1
|
Barnes DA, Janssen MJ, Yang H, Redegeld FA, Masereeuw R. An adverse outcome pathway for DNA adduct formation leading to kidney failure. Toxicology 2025; 515:154162. [PMID: 40268266 DOI: 10.1016/j.tox.2025.154162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/11/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
An Adverse Outcome Pathway (AOP) is a conceptual framework in toxicology and risk assessment that outlines the series of events from a chemical's molecular interaction to the resulting adverse health effect. This framework offers a structured approach to organizing biological knowledge, making it especially useful for understanding the mechanisms through which chemicals cause harm. Following a comprehensive analysis of the literature, an AOP was elucidated for key events linking DNA adduct formation, caused by compounds such as platinum anticancer drugs, to tubular necrosis, resulting in kidney failure. Currently, cisplatin, carboplatin and oxaliplatin are the three most utilised Pt-based drugs used globally for the treatment of cancer. The hydrolysis of platinum anticancer agents post-cellular uptake yields electrophilic intermediates that covalently bind to nucleophilic sites on DNA to form adducts that represent the molecular initiating event. When DNA repair mechanisms become unbalanced, the nephrotoxic response following the formation of DNA adducts leads to DNA damage and mitochondrial dysfunction. These events promote the generation and release of reaction oxygen species (ROS) to induce oxidative stress, causing cell death and inflammation. Upon detachment from the basement membrane, these compromised cells are subsequently deposited in the tubular lumen. Tubular obstruction and inflammatory responses to proximal tubule insult can lead to secondary toxicity and tubular necrosis, further exacerbating kidney injury and precipitating a progressive decline of renal function, finally resulting in kidney failure.
Collapse
Affiliation(s)
- D A Barnes
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands
| | - M J Janssen
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands
| | | | - F A Redegeld
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands
| | - R Masereeuw
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, the Netherlands.
| |
Collapse
|
2
|
Ke A, Yang W, Zhang W, Chen Y, Meng X, Liu J, Dai D. The cardiac glycoside periplocymarin sensitizes gastric cancer to ferroptosis via the ATP1A1-Src-YAP/TAZ-TFRC axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156804. [PMID: 40311597 DOI: 10.1016/j.phymed.2025.156804] [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: 03/18/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Targeting ferroptosis vulnerabilities in tumors has become an increasingly promising therapeutic strategy. While the regulatory effects of natural products on ferroptosis are progressively being elucidated, the role of cardiac glycosides in modulating ferroptosis remains poorly understood. PURPOSE This study aims to investigate the ferroptosis-sensitizing effects of periplocymarin (PPM), a cardiac glycoside derived from the traditional plant Periploca sepium, and to elucidate the underlying molecular mechanisms. METHODS The effects of PPM on ferroptosis regulation were comprehensively assessed through functional assays, followed by sequencing analysis to identify associated signaling pathways. Subsequent mechanistic validation experiments were conducted to confirm the upstream and downstream regulatory components involved in this ferroptosis-modulating axis. RESULTS PPM induced slow and mild apoptosis in gastric cancer cells through the inhibition of glycolysis. However, when combined with ferroptosis inducers, it promoted rapid and robust ferroptosis. In vivo, PPM sensitized gastric cancer xenografts to cisplatin-induced ferroptosis with no observable cardiotoxicity or renal impairment. Mechanistically, PPM targeted the α1 subunit of the Na+/K+-ATPase (ATP1A1), leading to the activation of Src, which subsequently induced tyrosine phosphorylation of YAP/TAZ in a Hippo-independent manner, promoting their nuclear translocation. The YAP/TAZ-TEAD transcriptional complex directly bound to the TFRC promoter region between nucleotides 401-409 upstream of the transcription start site, thereby activating TFRC transcription. This resulted in increased iron influx, elevated lipid peroxidation, and heightened sensitivity to ferroptosis. Notably, ATP1A1 was essential for ferroptosis resistance, as its knockdown mimicked the sensitizing effect of PPM on ferroptosis. Moreover, the oncogenic Src-YAP/TAZ-TFRC axis may have represented a ferroptosis vulnerability and a potential biomarker in ferroptosis therapy for cancer. Importantly, other cardiac glycosides targeting Na+/K+-ATPase, such as digitoxin and bufalin, also enhanced ferroptosis sensitivity in gastric cancer cells through activation of YAP/TAZ signaling. CONCLUSION Our findings establish the cardiac glycoside PPM as a novel ferroptosis sensitizer that targets ATP1A1 to activate the Src-YAP/TAZ-TFRC axis, providing mechanistic insights for repurposing cardiac glycosides as ferroptosis modulators in precision combinatorial cancer therapy.
Collapse
Affiliation(s)
- Angting Ke
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Weiguang Yang
- Department of Nephrology, the First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Wanchuan Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yibin Chen
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xiangyu Meng
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China
| | - Jie Liu
- Translational Research Experiment Department, Science Experiment Center, China Medical University, Shenyang 110122, China
| | - Dongqiu Dai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
3
|
Iglesias-Jiménez A, Artiaga G, Moreno-Gordaliza E, Milagros Gómez-Gómez M. Metallomic evaluation of selenium nanoparticles and selenomethionine for the attenuation of cisplatin-induced nephrotoxicity. Eur J Pharm Biopharm 2025; 212:114737. [PMID: 40345401 DOI: 10.1016/j.ejpb.2025.114737] [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: 10/28/2024] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Nephrotoxicity is one of the most limiting side effects in oncologic patients treated with cisplatin and is still clinically unresolved. In this work, chitosan-stabilised selenium nanoparticles (Ch-SeNPs) and selenomethionine (SeMet) have been evaluated as nephroprotectors of cisplatin using renal proximal tubule epithelial cells (RPTEC/TERT1) as a model. Moreover, the antineoplastic efficacy of cisplatin co-administered with these selenocompounds has been tested in cervical cancer cells (HeLa). Cell viability, cell localisation of Ch-SeNPs and changes in the morphology and cell ultrastructure, Pt and Se cellular internalisation and cisplatin binding to DNA, and speciation of Pt and Se in the cytosolic extracts were evaluated by MTT assays, transmission electron microscopy coupled to energy dispersive X-ray spectroscopy (TEM-EDS), inductively coupled plasma mass spectrometry (ICP-MS), and both size exclusion chromatography (SEC) and anion exchange chromatography (AEC) coupled to either ICP-MS or UV-Vis. Differences in the pharmacological activity of the two selenospecies were observed. SeMet exerted a moderate protection on kidney cells while reducing their degree of cisplatin intracellular accumulation and DNA binding in both cell lines, but the antitumour effect of cisplatin was not significantly altered. Conversely, Ch-SeNPs did not impair the Pt-drug uptake or DNA binding in any cell type; and even increased its antitumour effect, which might enable using lower doses of cisplatin without loss of anticancer efficacy, which would result in decreased risk of renotoxicity. Furthermore, cells incubated either with SeMet or SeNPs showed higher levels of selenoproteins, which might enhance cellular defences against the reactive oxygen species (ROS) involved in cisplatin renotoxicity. Hence, both selenocompounds are envisioned as potential coadjuvants to reduce the risk of kidney impairment in future treatments with cisplatin.
Collapse
Affiliation(s)
- Alejandro Iglesias-Jiménez
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain.
| | - Gema Artiaga
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain.
| | - Estefanía Moreno-Gordaliza
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain.
| | - M Milagros Gómez-Gómez
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain.
| |
Collapse
|
4
|
Poudel N, Zheng S, Skrypnyk N, Sung SSJ, Goggins E, Nash WT, Pavelec C, Yee M, Balogun I, Medina CB, Yao J, Rosin DL, Leitinger N, Ravichandran KS, Okusa MD. Proximal tubule pannexin 1 contributes to mitochondrial dysfunction and cell death during acute kidney injury. Am J Physiol Renal Physiol 2025; 328:F830-F849. [PMID: 40241514 DOI: 10.1152/ajprenal.00226.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/04/2024] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Pannexin 1 (Panx1) is a membrane-associated channel that, when activated, facilitates the release of small metabolites into the extracellular environment. These metabolites signal as damage-associated molecular patterns (DAMP) and initiate inflammation. Upregulation and activation of Panx1 is one of the early events during inflammatory injury. Animal models show that a lack of Panx1 is protective against acute kidney injury (AKI). How Panx1 modulates AKI is poorly understood. We utilized both in vivo and in vitro models of PANX1 overexpression to study mitochondrial function, cell death, and inflammation to evaluate how Panx1 contributes to AKI. We used two models of AKI, ischemia-reperfusion injury (IRI) and cisplatin-induced AKI (cis-AKI), in animals that overexpress PANX1 globally or specifically in the proximal tubule or in the endothelium. Cisplatin-induced injury was investigated in vitro in PANX1-overexpressing proximal tubule cells in culture. Both global and proximal tubule-specific overexpression of PANX1 exacerbated AKI, whereas endothelium-specific overexpression had no effect. Panx1-dependent metabolite release and alterations in the intracellular compartment in proximal tubules independently contributed to cell death in vitro. PANX1 overexpression impaired mitochondrial function and increased mitochondrial reactive oxygen species (ROS) production. PANX1 overexpression resulted in increased inflammation in the kidneys during cis-AKI. We showed that PANX1 overexpression resulted in overt renal injury during AKI that is in part mediated by reduced mitochondrial function, increased cell death, and inflammation. Selective strategies to inhibit Panx1 could help prevent or treat AKI.NEW & NOTEWORTHY Despite the huge medical, economical, and quality of life burden that AKI poses to patients, there are no Food and Drug Administration (FDA)-approved therapeutic or pharmaceutical interventions for AKI. Pannexin 1 (Panx1), which is upregulated in patients with AKI as well as in animals that develop experimental AKI, plays a crucial role in mediating both inflammation and cell death during AKI. Our findings suggest clinical interventions with molecules that inhibit Panx1 channel activity could improve outcomes in AKI patients.
Collapse
Affiliation(s)
- Nabin Poudel
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Shuqiu Zheng
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Nataliya Skrypnyk
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Sun-Sang J Sung
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Eibhlin Goggins
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - William T Nash
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Caitlin Pavelec
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, United States
| | - Marissa Yee
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Ishaq Balogun
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Cristopher B Medina
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States
| | - Junlan Yao
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| | - Diane L Rosin
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, United States
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, United States
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|
5
|
Lou Y, Zhou F, Shi D, Zhang Y, Dong J, Shen X, Chen X, Liu X. Discovery of Novel Potent Molecule for Treating Cisplatin-Induced Acute Kidney Injury Driven by Phenotypic Screening. J Med Chem 2025. [PMID: 40448635 DOI: 10.1021/acs.jmedchem.5c00954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Cisplatin-induced acute kidney injury (cis-AKI) is the primary organ toxicity that occurs during cisplatin-based cancer treatment, and currently, there is a lack of effective treatment methods. The occurrence of cis-AKI involves complex pathophysiological processes, therefore, phenotypic screening-based drug discovery may be more applicable for identifying lead compounds to treat cis-AKI. In this study, a cascading phenotypic screening was designed and implemented. Through this protocol, a potent compound, CX116, was obtained, which exhibits potent anti-inflammatory activity and effectively protects cells from cisplatin toxicity. Mechanism studies indicated that CX116 exerts its effects by inhibiting the inflammatory response, reducing oxidative stress, protecting mitochondrial function, and counteracting apoptosis. In vivo studies showed that CX116 bears acceptable toxicity and appropriate pharmacokinetic properties, and significantly protects renal tissue from cisplatin-induced damage in a cis-AKI animal model. In conclusion, CX116 is a highly promising active molecule and holds great potential for the development of therapeutic drugs for cis-AKI.
Collapse
Affiliation(s)
- Yan Lou
- School of Pharmacy, Anhui Medical University, Hefei 230032, P.R. China
| | - Feilong Zhou
- School of Pharmacy, Anhui Medical University, Hefei 230032, P.R. China
| | - Daxing Shi
- School of Pharmacy, Anhui Medical University, Hefei 230032, P.R. China
| | - Yu Zhang
- School of Pharmacy, Anhui Medical University, Hefei 230032, P.R. China
| | - Jin Dong
- School of Pharmacy, Anhui Medical University, Hefei 230032, P.R. China
| | - Xiaobao Shen
- Medical School, Fuyang Normal University, Fuyang 236037, P.R. China
| | - Xing Chen
- School of Pharmacy, Anhui Medical University, Hefei 230032, P.R. China
| | - Xinhua Liu
- School of Pharmacy, Anhui Medical University, Hefei 230032, P.R. China
| |
Collapse
|
6
|
Markley P, Kadhem S, Moore J. Abemaciclib-Induced Pseudo-AKI: an underrecognised finding. BMJ Case Rep 2025; 18:e261725. [PMID: 40449940 DOI: 10.1136/bcr-2024-261725] [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: 06/03/2025] Open
Abstract
Targeted therapy-induced acute kidney injury (AKI) can complicate oncologic treatment. We present a case of creatinine elevation induced by abemaciclib, a cyclin-dependent kinases 4 and 6 inhibitor used in advanced breast cancer which can also cause nephrotoxicity. A female patient in her mid-sixties was referred to nephrology for elevated serum creatinine after starting abemaciclib. Diagnostic workup, including blood urea nitrogen (BUN), serum cystatin C, urine protein measurement, and serum electrolytes revealed no evidence of renal dysfunction. This case emphasises the importance of distinguishing creatinine elevation from true nephrotoxicity to avoid unnecessary treatment adjustments. Abemaciclib and other targeted therapies can induce creatinine elevation by inhibiting proximal tubule secretory transporters, rather than through direct nephrotoxicity. By incorporating additional assessments such as BUN, serum cystatin C, and additional biomarkers including urinary neutrophil gelatinase-associated lipocalin and kidney injury molecule 1, clinicians can distinguish creatinine elevation from AKI to guide appropriate management of oncologic diseases.
Collapse
Affiliation(s)
- Payton Markley
- Medicine, The University of Kansas School of Medicine-Wichita, Wichita, Kansas, USA
| | - Salam Kadhem
- Kansas Nephrology Physicians, Wichita, Kansas, USA
| | | |
Collapse
|
7
|
Chen S, Zheng W, Wang Y, Zhao X, Deng W, Chai N. Paeoniflorin attenuates cisplatin induced ototoxicity by inhibiting ferroptosis mediated by HMGB1/NRF2/GPX4 pathway. Food Chem Toxicol 2025:115550. [PMID: 40374000 DOI: 10.1016/j.fct.2025.115550] [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: 02/13/2025] [Revised: 05/01/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Cisplatin-induced ototoxicity is a major dose-limiting complication in cancer therapy, profoundly diminishing quality of life. Paeoniflorin (PAE), a bioactive compound from Paeonia lactiflora, exhibits diverse pharmacological activities. This study aimed to evaluate the efficacy of PAE in countering cisplatin-induced ototoxicity and explore its molecular mechanisms. Cochlear hair cell injury models were established both in vitro and in vivo using cisplatin. Ferroptosis was induced in HEI-OC1 cells with RSL3, and HMGB1-overexpressing models were constructed to investigate its role. The findings indicated that PAE effectively alleviated cisplatin-induced hearing loss and cochlear cell damage in both in vitro and in vivo models. Moreover, PAE significantly mitigated the inflammatory response triggered by cisplatin exposure. Mechanistically, PAE reduced oxidative stress and ferroptosis by upregulating nuclear factor erythroid 2-related factor 2 (NRF2), solute carrier family 7 member 11 (SLC7A11), and glutathione peroxidase 4 (GPX4). Notably, PAE directly interacted with HMGB1 and suppressed its expression, thereby inhibiting HMGB1-mediated ferroptosis in cochlear cells. This study highlights PAE as a promising therapeutic candidate for preventing cisplatin-induced ototoxicity. By elucidating the role of PAE in modulating ferroptosis through HMGB1 and the NRF2/SLC7A11/GPX4 pathway, our findings provide new insights into potential strategies for mitigating cisplatin-induced hearing loss.
Collapse
Affiliation(s)
- Shaoli Chen
- Department of Otolaryngology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Zheng
- Department of Otolaryngology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yichao Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyu Zhao
- Department of Otolaryngology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenhao Deng
- Department of Otolaryngology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ni Chai
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
8
|
Albuquerque EL, Horowitz JG, Lee GC. Clinical safety of a 1-h infusion of cisplatin approach in a broad cancer population. J Oncol Pharm Pract 2025:10781552251340630. [PMID: 40356492 DOI: 10.1177/10781552251340630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
IntroductionCisplatin has ubiquitous use throughout many cancer types and is incorporated in various regimens. Nephrotoxicity is a well-known side effect of cisplatin, occurring in up to one third of patients. Early clinical data showed elevated peak concentrations are correlated with increased incidence of nephrotoxicity, suggesting prolonging infusion duration may abate this known toxicity. A recent study showed there was no difference in rates of acute kidney injury (AKI) between rapid (1-h) and standard (3-h) infusions of cisplatin, however, cisplatin was administered inpatient with prolonged and aggressive hydration methods. Therefore, we aimed to compare outcomes in patients who received rapid-infusion vs. standard-infusion cisplatin in the outpatient setting at our institution.MethodsThis single center, pre-post intervention study evaluated outpatient visits between January 2022 and March 2024. Cisplatin was administered over 3-h at Mays Cancer Center, UT Health San Antonio, prior to an institutional practice change to 1-h (rapid infusion) that took place February 2023. The co-primary outcomes were the maximum decrease in eGFR from baseline and the incidence of stage 1 or higher AKI. Secondary outcomes included rates of treatment modification and rates of healthcare resource utilization.ResultsA total of 53 patients with a chemotherapy regimen containing cisplatin met study criteria. Baseline characteristics were similar for the standard (n = 28) and rapid infusion (n = 25) groups. The maximum decrease in eGFR in the standard infusion vs. rapid infusion group did not differ (15.2 vs 18.0 mL/min/1.73 m2, respectively; p = 0.89). The incidence of stage 1 or higher AKI was not significantly different between the standard and rapid infusion cohorts (p = 0.13). No differences were identified for the secondary outcomes including rates of regimen modifications or hospitalizations due to AKI or time to AKI in 120 days after the first cisplatin dose.ConclusionIn this pre-post intervention analysis neither occurrence nor severity of nephrotoxicity appeared to be affected by the infusion rate of cisplatin in the outpatient setting. Rapid infusion of cisplatin over 1-h appears to be a feasible option without increased safety risk for patients.
Collapse
Affiliation(s)
- Esther L Albuquerque
- College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- Pharmacotherapy Education and Research Center, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Justin G Horowitz
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Grace C Lee
- College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- Pharmacotherapy Education and Research Center, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| |
Collapse
|
9
|
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
|
10
|
Park I, Kim S, Um YW, Kim HE, Lee JH, Kim S, Kim P, Jo YH. Intravital imaging of peritubular microcirculation impairment in cisplatin-induced acute kidney injury. JCI Insight 2025; 10:e178689. [PMID: 40198123 DOI: 10.1172/jci.insight.178689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025] Open
Abstract
Despite the accumulation of cisplatin in proximal tubules, direct visualization of the surrounding peritubular microcirculation, including its change in cisplatin-induced acute kidney injury (AKI), is lacking. Here, using fluorescence and cellular angiography through video-rate high-resolution intravital microscopy, progressive disturbance of peritubular microcirculation in cisplatin-induced AKI in mice was demonstrated. Fluorescence angiography revealed increasing perfusion defects, with a stepwise rise in time to peak (TTP), originating from capillaries surrounding S1 segments. Cellular angiography demonstrated a progressive decrease in the velocity and track length of individual erythrocytes during AKI progression, accompanied by a sequential decrease in the functional capillary ratio (FCR). Changes in the perfusion area, TTP, and FCR preceded significant changes in blood urea nitrogen and cystatin C, suggesting the potential for early diagnosis. Although neutrophil infiltration near proximal tubules increased throughout the progression, it did not cause obstruction of the peritubular microcirculation. Depletion of neutrophils increased mortality due to systemic side effects, whereas functional inactivation of neutrophils using an anti-CD11b antibody improved peritubular microcirculation in cisplatin-induced AKI. This approach enables direct visualization and quantification of peritubular microcirculation and immune cell dynamics, providing insights into renal pathophysiology and potential therapeutic strategies.
Collapse
Affiliation(s)
- Inwon Park
- Department of Emergency Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam, Republic of Korea
- Department of Emergency Medicine, and
- Disaster Medicine Research Center, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seonghye Kim
- Department of Emergency Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam, Republic of Korea
| | - Young Woo Um
- Department of Emergency Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam, Republic of Korea
| | - Hee Eun Kim
- Department of Emergency Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam, Republic of Korea
| | - Jae Hyuk Lee
- Department of Emergency Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam, Republic of Korea
- Department of Emergency Medicine, and
- Disaster Medicine Research Center, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sejoong Kim
- Division of Nephrology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Pilhan Kim
- Graduate School of Medical Science and Engineering, and
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - You Hwan Jo
- Department of Emergency Medicine, Seoul National University Bundang Hospital (SNUBH), Seongnam, Republic of Korea
- Department of Emergency Medicine, and
- Disaster Medicine Research Center, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
11
|
Sun X, Wei Z, Liu Y, Li F, Liu X, Ding Q, Ding Z, Liu J, Sun Q. Structural features and efficacy of polysaccharide EPS22 from Klebsiella sp. S22 in mitigating cisplatin-induced acute kidney injury. Int J Biol Macromol 2025; 311:143944. [PMID: 40328401 DOI: 10.1016/j.ijbiomac.2025.143944] [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: 02/21/2025] [Revised: 04/13/2025] [Accepted: 05/03/2025] [Indexed: 05/08/2025]
Abstract
Polysaccharides are a type of natural macromolecular biopolymer with various biological activities. In this study, the structural characteristics and biological activity of a novel polysaccharide EPS22 have been elucidated. EPS22, a homogeneous acidic polysaccharide, was produced by Klebsiella sp. S22 with the yield of 16.4 g/L and a molecular weight of 63,800 Da. The hexasaccharide repeating units of EPS22 were structured as follows: →2)[α-L-Rhap-(1→3)-β-D-Galp-(1→2)-α-L-Rhap-(1→ 3)]-α-D-Manp-(1→4)-β-D-GlcpA-(1→2)-α-D-Manp-(1→. EPS22 demonstrated a dose-dependent protective effect against cisplatin-induced acute kidney injury (AKI). EPS22 mitigated cisplatin-induced AKI by attenuating renal oxidative damage through the NRF2 pathway, diminishing inflammation through reduced cell infiltration and NF-κB pathway downregulation, and preventing renal cell apoptosis via the Bcl-2/Bax/Caspase-3 signaling pathway. Meanwhile, EPS22 could regulate the renal metabolic profile associated with kidney injury. These results indicate that EPS22 has potential pharmaceutical and functional food value in alleviating cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Xiaqing Sun
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu 233030, China; Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, Anhui, China
| | - Zhenxuan Wei
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu 233030, China; Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, Anhui, China
| | - Yuanyan Liu
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu 233030, China; Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, Anhui, China
| | - Fangting Li
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, Anhui, China; Department of Dermatology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Xuqin Liu
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, Anhui, China; Department of Dermatology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Qian Ding
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, Anhui, China
| | - Zhao Ding
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, Anhui, China
| | - Junhao Liu
- Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, Anhui, China.
| | - Qi Sun
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu 233030, China; Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu, Anhui, China.
| |
Collapse
|
12
|
Zhang J, Zhang M, Tatar M, Gong R. Keap1-independent Nrf2 regulation: A novel therapeutic target for treating kidney disease. Redox Biol 2025; 82:103593. [PMID: 40107017 PMCID: PMC11968292 DOI: 10.1016/j.redox.2025.103593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
The transcription factor NF-E2-related factor 2 (Nrf2) is a master regulator of antioxidant responses in mammals, where it plays a critical role in detoxification, maintaining cellular homeostasis, combating inflammation and fibrosis, and slowing disease progression. Kelch-like ECH-associated protein 1 (Keap1), an adaptor subunit of Cullin 3-based E3 ubiquitin ligase, serves as a critical sensor of oxidative and electrophilic stress, regulating Nrf2 activity by sequestering it in the cytoplasm, leading to its proteasomal degradation and transcriptional repression. However, the clinical potential of targeting the Keap1-dependent Nrf2 regulatory pathway has been limited. This is evidenced by early postnatal lethality in Keap1 knockout mice, as well as significant adverse events after pharmacological blockade of Keap1 in human patients with Alport syndrome as well as in those with type 2 diabetes mellitus and chronic kidney disease. The exact underlying mechanisms remain elusive, but may involve non-specific and systemic activation of the Nrf2 antioxidant response in both injured and normal tissues. Beyond Keap1-dependent regulation, Nrf2 activity is modulated by Keap1-independent mechanisms, including transcriptional, epigenetic, and post-translational modifications. In particular, GSK3β has emerged as a critical convergence point for these diverse signaling pathways. Unlike Keap1-dependent regulation, GSK3β-mediated Keap1-independent Nrf2 regulation does not affect basal Nrf2 activity but modulates its response at a delayed/late phase of cellular stress. This allows fine-tuning of the inducibility, magnitude, and duration of the Nrf2 response specifically in stressed or injured tissues. As one of the most metabolically active organs, the kidney is a major source of production of reactive oxygen and nitrogen species and also a vulnerable organ to oxidative damage. Targeting the GSK3β-mediated Nrf2 regulatory pathway represents a promising new approach for the treatment of kidney disease.
Collapse
Affiliation(s)
- Jiahui Zhang
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Mingzhuo Zhang
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Marc Tatar
- Division of Biology and Medicine, Brown University, Providence, RI, USA
| | - Rujun Gong
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA.
| |
Collapse
|
13
|
Shahrahmani F, Badamchizadeh S, Kaihani F, Alavi-Moghadam S, Keshtkari S, Rezaei-Tavirani M, Arjmand R, Larijani B, Arjmand B. Platinum-based chemotherapies-induced nephrotoxicity: mechanisms, potential treatments, and management. Int Urol Nephrol 2025; 57:1563-1583. [PMID: 39630371 DOI: 10.1007/s11255-024-04303-2] [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: 09/25/2024] [Accepted: 11/20/2024] [Indexed: 04/17/2025]
Abstract
Platinum-based chemotherapies are essential in the treatment of several malignancies. However, such medications can damage the kidneys, frequently leading to both acute and chronic kidney disease. Treatment becomes more difficult for such problems. Physicians may alter chemotherapy regimens and utilize kidney-protecting medications to lessen renal damage. New imaging techniques and biomarkers also aid in the early detection of renal issues. To effectively handle the mentioned situation, oncologists, nephrologists, and pharmacists must collaborate. However, additional study is still required to develop customized therapies, discover strategies to minimize kidney injury and produce new platinum medicines. Hereupon, the present review's authors are being sought to address the causes, prospective treatments, and management of nephrotoxicity caused by platinum-based chemotherapy.
Collapse
Affiliation(s)
- Fatemeh Shahrahmani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sana Badamchizadeh
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Keshtkari
- Department of Internal Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | | - Rasta Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
You R, Li Y, Jiang Y, Hu D, Gu M, Zhou W, Zhang S, Bai M, Yang Y, Zhang Y, Huang S, Jia Z, Zhang A. WWP2 deletion aggravates acute kidney injury by targeting CDC20/autophagy axis. J Adv Res 2025; 71:471-485. [PMID: 38909885 DOI: 10.1016/j.jare.2024.06.015] [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: 03/30/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024] Open
Abstract
INTRODUCTION Acute kidney injury (AKI) is associated with high morbidity and mortality rates. The molecular mechanisms underlying AKI are currently being extensively investigated. WWP2 is an E3 ligase that regulates cell proliferation and differentiation. Whether WWP2 plays a regulatory role in AKI remains to be elucidated. OBJECTIVES We aimed to investigate the implication of WWP2 in AKI and its underlying mechanism in the present study. METHODS We utilized renal tissues from patients with AKI and established AKI models in global or tubule-specific knockout (cKO) mice strains to study WWP2's implication in AKI. We also systemically analyzed ubiquitylation omics and proteomics to decipher the underlying mechanism. RESULTS In the present study, we found that WWP2 expression significantly increased in the tubules of kidneys with AKI. Global or tubule-specific knockout of WWP2 significantly aggravated renal dysfunction and tubular injury in AKI kidneys, whereas WWP2 overexpression significantly protected tubular epithelial cells against cisplatin. WWP2 deficiency profoundly affected autophagy in AKI kidneys. Further analysis with ubiquitylation omics, quantitative proteomics and experimental validation suggested that WWP2 mediated poly-ubiquitylation of CDC20, a negative regulator of autophagy. CDC20 was significantly decreased in AKI kidneys, and selective inhibiting CDC20 with apcin profoundly alleviated renal dysfunction and tubular injury in the cisplatin model with or without WWP2 cKO, indicating that CDC20 may serve as a downstream target of WWP2 in AKI. Inhibiting autophagy with 3-methyladenine blocked apcin's protection against cisplatin-induced renal tubular cell injury. Activating autophagy by rapamycin significantly protected against cisplatin-induced AKI in WWP2 cKO mice, whereas inhibiting autophagy by 3-methyladenine further aggravated apoptosis in cisplatin-exposed WWP2 KO cells. CONCLUSION Taken together, our data indicated that the WWP2/CDC20/autophagy may be an essential intrinsic protective mechanism against AKI. Further activating WWP2 or inhibiting CDC20 may be novel therapeutic strategies for AKI.
Collapse
Affiliation(s)
- Ran You
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yanwei Li
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China; School of Medicine, Southeast University, Nanjing, China
| | - Yuteng Jiang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; School of Medicine, Southeast University, Nanjing, China
| | - Dandan Hu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Menglei Gu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhou
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Shengnan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Mi Bai
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yunwen Yang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China; School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
15
|
Abd-Eldayem AM, Ali MF, Ahmed EA. Nebivolol rescued the liver and kidney from the coadministration of rivaroxaban and cisplatin by targeting inflammation, oxidative stress, and apoptosis in rats. Int Immunopharmacol 2025; 153:114486. [PMID: 40112601 DOI: 10.1016/j.intimp.2025.114486] [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: 10/30/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Cisplatin is among the most frequently utilized drugs for addressing malignant tumors, yet it can lead to organ harm, especially hepatotoxicity and nephrotoxicity. Furthermore, the anticoagulant rivaroxaban could potentially cause injury to the liver and kidneys. This research aimed to examine the protective benefits of nebivolol, known for its pleiotropic and tissue-protective characteristics, against the harmful effects of rivaroxaban and cisplatin on the liver and kidneys. Male rats received cisplatin and/or rivaroxaban, and we evaluated hepatotoxicity and nephrotoxicity by measuring serum concentrations of AST, ALT, LDH, albumin, bilirubin, creatinine, and blood urea. We also measured MDA, GSH, GPx, NO, TNF-α, and IL-6 in kidney and liver homogenates. Histopathological analysis was performed on liver and kidney tissue sections, and immunohistochemical detection of caspase 3 in liver tissue and NF-κB in kidney tissue was conducted. Our findings demonstrated that nebivolol supported the preservation of the liver and kidney structure and function by reducing the biochemical and pathological alterations caused by cisplatin and rivaroxaban. Nebivolol decreased the elevations in MDA, TNF-α, and IL-6 levels while maintaining GSH, GPx, and NO levels in liver and kidney tissues. Moreover, nebivolol lowered the levels of caspase-3 in the liver and NF-κB in the kidneys. In conclusion, our study indicates that nebivolol protects the liver and kidneys from the detrimental effects of cisplatin and rivaroxaban.
Collapse
Affiliation(s)
- Ahmed M Abd-Eldayem
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, 71515 Assiut. Egypt; Pharmacology department, College of Medicine, Fahad Bin Sultan University (FBSU), 15700, Tabuk 71454, Saudi Arabia.
| | - Marwa F Ali
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Assiut, Assiut University, Egypt
| | - Esraa A Ahmed
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, 71515 Assiut. Egypt
| |
Collapse
|
16
|
Elmorsy EM, Al Doghaither HA, Al-Ghafari AB, Ebrahim NA, Amer S. The flavonoid hyperoside attenuates the toxic effect of cisplatin on the human ovarian granulosa cells: in vitro model study. Toxicol Mech Methods 2025:1-11. [PMID: 40257416 DOI: 10.1080/15376516.2025.2491774] [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: 12/21/2024] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 04/22/2025]
Abstract
Premature ovarian insufficiency/failure is a well-known long-term risk of chemotherapy including CDDP in women. Granulosa cells (GCs) are an essential ovarian cell type that promotes oocyte growth and is crucial for ovarian reproductive function. Hyperoside (HYP) is a flavonoid known for its beneficial pharmacological properties, including anti-inflammatory and antiapoptotic effects. Hence the current work aimed to evaluate the potential cytoprotective impact of HYP on CDDP-induced cytotoxicity in a human ovarian GCs cell line model via a wide range of assays including MTT, hormones secretion, ATP and mitochondrial membrane potential, reactive oxygen species, lipid peroxidation as well as antioxidant enzymes, Caspases, and Akt kinase activities. Forty-eight-hour exposure to 5-10µM CDDP resulted in reduction of GCs viability in a dose-dependent manner. HYP (40 µM) was found to ameliorate this CDDP -induced effect on GCs viability. CDDP in a concentration-dependent way, dramatically reduced cellular ATP, mitochondrial activities, cellular progesterone, and estradiol secretion. It also increased oxidative stress markers, cytochrome c levels, caspase -3.-8.-9, and Bax/Bcl2 ratio with decreased Akt kinase activity and its coding genes expression. These cytotoxic effects of CDDP on the treated GCs, were mitigated to varying degrees by HYP (40 µM). In conclusion, CDDP-induced cytotoxic effects on GCs seem to be the possible underlying cellular and molecular mechanisms of CDDP-induced ovarian insufficiency/failure. The study also demonstrated the therapeutic potential of HYP in mitigating CDDP-induced ovarian injury. Further studies are warranted to investigate the potential benefit of HYP as an adjuvant to CDDP treatment protocols to avoid adverse ovarian effects.
Collapse
Affiliation(s)
- Ekramy M Elmorsy
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Huda A Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ayat B Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Neven A Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah, Saudi Arabia
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Saad Amer
- Translational Medical Sciences, School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby, United Kingdom
| |
Collapse
|
17
|
Wei J, Xie Z, Kuang X. Extracellular Vesicles in Renal Inflammatory Diseases: Revealing Mechanisms of Extracellular Vesicle-Mediated Macrophage Regulation. Int J Mol Sci 2025; 26:3646. [PMID: 40332144 PMCID: PMC12027779 DOI: 10.3390/ijms26083646] [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: 03/12/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Renal inflammatory diseases are a group of severe conditions marked by significant morbidity and mortality. Extracellular vesicles (EVs), as facilitators of intercellular communication, have been recognized as pivotal regulators of renal inflammatory diseases, significantly contributing to these conditions by modulating immune responses among other mechanisms. This review highlights the intricate mechanisms through which EVs modulate macrophage-kidney cell interactions by regulating macrophages, the principal immune cells within the renal milieu. This regulation subsequently influences the pathophysiology of renal inflammatory diseases such as acute kidney injury and chronic kidney disease. Furthermore, understanding these mechanisms offers novel opportunities to alleviate the severe consequences associated with renal inflammatory diseases. In addition, we summarize the therapeutic landscape based on EV-mediated macrophage regulatory mechanisms, highlighting the potential of EVs as biomarkers and therapeutic targets as well as the challenges and limitations of translating therapies into clinical practice.
Collapse
Affiliation(s)
- Jiatai Wei
- The Second Clinical Medical College, Nanchang University, Nanchang 330031, China; (J.W.); (Z.X.)
| | - Zijie Xie
- The Second Clinical Medical College, Nanchang University, Nanchang 330031, China; (J.W.); (Z.X.)
| | - Xiaodong Kuang
- Pathology Teaching and Research Office, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
18
|
Zhao Y, Zhao QQ, Fan SJ, Xu DY, Lin LM, Luo W, Ye BZ, Zou CP, Zhu H, Zhuang ZS, Zhao YJ, Liang G. JOSD2 alleviates acute kidney injury through deubiquitinating SIRT7 and negativity regulating SIRT7-NF-κB inflammatory pathway in renal tubular epithelial cells. Acta Pharmacol Sin 2025. [DOI: 10.1038/s41401-025-01546-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/17/2025] [Indexed: 06/09/2025]
|
19
|
Zaaba NE, Al-Salam S, Beegam S, Elzaki O, Aldhaheri F, Nemmar A, Ali BH, Nemmar A. Attenuation of cisplatin-induced acute kidney injury by sanguinarine: modulation of oxidative stress, inflammation, and cellular damage. Front Pharmacol 2025; 16:1567888. [PMID: 40242453 PMCID: PMC11999955 DOI: 10.3389/fphar.2025.1567888] [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: 01/28/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Introduction Cisplatin (CP)-induced acute kidney injury (AKI) is a significant side effect of CP chemotherapy, driven by oxidative stress and inflammation. Sanguinarine (SANG), an alkaloid from the rhizomes of Sanguinaria canadensis and poppy-fumaria species, exhibits antioxidant and anti-inflammatory properties. This study examined SANG's effect on CP-induced AKI in mice and its underlying mechanisms. Methods Mice were orally administered 5 mg/kg SANG for 10 days. On the seventh day, they received a single intraperitoneal CP injection (20 mg/kg) and were sacrificed on the 11th day. Results SANG significantly improved CP-induced decreases in body weight, water intake, urine volume, relative kidney weight, creatinine clearance, albumin-to-creatinine ratio, and plasma urea and creatinine levels. It also reduced elevated plasma neutrophil gelatinase-associated lipocalin, kidney injury molecule-1, cystatin C, and adiponectin levels, as well as renal markers of inflammation and oxidative stress induced by CP administration. SANG normalized kidney mitochondrial dysfunction, DNA damage, and apoptosis caused by CP. It also inhibited the CP-induced increase in the expression of phosphorylated nuclear factor-κB and autophagy markers in the kidney. Histological analysis showed that SANG reduced acute tubular necrosis and intraluminal protein accumulation due to CP. Discussion In conclusion, SANG mitigated CP-induced AKI by reducing inflammation, oxidative stress, DNA damage, apoptosis, and autophagy. Pending more comprehensive pharmacological and toxicological assessments, SANG may be regarded as a potential therapeutic agent for mitigating CP-induced AKI.
Collapse
Affiliation(s)
- Nur Elena Zaaba
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ozaz Elzaki
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Fatima Aldhaheri
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Anas Nemmar
- College of Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Badreldin H. Ali
- Emeritus Professor, Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Science, Sultan Qaboos University, Muscat, Oman
| | - Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
20
|
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
|
21
|
Fawzy MH, Moustafa YM, Khodeer DM, Saeed NM, El-Sayed NM. Doxepin as OCT2 inhibitor ameliorates inflammatory response and modulates PI3K/Akt signaling associated with cisplatin-induced nephrotoxicity in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3995-4008. [PMID: 39400714 PMCID: PMC11978677 DOI: 10.1007/s00210-024-03473-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/18/2024] [Indexed: 10/15/2024]
Abstract
Organic cationic transporter 2 (OCT2) was identified as the main transporter involved in the accumulation of cisplatin (CP) in the proximal tubular renal cells, resulting in nephrotoxicity. Doxepin (DOX) is a tricyclic agent with an inhibitory effect on OCT2. This study aimed to explore the possible mechanisms of the renoprotective role of DOX toward CP-induced nephrotoxicity. Rats were randomly divided into six groups: group 1, control; group 2, CP; groups 3, 4, and 5 were treated with graded doses of DOX (5, 10, and 20 mg/kg, respectively) intraperitoneally (ip) once daily for 10 consecutive days and group 6 was treated only with DOX (20 mg/kg). On the seventh day, a single injected dose of CP (10 mg/kg, ip) was given to the rats in groups 2-5. Seventy-two hours after CP injection, rats were sacrificed, and the kidneys were removed for histological and biochemical measurements. DOX ameliorated the CP-induced histopathological alterations. DOX significantly reduced the expression of OCT2, lipid peroxidation marker (MDA), and inflammatory cytokines, including TNF-α, IL-6, IL-1, IL-2, and IL-1β, and increased the activity of antioxidant enzymes. In addition, pre- and co-treatment with DOX significantly reduced the CP-mediated apoptotic effect by reducing the renal tissue expression of BAX and caspase-3 levels, upregulating the expression of Bcl-2, and modulating the phosphorylation of PI3K/Akt signaling cascade. DOX exerts a nephroprotective impact against CP-mediated nephrotoxicity via the inhibition of OCT2, suppression of inflammation, oxidative stress, and apoptotic markers, and modulation of PI3K/Akt signaling cascade.
Collapse
Affiliation(s)
- Mariam H Fawzy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Yasser M Moustafa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo, Badr City, Egypt
| | - Dina M Khodeer
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Noha M Saeed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Norhan M El-Sayed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
22
|
Strader M, Friedman G, Benain X, Camerlingo N, Sultana S, Shapira S, Aber N, Murray PT. Early and Sensitive Detection of Cisplatin-Induced Kidney Injury Using Novel Biomarkers. Kidney Int Rep 2025; 10:1175-1187. [PMID: 40303198 PMCID: PMC12034874 DOI: 10.1016/j.ekir.2025.01.035] [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: 08/04/2024] [Revised: 01/10/2025] [Accepted: 01/27/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction We evaluated a panel of novel urinary and serum biomarkers (BMs) for early and sensitive detection of cisplatin drug-induced kidney injury (DIKI) in patients with cancer, comparing their diagnostic accuracy with standard BMs (SBMs). Methods In this prospective exploratory observational study, 105 patients treated with cisplatin ("treated" with > 65 mg/m2/cycle), 20 non-cisplatin treated cancer controls ("nontreated"), and 34 "healthy" controls were enrolled. The treated group's serum and urine samples were collected predose, after 12 hours, and on days 1, 2, 4, 7, 14, and 21. SBMs and novel BMs (NBMs; 8 urinary, 1 serum) were measured, comparing accuracy, percent changes from baseline (PCFBs), and median time to peak values between treated patients and nontreated cancer controls. Blinded adjudication of the treated group's BM profiles occurred at 2 stages for DIKI diagnosis. Results All urinary NBMs had significant PCFBs in the treated group compared with the nontreated cancer control group; most accurately detected cisplatin exposure (area under the receiver operating characteristics [ROC] curve [AUROC] > 0.8). NBMs peaked earlier. In stage 1 adjudication (SBMs) of the treated group, PCFB of urinary NBMs showed no difference between DIKI (n = 24) and no-DIKI (n = 71) groups except for neutrophil gelatinase-associated lipocalin (NGAL) and cystatin C (CYSC). In treated participants, all BMs showed greater PCFBs than control groups, regardless of stage 1 DIKI adjudication. Stage 2 (SBMs and NBMs), DIKI incidence (n = 63) increased by 41%, with most BMs having an AUROC > 0.80 compared with the nontreated cancer control group. Conclusion NBMs accurately and timely detected cisplatin exposure and identified "sub-clinical" DIKI undetected by standard acute kidney injury (AKI) criteria, highlighting the limitations of current functional BMs in estimating the true DIKI incidence.
Collapse
Affiliation(s)
- Michael Strader
- Department of Medicine, School of Medicine, University College Dublin, Dublin, Ireland
| | - Gary Friedman
- Global Biometrics and Data Management, Pfizer, Inc., Cambridge, Massachusetts, USA
| | - Xavier Benain
- Biostastics and Programming, Sanofi S.A., Paris, France
| | - Nunzio Camerlingo
- Global Biometrics and Data Management, Pfizer, Inc., Cambridge, Massachusetts, USA
| | - Stefan Sultana
- Patient Safety Center of Excellence, AstraZeneca PLC, Cambridge, UK
| | - Shiran Shapira
- Health Promotion Center and Integrated Cancer Prevention Center, Tel Aviv Sourasky Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Nadir Aber
- Health Promotion Center and Integrated Cancer Prevention Center, Tel Aviv Sourasky Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Patrick T. Murray
- Department of Medicine, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
23
|
Alanazi ST, Salama SA, Althobaiti MM, Bakhsh A, Aljehani NM, Alanazi E, Alanazi MT, Musa A. Theaflavin alleviates cisplatin-induced nephrotoxicity: Targeting SIRT1/p53/FOXO3a/Nrf2 signaling and the NF-kB inflammatory cascade. Food Chem Toxicol 2025; 198:115334. [PMID: 39978533 DOI: 10.1016/j.fct.2025.115334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/09/2025] [Accepted: 02/18/2025] [Indexed: 02/22/2025]
Abstract
Cisplatin is a widely used chemotherapeutic agent. Nevertheless, a significant fraction of cisplatin-treated patients develops nephrotoxicity which limits cisplatin therapeutic implementation. The current work was devoted to investigate the potential nephroprotective impact of theaflavin against the cisplatin-induced nephrotoxicity using male Wistar rats as a mammalian model. The results indicated that theaflavin significantly improved the renal histopathological picture and glomerular filtration rate, along with reduced renal injury marker KIM-1, urinary albumin/creatinine ratio, serum creatinine, and urea. Mechanistically, theaflavin upregulated protein level of SIRT1 and downregulated the acetylated forms of the inflammatory transcription factor (TF) NF-kB, the antioxidant TF FOXO3a, and the pro-apoptotic TF p53 in the cisplatin-treated rats. Additionally, it upregulated the antioxidant TF Nrf2. In the same context, it suppressed the inflammatory responses, oxidative stress, and apoptosis. NF-kB nuclear translocation and levels of its responsive gene products IL-6 and TNF-α were suppressed. Lipids and DNA oxidation were reduced, and level of the antioxidant GSH and activity of the antioxidant enzymes SOD, GPx, and CAT were increased. The apoptotic markers caspase-3, BAX, and Bcl2 were modulated. Collectively, these findings highlight the nephroprotective competency of theaflavin against cisplatin-induced nephrotoxicity and underscore modulations of SIRT1, p53, FOXO3a, Nrf2, and NF-kB as potential targets.
Collapse
Affiliation(s)
- Samyah T Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Samir A Salama
- Division of Biochemistry, Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
| | - Musaad M Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Afnan Bakhsh
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Najla M Aljehani
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, 13316, Saudi Arabia
| | - Ebtisam Alanazi
- Cardiac Center King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Maha T Alanazi
- Medical Imaging, King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Arafa Musa
- Department of Pharmacognosy, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| |
Collapse
|
24
|
Hosseini S, Alavi Darzam I, Amirdosara M, Zangi M, Sahraei Z. Evaluating the effects of intravenous magnesium sulfate for prevention of colistin induced acute kidney injury: an open-label, placebo-controlled, block randomized clinical trial. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4559-4570. [PMID: 39503756 DOI: 10.1007/s00210-024-03583-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/28/2024] [Indexed: 04/10/2025]
Abstract
Colistin, has reinstated as a last-resort antibiotic despite its known nephrotoxicity. The aim of this study was to determine the potential nephroprotective effects of Magnesium (Mg) Sulfate during colistin therapy. This study was an open-label, placebo-controlled, block-randomized clinical trial conducted from January 2023 to February 2024 involving 87 patients eligible for colistin therapy. Patients were randomly assigned to receive either Mg sulfate (16 mEq in 100 mL of normal saline) or 100 mL of normal saline as placebo before each dose of colistin. The primary outcome of the study was the incidence of Acute Kidney Injury (AKI) during the first week of colistin therapy, while the secondary outcomes included colistin dose adjustments, length of stay in the ICU and hospital, and overall mortality. This study was registered in The Iranian Registry of Clinical Trials (IRCT20130917014693N15; 2023-01-12). A total of 87 patients (46 in Mg and 41 in control group) completed the study. Fourteen patients (30.43%) in the Mg group and twenty-one patients (51.21%) in the control group developed AKI during the first week of colistin therapy (p = 0.048). Although AKI incidence was not statistically different between the groups in unadjusted Cox regression model (HR =0.51, 95% CI =0.26-1.01, P =0.057), it became significant after adjusting for confounding factors (HR =0.40,95% CI =0.18-0.86, P =0.021). The length of hospital stay was 48.62 ± 18.82 and 44.82 ± 20.23 days for Mg and control groups respectively (p=0.373). In the Mg group, 25 out of 46 patients (54.34 %) and in the control group, 24 out of 41 patients (58.53%) eventually expired (p=0.694). This study indicates that Mg sulfate significantly reduces AKI rates and prevents hypomagnesemia, optimizing dosing and enhancing patient safety during colistin therapy.
Collapse
Affiliation(s)
- Sareh Hosseini
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ilad Alavi Darzam
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Amirdosara
- Department of Anesthesiology, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Zangi
- Department of Anesthesiology, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Sahraei
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Zhang S, Song H, Chang M, Lu Y, Liu S, Wu J, Liu Q, Pan Y, Du J, Yahaya BH, Liu Y, Lin J. MSC-EV-transmitted HSPA8 alleviates cisplatin-induced ovotoxicity by regulating the MGARP/PRDX2 axis. Int J Biol Macromol 2025; 304:140973. [PMID: 39952536 DOI: 10.1016/j.ijbiomac.2025.140973] [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: 07/23/2024] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Cisplatin (Cis) is among the most widely employed antitumour agents, although its clinical application is limited by self-induced multiple-organ toxicity. Previous studies have demonstrated the essential role of mitochondrial injury in the pathogenesis of Cis-induced ovotoxicity. Notably, mesenchymal stem cell-extracellular vesicles (MSC-EVs), potential cell-free therapeutic agents, exhibit pronounced advantages for the treatment of ovarian dysfunction. However, little is known about which core component contained in MSC-EVs plays a major role in repairing Cis-induced ovarian damage, and further, the potential mechanisms underlying the repair of mitochondrial damage remain unclear. Herein, our study first verified that MSC-EVs effectively ameliorate Cis-induced ovarian dysfunction by upregulating the level of mitochondrion-localized glutamic acid-rich protein (MGARP), after which MGARP repairs mitochondrial damage and inhibits cellular ROS production by combining with and suppressing the degradation of peroxiredoxin 2 (PRDX2) in granulosa cells (GCs). More importantly, our study further showed that heat shock protein family A member 8 (HSPA8) is indispensable for MenSC-EV-mediated improvement of Cis-induced ovotoxicity. This investigation provides novel insights into the molecular mechanisms by which MSCs alleviate Cis-induced ovotoxicity through improving mitochondrial dysfunction.
Collapse
Affiliation(s)
- Shenghui Zhang
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China; Henan Joint International Research Laboratory of Stem Cell Medicine, College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, China; Department of reproductive medicine, Zhoukou Central hospital, Zhoukou 46600, China
| | - Haofeng Song
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Mengyuan Chang
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Central Hospital, Xinxiang 453000, China
| | - Yilin Lu
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Shuyao Liu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Jie Wu
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Central Hospital, Xinxiang 453000, China
| | - Qin Liu
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China; Henan Joint International Research Laboratory of Stem Cell Medicine, College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Ying Pan
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Jiang Du
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Badrul Hisham Yahaya
- Department of Biomedical Sciences, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia.
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China; Henan Joint International Research Laboratory of Stem Cell Medicine, College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, China.
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China; Henan Joint International Research Laboratory of Stem Cell Medicine, College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
26
|
Yang Y, Wang P, Zhou K, Zhang W, Liu S, Ouyang J, Bai M, Ding G, Huang S, Jia Z, Zhang A. HUWE1-Mediated Degradation of MUTYH Facilitates DNA Damage and Mitochondrial Dysfunction to Promote Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412250. [PMID: 39921445 PMCID: PMC11967787 DOI: 10.1002/advs.202412250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/26/2025] [Indexed: 02/10/2025]
Abstract
The role of MUTYH, a DNA repair glycosylase in the pathogenesis of acute kidney injury (AKI) is unclear. In this study, it is found that MUTYH protein levels are significantly decreased in the kidneys of cisplatin- or folic acid (FA)-induced mouse AKI models and patients with AKI. MUTYH deficiency aggravates renal dysfunction and tubular injury following cisplatin and FA treatment, along with the accumulation of 7, 8-dihydro-8-oxoguanine (8-oxoG) and impairs mitochondrial function. Importantly, the overexpression of type 2 MUTYH (nuclear) significantly ameliorates cisplatin-induced apoptosis, oxidative stress, mitochondrial dysfunction, and DNA damage in vivo and in vitro. In contrast, overexpression of type 1 MUTYH (mitochondrial) shows a marginal effect against cisplatin-induced injury, indicating the chief role of type 2 MUTYH in antagonizing AKI. Interestingly, the results also indicate that the upregulation of the E3 ligase HUWE1 causes the ubiquitination and degradation of MUTYH in tubular epithelial cells. HUWE1 knockout or treatment with the HUWE1 inhibitor BI8622 significantly protect against cisplatin-induced AKI. Taken together, these results suggest that the ubiquitin E3 ligase HUWE1-mediates ubiquitination and degradation of MUTYH can aggravate DNA damage in the nucleus and mitochondria and promote AKI. Targeting the HUWE1/MUTYH pathway may be a potential strategy for AKI treatment.
Collapse
Affiliation(s)
- Yunwen Yang
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Peipei Wang
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Kaiqian Zhou
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Wen Zhang
- Department of NephrologyAffiliated Hospital of Integrated Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjing210028P. R. China
| | - Suwen Liu
- Department of PediatricsShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan250021P. R. China
| | - Jing Ouyang
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Mi Bai
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Guixia Ding
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Songming Huang
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Zhanjun Jia
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Aihua Zhang
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| |
Collapse
|
27
|
He J, Chen Y, Li Y, Feng Y. Molecular mechanisms and therapeutic interventions in acute kidney injury: a literature review. BMC Nephrol 2025; 26:144. [PMID: 40121405 PMCID: PMC11929251 DOI: 10.1186/s12882-025-04077-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Acute kidney injury (AKI) is a clinical challenge characterized by elevated morbidity and a substantial impact on individual health and socioeconomic factors. A comprehensive examination of the molecular pathways behind AKI is essential for its prevention and management. In recent years, vigorous research in the domain of AKI has concentrated on pathophysiological characteristics, early identification, and therapeutic approaches across many aetiologies and highlighted the principal themes of oxidative stress, inflammatory response, apoptosis, necrosis, and immunological response. This review comprehensively reviewed the molecular mechanisms underlying AKI, including oxidative stress, inflammatory pathways, immune cell-mediated injury, activation of the renin-angiotensin-aldosterone (RAAS) system, mitochondrial damage and autophagy, apoptosis, necrosis, etc. Inflammatory pathways are involved in the injuries in all four structural components of the kidney. We also summarized therapeutic techniques and pharmacological agents associated with the aforementioned molecular pathways. This work aims to clarify the molecular mechanisms of AKI thoroughly, offer novel insights for further investigations of AKI, and facilitate the formulation of efficient therapeutic methods to avert the progression of AKI.
Collapse
Affiliation(s)
- Jiajia He
- Department of Nephrology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yanqin Chen
- Department of Nephrology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yi Li
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Chengdu, 610072, China
| | - Yunlin Feng
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Chengdu, 610072, China.
| |
Collapse
|
28
|
Zou YX, Kantapan J, Wang HL, Li JC, Su HW, Dai J, Dechsupa N, Wang L. Iron-Quercetin complex enhances mesenchymal stem cell-mediated HGF secretion and c-Met activation to ameliorate acute kidney injury through the prevention of tubular cell apoptosis. Regen Ther 2025; 28:169-182. [PMID: 39802634 PMCID: PMC11720445 DOI: 10.1016/j.reth.2024.12.003] [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: 09/26/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
Background Acute kidney injury (AKI) is a life-threatening clinical syndrome with no effective treatment currently available. This study aims to investigate whether Iron-Quercetin complex (IronQ) pretreatment can enhance the therapeutic efficacy of Mesenchymal stem cells (MSCs) in AKI and explore the underlying mechanisms. Methods A cisplatin-induced AKI model was established in male C57BL/6 mice, followed by the intravenous administration of 1x10ˆ6 MSCs or IronQ-pretreated MSCs (MSCIronQ). Renal function, histology, and tubular cell apoptosis were analyzed three days post-treatment. In vitro, apoptosis was induced in mouse tubular epithelial cells (mTECs) using cisplatin, followed by treatment with MSCs or MSCIronQ conditioned medium (CM). Apoptosis was evaluated using TUNEL assay, RT-PCR, and western blotting. Furthermore, RNA sequencing (RNA-seq) was performed on MSCIronQ to explore the underlying mechanisms. Results Compared to MSC-treated AKI mice, those treated with MSCIronQ showed significantly improved renal function and histological outcomes, with reduced tubular cell apoptosis. A similar effect was observed in cisplatin-treated mTECs exposed to MSCIronQ-CM. Mechanistically, RNA-seq and subsequent validation revealed that IronQ treatment markedly upregulated the expression and secretion of hepatocyte growth factor (HGF) in MSCs. Furthermore, RNA interference or antibody-mediated neutralization of HGF effectively abolished the anti-apoptotic effects of MSCIronQ on mTECs. This mechanistic insight was reinforced by pharmacological inhibition of c-Met, the specific receptor of HGF, in both in vitro and in vivo models. Conclusions IronQ pretreatment enhances MSCs efficacy in AKI by promoting HGF expression and secretion, activating the HGF/c-Met pathway to suppress tubular cell apoptosis. These findings indicate that IronQ improves MSC-based therapies and offers insights into molecular mechanisms, supporting the development of better AKI treatments.
Collapse
Affiliation(s)
- Yuan-Xia Zou
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Research Center for Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Children's Diagnosis and Treatment Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Jiraporn Kantapan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Hong-Lian Wang
- Research Center for Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Jian-Chun Li
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Research Center for Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Hong-Wei Su
- Department of Urology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Jian Dai
- Research Center for Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Neurology, The Third People's Hospital, Luzhou, 646000, China
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Li Wang
- Research Center for Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
29
|
Liu F, Gu Z, Yi F, Liu X, Zou W, Xu Q, Yuan Y, Chen N, Tang J. Potential of Glycyrrhiza in the prevention of colitis-associated colon cancer. Fitoterapia 2025; 181:106398. [PMID: 39842555 DOI: 10.1016/j.fitote.2025.106398] [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: 10/16/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Glycyrrhiza, a legume native to the Mediterranean region, has a long history of ethnomedicinal use in China. Due to its antiviral, antibacterial, anti-inflammatory, antioxidant, antitumor, anti-ulcer, and hepatoprotective properties, Glycyrrhiza is widely utilized in the treatment of gastrointestinal disorders. THE AIM OF THE REVIEW The specific mechanisms of the main active constituents of glycyrrhiza in the treatment of inflammatory bowel disease, precancerous lesions and colorectal cancer at all stages of the colitis-associated colon cancer "Inflammation-Dysplasia-Cancer" sequence, as well as its pharmacokinetics, toxicology, formulation improvements, and application studies, are reviewed to provide new insights and perspectives on glycyrrhiza as a dietary supplement to treat and prevent colitis-associated colon cancer. MATERIALS AND METHODS Information on Glycyrrhiza was retrieved from electronic databases, including PubMed and Web of Science. RESULTS Glycyrrhiza is a well-established medicinal plant with significant potential for applications in both the food and pharmaceutical industries. Over 400 active constituents have been identified in Glycyrrhiza, including terpenoids, flavonoids, isoflavones, coumarins, and polyphenols. Numerous studies have demonstrated that Glycyrrhiza and its active compounds can inhibit the "Inflammation-Dysplasia-Cancer" progression of colitis-associated colon cancer by mitigating inflammatory bowel disease, reducing the number of intestinal precancerous lesions, and counteracting colorectal cancer. Furthermore, derivatives and nanocarriers are crucial for the effective treatment of colitis-associated colon cancer using Glycyrrhiza and its active constituents. CONCLUSION In conclusion, Glycyrrhiza is a plant with both medicinal and nutritional value, making it a potential food ingredient and dietary supplement for the treatment of colitis-associated colon cancer.
Collapse
Affiliation(s)
- Fang Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; North Sichuan Medical College, Nanchong, China.
| | - Zhili Gu
- North Sichuan Medical College, Nanchong, China
| | - Feiyang Yi
- North Sichuan Medical College, Nanchong, China
| | - Xue Liu
- North Sichuan Medical College, Nanchong, China
| | - Wenxuan Zou
- North Sichuan Medical College, Nanchong, China
| | - Qingxia Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yun Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Nianzhi Chen
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
30
|
Martinez SA, Karel IZ, Silvaroli JA, Ahmed E, Kim JY, Stayton A, Patel PS, Afjal MA, Horton T, Bohmer M, Vanichapol T, Sander V, Andrade GM, Allison CV, Mondal M, Thorson VC, Partey A, Nimkar K, Williams V, Quimby J, Ganesan L, Madhavan SM, Davidson AJ, Peterson BR, Adebiyi A, Rao R, Sweet DH, Singh P, Bennett KM, Zepeda-Orozco D, Mallipattu SK, Eisenmann ED, Sparreboom A, Rovin BH, Bajwa A, Pabla NS. Resazurin dye is an in vivo sensor of kidney tubular function. Kidney Int 2025; 107:508-520. [PMID: 39733791 PMCID: PMC11845305 DOI: 10.1016/j.kint.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 11/01/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024]
Abstract
Glomerular filtration rate (GFR) is the main functional index of kidney health and disease. Currently, no methods are available to directly measure tubular mass and function. Here, we report a serendipitous finding that the in vitro cell viability dye resazurin can be used in mice as an exogenous sensor of tubular function. Intravenously injected resazurin exhibited significant plasma protein binding and was found to mainly undergo tubular secretion. Mechanistic studies showed that the blue-colored, weakly fluorescent resazurin is taken up by tubular cells through organic anion transporters, followed by conversion to a highly fluorescent, pink-colored resorufin by mitochondrial and cytosolic reductases, converted to an orange-colored β-d-glucuronide with subsequent efflux into the urine. Here we report a simple method in which the intravenous injection of resazurin is followed by the measurement of fluorescent metabolites in the urine, providing a sensitive readout of tubular function. Three mouse models of acute kidney injury (rhabdomyolysis, bilateral ischemia-reperfusion injury, and cisplatin nephrotoxicity) were tested and the resazurin-based method was able to sensitively detect the loss of tubular function much earlier than the increase in serum creatinine levels. Strikingly, in mice with unilateral ischemia-reperfusion injury and genetic mutation-linked kidney hypoplasia (oligosyndactylism, a genetic model for congenital kidney hypoplasia), the resazurin-based method was able to detect loss of tubular mass and function despite normal GFR levels. Collectively, our findings establish the preclinical utility of resazurin as a sensitive exogenous marker of tubular function and support future examination in larger animals for potential clinical translation.
Collapse
Affiliation(s)
- Shirely Acosta Martinez
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Isaac Z Karel
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Josie A Silvaroli
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Eman Ahmed
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Amanda Stayton
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Prisha S Patel
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Mohammad Amir Afjal
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Thomas Horton
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Margaret Bohmer
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Thitinee Vanichapol
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Veronika Sander
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Gabriel Mayoral Andrade
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Corynne Vermillion Allison
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Milon Mondal
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Victoria C Thorson
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Alexandra Partey
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Kartik Nimkar
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Victoria Williams
- Division of Clinical Pharmacy Practice and Science, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Jessica Quimby
- Department of Veterinary Clinical Sciences, The Ohio State University Veterinary Medical Center, Columbus, Ohio, USA
| | - Latha Ganesan
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sethu M Madhavan
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Blake R Peterson
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Adebowale Adebiyi
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Reena Rao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA; Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Douglas H Sweet
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Prabhleen Singh
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, California, USA
| | - Kevin M Bennett
- Washington University in St. Louis, Mallinckrodt Institute of Radiology, St. Louis, Missouri, USA
| | - Diana Zepeda-Orozco
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sandeep K Mallipattu
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York, USA; Renal Section, Northport VA Medical Center, Northport, New York, USA
| | - Eric D Eisenmann
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Brad H Rovin
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Amandeep Bajwa
- Transplant Research Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| | - Navjot S Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
31
|
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
|
32
|
Tao S, Qi Y, Gao J, Yuan H, Wang R, Shen X, Wei G, Peng Z. Ameliorative Effect of Sipunculus nudus Hydrolysate on Cisplatin-Induced Nephrotoxicity by Mitigating Oxidative Stress, Inflammation and Apoptosis. Mar Drugs 2025; 23:100. [PMID: 40137286 PMCID: PMC11943811 DOI: 10.3390/md23030100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 03/27/2025] Open
Abstract
The present study investigated the protective effects and possible mechanisms of an ultrafiltration fraction of Sipunculus nudus hydrolysate (UFSH) on cisplatin-induced kidney damage in a mouse model. The results showed that UFSH significantly attenuated cisplatin-induced nephrotoxicity by inhibiting increases in blood urea nitrogen (BUN) and serum creatinine (SCr). Additionally, UFSH treatment significantly alleviated cisplatin-induced renal histopathological changes, such as significant dilation of renal tubules, cast formation, and tubular cell necrosis, as well as tubulointerstitial fibrosis. Moreover, UFSH decreased cisplatin-induced oxidative stress by increasing the activities of antioxidant enzymes SOD and GSH-Px, while reducing the malondialdehyde (MDA) level in the kidney. Furthermore, UFSH significantly inhibited cisplatin-induced increases in inflammatory cytokines, including Interleukin 1-beta (IL-1β), Interleukin-6 (IL-6), and Tumor necrosis factor-alpha (TNF-α). Western blotting revealed that UFSH inhibited the phosphorylation of the inflammation-associated MAPK/NF-κB signaling pathway, lowered the expression of the apoptosis-related protein Bax, and reversed the reduction in the anti-apoptotic Bcl-2 protein. This investigation demonstrated that UFSH can ameliorate cisplatin-induced nephrotoxicity by mitigating oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Susu Tao
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
| | - Yi Qi
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
| | - Jialong Gao
- College of Food Science & Technology, Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Zhanjiang 524088, China;
| | - Huafang Yuan
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Ruimin Wang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Xiaoqin Shen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Gang Wei
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| | - Zhilan Peng
- School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China; (S.T.); (Y.Q.); (G.W.)
- College of Food Science & Technology, Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Zhanjiang 524088, China;
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China; (H.Y.); (R.W.); (X.S.)
| |
Collapse
|
33
|
Akakpo JY, Abbott E, Woolbright BL, Ramachandran A, Rick SG, Wallace DP, Taylor JA. 4-Methylpyrazole-mediated inhibition of Cytochrome P450 2E1 protects renal epithelial cells, but not bladder cancer cells, from cisplatin toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.10.622845. [PMID: 39605334 PMCID: PMC11601237 DOI: 10.1101/2024.11.10.622845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Cisplatin is an effective chemotherapeutic drug for the treatment of bladder cancer, though cisplatin-induced nephrotoxicity (CIN) occurs in approximately 20-30% of patients, limiting its clinical use. Evidence has shown that cytochrome P450 2E1 (CYP2E1), a drug metabolism enzyme expressed in proximal tubules, mediates the production of reactive oxygen species (ROS) during cisplatin-induced injury. Previously, we showed that the repurposed drug 4-methylpyrazole (4MP; fomepizole) blocks CYP2E1 activity and prevents acetaminophen-induced liver injury. Here, we investigated the potential protective effects of 4MP against CIN. Male and female C57BL/6J mice were treated with a single 20 mg/kg dose of cisplatin for 3 days (acute) or 9 mg/kg/week for 4 weeks (repeated dosing regimen) with or without a co-treatment of 50 mg/kg 4MP. Our findings revealed that acute treatment with cisplatin induced severe histological tubular damage and elevated plasma BUN and creatinine levels in male mice, but not in female mice. This difference correlated with higher basal CYP2E1 expression in the kidneys of male mice compared to female mice. We also found that cisplatin increased renal CYP2E1 activity and that inhibition of CYP2E1 with 4MP significantly reduced cisplatin induced cell death in male mice and primary normal human kidney cells. By contrast, human bladder cancer cells do not express CYP2E1, and treatment with 4MP did not interfere with cisplatin anti-cancer effects in human bladder cancer HTB9 cells. This study highlights the critical role of CYP2E1 in CIN and suggests that its inhibition with 4MP in the kidney is a potential prophylactic therapeutic option to prevent CIN in bladder cancer patients without affecting its anti-neoplastic effect.
Collapse
|
34
|
Zahedi K, Barone S, Soleimani M. Evaluation of platinum drug toxicity resulting from polyamine catabolism. Methods Enzymol 2025; 715:93-116. [PMID: 40382156 DOI: 10.1016/bs.mie.2025.01.065] [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: 05/20/2025]
Abstract
Polyamines, spermidine (Spd) and Spermine (Spm), are polycations that serve a number of important biological functions. The tissue contents of polyamines are tightly regulated through their cellular import and export, as well as their metabolism (anabolism and catabolism). Polyamine catabolism in mediated via the spermidine/spermine N1-acetyltransferase (SAT1)/acetylpolyamine oxidase (APOX) cascade and oxidation of Spm by spermine oxidase (SMOX). The expression of SAT1 and SMOX increases in injured organs in response to trauma, ischemia/reperfusion, sepsis, and exposure to toxic compounds. Cisplatin is a highly effective chemotherapeutic agent that is used for the treatment of a variety of solid tumors. Its anti-tumor activity is mediated via its ability to form stable DNA adducts that inhibit the growth of actively proliferating cells. However, cisplatin also can lead to severe off-target deleterious effects (e.g., nephrotoxicity and ototoxicity), and because of such adverse effects the use of cisplatin has to be discontinued in many patients. Understanding and decoupling the therapeutic and toxic effects of cisplatin will lead to more effective use of this and other platinum-derived compounds in the treatment of cancer patients. Acute and chronic exposure to cisplatin in mice leads to severe renal tubular injuries and an increase in the expression of SAT1 and SMOX while the ablation of their genes in mice reduces the severity of nephrotoxic injuries caused by cisplatin. Furthermore, neutralization of the toxic by-products of polyamine degradation reduce the severity if cisplatin nephrotoxicity. These observations suggest that interventions targeting the adverse effects of enhanced polyamine catabolism may provide effective therapies by reducing the toxic effects of cisplatin without affecting its anti-neoplastic activity.
Collapse
Affiliation(s)
- Kamyar Zahedi
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center Albuquerque, NM, United States; Research Services, New Mexico Veterans Health Care System, Albuquerque, NM, United States.
| | - Sharon Barone
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center Albuquerque, NM, United States; Research Services, New Mexico Veterans Health Care System, Albuquerque, NM, United States
| | - Manoocher Soleimani
- Division of Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center Albuquerque, NM, United States; Research Services, New Mexico Veterans Health Care System, Albuquerque, NM, United States
| |
Collapse
|
35
|
Patel P, Patel S, Patel Y, Chudasama P, Soni S, Patel S, Raval M. Roflumilast mitigates cisplatin-induced nephrotoxicity by regulating TNF-α/TNFR1/TNFR2/Fas/Caspase mediated apoptosis and inflammatory signals. J Pharm Pharmacol 2025; 77:308-320. [PMID: 39566023 DOI: 10.1093/jpp/rgae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/23/2024] [Indexed: 11/22/2024]
Abstract
PURPOSE The study aimed to evaluate the effect of roflumilast on modulating TNF-α/Caspase mediated cellular signals in cisplatin-induced nephrotoxicity in rats. METHODS The rats (Male Wistar) were divided into five groups: normal control, disease control (cisplatin: 7 mg/kg i.p.), and cisplatin + roflumilast (0.25, 0.5, and 1 mg/kg b.w., p.o.). Cisplatin was administrated to rats on 0 day, and roflumilast treatment was started from the 6th-15th days. Blood and tissue were collected. Tissue was used to measure oxidative stress, such as malondialdehyde, superoxide dismutase, and catalase. Gene expression study involved real-time PCR of key genes linked with inflammation and apoptosis, i.e. Tnf-α, Tnfr1, Tnfr2, Fas, Nfkb, Casp3, Casp8, and Nrf2. FINDINGS Cisplatin showed decreased serum creatinine and urea, high albumin, and total protein. Cisplatin elevated the malondialdehyde and reduced superoxide dismutase and catalase activity. Cisplatin also attributed an overexpression of Tnf-α, Tnfr1, Tnfr2, Nfkb, Fas, Casp3, and Casp8, and a decrease in the Nrf2 gene. Roflumilast decreased creatinine and urea and increased albumin and total protein levels. Roflumilast also downregulated the expression of Tnf-α, Tnfr1, Tnfr2, Nfkb, Fas, Casp3, and Casp8 and upregulated the Nrf2 gene expression. CONCLUSION Roflumilast manifested as a potential reno-protective agent against cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Priyal Patel
- Department of Pharmacology, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Anand, Gujarat 388421, India
| | - Sandip Patel
- Department of Pharmacology, L.M. College of Pharmacy, Ahmedabad, Gujarat 380009, India
| | - Yash Patel
- Department of Clinical Pharmacy, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Anand, Gujarat 388421, India
| | - Piyush Chudasama
- Department of Research and Development, Sat-Kaival Hospital Pvt. Ltd, Anand, Gujarat 388001, India
| | - Shailesh Soni
- Department of Pathology, Muljibhai Patel Urological Hospital, Nadiad, Gujarat 387002, India
| | - Samir Patel
- Department of Pharmaceutical Chemistry and Analysis, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Anand, Gujarat 388421, India
| | - Manan Raval
- Department of Pharmacognosy and Phytochemistry, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Anand, Gujarat 388421, India
| |
Collapse
|
36
|
Teixeira AR, Mata D, Ferreira H, Paiva A, Pelayo MJ, Rafael C, Maurício J, Calisto R, Cassiano Neves M. Short Hydration Regimen in Cisplatin-Based Chemotherapy and Its Impact on Nephrotoxicity: A Unicentric Prospective Study. Cureus 2025; 17:e79774. [PMID: 40161090 PMCID: PMC11954581 DOI: 10.7759/cureus.79774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Nephrotoxicity is common when cisplatin is used and hydration during treatment is nephroprotective, but the optimal volume, composition, and duration are unknown. We reviewed our institutional intravenous hydration regimen (total 2500 mL in 5 hours versus previously used 3000 mL in 7 hours) in cisplatin >50 mg/m2 schemes and we aimed to confirm the safety of this change by prospectively evaluating the incidence of acute kidney injury (AKI). We included 105 patients in our cohort. However, for comparison, we only considered 96 cases and 191 controls (ratio 2:1), due to the impossibility of matching all cases. The proportion of patients without Kidney Disease Improving Global Outcomes (KDIGO) AKI stage ≥2 (serum creatinine ≥2.0-2.9 times baseline) was 100.0% (n=96) after the first treatment and 97.8% (n=88) after the second treatment in our cohort. There was no difference when compared with the historical cohort of patients who received the previous cisplatin hydration regimen (p=1 for the first treatment and p=0.92 for the second treatment). AKI was the reason for cisplatin discontinuation in three patients (2.9%). Our results support the safety of a shorter and lower volume intravenous hydration during cisplatin treatment, which is more comfortable for patients and allows a better health resource allocation.
Collapse
Affiliation(s)
- Ana Raquel Teixeira
- Medical Oncology, Instituto Português Oncologia do Porto Francisco Gentil, Porto, PRT
| | - Diana Mata
- Medical Oncology, Instituto Português Oncologia do Porto Francisco Gentil, Porto, PRT
| | - Hugo Ferreira
- Nephrology, Instituto Português Oncologia do Porto Francisco Gentil, Porto, PRT
| | - Ana Paiva
- Nephrology, Instituto Português Oncologia do Porto Francisco Gentil, Porto, PRT
| | - Maria J Pelayo
- Pharmaceutics, Instituto Português Oncologia do Porto Francisco Gentil, Porto, PRT
| | - Carla Rafael
- Nursing, Instituto Português Oncologia do Porto Francisco Gentil, Porto, PRT
| | - Joaquina Maurício
- Medical Oncology, Instituto Português Oncologia do Porto Francisco Gentil, Porto, PRT
| | - Rita Calisto
- Epidemiology, Instituto Português Oncologia do Porto Francisco Gentil, Porto, PRT
| | - Maria Cassiano Neves
- Medical Oncology, Instituto Português Oncologia do Porto Francisco Gentil, Porto, PRT
| |
Collapse
|
37
|
Bian Y, Dong J, Zhou Z, Zhou H, Xu Y, Zhang Q, Chen C, Pi J. The spatiotemporal and paradoxical roles of NRF2 in renal toxicity and kidney diseases. Redox Biol 2025; 79:103476. [PMID: 39724848 PMCID: PMC11732127 DOI: 10.1016/j.redox.2024.103476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Over 10% of the global population is at risk to kidney disorders. Nuclear factor erythroid-derived 2-related factor 2 (NRF2), a pivotal regulator of redox homeostasis, orchestrates antioxidant response that effectively counters oxidative stress and inflammatory response in a variety of acute pathophysiological conditions, including acute kidney injury (AKI) and early stage of renal toxicity. However, if persistently activated, NRF2-induced transcriptional cascade may disrupt normal cell signaling and contribute to numerous chronic pathogenic processes such as fibrosis. In this concise review, we assembled experimental evidence to reveal the cell- and pathophysiological condition-specific roles of NRF2 in renal chemical toxicity, AKI, and chronic kidney disease (CKD), all of which are closely associated with oxidative stress and inflammation. By incorporating pertinent research findings on NRF2 activators, we dissected the spatiotemporal roles of NRF2 in distinct nephrotoxic settings and kidney diseases. Herein, NRF2 exhibits diverse expression patterns and downstream gene profiles across distinct kidney regions and cell types, and during specific phases of nephropathic progression. These changes are directly or indirectly connected to altered antioxidant defense, damage repair, inflammatory response, regulated cell death and fibrogenesis, culminating ultimately in either protective or deleterious outcomes. The spatiotemporal and paradoxical characteristics of NRF2 in mitigating nephrotoxicity suggest that translational application of NRF2 activation strategy for prevention and interventions of kidney injury are unlikely to be straightforward - right timing and spatial precision must be taken into consideration.
Collapse
Affiliation(s)
- Yiying Bian
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China.
| | - Jize Dong
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Zhengsheng Zhou
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China
| | - Hua Zhou
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), China; Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, China
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, GA, 30322, USA
| | - Chengjie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China.
| | - Jingbo Pi
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic (China Medical University), China; Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
38
|
Li Z, Mao C, Zhao Y, Zhao Y, Yi H, Liu J, Liang J. The STING antagonist SN-011 ameliorates cisplatin induced acute kidney injury via suppression of STING/NF-κB-mediated inflammation. Int Immunopharmacol 2025; 146:113876. [PMID: 39709905 DOI: 10.1016/j.intimp.2024.113876] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/17/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
Acute kidney injury (AKI) is a critical clinical syndrome associated with both innate and adaptive immune responses and thus increases mortality. Nevertheless, specific therapeutics for AKI are scarce so far. Recent studies have revealed that knockout of STING alleviate AKI, suggesting that STING could be an attractive target for AKI therapy. SN-011, a promising STING inhibitor, has not been reported in studies of its anti-AKI activity. In this study, we sought to examine the effects of SN-011 on AKI and explore its underlying mechanism. Our findings indicate that SN-011 could modulate the NF-κB and MAPK pathways, suppress the expression of inflammatory factors, and decrease ROS release in the cisplatin-induced cell model. In addition, SN-011 blocked the nuclear translocation of NF-κB p65, further mitigating the inflammatory response. In vivo, SN-011 enhanced survival rates and alleviated renal dysfunction. According to gene set enrichment analysis of sequencing data from mouse kidneys, we further confirm that SN-011 modulates the NF-κB and MAPK pathways. Our study suggests that SN-011 could be an attractive anti-inflammatory agent for further anti-AKI research.
Collapse
Affiliation(s)
- Ziyang Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Can Mao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yixin Zhao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yanbin Zhao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Hanyu Yi
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Jin Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China.
| | - Jinqiang Liang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China.
| |
Collapse
|
39
|
Mora-Lagos B, Reyes ME, Lobos-Gonzalez L, Del Campo M, Buchegger K, Zanella L, Riquelme I, Ili CG, Brebi P. Maraviroc/cisplatin combination inhibits gastric cancer tumoroid growth and improves mice survival. Biol Res 2025; 58:4. [PMID: 39827154 PMCID: PMC11748569 DOI: 10.1186/s40659-024-00581-3] [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: 05/09/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a significant cancer-related cause of death worldwide. GC's most used chemotherapeutic regimen is based on platinum drugs such as cisplatin (CDDP). However, CDDP chemoresistance reduces the survival rate of advanced GC. The immune C-C chemokine receptor type 5 (CCR5) have been proposed as a pivotal factor in cancer progression since its blockade has been linked with antineoplastic effects on tumor cell proliferation; nevertheless, its role in the chemoresistance of GC has not been elucidated. This study aimed to determine the effects induced by the CCR5 using Maraviroc (MVC), a highly selective CCR5 antagonist, on CDDP-resistant AGS cells (AGS R-CDDP), tumoroids (3D tumor spheroids), and animal models. RESULTS The combined CDDP and MVC treatment reduced cell viability and inhibited tumoroid formation in AGS R-CDDP cells. The effects of the MVC/CDDP combination on apoptosis and cell cycle progression were correlated with the increase in CDDP (dose-dependent). The mRNA levels of C-C Motif Chemokine Ligand 5 (CCL5), the main ligand for CCR5, decreased significantly in cells treated with the MVC/CDDP combination. MVC in the MVC/CDDP combination improved the survival rate and biochemical parameters of CDDP-treated mice by reducing the side effects of CDDP alone. CONCLUSIONS This finding suggests that MVC/CDDP combination could be a potential complementary therapy for GC.
Collapse
Affiliation(s)
- Bárbara Mora-Lagos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Temuco, Chile
| | - María Elena Reyes
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Temuco, Chile
| | - Lorena Lobos-Gonzalez
- Centro de Medicina Regenerativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
- Advanced Center for Chronic Diseases, ACCDiS, Santiago, Chile
- Laboratorio de comunicaciones celulares, Instituto de Ciencias Biomédicas, iCBM, Universidad de Chile, Santiago, Chile
| | - Matías Del Campo
- Centro de Medicina Regenerativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
- Advanced Center for Chronic Diseases, ACCDiS, Santiago, Chile
| | - Kurt Buchegger
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Biomedical Research Consortium-Chile (BMRC), Santiago, Chile
| | - Louise Zanella
- Doctorado en Ciencias Médicas, Universidad de La Frontera, Temuco, Chile
- Núcleo Milenio de Sociomedicina, Santiago, Chile
| | - Ismael Riquelme
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Temuco, Chile
| | - Carmen Gloria Ili
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Biomedical Research Consortium-Chile (BMRC), Santiago, Chile.
| | - Priscilla Brebi
- Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Biomedical Research Consortium-Chile (BMRC), Santiago, Chile.
| |
Collapse
|
40
|
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
|
41
|
Iskandar M, Xiao Barbero M, Jaber M, Chen R, Gomez-Guevara R, Cruz E, Westerheide S. A Review of Telomere Attrition in Cancer and Aging: Current Molecular Insights and Future Therapeutic Approaches. Cancers (Basel) 2025; 17:257. [PMID: 39858038 PMCID: PMC11764024 DOI: 10.3390/cancers17020257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES As cells divide, telomeres shorten through a phenomenon known as telomere attrition, which leads to unavoidable senescence of cells. Unprotected DNA exponentially increases the odds of mutations, which can evolve into premature aging disorders and tumorigenesis. There has been growing academic and clinical interest in exploring this duality and developing optimal therapeutic strategies to combat telomere attrition in aging and cellular immortality in cancer. The purpose of this review is to provide an updated overview of telomere biology and therapeutic tactics to address aging and cancer. METHODS We used the Rayyan platform to review the PubMed database and examined the ClinicalTrial.gov registry to gain insight into clinical trials and their results. RESULTS Cancer cells activate telomerase or utilize alternative lengthening of telomeres to escape telomere shortening, leading to near immortality. Contrarily, normal cells experience telomeric erosion, contributing to premature aging disorders, such as Werner syndrome and Hutchinson-Gilford Progeria, and (2) aging-related diseases, such as neurodegenerative and cardiovascular diseases. CONCLUSIONS The literature presents several promising therapeutic approaches to potentially balance telomere maintenance in aging and shortening in cancer. This review highlights gaps in knowledge and points to the potential of these optimal interventions in preclinical and clinical studies to inform future research in cancer and aging.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sandy Westerheide
- Department of Molecular Biosciences, University of South Florida, 4202 East Fowler Avenue, ISA2015, Tampa, FL 33620, USA; (M.I.); (M.X.B.); (M.J.); (R.C.); (R.G.-G.); (E.C.)
| |
Collapse
|
42
|
Hao X, Gao Z, Hu M. Anti-tumor role and molecular mechanism of vanillic acid. Discov Oncol 2025; 16:20. [PMID: 39775208 PMCID: PMC11711440 DOI: 10.1007/s12672-025-01746-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Vanillic Acid (VA) is an aromatic acid extracted from traditional Chinese medicine such as Angelica sinensis and Panax ginseng, which has demonstrated potent anti-cancer activity, inhibiting the onset and progression of various malignant tumors. This review highlights the principal mechanism by which VA exerts its anticancer activity, including apoptosis induction, specifically promoting the generation of intracellular reactive oxygen species (ROS), which in turn triggers mitochondrial apoptosis. Furthermore, VA disrupts the cancer cell cycle, arresting most cancer cells at the G1 phase, curtails cell migration, invasion, angiogenesis, and potentiates the therapeutic efficacy of chemotherapeutic drugs, all while minimizing adverse reactions. This paper offers a comprehensive review of VA's anti-tumor effects and underlying mechanisms, aiming to provide some references for scientists and clinical physicians in the research of anti-tumor therapeutic strategies.
Collapse
Affiliation(s)
- Xunxing Hao
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shangdong, China
| | - Zhixiao Gao
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mingzhe Hu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
43
|
Famurewa AC, Prabhune NM, Prabhu S. Natural product mitigation of ferroptosis in platinum-based chemotherapy toxicity: targeting the underpinning oxidative signaling pathways. J Pharm Pharmacol 2025; 77:1-17. [PMID: 39485898 DOI: 10.1093/jpp/rgae132] [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: 04/21/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVES Platinum-based anticancer chemotherapy (PAC) represents a cornerstone in cancer treatment, retaining its status as the gold standard therapy. However, PAC's efficacy is countered by significant toxicities, such as nephrotoxicity, ototoxicity, and neurotoxicity. Recent studies have linked these toxicities to ferroptosis, characterized by iron accumulation, reactive oxygen species generation, and lipid peroxidation. This review explores the mechanisms underlying PAC-induced toxicities, focusing on the involvement of ferroptosis with three major PAC drugs-cisplatin, carboplatin, and oxaliplatin. Further, we provide a comprehensive analysis of the natural product mitigation of PAC-induced ferroptotic toxicity. KEY FINDINGS The mechanistic role of ferroptosis in cisplatin- and oxaliplatin-induced toxicities has been investigated, while studies on carboplatin-induced ferroptotic toxicities are lacking. Natural compounds targeting molecular pathways of ferroptosis have been explored to mitigate PAC-induced ferroptotic toxicity. CONCLUSION While ferroptosis in cisplatin- and oxaliplatin-induced toxicities has been investigated, there remains a notable dearth of studies examining its involvement in carboplatin-induced toxicities. Hence, further exploration is warranted to define the role of ferroptosis in carboplatin-induced toxicities, and its further mitigation. Moreover, in-depth mechanistic evaluation is necessary to establish natural products evaluated against PAC-induced ferroptosis, as PAC adjuvants.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University, Ikwo 482103, Ebonyi State, Nigeria
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Nupura Manish Prabhune
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Sudharshan Prabhu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
44
|
Wang Z, Kregel M, Meijers JL, Franch J, Cuijpers VMJI, Ahlers D, Karst U, Slootweg P, van der Geest IC, Leeuwenburgh SC, van den Beucken JJ. Cisplatin-functionalized dual-functional bone substitute granules for bone defect treatment after bone tumor resection. Acta Biomater 2025; 191:158-176. [PMID: 39551330 DOI: 10.1016/j.actbio.2024.11.020] [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: 08/12/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Invasive bone tumors pose a significant healthcare challenge, often requiring systemic chemotherapy and limb salvage surgery. However, these strategies are hampered by severe side effects, complex post-resection bone defects, and high local recurrence rates. To address this, we developed dual-functional bone substitute biomaterials by functionalizing commercially available bone substitute granules (Bio-Oss® and MBCP®+) with the established anticancer agent cisplatin. Physicochemical characterization revealed that Bio-Oss® granules possess a higher surface area and lower crystallinity compared to MBCP®+ granules, which enhances their capacity for cisplatin adsorption and release. In co-cultures with metastatic breast and prostate cancer cells (MDA-MB-231 and PC3) and bone marrow stromal cells (hBMSCs), cisplatin-functionalized granules and their releasates exhibited dose-dependent cytotoxic effects on cancer cells while having less impact on hBMSCs. Furthermore, investigations on the mechanism of action indicated that cisplatin induced significant cell cycle arrest and apoptosis in MDA-MB-231 and PC3 cells, contrasting with minimal effects on hBMSCs. In a rat femoral condyle defect model, cisplatin-functionalized granules did not evoke adverse effects on bone tissue ingrowth or new bone formation. Importantly, local application of cisplatin-functionalized granules resulted in negligible cisplatin accumulation without signs of apoptotic damage in kidneys and livers. Taken together, we here provide hard evidence that cisplatin-functionalized granules maintain a favorable balance between biosafety, anticancer efficacy, and bone regenerative capacity. Consequently, loading granular bone substitutes with cisplatin holds promise for local treatment of bone defects following bone tumor resections, presenting a safe and potentially more effective alternative to systemic cisplatin administration. STATEMENT OF SIGNIFICANCE: Current treatments in combating malignant bone tumors are hampered by severe side effects, high local tumor recurrence, and complex bone defects after surgery. This study explores a facile manufacturing method to render two types of commercially available bone substitute granules (Bio-Oss® and MBCP®+) suitable for local delivery of cisplatin. The use of cisplatin-functionalized granules has shown promising results both in killing cancer cells in a dose-dependent manner and in aiding bone regeneration. Importantly, this local treatment strategy avoids the systemic toxicity associated with traditional chemotherapy to excretory organs. This dual-functional strategy represents a significant advancement in bone cancer treatment, offering a safe and more efficient alternative that could improve outcomes for patients following bone tumor resection.
Collapse
Affiliation(s)
- Zhule Wang
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands
| | - Mark Kregel
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - Jean-Luc Meijers
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - Jordi Franch
- Department of Small Animal Medicine and Surgery, Veterinary School, Universitat Autonoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Vincent M J I Cuijpers
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - David Ahlers
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Piet Slootweg
- Department of Pathology, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, the Netherlands
| | - Ingrid Cm van der Geest
- Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands; Department of Orthopedics, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, the Netherlands
| | - Sander Cg Leeuwenburgh
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands
| | - Jeroen Jjp van den Beucken
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands.
| |
Collapse
|
45
|
Chaturvedi A, Sehrawat A, Mopidevi TR, Parthasarthy KM, Gupta D, Singh A. Comparison of the Effect of Weekly Cisplatin Versus Three Weekly Cisplatin in Concurrent Chemoradiotherapy of Head and Neck Cancer: A Pilot Study. Indian J Otolaryngol Head Neck Surg 2025; 77:356-370. [PMID: 40070996 PMCID: PMC11890838 DOI: 10.1007/s12070-024-05190-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/31/2024] [Indexed: 03/14/2025] Open
Abstract
Patients with squamous cell carcinoma of the head and neck (HNSCC) often present with advanced locoregional disease. Combined modality treatment with definitive concurrent chemoradiotherapy (CTRT) has become the standard of care for these cancers. We compared the efficacy and toxicity profile of weekly cisplatin regime compared to a three-weekly cisplatin regime in the setting of definitive CTRT for locally advanced HNSCC (LAHNSCC). 51 patients were enrolled in the study between May 2016 and June 2019. In arm A, 25 patients (n = 25) were given three weekly cisplatin at dose of 100 mg/m2 on days 1, 22 and 43 (D1, D22, D43) concurrent with radiotherapy. In arm B, 26 patients (n = 26) were given weekly cisplatin at dose of 35 mg/m2. Primary end points of the study were locoregional control (LRC), distant metastasis free survival (DMFS) and disease free survival (DFS). Adverse effects / toxicity was the secondary end point. Out of the 51 total patients, 49 patients underwent response evaluation at 3-month follow up, after completion of definitive CTRT. Two patients expired in the 3 weekly cisplatin arm before response assessment. On MRI, total 29 patients had complete clinicoradiologic response (cCR), 14 in weekly cisplatin arm and 15 in three weekly cisplatin arm. Only these 29 patients showing cCR were eligible for evaluation of LRC, DMFS and DFS at 2 years. Median follow up period was 20 months. At the time analysis 1 patient in 3 weekly arm and 2 patients in weekly arm expired. All of them had disease relapse during follow up. After 2 years of follow up, overall 89.7% (n = 26) patients had LRC. 92.86% (n = 13) in 3 weekly arm and 86.15% (n = 13) in weekly arm had LRC after 2 years. DMFS at 2 years was 86.15% (n = 13) in 3 weekly arm in comparison to 78.57% (n = 11) in weekly arm. DFS at 2 years was 77.92% (n = 11) in 3 weekly arm versus 66.67% (n = 10) in weekly arm. LAHNSCC patients with ECOG PS of 0-2, should be offered 3 weekly cisplatin regime in the setting of definitive CTRT as preferred regime in comparison to weekly cisplatin in view of trend favoring better response in terms of LRC, DMFS and DFS. Acute toxicities are more common in 3 weekly cisplatin arm as compared to weekly cisplatin arm but most of the toxicities are grade 1 or 2 which can be easily managed. This is a pilot study and randomized trials are needed to confirm the findings.
Collapse
Affiliation(s)
| | - Amit Sehrawat
- All India Institute of Medical Sciences, Rishikesh, Uttarakhand India
| | | | | | - Deni Gupta
- Dharamshila Narayana Superspeciality Hospital, New Delhi, India
| | - Anuj Singh
- National Institute of Medical Science and Research, Jaipur, Rajasthan India
| |
Collapse
|
46
|
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
|
47
|
Saberian E, Jenčová J, Jenča A, Jenča A, Petrášová A, Jenča J, akbarzadehkhayavi A. Combination Therapy of Curcumin and Cisplatin Encapsulated in Niosome Nanoparticles for Enhanced Oral Cancer Treatment. Indian J Clin Biochem 2025; 40:59-66. [PMID: 39835233 PMCID: PMC11741963 DOI: 10.1007/s12291-024-01279-9] [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: 09/25/2024] [Accepted: 10/21/2024] [Indexed: 01/22/2025]
Abstract
Oral cavity cancer poses a significant health threat due to its aggressive nature and limited responsiveness to traditional therapies like chemotherapy and radiation, highlighting the need for more effective treatment options. To address this, researchers have explored a novel approach using niosome nanoparticles to co-encapsulate curcumin (CUR) and cisplatin (Cis), to enhance therapeutic efficacy. While CUR has anti-cancer properties, its poor bioavailability limits its effectiveness. Cis, on the other hand, is hindered by severe side effects and resistance. A dual-drug delivery system that encapsulates both CUR and Cis in niosome nanoparticles seeks to leverage the synergistic effects of these agents to improve treatment outcomes. The study synthesized Cis and CUR co-loaded nanoparticles (Cis/CUR-NPs) using reverse microemulsion and film dispersion methods, resulting in nanoparticles with an average size of 220.9 nm and a consistent size distribution. In vitro experiments demonstrated that the nanosized Cis/CUR-NPs could release both Cis and CUR, achieving a synergistic effect on OECM-1 cells at an optimal ratio (1:6) of the two drugs. Overall, the findings suggest that Cis/CUR-NPs offer a promising and effective strategy for leveraging the synergistic effects of Cis and CUR in treating oral cancer.
Collapse
Affiliation(s)
- Elham Saberian
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | - Janka Jenčová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | - Adriána Petrášová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | - Jozef Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice, Pavol Jozef Šafárik University, Kosice, Slovakia
| | | |
Collapse
|
48
|
Li N, Shi J, Chen Z, Dong Z, Ma S, Li Y, Huang X, Li X. In silico prediction of drug-induced nephrotoxicity: current progress and pitfalls. Expert Opin Drug Metab Toxicol 2025; 21:203-215. [PMID: 39360665 DOI: 10.1080/17425255.2024.2412629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 09/05/2024] [Accepted: 10/01/2024] [Indexed: 10/04/2024]
Abstract
INTRODUCTION Due to its role in absorption and metabolism, the kidney is an important target for drug toxicity. Drug-induced nephrotoxicity (DIN) presents a significant challenge in clinical practice and drug development. Conventional methods for assessing nephrotoxicity have limitations, highlighting the need for innovative approaches. In recent years, in silico methods have emerged as promising tools for predicting DIN. AREAS COVERED A literature search was performed using PubMed and Web of Science, from 2013 to February 2023 for this review. This review provides an overview of the current progress and pitfalls in the in silico prediction of DIN, which discusses the principles and methodologies of computational models. EXPERT OPINION Despite significant advancements, this review identified issues accentuates the pivotal imperatives of data fidelity, model optimization, interdisciplinary collaboration, and mechanistic comprehension in sculpting the vista of DIN prediction. Integration of multiple data sources and collaboration between disciplines are essential for improving predictive models. Ultimately, a holistic approach combining computational, experimental, and clinical methods will enhance our understanding and management of DIN.
Collapse
Affiliation(s)
- Na Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Juan Shi
- Department of Clinical Pharmacy, The First People's Hospital of Jinan, Jinan, China
| | - Zhaoyang Chen
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Zhonghua Dong
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Shiyu Ma
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Xin Huang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Xiao Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| |
Collapse
|
49
|
Yoshioka H, Yokota S, Torimoto S, Horita H, Tsukiboshi Y, Maeda T, Miura N. Cry2 Alleviates Cisplatin-Induced Cytotoxicity in Mouse Renal Cortex Tubular Cell Lines. Biol Pharm Bull 2025; 48:390-398. [PMID: 40254427 DOI: 10.1248/bpb.b24-00811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Cisplatin is a platinum-based drug that is widely used to treat various types of cancer. However, cisplatin is known to cause severe adverse effects, such as nephrotoxicity and ototoxicity. Clock genes, such as Bmal1 and Clock, regulate cisplatin-related homeostasis genes, such as Oct2 and Mate1. Although these clock genes may be involved in cisplatin-induced nephrotoxicity, their associations with other clock genes remain unclear. The aim of the present study was to investigate whether seven clock genes (Ciart, cryptochrome 1 (Cry1), Cry2, Npas2, Per1, Per2, and Per3) regulate cisplatin-induced renal toxicity in a renal cortex tubule cell line (MuRTE61). Cisplatin treatment decreases MuRTE61 cell viability in a dose-dependent manner. Cry2 expression levels increased after treatment with cisplatin for 24 h. Notably, Cry2 overexpression alleviated cisplatin-induced suppression of cell proliferation, apoptosis, and platinum content in MuRTE61 cells. Moreover, Cry2 overexpression upregulated the efflux-related transporters (Atp7a and Mrp2). These results suggest that Cry2 protects against cisplatin toxicity by reducing Pt accumulation and increasing the expression of Atp7a and Mrp2.
Collapse
Affiliation(s)
- Hiroki Yoshioka
- Department of Pharmacy, Gifu University of Medical Science, 4-3-3 Nijigaoka, Kani, Gifu 509-0293, Japan
- Department of Hygiene, Kitasato University School of Medicine, 1-15-1 Kitazato, Minami-ku, Sagamihara 252-0374, Japan
| | - Satoshi Yokota
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences, 3-25-26 Tono-machi, Kawasaki-ku, Kawasaki 210-9501, Japan
| | - Shintaro Torimoto
- Department of Pharmacy, Gifu University of Medical Science, 4-3-3 Nijigaoka, Kani, Gifu 509-0293, Japan
| | - Hanane Horita
- Department of Pharmacy, Gifu University of Medical Science, 4-3-3 Nijigaoka, Kani, Gifu 509-0293, Japan
| | - Yosuke Tsukiboshi
- Department of Pharmacy, Gifu University of Medical Science, 4-3-3 Nijigaoka, Kani, Gifu 509-0293, Japan
| | - Tohru Maeda
- College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyama-ku, Nagoya 463-8521, Japan
| | - Nobuhiko Miura
- Department of Health Science, Yokohama University of Pharmacy, 601 Matano-cho, Totsuka-ku, Yokohama 245-2006, Japan
| |
Collapse
|
50
|
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
|