Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.730
Filter
1.
Physiol Res ; 73(3): 405-413, 2024 07 17.
Article in English | MEDLINE | ID: mdl-39027957

ABSTRACT

Cisplatin is a widely used chemotherapy drug for the treatment of various cancers. However, although cisplatin is effective in targeting cancer cells, it has severe side effects including skeletal muscle atrophy. In this study, we aimed to characterize the role of Dihydromyricetin in cisplatin-induced muscle atrophy in mice. 5-week-old male C57BL/6 mice were treated with Dihydromyricetin for 14 days orally followed by in intraperitoneally cisplatin administration for 6 days. Gastrocnemius muscles were isolated for the following experiments. Antioxidative stress were determined by peroxidative product malondialdehyde (MDA) and antioxidants superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities. Quadriceps muscle mass and grip strength were significantly restored by Dihydromyricetin in a dose-dependent manner. Moreover, muscle fibers were improved in Dihydromyricetin treated group. Excessive skeletal muscle E3 ubiquitin-protein ligases in cisplatin group were significantly repressed by Dihydromyricetin treatment. Dihydromyricetin significantly reduced oxidative stress induced by cisplatin by decreasing MDA level and restored SOD and GPx activities. In addition, ferroptosis was significantly reduced by Dihydromyricetin characterized by reduced iron level and ferritin heavy chain 1 and improved Gpx4 level. The present study demonstrated that Dihydromyricetin attenuated cisplatin-induced muscle atrophy by reducing skeletal muscle E3 ubiquitin-protein ligases, oxidative stress, and ferroptosis.


Subject(s)
Cisplatin , Ferroptosis , Flavonols , Mice, Inbred C57BL , Muscular Atrophy , Oxidative Stress , Animals , Male , Flavonols/pharmacology , Flavonols/therapeutic use , Muscular Atrophy/chemically induced , Muscular Atrophy/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/prevention & control , Muscular Atrophy/drug therapy , Ferroptosis/drug effects , Cisplatin/toxicity , Mice , Oxidative Stress/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Antineoplastic Agents/toxicity , Antioxidants/pharmacology
2.
Ren Fail ; 46(2): 2378212, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39011587

ABSTRACT

PURPOSE: The present study investigated the nephron-testicular protective effects of sesamin against cisplatin (CP)-induced acute renal and testicular injuries. METHODS: Thirty-two male Wistar rats were allocated to receive carboxymethylcellulose (0.5%, as sesamin vehicle), CP (a single i.p. 5 mg/kg dose), CP plus sesamin at 10 or 20 mg/kg orally for 10 days. RESULTS: Data analysis showed significant increases in serum urea, creatinine, interleukin (IL)-1, IL-6, and tumor necrosis factor-α (TNF-α), as well as renal and testicular tissue malondialdehyde and nitric-oxide concentrations in CP-intoxicated rats in comparison to control animals. On the contrary, rats treated with CP only exhibited significantly lower (p < .05) serum testosterone, tissue glutathione, and activities of endogenous antioxidant enzymes compared to control rats. Histopathologically examining CP-intoxicated rats' tissues using H&E and PAS stains showed atrophied glomeruli, interstitial inflammatory cells, atypic tubular epithelium with focal apoptosis, and reduced mucopolysaccharide content. Further, immunohistochemical staining of the same group revealed an increase in p53 and cyclooxygenase-II (Cox-II) expression in renal and testicular tissues. Treatment with sesamin alleviated almost all the changes mentioned above in a dose-dependent manner, with the 20 mg/kg dose restoring several parameters' concentrations to normal ranges. CONCLUSIONS: In brief, sesamin could protect the kidneys and testes against CP toxicity through its antioxidant, anti-inflammatory, and anti-apoptotic effects.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Apoptosis , Cisplatin , Dioxoles , Kidney , Lignans , Rats, Wistar , Testis , Animals , Male , Lignans/pharmacology , Lignans/therapeutic use , Cisplatin/toxicity , Cisplatin/adverse effects , Rats , Dioxoles/pharmacology , Antioxidants/pharmacology , Testis/drug effects , Testis/pathology , Testis/metabolism , Apoptosis/drug effects , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Anti-Inflammatory Agents/pharmacology , Oxidative Stress/drug effects , Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Antineoplastic Agents/toxicity
3.
Drug Chem Toxicol ; 47(4): 404-415, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38949608

ABSTRACT

Although the presence of nitro groups in chemicals can be recognized as structural alerts for mutagenicity and carcinogenicity, nitroaromatic compounds have attracted considerable interest as a class of agents that can serve as source of potential new anticancer agents. In the present study, the in vitro cytotoxicity, genotoxicity, and mutagenicity of three synthetic ortho-nitrobenzyl derivatives (named ON-1, ON-2 and ON-3) were evaluated by employing human breast and ovarian cancer cell lines. A series of biological assays was carried out with and without metabolic activation. Complementarily, computational predictions of the pharmacokinetic properties and druglikeness of the compounds were performed in the Swiss ADME platform. The MTT assay showed that the compounds selectively affected selectively the cell viability of cancer cells in comparison with a nontumoral cell line. Additionally, the metabolic activation enhanced cytotoxicity, and the compounds affected cell survival, as demonstrated by the clonogenic assay. The comet assay, the cytokinesis-block micronucleus assay, and the immunofluorescence of the γ-H2AX foci formation assay have that the compounds caused chromosomal damage to the cancer cells, with and without metabolic activation. The results obtained in the present study showed that the compounds assessed were genotoxic and mutagenic, inducing double-strand breaks in the DNA structure. The high selectivity indices observed for the compounds ON-2 and ON-3, especially after metabolic activation with the S9 fraction, must be highlighted. These experimental biological results, as well as the theoretical properties predicted for the compounds have shown that they are promising anticancer candidates to be exploited in additional studies.


Subject(s)
Activation, Metabolic , Antineoplastic Agents , Cell Survival , DNA Damage , Humans , Cell Survival/drug effects , Antineoplastic Agents/toxicity , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , DNA Damage/drug effects , Cell Line, Tumor , Micronucleus Tests , Mutagens/toxicity , Comet Assay , Mutagenicity Tests , Female , Nitrobenzenes/toxicity , Nitrobenzenes/chemistry , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Dose-Response Relationship, Drug
4.
Sci Adv ; 10(30): eadk9878, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39047106

ABSTRACT

Cisplatin is a widely used anticancer drug with notable side effects including ototoxicity and nephrotoxicity. Macrophages, the major resident immune cells in the cochlea and kidney, are important drivers of both inflammatory and tissue repair responses. To investigate the roles of macrophages in cisplatin-induced toxicities, we used PLX3397, a U.S. Food and Drug Administration-approved inhibitor of the colony-stimulating factor 1 receptor, to eliminate tissue-resident macrophages. Mice treated with cisplatin alone had considerable hearing loss (ototoxicity) and kidney injury (nephrotoxicity). Macrophage ablation resulted in significantly reduced hearing loss and had greater outer hair cell survival. Macrophage ablation also protected against cisplatin-induced nephrotoxicity, as evidenced by markedly reduced tubular injury and fibrosis. Mechanistically, our data suggest that the protective effect of macrophage ablation against cisplatin-induced ototoxicity and nephrotoxicity is mediated by reduced platinum accumulation in both the inner ear and the kidney. Together, our data indicate that ablation of tissue-resident macrophages represents an important strategy for mitigating cisplatin-induced ototoxicity and nephrotoxicity.


Subject(s)
Cisplatin , Macrophages , Ototoxicity , Cisplatin/adverse effects , Cisplatin/toxicity , Animals , Macrophages/drug effects , Macrophages/metabolism , Ototoxicity/etiology , Ototoxicity/prevention & control , Mice , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Antineoplastic Agents/adverse effects , Antineoplastic Agents/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Kidney Diseases/pathology , Cochlea/drug effects , Cochlea/metabolism , Cochlea/pathology , Mice, Inbred C57BL , Aminopyridines , Pyrroles
5.
PeerJ ; 12: e17687, 2024.
Article in English | MEDLINE | ID: mdl-39056050

ABSTRACT

Background: Cisplatin (CIS) is a broad-spectrum anticancer drug, with cytotoxic effects on either malignant or normal cells. We aimed to evaluate the hepatotoxicity in rats caused by CIS and its amelioration by the co-administration of either curcumin or resveratrol. Materials and Methods: Forty adult male rats divided into four equal groups: (control group): rats were given a saline solution (0.9%) once intraperitoneally, daily for the next 28 days; (cisplatin group): rats were given a daily oral dose of saline solution (0.9%) for 28 days after receiving a single dose of cisplatin (3.3 mg/kg) intraperitoneally for three successive days; (CIS plus curcumin/resveratrol groups): rats received the same previous dose of cisplatin (3.3 mg/kg) daily for three successive days followed by oral administration of either curcumin/resveratrol solution at a dose of (20 mg/kg) or (10 mg/kg) consequently daily for 28 days. Different laboratory tests (ALT, AST, ALP, bilirubin, oxidative stress markers) and light microscopic investigations were done. Results: Administration of CIS resulted in hepatotoxicity in the form of increased liver enzymes, oxidative stress markers; degenerative and apoptotic changes, the co-administration of CIS with either curcumin or resveratrol improved hepatotoxicity through improved microscopic structural changes, reduction in liver enzymes activity, decreased oxidative stress markers, improved degenerative, and apoptotic changes in liver tissues. Conclusion: Co-administration of either curcumin or resveratrol with cisplatin treatment could ameliorate hepatotoxicity caused by cisplatin in rats via anti-inflammatory and oxidative stress-apoptotic pathways.


Subject(s)
Apoptosis , Chemical and Drug Induced Liver Injury , Cisplatin , Curcumin , Oxidative Stress , Resveratrol , Animals , Resveratrol/pharmacology , Resveratrol/administration & dosage , Cisplatin/toxicity , Cisplatin/administration & dosage , Curcumin/pharmacology , Curcumin/administration & dosage , Oxidative Stress/drug effects , Male , Rats , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/etiology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/toxicity , Liver/drug effects , Liver/metabolism , Liver/pathology , Antioxidants/pharmacology , Antioxidants/administration & dosage , Stilbenes/administration & dosage , Stilbenes/pharmacology , Stilbenes/therapeutic use , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/administration & dosage , Rats, Wistar
6.
Rev Assoc Med Bras (1992) ; 70(7): e20240136, 2024.
Article in English | MEDLINE | ID: mdl-39045937

ABSTRACT

OBJECTIVE: Cisplatin, a widely used anticancer agent, induces hepatotoxicity alongside organ damage. Understanding Cisplatin's toxicity mechanism and developing preventive measures are crucial. Our study explores Myricetin, a flavonoid, for its protective effects against Cisplatin-induced hepatotoxicity. METHODS: In our study, a total of 32 Wistar albino male rats were utilized, which were categorized into four distinct groups: Control, Myricetin, Cisplatin, and Myricetin+Cisplatin. For the histological assessment of hepatic tissues, hematoxylin-eosin and periodic acid Schiff staining were employed, alongside immunohistochemical measurements of TNF-α, interleukin-17, and interleukin-6 immunoreactivity. Additionally, aspartate transaminase and alanine transaminase values were examined by biochemical analysis. RESULTS: In the histological evaluation of the tissues, a normal healthy cell structure and a strong periodic acid Schiff (+) reaction were observed in the hepatocyte cells in the tissues of the Control and Myricetin groups, while intense eosinophilia, minimal vacuolization, congestion, and sinusoidal expansions were observed in the hematoxylin-eosin stainings, and a decrease in the positive reaction in the periodic acid Schiff staining was observed in the Cisplatin group. Consistent with these histological findings, an increase in TNF-α, interleukin-17, and interleukin-6 expressions (p<0.0001) and a concomitant increase in aspartate transaminase and alanine transaminase values were observed in the Cisplatin group. In the group protected by Myricetin, a significant improvement was observed in all these histological and biochemical values. CONCLUSION: Cisplatin induces notable histopathological alterations in the liver. In this context, Myricetin exhibits the potential to alleviate Cisplatin-induced damage by modulating histological parameters and biochemical processes.


Subject(s)
Alanine Transaminase , Antineoplastic Agents , Aspartate Aminotransferases , Chemical and Drug Induced Liver Injury , Cisplatin , Flavonoids , Interleukin-6 , Rats, Wistar , Tumor Necrosis Factor-alpha , Animals , Flavonoids/pharmacology , Flavonoids/therapeutic use , Cisplatin/toxicity , Male , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/pathology , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Antineoplastic Agents/adverse effects , Antineoplastic Agents/toxicity , Interleukin-6/analysis , Interleukin-6/metabolism , Liver/drug effects , Liver/pathology , Rats , Interleukin-17/metabolism , Immunohistochemistry
7.
Biochem Pharmacol ; 226: 116369, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38880358

ABSTRACT

Nephrotoxicity is a major constraint of cisplatin application in many solid tumors. Since the lack of preventive strategies, the necessity exists to identify critical molecular targets involved in cisplatin nephrotoxicity. The Purinergic ligand-gcotedion channel 7 receptor (P2X7R) is a ligand-gated ion channel that is predominantly implicated in inflammation and cell death. Our aim is to investigate the role P2X7R in cisplatin-induced acute and chronic kidney injury, as well as the underlying mechanism. In this study, we found that cisplatin can cause an increase in the expression of P2X7R in mouse kidney tissue, and P2X7R knockout can alleviate acute renal function damage caused by cisplatin, as well as the expression of kidney injury molecule 1 (KIM-1) and interleukin-18 (IL-18). Cisplatin can cause an increase in the expression of nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome in mouse kidney tissue. Compared with wild-type mice, P2X7R -/- mice showed decreased expression of NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), cleaved Caspase-1, and cleaved IL-1ß in kidney tissue after cisplatin administration, and the apoptosis of renal tubular epithelial cells were also decreased. In addition, we also found that NLRP3 knockout can improve cisplatin induced degeneration, detachment, and necrosis of renal tubular epithelial cells. Furthermore, P2X7R -/- mice also showed reduced renal fibrosis and better long-term renal prognosis. In conclusion, our study identified that P2X7R knockout can improve cisplatin induced acute renal injury and chronic renal fibrosis by inhibiting the activation of NLRP3 inflammasome.


Subject(s)
Acute Kidney Injury , Cisplatin , Inflammasomes , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Purinergic P2X7 , Animals , Cisplatin/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/deficiency , Mice , Inflammasomes/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Acute Kidney Injury/pathology , Male , Antineoplastic Agents/toxicity
8.
Food Chem Toxicol ; 190: 114791, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38849045

ABSTRACT

AIM: Bortezomib is a proteasome inhibitor antineoplastic agent that was the first to be approved for cancer treatment. One of bortezomib's most prominent dose-limiting effects is nephrotoxicity; the underlying mechanism is believed to be oxidative stress. Chrysin is a compound found actively in honey and many plant species and stands out with its antioxidant properties. The present study aimed to determine the ameliorative effects of chrysin in bortezomib-induced nephrotoxicity. MATERIAL-METHOD: Thirty-five male Wistar rats were divided into control, BTZ, CHR, BTZ + CHR25, and BTZ + CHR50. Biochemical, molecular, Western blot, and histological methods analyzed renal function indicators, oxidative stress, endoplasmic reticulum stress, inflammation, apoptosis, and damage pathways. RESULTS: Chrysin decreased oxidative stress by reducing oxidants (MDA) and increasing antioxidants (SOD, CAT, Gpx, GSH, Nrf-2, HO-1, NQO1). Chrysin reduced endoplasmic reticulum stress by decreasing ATF-6, PERK, IRE1, and GRP-78 levels. Chrysin reduced inflammation damage by inhibiting the NF-κB pathway. Chrysin exhibited protective properties against apoptotic damage by decreasing Bax and Caspase-3 levels and increasing Bcl-2 levels. In addition, chrysin improved renal function and structural integrity and exhibited healing properties against toxic damage in tissue structure. CONCLUSION: Overall, chrysin exhibited an ameliorative effect against bortezomib-induced nephrotoxicity.


Subject(s)
Apoptosis , Bortezomib , Endoplasmic Reticulum Stress , Flavonoids , Inflammation , Kidney , Oxidative Stress , Rats, Wistar , Animals , Flavonoids/pharmacology , Male , Endoplasmic Reticulum Stress/drug effects , Oxidative Stress/drug effects , Apoptosis/drug effects , Bortezomib/pharmacology , Rats , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Inflammation/drug therapy , Inflammation/chemically induced , Autophagy/drug effects , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Kidney Diseases/prevention & control , Kidney Diseases/metabolism , Kidney Diseases/pathology , Antioxidants/pharmacology , Antineoplastic Agents/toxicity
9.
Food Chem Toxicol ; 190: 114792, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38849049

ABSTRACT

Cisplatin is an effective chemotherapy agent against various solid malignancies; however, it is associated with irreversible bilateral sensorineural hearing loss, emphasizing the need for drug development to prevent this complication, with the current options being very limited. Rho-associated coiled-coil-containing protein kinase (ROCK) is a serine-threonine protein kinase involved in various cellular processes, including apoptosis regulation. In this study, we used a transgenic zebrafish model (Brn3C: EGFP) in which hair cells within neuromasts are observed in green under fluorescent microscopy without the need for staining. Zebrafish larvae were exposed to cisplatin alone or in combination with various concentrations of Y-27632, a potent ROCK inhibitor. Hair cell counts, apoptosis assessments using the terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling assay, FM1-43FX labeling assay and behavioral analyses (startle response and rheotaxis) were performed to evaluate the protective effects of Y-27632 against cisplatin-induced ototoxicity. Cisplatin treatment reduced the number of hair cells in neuromasts, induced apoptosis, and impaired zebrafish larval behaviors. Y-27632 demonstrated a dose-dependent protective effect against cisplatin-induced hair cell loss and apoptosis. These findings suggest that Y-27632, as a ROCK inhibitor, mitigates cisplatin-induced hair cell loss and associated ototoxicity in zebrafish.


Subject(s)
Amides , Apoptosis , Cisplatin , Ototoxicity , Pyridines , Zebrafish , Animals , Cisplatin/toxicity , Amides/pharmacology , Pyridines/pharmacology , Ototoxicity/prevention & control , Apoptosis/drug effects , Animals, Genetically Modified , Antineoplastic Agents/toxicity , Hair Cells, Auditory/drug effects , Larva/drug effects , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , Disease Models, Animal
10.
Toxicol Appl Pharmacol ; 489: 117018, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38945373

ABSTRACT

Colitis-associated cancer (CAC) is an aggressive subtype of colorectal cancer that can develop in ulcerative colitis patients and is driven by chronic inflammation and oxidative stress. Current chemotherapy for CAC, based on 5-fluorouracil and oxalipltin, is not fully effective and displays severe side effects, prompting the search for alternative therapies. Dimethylfumarate (DMF), an activator of the nuclear factor erythroid 2-related factor 2 (NRF2), is a potent antioxidant and immunomodelatrory drug used in the treatment of multiple sclerosis and showed a strong anti-inflammatory effect on experimental colitis. Here, we investigated the chemotherapeutic effect of DMF on an experimental model of CAC. Male NMRI mice were given two subcutaneous injections of 1,2 Dimethylhydrazine (DMH), followed by three cycles of dextran sulfate sodium (DSS). Low-dose (DMF30) and high-dose of DMF (DMF100) or oxaliplatin (OXA) were administered from the 8th to 12th week of the experiment, and then the colon tissues were analysed histologically and biochemically. DMH/DSS induced dysplastic aberrant crypt foci (ACF), oxidative stress, and severe colonic inflammation, with a predominance of pro-inflammatory M1 macrophages. As OXA, DMF30 reduced ACF multiplicity and crypt dysplasia, but further restored redox status, and reduced colitis severity by shifting macrophages towards the anti-inflammatory M2 phenotype. Surprisingly, DMF100 exacerbated ACF multiplicity, oxidative stress, and colon inflammation, likely through NRF2 and p53 overexpression in colonic inflammatory cells. DMF had a dual effect on CAC. At low dose, DMF is chemotherapeutic and acts as an antioxidant and immunomodulator, whereas at high dose, DMF is pro-oxidant and exacerbates colitis-associated cancer.


Subject(s)
Colitis-Associated Neoplasms , Dextran Sulfate , Dimethyl Fumarate , Macrophages , Oxidative Stress , Animals , Dimethyl Fumarate/pharmacology , Oxidative Stress/drug effects , Male , Mice , Macrophages/drug effects , Macrophages/metabolism , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/drug therapy , Colitis-Associated Neoplasms/prevention & control , Dextran Sulfate/toxicity , NF-E2-Related Factor 2/metabolism , Antioxidants/pharmacology , Colon/drug effects , Colon/pathology , Colon/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Disease Models, Animal , Anti-Inflammatory Agents/pharmacology , Aberrant Crypt Foci/pathology , Dose-Response Relationship, Drug , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity
11.
Toxicology ; 506: 153840, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38830481

ABSTRACT

Cisplatin (CDDP) is administered as an anticancer drug across a broad spectrum of cancer treatments, but it causes severe renal damage. Several studies have attempted to elucidate the cause of CDDP-induced renal injury, but the detailed mechanism remains unclear. We previously found that S3 cells are more sensitive to CDDP than S1 and S2 cells by using immortalized cells derived from S1, S2, and S3 segments of proximal tubules. In this study, we investigated the potential contribution of reactive oxygen species (ROS) to the sensitivity of S3 cells to CDDP. The results showed that S3 cells have high sensitivity to CDDP, paraquat (PQ) and three ROS substances. To examine the mechanisms underlying the sensitivity to ROS in S3 cells, we compared the cellular responses of CDDP- and PQ-exposed S3 cells. The results indicated that the levels of intracellular ROS and lipid peroxides were increased in S3 cells after CDDP and PQ exposure. The intracellular levels of antioxidant proteins such as thioredoxin, thioredoxin reductase 1 and glutathione peroxidase 4 were also increased by exposure to PQ, but these proteins were decreased by CDDP exposure in S3 cells. Furthermore, the levels of intracellular free Fe2+ were increased by CDDP exposure only in S3 cells but not S1 or S2 cells, and cytotoxicity by exposure to CDDP in S3 cells was suppressed by ferroptosis inhibitors. These results suggested that the induction of ferroptosis due to the ROS production through attenuation of the antioxidant system and elevated free Fe2+ is partly responsible for the sensitivity of S3 cells to CDDP.


Subject(s)
Antineoplastic Agents , Cisplatin , Ferroptosis , Kidney Tubules, Proximal , Reactive Oxygen Species , Cisplatin/toxicity , Cisplatin/pharmacology , Ferroptosis/drug effects , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Reactive Oxygen Species/metabolism , Antineoplastic Agents/toxicity , Antineoplastic Agents/pharmacology , Animals , Paraquat/toxicity , Cell Line , Cell Line, Transformed , Mice , Cell Survival/drug effects
12.
Sci Total Environ ; 943: 173668, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38839013

ABSTRACT

This study investigates the chronic impact of two of the most widely consumed antineoplastic drugs, Ifosfamide (IF) and Cisplatin (CDDP), on the bivalve species Mytilus galloprovincialis under current (17 °C) and predicted warming conditions (21 °C). Accompanying the expected increase in worldwide cancer incidence, antineoplastics detection in the aquatic environment is also expected to rise. Mussels were exposed to varying concentrations of IF (10, 100, 500 ng/L) and CDDP (10, 100, 1000 ng/L) for 28 days. Biochemical analyses focused on metabolic, antioxidant and biotransformation capacities, cellular damage, and neurotoxicity. Results showed temperature-dependent variations in biochemical responses. Metabolic capacity remained stable in mussels exposed to IF, while CDDP exposure increased it at 1000 ng/L for both temperatures. Antioxidant enzyme activities were unaffected by IF, but CDDP activated them, particularly at 21 °C. Biotransformation capacity was unchanged by IF but enhanced by CDDP. Nevertheless, cellular damage occurred at CDDP concentrations above 100 ng/L, regardless of temperature. Integrated biomarker responses highlighted CDDP's greater impact, emphasizing the critical role of temperature in shaping organismal responses and underscoring the complexity of environmental stressor interactions.


Subject(s)
Antineoplastic Agents , Cisplatin , Ifosfamide , Mytilus , Temperature , Water Pollutants, Chemical , Animals , Cisplatin/toxicity , Mytilus/physiology , Mytilus/drug effects , Ifosfamide/toxicity , Water Pollutants, Chemical/toxicity , Antineoplastic Agents/toxicity
13.
Sci Rep ; 14(1): 14004, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38890434

ABSTRACT

Cisplatin is an effective chemotherapeutic agent widely used for the treatment of various solid tumors. However, cisplatin has an important limitation in its use; currently, there is no method to ameliorate cisplatin-induced acute kidney injury (AKI). Thrombomodulin (TM) is well known not only for its role as a cofactor in the clinically important natural anticoagulation pathway but also for its anti-inflammatory properties. Here, we investigated the effects of TM in cisplatin-induced AKI. In mice intraperitoneally injected with 15 mg/kg cisplatin, TM (10 mg/kg) or PBS was administered intravenously at 24 h after cisplatin injection. TM significantly attenuated cisplatin-induced nephrotoxicity with the suppressed elevation of blood urea nitrogen and serum creatinine, and reduced histological damages. Actually, TM treatment significantly alleviated oxidative stress-induced apoptosis by reducing reactive oxygen species (ROS) levels in cisplatin-treated renal proximal tubular epithelial cells (RPTECs) in vitro. Furthermore, TM clarified cisplatin-induced apoptosis by reducing caspase-3 levels. In addition, TM attenuated the endoplasmic reticulum (ER) stress signaling pathway in both renal tissues and RPTECs to protect the kidneys from cisplatin-induced AKI. These findings suggest that TM is a potential protectant against cisplatin-induced nephrotoxicity through suppressing ROS generation and ER stress in response to cisplatin.


Subject(s)
Acute Kidney Injury , Apoptosis , Cisplatin , Endoplasmic Reticulum Stress , Oxidative Stress , Reactive Oxygen Species , Thrombomodulin , Cisplatin/adverse effects , Animals , Thrombomodulin/metabolism , Endoplasmic Reticulum Stress/drug effects , Oxidative Stress/drug effects , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , Mice , Reactive Oxygen Species/metabolism , Male , Apoptosis/drug effects , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/toxicity , Mice, Inbred C57BL , Blood Urea Nitrogen , Signal Transduction/drug effects , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology
14.
J Ethnopharmacol ; 333: 118405, 2024 Oct 28.
Article in English | MEDLINE | ID: mdl-38844249

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ecliptea herba, a traditional Chinese herbal medicine for hair loss, was first recorded in the Tang Dynasty's 'Qian Jin Yue Ling', of which the active ingredients and mechanisms of action in the treatment of chemotherapy-induced hair loss remain poorly investigated. AIM OF THE STUDY: To investigate the effects of the petroleum ether extract of Eclipta (PEE) on alopecia and follicle damage and elucidate its potential therapeutic mechanisms using the integration of network pharmacology, bioinformatics, and experimental validation. MATERIALS AND METHODS: UPLC-MS was used to analyse the chemical composition of PEE. A network pharmacology approach was employed to establish the 'components-targets-pathways' network of PEE to explore potential therapeutic pathways and targets. Molecular docking was used for validation, and the mechanism of PEE in treating chemotherapy-induced alopecia (CIA) was elucidated using in vitro and in vivo on CIA models. RESULTS: UPLC-MS analysis of PEE revealed 185 components, while network pharmacology and molecular docking analyses revealed potential active compounds and their target molecules, suggesting the involvement of core genes, such as TP53, ESR1, AKT1, IL6, TNF, and EGFR. The key components included wedelolactone, dimethyl-wedelolactone, luteoloside, linarin, and hispidulin. In vivo, PEE promoted hair growth, restored the number of hair follicles, and reduced follicle apoptosis. Conversely, in vitro, PEE enhanced cell viability, reduced apoptosis, and protected HaCaT cells from damage induced by 4-hydroperoxycyclophosphamide (4-HC). CONCLUSIONS: PEE alleviated hair follicle damage in CIA mice by inhibiting the P53/Fas pathway, which may be associated with inhibiting hair follicle cell apoptosis. This study provides a novel therapeutic strategy for treating cyclophosphamide-induced hair loss.


Subject(s)
Alopecia , Eclipta , Molecular Docking Simulation , Network Pharmacology , Tumor Suppressor Protein p53 , Alopecia/chemically induced , Alopecia/drug therapy , Animals , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Eclipta/chemistry , Mice , Plant Extracts/pharmacology , Plant Extracts/chemistry , Hair Follicle/drug effects , Hair Follicle/metabolism , Male , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Apoptosis/drug effects , HaCaT Cells , Cyclophosphamide/toxicity , Alkanes
15.
Biochem Biophys Res Commun ; 725: 150266, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38878759

ABSTRACT

Cisplatin (CDDP) is a platinum-based anticancer drug widely prescribed for its effectiveness in treating various forms of cancer. However, its major side effect is nephrotoxicity. Although several methods have been developed to mitigate CDDP-induced nephrotoxicity, an optimal approach has yet to be established. This study aimed to investigate the "chronotoxicity" of CDDP as a potential strategy to reduce its side effects. Male ICR mice were treated with CDDP (20 mg/kg, intraperitoneal injection, one shot) at zeitgeber time (ZT) 2 or ZT14 (light or dark phase). After 72 h, we collected plasma and kidney and evaluated several markers. We found that body weight change between ZT2 and ZT14 by CDDP was comparable. In contrast, many toxicological factors, such as plasma blood urine nitrogen, plasma creatinine, renal oxidative stress (malondialdehyde), DNA damage (γH2AX), acute kidney injury biomarker (KIM-1), and inflammation (Tnfα), were significantly induced at ZT14 compared to than that of ZT2. Our present data suggested that chronotoxicology might provide beneficial information on the importance of administration timings for toxic evaluations and unacceptable side effects.


Subject(s)
Antineoplastic Agents , Circadian Rhythm , Cisplatin , Kidney , Mice, Inbred ICR , Animals , Cisplatin/toxicity , Male , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Antineoplastic Agents/toxicity , Antineoplastic Agents/adverse effects , Mice , Circadian Rhythm/drug effects , Oxidative Stress/drug effects , DNA Damage/drug effects , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/pathology
16.
PeerJ ; 12: e17485, 2024.
Article in English | MEDLINE | ID: mdl-38854800

ABSTRACT

Background: Cisplatin is a commonly used nephrotoxic drug and can cause acute kidney injury (AKI). In the present study, isobaric tags for relative and absolute quantification (iTRAQ) and parallel reaction monitoring (PRM)-based comparative proteomics were used to analyze differentially expressed proteins (DEPs) to determine the key molecular mechanism in mice with cisplatin-induced AKI in the presence or absence of SIS3, a specific p-smad3 inhibitor, intervention. Methods: The cisplatin-induced AKI mouse model was established and treated with SIS3. We used iTRAQ to search for DEPs, PRM to verify key DEPs and combined Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) for bioinformatics analysis. We then assessed lipid deposition, malondialdehyde (MDA) and reactive oxygen species (ROS) and detected the expression of SREBF1, SCD1, CPT1A, PPARα and NDRG1 in vitro. Results: Proteomic analysis showed that the identified DEPs were mainly enriched in energy metabolism pathways, especially in lipid metabolism. When SIS3 was applied to inhibit the phosphorylation of Smad3, the expression of NDRG1 and fatty acid oxidation key proteins CPT1A and PPARα increased, the expression of lipid synthesis related proteins SREBF1 and SCD1 decreased and the production of lipid droplets, MDA and ROS decreased. Conclusion: SIS3 alleviates oxidative stress, reduces lipid accumulation and promotes fatty acid oxidation through NDRG1 in cisplatin-induced AKI. Our study provides a new candidate protein for elucidating the molecular mechanisms of fatty acid metabolism disorders in cisplatin-induced acute kidney injury.


Subject(s)
Acute Kidney Injury , Cisplatin , Proteomics , Cisplatin/adverse effects , Cisplatin/toxicity , Animals , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Proteomics/methods , Mice , Disease Models, Animal , Male , Smad3 Protein/metabolism , Smad3 Protein/genetics , Lipid Metabolism/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Antineoplastic Agents/adverse effects , Antineoplastic Agents/toxicity
17.
Environ Toxicol ; 39(8): 4184-4195, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38708805

ABSTRACT

Small extracellular vesicles (sEVs) secreted by human umbilical cord have therapeutic effects on various degenerative diseases. However, the characteristics and potential functions of human umbilical cord mesenchymal stem cells (huMSCs)-derived sEVs, especially the role of premature ovarian failure (POF), are poorly understood. Here, we isolated and characterized huMSCs and their sEVs. huMSCs highly expressed CD73, CD90, and CD105. huMSC-sEVs showed typical exosomal features, highly expressing CD9, TSG101, and CD63. It was shown that huMSC-sEVs could be taken up by granulosa cells (GCs) and damaged ovarian tissue, which increased the levels of hormone secretion and reduced GCs apoptosis. We further confirmed that the levels of follicle-stimulating hormone in rat serum decreased dramatically, while the levels of estrogen (E2)and anti-mullerian hormone (AMH) increased significantly with the treatment of huMSC-sEVs. Meanwhile, huMSC-sEVs treatment greatly reduced cell apoptosis and autophagy, while increased the phosphorylation levels of p-PI3K and p-Akt. Therefore, treatment with huMSC-sEVs significantly inhibited GCs apoptosis, improved ovarian morphology, promoted follicular development, inhibited follicular over-atresia, and improved ovarian reserve capacity in POF rats. Our study verified that activation of PI3K/Akt signaling pathway and regulation of cellular autophagy, thus reducing GCs death, are the mechanisms by which huMSC-sEVs restore ovarian tissue function.


Subject(s)
Apoptosis , Cisplatin , Extracellular Vesicles , Granulosa Cells , Mesenchymal Stem Cells , Ovary , Primary Ovarian Insufficiency , Umbilical Cord , Female , Mesenchymal Stem Cells/drug effects , Animals , Extracellular Vesicles/drug effects , Extracellular Vesicles/metabolism , Humans , Umbilical Cord/cytology , Primary Ovarian Insufficiency/chemically induced , Cisplatin/toxicity , Apoptosis/drug effects , Rats , Ovary/drug effects , Ovary/pathology , Granulosa Cells/drug effects , Rats, Sprague-Dawley , Antineoplastic Agents/toxicity
18.
Biomed Pharmacother ; 175: 116752, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38761425

ABSTRACT

The gut microbiota has been reported to be perturbed by chemotherapeutic agents and to modulate side effects. However, the critical role of ß-hydroxybutyrate (BHB) in the regulation of the gut microbiota and the pathogenesis of chemotherapeutic agents related nephrotoxicity remains unknown. We conducted a comparative analysis of the composition and function of gut microbiota in healthy, cisplatin-challenged, BHB-treated, and high-fat diet-treated mice using 16 S rDNA gene sequencing. To understand the crucial involvement of intestinal flora in BHB's regulation of cisplatin -induced nephrotoxicity, we administered antibiotics to deplete the gut microbiota and performed fecal microbiota transplantation (FMT) before cisplatin administration. 16 S rDNA gene sequencing analysis demonstrated that both endogenous and exogenous BHB restored gut microbiota dysbiosis and cisplatin-induced intestinal barrier disruption in mice. Additionally, our findings suggested that the LPS/TLR4/NF-κB pathway was responsible for triggering renal inflammation in the gut-kidney axis. Furthermore, the ablation of the gut microbiota ablation using antibiotics eliminated the renoprotective effects of BHB against cisplatin-induced acute kidney injury. FMT also confirmed that administration of BHB-treated gut microbiota provided protection against cisplatin-induced nephrotoxicity. This study elucidated the mechanism by which BHB affects the gut microbiota mediation of cisplatin-induced nephrotoxicity by inhibiting the inflammatory response, which may help develop novel therapeutic approaches that target the composition of the microbiota.


Subject(s)
3-Hydroxybutyric Acid , Acute Kidney Injury , Cisplatin , Dysbiosis , Gastrointestinal Microbiome , Mice, Inbred C57BL , Animals , Cisplatin/adverse effects , Gastrointestinal Microbiome/drug effects , Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Male , Dysbiosis/chemically induced , Mice , 3-Hydroxybutyric Acid/pharmacology , Kidney/drug effects , Fecal Microbiota Transplantation , Diet, High-Fat/adverse effects , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Protective Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/adverse effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents/toxicity
19.
Biomed Pharmacother ; 175: 116797, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38776675

ABSTRACT

Cisplatin (CIS) stands as one of the most effective chemotherapy drugs currently available. Despite its anticancer properties, the clinical application of CIS is restricted due to nephrotoxicity. Our research aimed to specify the impact of ketotifen fumarate (KET) against nephrotoxicity induced by CIS in mice. Male NMRI mice were treated with KET (0.4, 0.8, and 1.6 mg/kg, ip) for seven days. On the fourth day of the study, a single dose of CIS (13 mg/kg, ip) was administered, and the mice were sacrificed on the eighth day. The results indicated that administration of KET attenuated CIS-induced elevation of BUN and Cr in the serum, as well as renal KIM-1 levels. This improvement was accompanied by a significant reduction in kidney tissue damage, which was supported by histopathological examinations. Likewise, the decrease in the ratio of GSH to GSSG and antioxidant enzyme activities (CAT, SOD, and GPx), and the increase in lipid peroxidation marker (TBARS) were reversed in KET-treated mice. The ELISA results revealed that KET-treated mice ameliorated CIS-induced elevation in the renal levels of TNF-α, IL-1ß, and IL-18. Western blot analysis exhibited that KET suppressed the activation of the transcription factor NF-κB and the NLRP3 inflammasome in the kidney of CIS-treated mice. Moreover, KET treatment reversed the changes in the protein expression of markers related to apoptosis (Bax, Bcl2, Caspase-3, and p53). Interestingly, KET significantly enhanced the cytotoxicity of CIS in HeLa cells. In conclusion, this study provides valuable insights into the promising effects of KET in mitigating CIS-induced nephrotoxicity.


Subject(s)
Acute Kidney Injury , Caspase 1 , Caspase 3 , Cisplatin , Ketotifen , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , bcl-2-Associated X Protein , Animals , Cisplatin/toxicity , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction/drug effects , Mice , NF-kappa B/metabolism , Caspase 1/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Caspase 3/metabolism , Humans , Ketotifen/pharmacology , bcl-2-Associated X Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis/drug effects , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , HeLa Cells , Oxidative Stress/drug effects
20.
Hear Res ; 447: 109013, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718672

ABSTRACT

Cisplatin, a highly effective chemotherapeutic drug for various human cancers, induces irreversible sensorineural hearing loss as a side effect. Currently there are no highly effective clinical strategies for the prevention of cisplatin-induced ototoxicity. Previous studies have indicated that short-term cisplatin ototoxicity primarily affects the outer hair cells of the cochlea. Therefore, preventing the entry of cisplatin into hair cells may be a promising strategy to prevent cisplatin ototoxicity. This study aimed to investigate the entry route of cisplatin into mouse cochlear hair cells. The competitive inhibitor of organic cation transporter 2 (OCT2), cimetidine, and the sensory mechanoelectrical transduction (MET) channel blocker benzamil, demonstrated a protective effect against cisplatin toxicity in hair cells in cochlear explants. Sensory MET-deficient hair cells explanted from Tmc1Δ;Tmc2Δ mice were resistant to cisplatin toxicity. Cimetidine showed an additive protective effect against cisplatin toxicity in sensory MET-deficient hair cells. However, in the apical turn, cimetidine, benzamil, or genetic ablation of sensory MET channels showed limited protective effects, implying the presence of other entry routes for cisplatin to enter the hair cells in the apical turn. Systemic administration of cimetidine failed to protect cochlear hair cells from ototoxicity caused by systemically administered cisplatin. Notably, outer hair cells in MET-deficient mice exhibited no apparent deterioration after systemic administration of cisplatin, whereas the outer hair cells in wild-type mice showed remarkable deterioration. The susceptibility of mouse cochlear hair cells to cisplatin ototoxicity largely depends on the sensory MET channel both ex vivo and in vivo. This result justifies the development of new pharmaceuticals, such as a specific antagonists for sensory MET channels or custom-designed cisplatin analogs which are impermeable to sensory MET channels.


Subject(s)
Antineoplastic Agents , Cimetidine , Cisplatin , Mechanotransduction, Cellular , Organic Cation Transporter 2 , Ototoxicity , Cisplatin/toxicity , Animals , Ototoxicity/prevention & control , Ototoxicity/metabolism , Ototoxicity/physiopathology , Mechanotransduction, Cellular/drug effects , Organic Cation Transporter 2/metabolism , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/antagonists & inhibitors , Cimetidine/pharmacology , Antineoplastic Agents/toxicity , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Hair Cells, Auditory, Outer/metabolism , Mice, Inbred C57BL , Mice , Membrane Proteins
SELECTION OF CITATIONS
SEARCH DETAIL