Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.232
Filtrar
1.
J Biochem Mol Toxicol ; 38(8): e23768, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39015062

RESUMO

Nephrotoxicity remains a major adverse reaction of the anticancer drug cisplatin (CDDP) chemotherapy, which is an important risk factor for chronic renal disease. Ginsenoside Rh2 from Panax ginseng has been shown to protect against CDDP-induced nephrotoxicity in vivo, but its pharmacological effect on renal tubular epithelial cells is not clearly understood. This study examined the molecular mechanisms underlying the nephroprotective effects of Rh2 on CDDP-induced HK-2 cells and acute kidney injury (AKI) mice. As a result of Rh2 treatment, CDDP-induced HK-2 cells showed increased cell viability and reduced lactate dehydrogenase release. Moreover, Rh2 ameliorated CDDP-induced mitochondrial membrane potential, increased antioxidant enzyme activities, and reduced pro-inflammatory cytokine expression to reduce damage. Rh2 inhibited apoptosis and enhanced the antioxidant capacity of HK-2 cells by reducing proteins associated with endoplasmic reticulum (ER) stress, as well as by attenuating tunicamycin-induced ER stress. In addition, treatment of CDDP-induced AKI mice with Rh2 substantially reduced blood urea nitrogen and serum creatinine levels, attenuated histological damage of kidney. Further, Rh2 also improved kidney function by inhibiting ER stress to support in vitro findings. These results consistently demonstrated that Rh2 protects renal tubular epithelial cells from CDDP-induced nephrotoxicity and apoptosis by restoring ER homeostasis, which might suggest a therapeutic potential and providing new insights into AKI alternative therapies.


Assuntos
Injúria Renal Aguda , Cisplatino , Estresse do Retículo Endoplasmático , Células Epiteliais , Ginsenosídeos , Túbulos Renais , Ginsenosídeos/farmacologia , Cisplatino/efeitos adversos , Cisplatino/toxicidade , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Animais , Camundongos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/patologia , Túbulos Renais/metabolismo , Humanos , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/tratamento farmacológico , Masculino , Linhagem Celular , Apoptose/efeitos dos fármacos , Camundongos Endogâmicos C57BL
2.
Rev Assoc Med Bras (1992) ; 70(7): e20240136, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39045937

RESUMO

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.


Assuntos
Alanina Transaminase , Antineoplásicos , Aspartato Aminotransferases , Doença Hepática Induzida por Substâncias e Drogas , Cisplatino , Flavonoides , Interleucina-6 , Ratos Wistar , Fator de Necrose Tumoral alfa , Animais , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Cisplatino/toxicidade , Masculino , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Doença Hepática Induzida por Substâncias e Drogas/patologia , Alanina Transaminase/sangue , Aspartato Aminotransferases/sangue , Antineoplásicos/efeitos adversos , Antineoplásicos/toxicidade , Interleucina-6/análise , Interleucina-6/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Ratos , Interleucina-17/metabolismo , Imuno-Histoquímica
3.
Physiol Res ; 73(3): 405-413, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39027957

RESUMO

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.


Assuntos
Cisplatino , Ferroptose , Flavonóis , Camundongos Endogâmicos C57BL , Atrofia Muscular , Estresse Oxidativo , Animais , Masculino , Flavonóis/farmacologia , Flavonóis/uso terapêutico , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/patologia , Atrofia Muscular/metabolismo , Atrofia Muscular/prevenção & controle , Atrofia Muscular/tratamento farmacológico , Ferroptose/efeitos dos fármacos , Cisplatino/toxicidade , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Músculo Esquelético/metabolismo , Antineoplásicos/toxicidade , Antioxidantes/farmacologia
4.
Ren Fail ; 46(2): 2378212, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39011587

RESUMO

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.


Assuntos
Anti-Inflamatórios , Antioxidantes , Apoptose , Cisplatino , Dioxóis , Rim , Lignanas , Ratos Wistar , Testículo , Animais , Masculino , Lignanas/farmacologia , Lignanas/uso terapêutico , Cisplatino/toxicidade , Cisplatino/efeitos adversos , Ratos , Dioxóis/farmacologia , Antioxidantes/farmacologia , Testículo/efeitos dos fármacos , Testículo/patologia , Testículo/metabolismo , Apoptose/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/patologia , Rim/metabolismo , Anti-Inflamatórios/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/patologia , Injúria Renal Aguda/metabolismo , Antineoplásicos/toxicidade
5.
Int J Mol Sci ; 25(11)2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38892427

RESUMO

Neurotoxicity is a major obstacle in the effectiveness of Cisplatin in cancer chemotherapy. In this process, oxidative stress and inflammation are considered to be the main mechanisms involved in brain and lung toxicity. The aim of the present work was to study the influence of the amount of protein on some oxidative parameters in the brain and lungs of rats treated with Cisplatin (CP) and N-Acetylcysteine (NAC) as neuroprotectors. Four groups of Wistar rats, each containing six animals, were fed with a protein diet at 7% for 15 days. Thereafter, the groups were given either a unique dose of CP® 5 mg/kg or NAC® 5 mg/kg as follows: group 1 (control), NaCl 0.9% vehicle; group 2, CP; group 3, NAC; and group 4, NAC + CP. The animals were sacrificed immediately after the treatments. Blood samples were collected upon sacrifice and used to measure blood triglycerides and glucose. The brain and lungs of each animal were obtained and used to assay lipid peroxidation (TBARS), glutathione (GSH), serotonin metabolite (5-HIAA), catalase, and the activity of Ca+2, and Mg+2 ATPase using validated methods. TBARS, H2O2, and GSH were found to be significantly decreased in the cortex and cerebellum/medulla oblongata of the groups treated with CP and NAC. The total ATPase showed a significant increase in the lung and cerebellum/medulla oblongata, while 5-HIAA showed the same tendency in the cortex of the same group of animals. The increase in 5-HIAA and ATPase during NAC and CP administration resulted in brain protection. This effect could be even more powerful when membrane fluidity is increased, thus proving the efficacy of combined NAC and CP drug therapy, which appears to be a promising strategy for future chemotherapy in malnourished patients.


Assuntos
Acetilcisteína , Cisplatino , Pulmão , Ratos Wistar , Animais , Cisplatino/efeitos adversos , Cisplatino/toxicidade , Acetilcisteína/farmacologia , Ratos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Masculino , Cérebro/efeitos dos fármacos , Cérebro/metabolismo , Glutationa/metabolismo , Fármacos Neuroprotetores/farmacologia , Antineoplásicos/efeitos adversos
6.
Biochem Biophys Res Commun ; 725: 150266, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-38878759

RESUMO

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.


Assuntos
Antineoplásicos , Ritmo Circadiano , Cisplatino , Rim , Camundongos Endogâmicos ICR , Animais , Cisplatino/toxicidade , Masculino , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Antineoplásicos/toxicidade , Antineoplásicos/efeitos adversos , Camundongos , Ritmo Circadiano/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Nefropatias/patologia
7.
Sci Total Environ ; 943: 173668, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-38839013

RESUMO

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.


Assuntos
Antineoplásicos , Cisplatino , Ifosfamida , Mytilus , Temperatura , Poluentes Químicos da Água , Animais , Cisplatino/toxicidade , Mytilus/fisiologia , Mytilus/efeitos dos fármacos , Ifosfamida/toxicidade , Poluentes Químicos da Água/toxicidade , Antineoplásicos/toxicidade
8.
PeerJ ; 12: e17485, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38854800

RESUMO

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.


Assuntos
Injúria Renal Aguda , Cisplatino , Proteômica , Cisplatino/efeitos adversos , Cisplatino/toxicidade , Animais , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Proteômica/métodos , Camundongos , Modelos Animais de Doenças , Masculino , Proteína Smad3/metabolismo , Proteína Smad3/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Antineoplásicos/efeitos adversos , Antineoplásicos/toxicidade
9.
Toxicology ; 506: 153840, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38830481

RESUMO

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.


Assuntos
Antineoplásicos , Cisplatino , Ferroptose , Túbulos Renais Proximais , Espécies Reativas de Oxigênio , Cisplatino/toxicidade , Cisplatino/farmacologia , Ferroptose/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Espécies Reativas de Oxigênio/metabolismo , Antineoplásicos/toxicidade , Antineoplásicos/farmacologia , Animais , Paraquat/toxicidade , Linhagem Celular , Linhagem Celular Transformada , Camundongos , Sobrevivência Celular/efeitos dos fármacos
10.
Food Chem Toxicol ; 190: 114792, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38849049

RESUMO

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.


Assuntos
Amidas , Apoptose , Cisplatino , Ototoxicidade , Piridinas , Peixe-Zebra , Animais , Cisplatino/toxicidade , Amidas/farmacologia , Piridinas/farmacologia , Ototoxicidade/prevenção & controle , Apoptose/efeitos dos fármacos , Animais Geneticamente Modificados , Antineoplásicos/toxicidade , Células Ciliadas Auditivas/efeitos dos fármacos , Larva/efeitos dos fármacos , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Modelos Animais de Doenças
11.
Physiol Res ; 73(2): 227-237, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38710058

RESUMO

Nephrotoxicity as a cause of acute kidney injury (AKI) induced by cisplatin (CP), limits its usefulness as an anticancer agent. Diminazene, an angiotensin converting enzyme 2 activator, exhibited renoprotective properties on rat models of kidney diseases. This research aims to investigate the salutary effect of diminazene in comparison with lisinopril or valsartan in CP-induced AKI. The first and second groups of rats received oral vehicle (distilled water) for 9 days, and saline injection or intraperitoneal CP (6 mg/kg) on day 6, respectively. Third, fourth, and fifth groups received intraperitoneal injections of CP on day 6 and diminazene (15 mg/kg/day, orally), lisinopril (10 mg/kg/day, orally), or valsartan (30 mg/kg/day, orally), for 9 days, respectively. 24h after the last day of treatment, blood and kidneys were removed under anesthesia for biochemical and histopathological examination. Urine during the last 24 h before sacrificing the rats was also collected. CP significantly increased plasma urea, creatinine, neutrophil gelatinase-associated lipocalin, calcium, phosphorus, and uric acid. It also increased urinary albumin/creatinine ratio, N-Acetyl-beta-D-Glucosaminidase/creatinine ratio, and reduced creatinine clearance, as well the plasma concentrations of inflammatory cytokines [plasma tumor necrosis factor-alpha, and interleukin-1beta], and significantly reduced antioxidant indices [catalase, glutathione reductase , and superoxide dismutase]. Histopathologically, CP treatment caused necrosis of renal tubules, tubular casts, shrunken glomeruli, and increased renal fibrosis. Diminazine, lisinopril, and valsartan ameliorated CP-induced biochemical and histopathological changes to a similar extent. The salutary effect of the three drugs used is, at least partially, due to their anti-inflammatory and antioxidant effects. Keywords: Cisplatin, Diminazene, ACE2 activator, Lisinopril, Valsartan, Acute kidney injury.


Assuntos
Injúria Renal Aguda , Cisplatino , Diminazena , Lisinopril , Ratos Wistar , Valsartana , Animais , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/tratamento farmacológico , Lisinopril/farmacologia , Cisplatino/toxicidade , Valsartana/farmacologia , Masculino , Diminazena/análogos & derivados , Diminazena/farmacologia , Diminazena/uso terapêutico , Ratos , Antineoplásicos/toxicidade , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Rim/efeitos dos fármacos , Rim/patologia , Rim/metabolismo
12.
Int J Mol Sci ; 25(10)2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38791162

RESUMO

Early detection of drug-induced kidney injury is essential for drug development. In this study, multiple low-dose aristolochic acid (AA) and cisplatin (Cis) injections increased renal mRNA levels of inflammation, fibrosis, and renal tubule injury markers. We applied a serum amyloid A3 (Saa3) promoter-driven luciferase reporter (Saa3 promoter-luc mice) to these two tubulointerstitial nephritis models and performed in vivo bioluminescence imaging to monitor early renal pathologies. The bioluminescent signals from renal tissues with AA or CIS injections were stronger than those from normal kidney tissues obtained from normal mice. To verify whether the visualized bioluminescence signal was specifically generated by the injured kidney, we performed in vivo bioluminescence analysis after opening the stomachs of Saa3 promoter-luc mice, and the Saa3-mediated bioluminescent signal was specifically detected in the injured kidney. This study showed that Saa3 promoter activity is a potent non-invasive indicator for the early detection of drug-induced nephrotoxicity.


Assuntos
Ácidos Aristolóquicos , Luciferases , Regiões Promotoras Genéticas , Proteína Amiloide A Sérica , Animais , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/metabolismo , Camundongos , Luciferases/metabolismo , Luciferases/genética , Ácidos Aristolóquicos/toxicidade , Genes Reporter , Cisplatino/toxicidade , Cisplatino/efeitos adversos , Medições Luminescentes/métodos , Masculino , Nefropatias/induzido quimicamente , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Rim/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
13.
J Pharm Pharmacol ; 76(7): 884-896, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38708970

RESUMO

OBJECTIVES: Acute kidney injury (AKI) caused by cisplatin (CDDP) is a complex, critical illness with no effective or specific treatment. The purpose of the study was to assess the protective effect of protopanaxadiol (PPD) on the kidneys in CDDP-induced AKI models and its possible mechanisms. METHODS: In vitro, the protection of PPD was assessed in HK-2. KM mice were injected with CDDP to induce AKI models in vivo. The determination of blood urea nitrogen and serum creatinine (SCr) was performed, and pathological changes were examined by histopathological examination. Immunostaining and western blot analyses were used to analyze the expression levels of proteins. RESULTS: PPD can increase the viability of HK-2 cells damaged by CDDP, improve cell morphology, and alleviate the symptoms of AKI in mice. In addition, PPD can down-regulate the protein expression of TRF and up-regulate the protein expression of Ferritin heavy chain, Glutathione peroxidase 4, and ferroptosis suppressor protein 1 reduce the iron content in cells and kidney tissues, and restore the antioxidant defense system. CONCLUSION: PPD has an inhibitory effect on cisplatin-induced nephrotoxicity, which may be related to the inhibition of ferroptosis by regulating iron metabolism and lipid peroxidation.


Assuntos
Injúria Renal Aguda , Cisplatino , Ferroptose , Sapogeninas , Cisplatino/toxicidade , Animais , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/metabolismo , Ferroptose/efeitos dos fármacos , Camundongos , Sapogeninas/farmacologia , Humanos , Masculino , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Modelos Animais de Doenças , Linhagem Celular , Peroxidação de Lipídeos/efeitos dos fármacos , Ferro/metabolismo , Antioxidantes/farmacologia , Sobrevivência Celular/efeitos dos fármacos
14.
Analyst ; 149(13): 3596-3606, 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38767610

RESUMO

Real-time and non-invasive assessment of tissue health is crucial for maximizing the potential of microphysiological systems (MPS) for drug-induced nephrotoxicity screening. Although impedance has been widely considered as a measure of the barrier function, it has not been incorporated to detect cell detachment in MPS with top and bottom microfluidic channels separated by a porous membrane. During cell delamination from the porous membrane, the resistance between both channels decreases, while capacitance increases, allowing the detection of such detachment. Previously reported concepts have solely attributed the decrease in the resistance to the distortion of the barrier function, ignoring the resistance and capacitance changes due to cell detachment. Here, we report a two-channel MPS with integrated indium tin oxide (ITO) electrodes capable of measuring impedance in real time. The trans-epithelial electrical resistance (TEER) and tissue reactance (capacitance) were extracted from the impedance profiles. We attributed the anomalous initial increase observed in TEER, upon cisplatin administration, to the distortion of tight junctions. Cell detachment was captured by sudden jumps in capacitance. TEER profiles illuminated the effects of cisplatin and cimetidine treatments in a dose-dependent and polarity-dependent manner. The correspondence between TEER and barrier function was validated for a continuous tissue using the capacitance profiles. These results demonstrate that capacitance can be used as a real-time and non-invasive indicator of confluence and will support the accuracy of the drug-induced cytotoxicity assessed by TEER profiles in the two-channel MPS for the barrier function of a cell monolayer.


Assuntos
Cisplatino , Impedância Elétrica , Túbulos Renais Proximais , Cisplatino/toxicidade , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/patologia , Animais , Compostos de Estanho/química , Compostos de Estanho/toxicidade , Cinética , Cimetidina/farmacologia , Adesão Celular/efeitos dos fármacos , Eletrodos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Linhagem Celular , Humanos , Junções Íntimas/efeitos dos fármacos
15.
Neuroreport ; 35(10): 657-663, 2024 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-38813907

RESUMO

Cisplatin-induced cognitive impairment (chemobrain) affects a considerable percentage of cancer patients and has no established pharmacological treatment. Chemobrain can be associated with neuroinflammation and oxidative stress. Melatonin, a pineal hormone, is known to have antioxidant, anti-inflammatory and neuroprotective potential. In this study, we investigated cisplatin-induced cognitive impairment in rats and whether melatonin can improve or reverse this impairment. Behavioral testing involved measuring working memory using the novel location recognition test (NLRT) under conditions of cisplatin or cisplatin + melatonin treatment, followed by the collection of rats' brains. The brains were subsequently stained with Golgi-Cox stain and then the hippocampus area CA3 of each one was examined, and dendritic spine density was calculated. Treatment with cisplatin resulted in deficits in the rats' performance in the NLRT (P < 0.05). These deficits were prevented by the coadministration of melatonin (P < 0.05). Cisplatin also reduced the density of dendritic spines in the hippocampus (P < 0.0001), specifically CA3 area, while the coadministration of melatonin significantly reversed this reduction (P < 0.001). This study showed that melatonin can ameliorate cisplatin-induced spatial memory deficits and dendritic spines density abnormalities in rats. Given that melatonin is a safe and wildly used supplement, it is feasible to explore its use as a palliative intervention in cancer treatment.


Assuntos
Cisplatino , Espinhas Dendríticas , Hipocampo , Melatonina , Animais , Melatonina/farmacologia , Cisplatino/toxicidade , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , Masculino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/metabolismo , Ratos , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/prevenção & controle , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/patologia , Antineoplásicos/toxicidade , Fármacos Neuroprotetores/farmacologia , Antioxidantes/farmacologia , Ratos Wistar , Comprometimento Cognitivo Relacionado à Quimioterapia , Memória de Curto Prazo/efeitos dos fármacos
16.
Environ Toxicol ; 39(8): 4184-4195, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38708805

RESUMO

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.


Assuntos
Apoptose , Cisplatino , Vesículas Extracelulares , Células da Granulosa , Células-Tronco Mesenquimais , Ovário , Insuficiência Ovariana Primária , Cordão Umbilical , Feminino , Células-Tronco Mesenquimais/efeitos dos fármacos , Animais , Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/metabolismo , Humanos , Cordão Umbilical/citologia , Insuficiência Ovariana Primária/induzido quimicamente , Cisplatino/toxicidade , Apoptose/efeitos dos fármacos , Ratos , Ovário/efeitos dos fármacos , Ovário/patologia , Células da Granulosa/efeitos dos fármacos , Ratos Sprague-Dawley , Antineoplásicos/toxicidade
17.
Toxicol In Vitro ; 99: 105852, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38789064

RESUMO

Cisplatin is an effective chemotherapeutic agent; however, ototoxicity is one of its negative effects that greatly limits the use of cisplatin in clinical settings. Previous research has shown that the most important process cisplatin damage to inner ear cells, such as hair cells (HCs), is the excessive production and accumulation of ROS. Schisandrin B (SchB), is a low-toxicity, inexpensive, naturally occurring antioxidant with a variety of pharmacological effects. Therefore, the potential antioxidant effects of SchB may be useful for cisplatin ototoxicity treatment. In this study, the effects of SchB on cochlear hair cell viability, ROS levels, and expression of apoptosis-related molecules were evaluated by CCK-8, immunofluorescence, flow cytometry, and qRT-PCR, as well as auditory brainstem response (ABR) and dysmorphic product otoacoustic emission (DPOAE) tests to assess the effects on inner ear function. The results showed that SchB treatment increased cell survival, prevented apoptosis, and reduced cisplatin-induced ROS formation. SchB treatment reduced the loss of cochlear HCs caused by cisplatin in exosome culture. In addition, SchB treatment attenuated cisplatin-induced hearing loss and HC loss in mice. This study demonstrates the ability of SchB to inhibit cochlear hair cell apoptosis and ROS generation and shows its potential therapeutic effect on cisplatin ototoxicity.


Assuntos
Antineoplásicos , Apoptose , Sobrevivência Celular , Cisplatino , Ciclo-Octanos , Células Ciliadas Auditivas Internas , Lignanas , Estresse Oxidativo , Compostos Policíclicos , Espécies Reativas de Oxigênio , Cisplatino/toxicidade , Ciclo-Octanos/farmacologia , Compostos Policíclicos/farmacologia , Compostos Policíclicos/toxicidade , Animais , Apoptose/efeitos dos fármacos , Lignanas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Antineoplásicos/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Células Ciliadas Auditivas Internas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Substâncias Protetoras/farmacologia , Antioxidantes/farmacologia , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Masculino , Ototoxicidade/prevenção & controle
18.
Biomed Pharmacother ; 175: 116797, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38776675

RESUMO

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.


Assuntos
Injúria Renal Aguda , Caspase 1 , Caspase 3 , Cisplatino , Cetotifeno , NF-kappa B , Proteína 3 que Contém Domínio de Pirina da Família NLR , Transdução de Sinais , Proteína X Associada a bcl-2 , Animais , Cisplatino/toxicidade , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Transdução de Sinais/efeitos dos fármacos , Camundongos , NF-kappa B/metabolismo , Caspase 1/metabolismo , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Caspase 3/metabolismo , Humanos , Cetotifeno/farmacologia , Proteína X Associada a bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Apoptose/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/patologia , Rim/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Células HeLa , Estresse Oxidativo/efeitos dos fármacos
19.
Ren Fail ; 46(1): 2354918, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38757723

RESUMO

Cisplatin is a particularly potent antineoplastic drug. However, its usefulness is restricted due to the induction of nephrotoxicity. More recent research has indicated that ß-hydroxybutyrate (ß-HB) protects against acute or chronic organ damage as an efficient healing agent. Nonetheless, the therapeutic mechanisms of ß-HB in acute kidney damage caused by chemotherapeutic drugs remain unclear. Our study developed a model of cisplatin-induced acute kidney injury (AKI), which involved the administration of a ketogenic diet or ß-HB. We analyzed blood urea nitrogen (BUN) and creatinine (Cr) levels in serum, and used western blotting and immunohistochemical staining to assess ferroptosis and the calcium/calmodulin-dependent kinase kinase 2 (Camkk2)/AMPK pathway. The mitochondrial morphology and function were examined. Additionally, we conducted in vivo and in vitro experiments using selective Camkk2 inhibitor or activator to investigate the protective mechanism of ß-HB on cisplatin-induced AKI. Exogenous or endogenous ß-HB effectively alleviated cisplatin-induced abnormally elevated levels of BUN and Cr and renal tubular necrosis in vivo. Additionally, ß-HB reduced ferroptosis biomarkers and increased the levels of anti-ferroptosis biomarkers in the kidney. ß-HB also improved mitochondrial morphology and function. Moreover, ß-HB significantly attenuated cisplatin-induced cell ferroptosis and damage in vitro. Furthermore, western blotting and immunohistochemical staining indicated that ß-HB may prevent kidney injury by regulating the Camkk2-AMPK pathway. The use of the Camkk2 inhibitor or activator verified the involvement of Camkk2 in the renal protection by ß-HB. This study provided evidence of the protective effects of ß-HB against cisplatin-induced nephrotoxicity and identified inhibited ferroptosis and Camkk2 as potential molecular mechanisms.


ß-HB protects against cisplatin-induced renal damage both in vivo and in vitro.Moreover, ß-HB is effective in attenuating cisplatin-induced lipid peroxidation and ferroptosis.The regulation of energy metabolism, as well as the treatment involving ß-HB, is associated with Camkk2.


Assuntos
Ácido 3-Hidroxibutírico , Injúria Renal Aguda , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina , Cisplatino , Ferroptose , Cisplatino/efeitos adversos , Cisplatino/toxicidade , Animais , Ferroptose/efeitos dos fármacos , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Masculino , Camundongos , Ácido 3-Hidroxibutírico/farmacologia , Modelos Animais de Doenças , Rim/efeitos dos fármacos , Rim/patologia , Rim/metabolismo , Antineoplásicos/toxicidade , Antineoplásicos/efeitos adversos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Nitrogênio da Ureia Sanguínea , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Creatinina/sangue , Humanos
20.
Hear Res ; 447: 109022, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38705005

RESUMO

The disruption of ribbon synapses in the cochlea impairs the transmission of auditory signals from the cochlear sensory receptor cells to the auditory cortex. Although cisplatin-induced loss of ribbon synapses is well-documented, and studies have reported nitration of cochlear proteins after cisplatin treatment, yet the underlying mechanism of cochlear synaptopathy is not fully understood. This study tests the hypothesis that cisplatin treatment alters the abundance of cochlear synaptosomal proteins, and selective targeting of nitrative stress prevents the associated synaptic dysfunction. Auditory brainstem responses of mice treated with cisplatin showed a reduction in amplitude and an increase in latency of wave I, indicating cisplatin-induced synaptic dysfunction. The mass spectrometry analysis of cochlear synaptosomal proteins identified 102 proteins that decreased in abundance and 249 that increased in abundance after cisplatin treatment. Pathway analysis suggested that the dysregulated proteins were involved in calcium binding, calcium ion regulation, synapses, and endocytosis pathways. Inhibition of nitrative stress by co-treatment with MnTBAP, a peroxynitrite scavenger, attenuated cisplatin-induced changes in the abundance of 27 proteins. Furthermore, MnTBAP co-treatment prevented the cisplatin-induced decrease in the amplitude and increase in the latency of wave I. Together, these findings suggest a potential role of oxidative/nitrative stress in cisplatin-induced cochlear synaptic dysfunction.


Assuntos
Cisplatino , Cóclea , Potenciais Evocados Auditivos do Tronco Encefálico , Proteômica , Sinapses , Sinaptossomos , Cisplatino/toxicidade , Cisplatino/farmacologia , Animais , Cóclea/efeitos dos fármacos , Cóclea/metabolismo , Cóclea/patologia , Cóclea/fisiopatologia , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/patologia , Sinaptossomos/metabolismo , Sinaptossomos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Camundongos Endogâmicos CBA , Masculino , Ototoxicidade/metabolismo , Ototoxicidade/fisiopatologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA