprotective effects on cisplatin-induced AKI. Cisplatin treatment was found to reduce cell viability in proximal tubular cells via oxidative stress and inflammation. Cisplatin-induced cytotoxicity, however, was suppressed by the CO-RBC treatment. The intraperitoneal administration of cisplatin caused an elevation in the blood urea nitrogen and serum creatinine levels. The administration of CO-RBC significantly suppressed these elevations. Furthermore, the administration of CO-RBC also reduced the deterioration of renal histology and tubular cell injury through its antioxidant and anti-inflammatory effects in cisplatin-induced AKI mice. Thus, our data suggest that CO-RBC has the potential to substantially prevent the onset of cisplatin-induced AKI, which, in turn, may improve the usefulness of cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Taisei Nagasaki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (T.N.); (H.Y.); (K.N.); (K.K.); (N.W.); (I.N.); (R.I.); (H.W.)
| | - Hitoshi Maeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (T.N.); (H.Y.); (K.N.); (K.K.); (N.W.); (I.N.); (R.I.); (H.W.)
| | - Hiroki Yanagisawa
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (T.N.); (H.Y.); (K.N.); (K.K.); (N.W.); (I.N.); (R.I.); (H.W.)
| | - Kento Nishida
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (T.N.); (H.Y.); (K.N.); (K.K.); (N.W.); (I.N.); (R.I.); (H.W.)
| | - Kazuki Kobayashi
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (T.N.); (H.Y.); (K.N.); (K.K.); (N.W.); (I.N.); (R.I.); (H.W.)
| | - Naoki Wada
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (T.N.); (H.Y.); (K.N.); (K.K.); (N.W.); (I.N.); (R.I.); (H.W.)
| | - Isamu Noguchi
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (T.N.); (H.Y.); (K.N.); (K.K.); (N.W.); (I.N.); (R.I.); (H.W.)
| | - Ryotaro Iwakiri
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (T.N.); (H.Y.); (K.N.); (K.K.); (N.W.); (I.N.); (R.I.); (H.W.)
| | - Kazuaki Taguchi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan;
| | - Hiromi Sakai
- Department of Chemistry, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan;
| | - Junji Saruwatari
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan;
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (T.N.); (H.Y.); (K.N.); (K.K.); (N.W.); (I.N.); (R.I.); (H.W.)
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (T.N.); (H.Y.); (K.N.); (K.K.); (N.W.); (I.N.); (R.I.); (H.W.)
| |
Collapse
|
44
|
Hou YY, Qi SM, Leng J, Shen Q, Tang S, Zhang JT, Hu JN, Jiang S, Li W. Lobetyolin, a Q-marker isolated from Radix Platycodi, exerts protective effects on cisplatin-induced cytotoxicity in HEK293 cells. J Nat Med 2023; 77:721-734. [PMID: 37353674 DOI: 10.1007/s11418-023-01714-w] [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: 10/12/2022] [Accepted: 05/28/2023] [Indexed: 06/25/2023]
Abstract
This study investigated the protective effect of lobetyolin (LBT), a Q-marker isolated from the roots of Platycodon grandiflorum (Radix Platycodi), against cisplatin-induced cytotoxicity in human embryonic kidney (HEK293) cells. Results showed that LBT at 20 μM significantly prevented cisplatin-induced cytotoxicity by improving the viability of HEK293 cells, decreasing levels of MDA, and decreasing GSH content triggered by cisplatin. It also suppressed reactive oxygen species (ROS) levels. Molecular docking analysis revealed a strong binding affinity between LBT and the NF-κB protein, with a docking fraction of - 6.5 kcal/mol. These results provide compelling evidence suggesting a potential link between the visualization analysis of LBT and its protective mechanism, specifically implicating the NF-κB signaling pathway. LBT also reduced the expression level of tumor necrosis factor-alpha (TNF-α), phosphorylation NF-κB and IκBα in HEK293 cells which were increased by cisplatin exposure, leading to inhibition of inflammation. Furthermore, western blotting showed that LBT antagonized the up-regulation of Bax, cleaved caspase 3, 8, and 9 expression and inhibited the MAPK signaling pathway by down-regulating phosphorylation JNK, ERK, and p38, partially ameliorating cisplatin-induced cytotoxicity in HEK293 cells. Therefore, these results indicate that LBT has potentially protected renal function by inhibiting inflammation and apoptosis.
Collapse
Affiliation(s)
- Yun-Yi Hou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Si-Min Qi
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Jing Leng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Qiong Shen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Shan Tang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Jun-Nan Hu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Shuang Jiang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China.
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China.
- National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun, 130118, China.
| |
Collapse
|
45
|
Ibrahim D, Halboup A, Al Ashwal M, Shamsher A. Ameliorative Effect of Olea europaea Leaf Extract on Cisplatin-Induced Nephrotoxicity in the Rat Model. Int J Nephrol 2023; 2023:2074498. [PMID: 37497380 PMCID: PMC10368505 DOI: 10.1155/2023/2074498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/29/2023] [Accepted: 07/05/2023] [Indexed: 07/28/2023] Open
Abstract
Background Olea europaea leaf extract (OELE) has potential health benefits and protects against cytotoxicity. This study investigated the possible ameliorative effect of OELE on cisplatin-induced nephrotoxicity in rats. Methods Rats were assigned into six groups; two groups received 150 mg/kg or 300 mg/kg of OELE, one group received a single dose of cisplatin (6 mg/kg) IP on the first day of the experiment, two groups received a single dose of cisplatin 150 mg/kg or 300 mg/kg of OELE on the first day then starting from the fifth day for 10 consecutive days, and one group acted as a control. Results and Conclusion. The findings showed that cisplatin-induced nephrotoxicity was evidenced by a significant increase in serum creatinine blood urea nitrogen (BUN) and a significant decrease in estimated creatinine clearance and potassium level, which corresponded with the alterations in the histopathology of the renal tissue. OELE significantly ameliorated the nephrotoxic effects of cisplatin as dose-dependent.
Collapse
Affiliation(s)
- Doa'a Ibrahim
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, University of Science and Technology, Sana'a, Yemen
| | - Abdulsalam Halboup
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, University of Science and Technology, Sana'a, Yemen
- Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Mohammed Al Ashwal
- Department of Pharmacology, Faculty of Pharmacy, University of Science and Technology, Sana'a, Yemen
| | - Amani Shamsher
- Department of Histopathology, University of Science and Technology Hospital, Sana'a, Yemen
| |
Collapse
|
46
|
Shati AA, Alkabli J, Alfaifi MY, Elbehairi SEI, Elshaarawy RFM, Serag WM, Hassan YA. Comparison of the ameliorative roles of crab chitosan nanoparticles and mesenchymal stem cells against cisplatin-triggered nephrotoxicity. Int J Biol Macromol 2023:124985. [PMID: 37230447 DOI: 10.1016/j.ijbiomac.2023.124985] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
AIM In the present investigation, we compared the effects of mesenchymal stem cells extracted from bone marrow (BMSCs) and crab chitosan nanoparticles (CCNPs) on renal fibrosis in cisplatin (CDDP)-induced kidney injury rats. MATERIAL AND METHODS 90 male Sprague-Dawley (SD) rats were divided into two equal groups and alienated. Group I was set into three subgroups: the control subgroup, the CDDP-infected subgroup (acute kidney injury), and the CCNPs-treated subgroup. Group II was also divided into three subgroups: the control subgroup, the CDDP-infected subgroup (chronic kidney disease), and the BMSCs-treated subgroup. Through biochemical analysis and immunohistochemical research, the protective effects of CCNPs and BMSCs on renal function have been identified. RESULTS CCNPs and BMSC treatment resulted in a substantial rise in GSH and albumin and a decrease in KIM-1, MDA, creatinine, urea, and caspase-3 when compared to the infected groups (p < 0.05). CONCLUSION According to the current research, chitosan nanoparticles and BMSCs may be able to reduce renal fibrosis in acute and chronic kidney diseases caused by CDDP administration, with more improvement of kidney damage resembling normal cells after CCNPs administration.
Collapse
Affiliation(s)
- Ali A Shati
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - J Alkabli
- Department of Chemistry, College of Sciences and Arts - Alkamil, University of Jeddah, Jeddah 23218, Saudi Arabia
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Serag Eldin I Elbehairi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia; Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company), 51 Wezaret El-Zeraa St., Agouza, Giza, Egypt
| | - Reda F M Elshaarawy
- Department of Chemistry, Faculty of Science, Suez University, 43533 Suez, Egypt; Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany.
| | - Waleed M Serag
- Department of Chemistry, Faculty of Science, Suez University, 43533 Suez, Egypt
| | - Yasser A Hassan
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
47
|
Alrfaei BM, Almutairi AO, Aljohani AA, Alammar H, Asiri A, Bokhari Y, Aljaser FS, Abudawood M, Halwani M. Electrolytes Play a Role in Detecting Cisplatin-Induced Kidney Complications and May Even Prevent Them-Retrospective Analysis. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59050890. [PMID: 37241122 DOI: 10.3390/medicina59050890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023]
Abstract
Background and Objective: Cisplatin is a chemotherapy drug used to treat several types of malignancies. It is a platinum-based compound that interferes with cell division and DNA replication. Cisplatin has been associated with renal damage. This study evaluates the early detection of nephrotoxicity through routine laboratory tests. Materials and Methods: This is a retrospective chart review based on the Saudi Ministry of National Guard Hospital (MNGHA). We evaluated deferential laboratory tests for cancer patients treated with cisplatin between April 2015 and July 2019. The evaluation included age, sex, WBC, platelets, electrolytes, co-morbidities and interaction with radiology. Results: The review qualified 254 patients for evaluation. Around 29 patients (11.5%) had developed kidney function abnormality. These patients presented with abnormally low magnesium 9 (31%), potassium 6 (20.7%), sodium 19 (65.5%) and calcium 20 (69%). Interestingly, the whole sample size had abnormal electrolytes presenting magnesium 78 (30.8%), potassium 30 (11.9%), sodium 147 (58.1%) and calcium 106 (41.9%). Some pathological features were detected, such as hypomagnesemia, hypocalcemia and hypokalemia. In addition, infections that needed antibiotics were dominant in patients treated with cisplatin alone, representing 50% of this group. Conclusions: We report that an average of 15% of patients with electrolyte abnormalities develop renal toxicity and reduced function. Moreover, electrolytes may serve as an early indicator for renal damage as part of chemotherapy complication. This indication represents 15% of renal toxicity cases. Changes in electrolyte levels have been reported with cisplatin. Specifically, it has been linked to hypomagnesemia, hypocalcemia and hypokalemia. This study will help reduce the risk of dialysis or the need for kidney transplant. It is also important to manage any underlying conditions and control patients' intake of electrolytes.
Collapse
Affiliation(s)
- Bahauddeen M Alrfaei
- Cellular Therapy and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh 14611, Saudi Arabia
| | - Abdulaziz O Almutairi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh 14611, Saudi Arabia
| | - Alaa A Aljohani
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh 14611, Saudi Arabia
| | - Hajar Alammar
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh 14611, Saudi Arabia
| | - Abdulaziz Asiri
- Faculty of Applied Medical Sciences, University of Bisha, 255, Al Nakhil, Bisha 67714, Saudi Arabia
| | - Yahya Bokhari
- Department of AI and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
- Department of Health Informatics, College of Public Health and Health Informatics, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11426, Saudi Arabia
| | - Feda S Aljaser
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12371, Saudi Arabia
| | - Manal Abudawood
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12371, Saudi Arabia
| | - Majed Halwani
- Nanomedicine Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Saudi Arabia
| |
Collapse
|
48
|
Sharaf G, El Morsy EM, El-Sayed EK. Augmented nephroprotective effect of liraglutide and rabeprazole via inhibition of OCT2 transporter in cisplatin-induced nephrotoxicity in rats. Life Sci 2023; 321:121609. [PMID: 36958435 DOI: 10.1016/j.lfs.2023.121609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
AIMS Cisplatin, a widely used anticancer treatment, has a marked nephrotoxic effect. This nephrotoxic effect is linked to the triggering of oxidative stress, inflammation, activation of mitogen-activated protein kinase (MAPK) pathway as well as apoptosis. The purpose of the present research was to examine the possible ameliorative effect of liraglutide and/or rabeprazole on cisplatin-induced nephrotoxicity in rats and to underline the potential molecular pathways involved. MAIN METHODS Rats were divided into five groups: Control, cisplatin, liraglutide (200 μg/kg/day, i.p), rabeprazole (10 mg/kg/day, orally) and liraglutide + rabeprazole combination groups. All treatments were given for 7 days. Cisplatin was given as a single dose (7 mg/kg, i.p) at day 4 to induce nephrotoxicity in all groups except the control group. KEY FINDINGS Treatment with liraglutide and/or rabeprazole prior to cisplatin maintained the function and morphology of kidney via decreasing cisplatin renal uptake by significant inhibition of OCT2. Besides, they showed a significant increase in GLP-1 receptor expression. Liraglutide and/or rabeprazole significantly attenuated the levels of TNF-α. ICAM, NF-κB, and downregulated MAPK pathway proteins such as JNK, and ERK1/2. Moreover, they maintained oxidant antioxidant balance by decreasing MDA level and increasing GSH level and CAT activity. Additionally, liraglutide and/or rabeprazole exhibited antiapoptotic effect evidenced by the decreased caspase-3 level and Bax expression and the increased Bcl-2 expression. SIGNIFICANCE The current study showed that both liraglutide and rabeprazole exerted a nephroprotective effect against cisplatin-induced renal toxicity in rats. Interestingly, co-treatment with both drugs showed an augmented effect.
Collapse
Affiliation(s)
- Gehad Sharaf
- Nasr Hospital Health Insurance, Helwan, Cairo, Egypt.
| | - E M El Morsy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| | - Elsayed K El-Sayed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt.
| |
Collapse
|
49
|
Alassaf N, Attia H. Autophagy and necroptosis in cisplatin-induced acute kidney injury: Recent advances regarding their role and therapeutic potential. Front Pharmacol 2023; 14:1103062. [PMID: 36794281 PMCID: PMC9922871 DOI: 10.3389/fphar.2023.1103062] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Cisplatin (CP) is a broad-spectrum antineoplastic agent, used to treat many different types of malignancies due to its high efficacy and low cost. However, its use is largely limited by acute kidney injury (AKI), which, if left untreated, may progress to cause irreversible chronic renal dysfunction. Despite substantial research, the exact mechanisms of CP-induced AKI are still so far unclear and effective therapies are lacking and desperately needed. In recent years, necroptosis, a novel subtype of regulated necrosis, and autophagy, a form of homeostatic housekeeping mechanism have witnessed a burgeoning interest owing to their potential to regulate and alleviate CP-induced AKI. In this review, we elucidate in detail the molecular mechanisms and potential roles of both autophagy and necroptosis in CP-induced AKI. We also explore the potential of targeting these pathways to overcome CP-induced AKI according to recent advances.
Collapse
Affiliation(s)
- Noha Alassaf
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia,*Correspondence: Noha Alassaf,
| | - Hala Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia,Department of Biochemistry, College of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
50
|
Multi-Mechanistic and Therapeutic Exploration of Nephroprotective Effect of Traditional Ayurvedic Polyherbal Formulation Using In Silico, In Vitro and In Vivo Approaches. Biomedicines 2023; 11:biomedicines11010168. [PMID: 36672676 PMCID: PMC9855918 DOI: 10.3390/biomedicines11010168] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Based on traditional therapeutic claims, NEERI KFT (a traditional Ayurvedic polyherbal preparation) has been innovatively developed in recent time on the decades of experience for treating kidney dysfunction. Due to the lack of scientific evidence, the present investigations are needed to support the rationale use of NEERI KFT. Considering the facts, the study investigated the nephroprotective effect of NEERI KFT against kidney dysfunction using in silico, in vitro and in vivo approaches. In this study, phytochemical and network pharmacology studies were performed for the developed formulation to evaluate the molecular mechanism of NEERI KFT in the amelioration of kidney disease. In vitro nephroprotective and antioxidant effect of NEERI KFT was determined on HEK 293 cells against cisplatin-induced cytotoxicity and oxidative stress. In vivo nephroprotective effect of NEERI KFT was determined against cisplatin-induced nephrotoxicity in Wistar rats, via assessing biochemical markers, antioxidant enzymes and inflammatory cytokines such as TNF-α, IL-1β, CASP-3, etc. The results showed that the compounds such as gallic acid, caffeic acid and ferulic acid are the major constituents of NEERI KFT, while network pharmacology analysis indicated a strong interaction between polyphenols and several genes (CASPs, ILs, AGTR1, AKT, ACE2, SOD1, etc.) involved in the pathophysiology of kidney disease. In vivo studies showed a significant (p < 0.05) ameliorative effect on biochemical markers and antioxidant enzymes (SOD, CAT, GSH, etc.), and regulates inflammatory cytokine (TNF-α, IL-1β, CASP-3) expression in kidney tissue. Hence, it can be concluded that NEERI KFT subsequently alleviates renal dysfunction mediated by cisplatin via attenuating oxidative and inflammatory stress, thus preserving the normalcy of kidney function.
Collapse
|