Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 574
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
FASEB J ; 38(5): e23550, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38466338

RESUMO

Breast cancer is the most prevalent malignant tumor in women. Adriamycin (ADR) is a primary chemotherapy drug, but resistance limits its effectiveness. Ferroptosis, a newly identified cell death mechanism, involves the transferrin receptor (TFRC), closely linked with tumor cells. This study aimed to explore TFRC and ferroptosis's role in breast cancer drug resistance. Bioinformatics analysis showed that TFRC was significantly downregulated in drug-resistant cell lines, and patients with low TFRC expression might demonstrate a poor chemotherapeutic response to standard treatment. High expression of TFRC was positively correlated with most of the ferroptosis-related driver genes. The research findings indicate that ferroptosis markers were higher in breast cancer tissues than in normal ones. In chemotherapy-sensitive cases, Ferrous ion (Fe2+ ) and malondialdehyde (MDA) levels were higher than in resistant cases (all p < .05). TFRC expression was higher in breast cancer than in normal tissue, especially in the sensitive group (all p < .05). Cytological experiments showed increased hydrogen peroxide (H2 O2 ) after ADR treatment in both sensitive and resistant cells, with varying MDA changes (all p < .05). Elevating TFRC increased Fe2+ and MDA in ADR-resistant cells, enhancing their sensitivity to ADR. However, TFRC upregulation combined with ADR increased proliferation and invasiveness in resistant cell lines (all p < .05). In conclusion, ADR resistance to breast cancer is related to the regulation of iron ion-mediated ferroptosis by TFRC. Upregulation of TFRC in ADR-resistant breast cancer cells activates ferroptosis and reverses ADR chemotherapy resistance of breast cancer.


Assuntos
Neoplasias da Mama , Ferroptose , Feminino , Humanos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Doxorrubicina/farmacologia , Receptores da Transferrina/genética , Transferrina
2.
FASEB J ; 38(15): e23876, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-39120539

RESUMO

Breast cancer is a common malignant tumor in women. Ferroptosis, a programmed cell death pathway, is closely associated with breast cancer and its resistance. The transferrin receptor (TFRC) is a key factor in ferroptosis, playing a crucial role in intracellular iron accumulation and the occurrence of ferroptosis. This study investigates the influence and significance of TFRC and its upstream transcription factor hypoxia-inducible factor-1α (HIF1α) on the efficacy of neoadjuvant therapy in breast cancer. The differential gene obtained from clinical samples through genetic sequencing is TFRC. Bioinformatics analysis revealed that TFRC expression in breast cancer was significantly greater in breast cancer tissues than in normal tissues, but significantly downregulated in Adriamycin (ADR)-resistant tissues. Iron-responsive element-binding protein 2 (IREB2) interacts with TFRC and participates in ferroptosis. HIF1α, an upstream transcription factor, positively regulates TFRC. Experimental results indicated higher levels of ferroptosis markers in breast cancer tissue than in normal tissue. In the TAC neoadjuvant regimen-sensitive group, iron ion (Fe2+) and malondialdehyde (MDA) levels were greater than those in the resistant group (all p < .05). Expression levels of TFRC, IREB2, FTH1, and HIF1α were higher in breast cancer tissue compared to normal tissue. Additionally, the expression of the TFRC protein in the TAC neoadjuvant regimen-sensitive group was significantly higher than that in the resistant group (all p < .05), while the difference in the level of expression of IREB2 and FTH1 between the sensitive and resistant groups was not significant (p > .05). The dual-luciferase assay revealed that HIF1α acts as an upstream transcription factor of TFRC (p < .05). Overexpression of HIF1α in ADR-resistant breast cancer cells increased TFRC, Fe2+, and MDA content. After ADR treatment, the cell survival rate decreased significantly, and ferroptosis could be reversed by the combined application of Fer-1 (all p < .05). In conclusion, ferroptosis and chemotherapy resistance are correlated in breast cancer. TFRC is a key regulatory factor influenced by HIF1α and is associated with chemotherapy resistance. Upregulating HIF1α in resistant cells may reverse resistance by activating ferroptosis through TFRC overexpression.


Assuntos
Neoplasias da Mama , Doxorrubicina , Resistencia a Medicamentos Antineoplásicos , Ferroptose , Subunidade alfa do Fator 1 Induzível por Hipóxia , Receptores da Transferrina , Feminino , Humanos , Pessoa de Meia-Idade , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Quimioterapia Adjuvante/métodos , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Ferroptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Células MCF-7 , Receptores da Transferrina/metabolismo , Receptores da Transferrina/genética , Regulação para Cima
3.
Funct Integr Genomics ; 24(1): 22, 2024 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-38306005

RESUMO

MHY1485 is an mTOR activator that inhibits the autophagy process by inhibiting the fusion between autophagosomes and lysosomes. This study aimed to explore the role and mechanism of MHY1485 in hepatocellular carcinoma (HCC) and to provide an in-depth understanding of the mechanisms of autophagy regulation in relation to adriamycin (ADM) resistance, as well as the development of a molecularly targeted autophagy-modulating approach. Here, ADM was used to treat HepG2 cells and construct an ADM-resistant cell model. The HepG2/ADM cell line and HepG2 cells were treated with MHY1485 and ADM, respectively, and the proliferation and apoptosis of HCC cells were detected using CCK8, clone formation, flow cytometry, and 5-ethynyl-2'-deoxyuridine staining assays. Ki-67, mTOR phosphorylation, and LC3A expression were detected by IF staining; the expression or phosphorylation levels of autophagy-related proteins (i.e., GLUT1, PGI, PFK, END, and MTHFD2) and apoptosis-related proteins (caspase-3, caspase-8, and caspase-9) were detected by qPCR and western blotting. The number of autophagosomes was determined by monodansylcadaverine staining. Our results showed that MHY1485 can inhibit the proliferation and growth of liver cancer cells, and that MHY1485 combined with ADM can effectively inhibit the tolerance of HepG2/ADM cells to ADM and enhance the efficacy of ADM. The results of the detection of the autophagy-related protein LC3A also indicated that MHY1485 activates mTOR and can affect the phosphorylation level of ULK1, inhibit autophagy, and enhance the sensitivity of liver cancer cells to adriamycin. In summary, MHY1485 can enhance the sensitivity of adriamycin-resistant cells to adriamycin by activating mTOR and blocking the autophagy process in cells; therefore, mTOR may become a potential target for the treatment of liver cancer.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Morfolinas , Triazinas , Humanos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Doxorrubicina/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Células Hep G2 , Apoptose , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Autofagia , Proliferação de Células , Linhagem Celular Tumoral
4.
Mol Cell Biochem ; 479(1): 73-83, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36995547

RESUMO

Adriamycin (ADR) causes irreversible damage to the heart, leading to ADR-induced cardiomyopathy (ACM). Angiotensin-(1-9) [Ang-(1-9)] is a peptide from the counter-regulatory renin-angiotensin system, but the effects on ACM is unclear. Our study was aimed to explore the effects and underlying molecular mechanisms of Ang-(1-9) against ACM in Wistar rats. Rats were injected intraperitoneally with ADR via six equal doses (each containing 2.5 mg/kg) within a period of 2 weeks to induce ACM. After 2 weeks of ADR treatment, the rats were treated with Ang-(1-9) (200 ng/kg/min) or angiotensin type 2 receptor (AT2R) antagonist PD123319 (100 ng/kg/min) for 4 weeks. Although Ang-(1-9) treatment did not influence blood pressure, it significantly improved left ventricular function and remodeling in ADR-treated rats, by inhibiting collagen deposition, the expression of TGF-ß1, inflammatory response, cardiomyocyte apoptosis and oxidative stress. Moreover, Ang-(1-9) reduced ERK1/2 and P38 MAPK phosphorylation. The therapeutic effects of Ang-(1-9) were blocked by the AT2R antagonist PD123319, which also offset the down-regulation protein expression of pERK1/2 and pP38 MAPK induced by Ang-(1-9). These data suggest that Ang-(1-9) improved left ventricular function and remodeling in ADR-treated rats by an AT2R/ ERK1/2 and P38 MAPK-dependent mechanism. Thus, the Ang-(1-9)/AT2R axis may provide a novel and promising target to the prevention and treatment of ACM.


Assuntos
Cardiomiopatias , Receptor Tipo 2 de Angiotensina , Ratos , Animais , Receptor Tipo 2 de Angiotensina/metabolismo , Ratos Wistar , Doxorrubicina/efeitos adversos , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/prevenção & controle , Angiotensina II/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno , Receptor Tipo 1 de Angiotensina
5.
Cell Biol Int ; 48(9): 1301-1312, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39021301

RESUMO

Adriamycin (ADR) is widely used against breast cancer, but subsequent resistance always occurs. YAP, a downstream protein of angiomotin (AMOT), importantly contributes to ADR resistance, whereas the mechanism is largely unknown. MCF-7 cells and MDA-MB-231 cells were used to establish ADR-resistant cell. Then, mRNA and protein expressions of AMOT and YAP expressions were determined. After AMOT transfection alone or in combination with YAP, the sensitivity of the cells to ADR were evaluated in vitro by examining cell proliferation, apoptosis, and cell cycle, as well as in vivo by examining tumor growth. Additionally, the expressions of proteins in YAP pathway were determined in AMOT-overexpressing cells. In the ADR-resistant cells, the expression of AMOT was decreased while YAP was increased, respectively, and the nucleus localization of YAP was increased at the same time. After AMOT overexpression, these were inhibited, whereas the cell sensitivity to ADR was enhanced. However, the AMOT-induced changes were significantly suppressed by YAP knockdown. The consistent results in vivo showed that AMOT enhanced the inhibition of ADR on tumor growth, and inhibited YAP signaling, evidenced by decreased levels of YAP, CycD1, and p-ERK. Our data revealed that decreased AMOT contributed to ADR resistance in breast cancer cells, which was importantly negatively mediated YAP. These observations provide a potential therapy against breast cancer with ADR resistance.


Assuntos
Angiomotinas , Apoptose , Neoplasias da Mama , Proliferação de Células , Doxorrubicina , Resistencia a Medicamentos Antineoplásicos , Camundongos Nus , Proteínas de Sinalização YAP , Humanos , Doxorrubicina/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Proliferação de Células/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Células MCF-7 , Proteínas de Sinalização YAP/metabolismo , Animais , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Camundongos Endogâmicos BALB C , Antibióticos Antineoplásicos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos
6.
Mol Biol Rep ; 51(1): 930, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39174728

RESUMO

BACKGROUND: Among the three most used anticancer drugs, cyclophosphamide, Adriamycin, and 5-Fluorouracil (CAF), the most significant outcome is chemobrain, caused by increased oxidative stress, inflammatory insult, and mitochondrial dysfunction. OBJECTIVE: In this study, endogenous antioxidant coenzyme Q10 (CoQ10) was evaluated for its neuroprotective effects in CICI. MATERIALS AND METHODS: The chemobrain was induced in Swiss albino female mice by administering CAF (40 + 4 + 25 mg/kg) intraperitoneal (i.p.) in three cycles (single injection per week) followed by treatment with CoQ10 (40 mg/kg; p.o.) for up to 3 weeks followed by behavioral, biochemical, molecular and histopathological analysis. RESULTS: Treatment with CoQ10 significantly improved cognition by improving exploring time in novel objects recognition test followed by increasing the time spent in the target quadrant in MWM test as compared to CAF-treated animals. Moreover, CoQ10 demonstrated antioxidant properties by reducing the expression of LPO while increasing levels of GSH, SOD, and catalase as compared to CAF-treated animals. While the levels of AChEs were significantly reduced after CoQ10 treatment in CAF-treated animals. In terms of its mechanism, it effectively counteracted the pro-inflammatory substances (TNF-α and IL-1ß) triggered by CAF while also enhancing the levels of anti-inflammatory markers (IL-10 and Nrf2). Moreover, CoQ10 showed mitochondrial enhancers and it improved the level of Complex (I, II, and IV). Besides that, mitochondrial morphological analysis was done by TEM, and neuronal morphology along with quantification analysis was performed by H&E staining using Image J software to confirm the neuroprotective effect of CoQ10 over CAF-induced cognitive impairment. CONCLUSION: This study suggests CoQ10 can protect the mitochondria by imposing antioxidant, and anti-inflammatory properties, which could be a potential therapy for CICI.


Assuntos
Antioxidantes , Estresse Oxidativo , Ubiquinona , Animais , Ubiquinona/análogos & derivados , Ubiquinona/farmacologia , Ubiquinona/uso terapêutico , Camundongos , Feminino , Antioxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Comprometimento Cognitivo Relacionado à Quimioterapia/tratamento farmacológico , Comprometimento Cognitivo Relacionado à Quimioterapia/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Doxorrubicina/efeitos adversos , Fluoruracila/efeitos adversos , Fluoruracila/farmacologia , Modelos Animais de Doenças , Antineoplásicos/farmacologia , Antineoplásicos/efeitos adversos , Ciclofosfamida/efeitos adversos , Ciclofosfamida/farmacologia
7.
J Biochem Mol Toxicol ; 38(10): e23855, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39328005

RESUMO

Adriamycin is an anticancer anthracycline drug that inhibits the progression of topoisomerase II activity and causes apoptosis. The effective clinical application of the drug is very much limited by its adverse drug reactions on various tissues. Most importantly, Adriamycin causes cardiomyopathy, one of the life-threatening complications of the drug. Altered expression of PPARγ in adipocytes inhibited the glucose and fatty acids uptake by down regulating GLUT4 and CD36 expression and causes cardiotoxicity. Therefore, the influence of Adriamycin in cardiac ailments was investigated in vivo and in vitro. Adriamycin treated rats showed altered ECG profile, arrhythmic heartbeat with the elevated levels of CRP and LDH. Dysregulated lipid profiles with elevated levels of cholesterol and triglycerides were also observed. Possibilities of cardiac problems due to cardiomyopathy were analyzed through histopathology. Adriamycin treated rats showed no signs for atheromatous plaque formation in aorta but disorganized cardiomyocytes with myofibrillar loss and inflammation in heart tissue, indicative of cardiomyopathy. Reduced levels of antioxidant enzymes confirmed the incidence of oxidative stress. Adriamycin treatment significantly reduced glucose and insulin levels, creating energy demand due to decreased glucose and insulin levels with increased fatty acid accumulation, ultimately resulting in oxidative stress mediated cardiomyopathy. Since PPARs play a vital role in regulating oxidative stress, the effect of Adriamycin on PPARγ was analyzed by western blot. Adriamycin downregulated PPARγ in a dose-dependent manner in H9C2 cells in vitro. Overall, our study suggests that Adriamycin alters glucose and lipid metabolism via PPARγ inhibition that leads to oxidative stress and cardiomyopathy that necessitates a different therapeutic approach.


Assuntos
Cardiomiopatias , Doxorrubicina , PPAR gama , Animais , Masculino , Ratos , Antibióticos Antineoplásicos/efeitos adversos , Antibióticos Antineoplásicos/toxicidade , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Linhagem Celular , Doxorrubicina/efeitos adversos , Metabolismo Energético/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , PPAR gama/antagonistas & inibidores , PPAR gama/metabolismo , Ratos Wistar
8.
Kidney Blood Press Res ; 49(1): 81-90, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38185119

RESUMO

INTRODUCTION: Sodium-glucose cotransporter 2 (SGLT2) inhibitors target SGLT2 in renal proximal tubules and promote glycosuria in type 2 diabetes mellitus in humans and animal models, resulting in reduced blood glucose levels. Although clinical trials have shown that SGLT2 inhibitors attenuate the progression of chronic kidney disease, there have been concerns regarding SGLT2-induced acute kidney injury. In this study, we investigated the effect of SGLT2 inhibitors on adriamycin-induced kidney injury in mice. METHODS: Seven-week-old balb/c mice were injected with adriamycin 11.5 mg/kg via the tail vein. Additionally, dapagliflozin was administered via gavage for 2 weeks. The mice were divided into five groups: vehicle, dapagliflozin 3 mg/kg, adriamycin, adriamycin plus dapagliflozin 1 mg/kg, and adriamycin plus dapagliflozin 3 mg/kg. RESULTS: Adriamycin injection reduced the body weight and food and water intakes. Dapagliflozin also decreased the body weight and food and water intakes. Fasting blood glucose and urine volume were not altered by either adriamycin or dapagliflozin. Once adriamycin-induced kidney injury had developed, there were no differences in systolic blood pressure among the groups. Dapagliflozin did not alleviate proteinuria in adriamycin-induced kidney injury. Adriamycin induced significant glomerular and interstitial injury, but dapagliflozin did not attenuate these changes in renal injury. Interestingly, SGLT2 expressions were different between the cortex and medulla of kidneys by dapagliflozin treatment. Dapagliflozin increased SGLT2 expression in medulla, not in cortex. CONCLUSION: Dapagliflozin had no effect on proteinuria or inflammatory changes such as glomerular and tubular damages in adriamycin-induced kidney injury. Our study suggests that dapagliflozin does not protect against adriamycin-induced kidney injury. More experimental studies regarding the effects of SGLT2 inhibitors on various kidney diseases are needed to clarify the underlying mechanisms.


Assuntos
Diabetes Mellitus Tipo 2 , Glucosídeos , Insuficiência Renal Crônica , Inibidores do Transportador 2 de Sódio-Glicose , Humanos , Camundongos , Animais , Transportador 2 de Glucose-Sódio/farmacologia , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico , Doxorrubicina , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Rim/metabolismo , Compostos Benzidrílicos/farmacologia , Compostos Benzidrílicos/uso terapêutico , Insuficiência Renal Crônica/metabolismo , Proteinúria/tratamento farmacológico , Peso Corporal , Água/metabolismo
9.
Mol Cell Proteomics ; 21(2): 100187, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34922009

RESUMO

Drug resistance is a critical obstacle to effective treatment in patients with chronic myeloid leukemia. To understand the underlying resistance mechanisms in response to imatinib mesylate (IMA) and adriamycin (ADR), the parental K562 cells were treated with low doses of IMA or ADR for 2 months to generate derivative cells with mild, intermediate, and severe resistance to the drugs as defined by their increasing resistance index. PulseDIA-based (DIA [data-independent acquisition]) quantitative proteomics was then employed to reveal the proteome changes in these resistant cells. In total, 7082 proteins from 98,232 peptides were identified and quantified from the dataset using four DIA software tools including OpenSWATH, Spectronaut, DIA-NN, and EncyclopeDIA. Sirtuin signaling pathway was found to be significantly enriched in both ADR-resistant and IMA-resistant K562 cells. In particular, isocitrate dehydrogenase (NADP(+)) 2 was identified as a potential drug target correlated with the drug resistance phenotype, and its inhibition by the antagonist AGI-6780 reversed the acquired resistance in K562 cells to either ADR or IMA. Together, our study has implicated isocitrate dehydrogenase (NADP(+)) 2 as a potential target that can be therapeutically leveraged to alleviate the drug resistance in K562 cells when treated with IMA and ADR.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva , Proteômica , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Humanos , Mesilato de Imatinib/farmacologia , Mesilato de Imatinib/uso terapêutico , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo
10.
Int Endod J ; 57(2): 195-207, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38071432

RESUMO

AIM: This study aimed to investigate the potential protective effects of N-acetyl-L-cysteine (NAC) against apical periodontitis (AP) in rats with adriamycin (ADR)-induced kidney and heart diseases. METHODOLOGY: Fourty-eight Wistar albino rats were divided into six groups: (1) Control group, (2) ADR group (1 mg/kg/day ip for 10 days), (3) AP Group (1st mandibular molar tooth), (4) AP + ADR Group, (5) AP + NAC group (150 mg/kg/day ip), and (6) AP + ADR + NAC group. After 3 weeks, the rats were decapitated and blood and tissue samples (heart, kidney, and jaw) were collected. Tissue samples were evaluated by biochemical (inflammatory cytokines and hemodynamic parameters) and radiological analyses. One-way anova with Tukey post hoc tests was used to compare data, considering p < .05 as statistically significant. RESULTS: The serum levels of TNF-α, IL-1ß, BUN, Creatinine, CK, and LDH were elevated in the test groups compared with the control group, and treatment with NAC reduced these levels (p < .05). Heart and kidney tissue analysis showed a higher heart-to-body weight ratio (HW/BW) and kidney-to-body weight ratio (KW/BW) in the test groups compared with the control group (p < .05). No significant differences in HW/BW and KW/BW were found between the control and AP + NAC groups. Volumetric apical bone resorption analysis showed an increase in periapical radiolucencies in AP-induced groups indicating apical periodontitis. NAC treatment reduced the total area and volume of resorption cavities (p < .05). CONCLUSIONS: The results suggest that NAC's antioxidant and anti-inflammatory effects can reduce adriamycin-mediated heart and kidney damage and may have a positive effect on apical periodontitis in individuals with nephropathy and cardiomyopathy.


Assuntos
Cardiomiopatias , Periodontite Periapical , Ratos , Animais , Ratos Wistar , Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Doxorrubicina , Periodontite Periapical/induzido quimicamente , Periodontite Periapical/tratamento farmacológico , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/tratamento farmacológico , Peso Corporal
11.
Int J Mol Sci ; 25(11)2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38891998

RESUMO

Approximately 30% of steroid-resistant nephrotic syndromes are attributed to monogenic disorders that involve 27 genes. Mutations in KANK family members have also been linked to nephrotic syndrome; however, the precise mechanism remains elusive. To investigate this, podocyte-specific Kank1 knockout mice were generated to examine phenotypic changes. In the initial assessment under normal conditions, Kank1 knockout mice showed no significant differences in the urinary albumin-creatinine ratio, blood urea nitrogen, serum creatinine levels, or histological features compared to controls. However, following kidney injury with adriamycin, podocyte-specific Kank1 knockout mice exhibited a significantly higher albumin-creatinine ratio and a significantly greater sclerotic index than control mice. Electron microscopy revealed more extensive foot process effacement in the knockout mice than in control mice. In addition, KANK1-deficient human podocytes showed increased detachment and apoptosis following adriamycin exposure. These findings suggest that KANK1 may play a protective role in mitigating podocyte damage under pathological conditions.


Assuntos
Proteínas do Citoesqueleto , Doxorrubicina , Camundongos Knockout , Podócitos , Animais , Humanos , Masculino , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose , Proteínas do Citoesqueleto/metabolismo , Proteínas do Citoesqueleto/genética , Síndrome Nefrótica/metabolismo , Síndrome Nefrótica/genética , Síndrome Nefrótica/patologia , Podócitos/metabolismo , Podócitos/patologia , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética
12.
Rev Invest Clin ; 76(1): 6-17, 2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-38253021

RESUMO

Background: Adriamycin resistance remains an obstacle to gastric cancer chemotherapy treatment. Objective: The objective of this study was to study the role and mechanism of transcription factor E2F7 in sensitivity to ADM chemotherapeutic agents in gastric cancer. Methods: Cell viability and cell sensitivity were assessed by CCK-8 and IC50 values of ADM were calculated. The impact of ADM on cellular proliferative capacity was assessed through colony formation assay. The binding relationship between E2F7 and PKMYT1 was then verified by dual luciferase assay and chromatin immunoprecipitation assay. ERK1/ERK2 and p-ERK1/p-ERK2 protein expression levels were detected by western blot. Results: In both gastric cancer tissue and ADM-resistant cells, a conspicuous upregulation of E2F7 and PKMYT1 was observed. Upregulated PKMYT1 was notably enriched in the MAPK signaling pathway. Enhanced levels of E2F7 were shown to not only drive gastric cancer cell proliferation but also engender a reduction in the sensitivity of these cells to ADM. Furthermore, PKMYT1 emerged as a downstream target of E2F7. Activation of E2F7 culminated in the transcriptional upregulation of PKMYT1, and silencing E2F7 reversed the inhibitory impact of PKMYT1 overexpression on ADM sensitivity in gastric cancer cells. Conclusion: E2F7/PKMYT1 axis might promote the proliferation and partially inhibit ADM sensitivity of gastric cancer cells by activating the MAPK pathway.


Assuntos
MicroRNAs , Neoplasias Gástricas , Humanos , Doxorrubicina/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Fatores de Transcrição/metabolismo , Linhagem Celular Tumoral , Transdução de Sinais , MicroRNAs/metabolismo , Regulação Neoplásica da Expressão Gênica , Fator de Transcrição E2F7/genética , Fator de Transcrição E2F7/metabolismo , Proteínas de Membrana/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo
13.
Arch Biochem Biophys ; 736: 109523, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36682704

RESUMO

OBJECTIVE: Acute myeloid leukemia (AML) remains a common hematopoietic malignancy, and drug resistance greatly blunts the efficacy of chemotherapy in AML treatment. Adriamycin (ADM, also called doxorubicin), is one of the most widely used chemotherapeutics for treating cancers. Herein, we studied the molecular mechanisms underlying microRNA-188-5p (miR-188-5p)-mediated ADM resistance in AML. METHODS: Differentially expressed miRNAs were screened in normal and malignant hematopoietic cells by bioinformatics tools. MiR-188-5p expression in primary bone marrow CD34+ cells and AML cells was evaluated. AML/ADM cells were established using THP-1 and Kasumi-1 cells. The effect of miR-188-5p on the drug resistance in AML/ADM cells was examined by delivery of miR-188-5p-inhibitor. The binding relationship between TET1 and miR-188-5p was analyzed by ChIP, and the downstream target of miR-188-5p was predicted by bioinformatics analysis and validated by dual-luciferase assay. Finally, rescue experiments were carried out in vitro and in vivo. RESULTS: miR-188-5p was highly expressed in AML cells, and miR-188-5p-inhibitor sensitized the AML/ADM cells to ADM. Inhibition of TET1 reduced miR-188-5p promoter hydroxymethylation and downregulated miR-188-5p. miR-188-5p bound to the 3'UTR of PTEN to inhibit PTEN expression, and the PI3K/AKT signaling was activated upon inhibition of PTEN. Suppression of PTEN conferred resistance again to AML/ADM cells in the presence of miR-188-5p inhibitor. CONCLUSION: TET1 elevates miR-188-5p expression by promoting miR-188-5p promoter hydroxymethylation, and miR-188-5p inhibits PTEN expression to induce PI3K/AKT signaling pathway activation, leading to ADM resistance in AML.


Assuntos
Leucemia Mieloide Aguda , MicroRNAs , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , MicroRNAs/genética , MicroRNAs/metabolismo , Leucemia Mieloide Aguda/genética , Doxorrubicina/farmacologia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proliferação de Células , Oxigenases de Função Mista/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo
14.
Cell Biol Toxicol ; 39(4): 1735-1752, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36576707

RESUMO

Adriamycin is widely used as a chemotherapeutic strategy for advanced hepatocellular carcinoma (HCC). However, the clinical response was disappointing because of the acquired drug resistance with long-term usage. Revealing the underlying mechanism could provide promising therapeutics for the drug-resistant patients. The recently identified linc-ROR (long intergenic non-protein-coding RNA, regulator of reprogramming) has been found to be an oncogene in various cancers, and it also demonstrated to mediate drug resistance and metastasis. We thereby wonder whether this lincRNA could mediate adriamycin chemoresistance in HCC. In this study, linc-ROR was found to be upregulated in adriamycin-resistant HCC cells. And its overexpression accelerated epithelial-mesenchymal transition (EMT) program and adriamycin resistance. Conversely, its silence suppressed EMT and made HCC cells sensitize to adriamycin in vitro and in vivo. Further investigation revealed that linc-ROR physically interacted with AP-2α, mediated its stability by a post-translational modification manner, and sequentially activated Wnt/ß-catenin pathway. Furthermore, linc-ROR expression was positively associated with ß-catenin expression in human clinical specimens. Taken together, linc-ROR promoted tumorigenesis and adriamycin resistance in HCC via a linc-ROR/AP-2α/Wnt/ß-catenin axis, which could be developed as a potential therapeutic target for the adriamycin-resistant patients.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , beta Catenina/genética , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Doxorrubicina/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , RNA Longo não Codificante/genética
15.
J Fluoresc ; 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37831353

RESUMO

In this work, a facile and fast aqueous-phase synthetic method is proposed to prepare water-soluble ZnS quantum dots stabilized simultaneously with glutathione and L-cysteine (ZnS QDs-GSH/L-Cys). As-synthesized ZnS QDs-GSH/L-Cys were monodispersed spherical nanocrystals with a mean diameter of 5.0 ± 0.7 nm. Besides, the obtained ZnS QDs-GSH/L-Cys emitted more intensive blue fluorescence and exhibited an improved stability in aqueous solution compared with ZnS quantum dots merely stabilized with GSH (ZnS QDs-GSH). Interestingly, Adriamycin, a representative anticancer drug, was added into the solution of ZnS QDs-GSH/L-Cys, the blue fluorescence of ZnS QDs-GSH/L-Cys was greatly enhanced instead of being quenched, which indicated that ZnS QDs-GSH/L-Cys can be used as an enhanced-fluorescence nanoprobe for determining Adriamycin. The observed fluorescent enhancement could be attributed to the blocking of photoinduced electron transfer (PET) in ZnS QDs-GSH/L-Cys due to the electrostatic interaction between the -COO- groups on the surface of quantum dots and the -NH3+ groups in Adriamycin, followed by the coordination interaction among ZnS QDs-GSH/L-Cys and Adriamycin. The fluorescence intensity of ZnS QDs-GSH/L-Cys presented a good linear response with the concentration of Adriamycin ranging from 2.0 to 20 µg•mL-1. The proposed fluorescent nanoprobe exhibited an excellent sensitivity with the LOD of 0.1 µg•mL-1 and a good accuracy for detecting Adriamycin.

16.
Acta Pharmacol Sin ; 44(3): 573-583, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36056082

RESUMO

Long-term treatment with adriamycin (ADR) is associated with higher incidences of cumulative cardiotoxicity manifest as heart failure. ADR-induced cardiomyopathy is characterized by extensive fibrosis that is caused by cardiac fibroblast activation. To date, however, no specific treatment is available to alleviate ADR-induced cardiotoxicity. Protein arginine methyltransferase 5 (PRMT5), a major enzyme responsible for methylation of arginine, regulates numerous cellular processes such as cell differentiation. In the present study we investigated the role of PRMT5 in cardiac fibrosis. Mice were administered ADR (3 mg/kg, i.p., every 2 days) for 2 weeks. We showed that aberrant PRMT5 expression was largely co-localized with α-SMA-positive activated cardiac fibroblasts in ADR-injected mice and in ADR-treated cardiac fibroblasts in vitro. PRMT5-overexpression exacerbated, whereas PRMT5 knockdown alleviated ADR-induced cardiac fibrosis in vivo and TGF-ß1-induced cardiac fibroblast activation in vitro. We demonstrated that PRMT5-overexpression enhanced methylated-Smad3 levels in vivo and in vitro. Pretreatment with a specific PRMT5 inhibitor EPZ015666 (5 nM) or overexpression of a catalytically inactive mutant of PRMT5, PRMT5(E444Q), reduced PRMT5-induced methylation of Smad3, thus suppressing PRMT5-mediated cardiac fibroblast activation in vitro. Furthermore, ADR activated cardiac fibroblasts was depending on autocrine TGF-ß1. Taken together, our results demonstrate that PRMT5 promotes ADR-induced cardiac fibrosis via activating cardiac fibroblasts, suggesting that it may be a potential therapeutic target of ADR-caused cardiotoxicity.


Assuntos
Cardiomiopatias , Fator de Crescimento Transformador beta1 , Camundongos , Animais , Fator de Crescimento Transformador beta1/metabolismo , Doxorrubicina , Cardiotoxicidade/metabolismo , Fibrose , Fibroblastos/metabolismo , Cardiomiopatias/patologia , Proteína Smad3/metabolismo
17.
Skin Res Technol ; 29(1): e13259, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36704890

RESUMO

BACKGROUND: Targeting CD20+ melanoma cancer stem cells (CSCs) subset is essential for treating melanoma. Anti-CD20 aptamer-modified exosomes (ACEXO) loaded with Adriamycin could be a therapeutic strategy for targeting CSCs. MATERIALS AND METHODS: Exosomes loaded with Adriamycin were modified with anti-CD20 aptamer and characterized by size and molecular markers using transmission electron microscope and dynamic light scattering. The uptake of ACEXO into CD20+ cells was checked, and its cytotoxicities in CD20+ melanoma cells, HEK 293T, and 3T3 cells were evaluated. At the same time, the in vivo distribution of ACEXO in the tumor-bearing mice model was determined. RESULTS: The particle size of the exosome is about 80-100 nm. Western blot analysis showed that they expressed the characteristic exosome markers: CD9 and CD63. Quantitative analysis of the mean fluorescence intensity after 4 h incubation showed that ACEXO significantly improved Adriamycin uptake. Notably, the ACEXO killed only CD20+ melanoma cells. In addition, they exhibited good biocompatibility with both 293T and 3T3 cells at all doses. After intravenous injection, exosome distribution data showed that ACEXO's accumulation in the tumor is higher than anti-CD20-modified exosomes (AEXO)'s at all time points, and the accumulation increased as time prolonged. Addition of ACEXO reduces the number of tumorspheres in A375 or WM266-4 cells compared to untreated controls or AEXO-treated group. More important, while treating melanoma tumor-bearing mice, ACEXO-treated group showed the lowest tumor weight without body weight loss. CONCLUSION: ACEXO loaded with Adriamycin could suppress tumor cell growth in vitro and in vivo, probably by targeting CD20+ melanoma CSCs.


Assuntos
Exossomos , Melanoma , Humanos , Animais , Camundongos , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Exossomos/patologia , Melanoma/tratamento farmacológico , Melanoma/patologia , Células-Tronco/patologia
18.
Ecotoxicol Environ Saf ; 256: 114778, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36989556

RESUMO

Adriamycin (ADR), one of the most effective broad-spectrum antitumor chemotherapeutic agents in clinical practice, is used to treat solid tumors as well as hematological malignancies in adults and children. However, long-term ADR use causes several adverse reactions, including time- and dose-dependent cardiotoxicity, which limit its clinical application. In addition, the mechanism by which ADR induces cardiotoxicity remains unclear. Therefore, we used zebrafish as animal models to evaluate ADR toxicity during embryonic heart development owing to the similarity of this process in zebrafish to that in humans. Exposure of zebrafish embryos to 1.25, 2.5, and 5 mg/L ADR induced abnormal embryonic development, with the occurrence of cardiac malformations, pericardial edema, decreased movement speed and activity, and increased distance between the venous sinus and the arterial bulb (SV-BA). ADR exposure induced dysregulated cardiogenesis during the precardiac mesoderm formation period. We also observed irregular expression of cardiac-related genes, an upregulation of apoptotic gene expression, and a dose-dependent increase in oxidative stress levels. Furthermore, oxidative stress-induced apoptosis exerted deleterious effects on cardiac development in zebrafish embryos, and treatment with astaxanthin (ATX) alleviated these heart defects. ADR- and Wnt pathway-related genes exhibited good energy and spatial matching, and ADR upregulated the Wnt signaling pathway in zebrafish. Moreover, IWR-1 effectively alleviated ADR-induced heart defects. In conclusion, we demonstrated that the toxic effects of ADR on cardiac development in zebrafish embryos could provide a theoretical basis for explaining the pathogenesis of ADR-induced cardiotoxicity, which occurs through the upregulation of oxidative stress and Wnt signaling pathway, as well as its prevention and treatment in humans. These findings will help develop effective treatment strategies to combat ADR-induced cardiotoxicity and broaden the application of ADR for clinical practice.


Assuntos
Cardiotoxicidade , Cardiopatias Congênitas , Animais , Criança , Humanos , Cardiotoxicidade/metabolismo , Peixe-Zebra/metabolismo , Doxorrubicina/toxicidade , Simulação de Acoplamento Molecular , Coração , Estresse Oxidativo , Embrião não Mamífero
19.
Drug Chem Toxicol ; 46(2): 219-225, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34965830

RESUMO

Male reproductive toxicity is a well-established side effect of the chemotherapeutic drug adriamycin (ADR). Sildenafil (SIL) is a phosphodiesterase inhibitor known to enhance the chemosensitivity of cancer cells to ADR. However, there is a scarcity of information on the effect of SIL on ADR-induced testicular toxicity. In this study, SIL (5, 10, or 20 mg/kg/day) was administered to male rats for 7 days, followed by a single intraperitoneal injection of ADR (20 mg/kg) on day 7. Control rats received either ADR, SIL, or normal saline for 7 days. Epididymal sperm were collected from the testes to assess the effects on sperm quality, quantity, and serum testosterone concentration was also determined. ADR treatment caused a decrease in sperm motility and elevated the percentage of sperms with tail defects which worsened in combination with SIL (20 mg/kg). Furthermore, ADR alone or in combination with SIL dose-dependently increased total sperm abnormalities. SIL (20 mg/kg) plus ADR also decreased sperm count and lowered testosterone level compared to ADR-only rats. In conclusion, exposure of rats to SIL before ADR treatment has the potential to worsen ADR-induced testicular toxicity.


Assuntos
Doxorrubicina , Testículo , Ratos , Masculino , Animais , Doxorrubicina/toxicidade , Citrato de Sildenafila/toxicidade , Motilidade dos Espermatozoides , Sêmen , Testosterona
20.
Zhongguo Zhong Yao Za Zhi ; 48(3): 744-751, 2023 Feb.
Artigo em Zh | MEDLINE | ID: mdl-36872238

RESUMO

This study analyzes the impact of echinacoside(ECH) in the proliferation, metastasis and adriamycin(ADR) resistance of breast cancer(BC) MCF-7 cells via the modulation of aldo-keto reductase family 1 member 10(AKR1B10)/extracellular signal-regulated kinase(ERK) pathway. The chemical structure of ECH was firstly confirmed. MCF-7 cells were treated with different concentration(0, 10, 20, 40 µg·mL~(-1)) of ECH for 48 h. Western blot was used to analyze expression of AKR1B10/ERK pathway-associated proteins and cell counting kit-8(CCK-8) assay to determine cell viability. MCF-7 cells were collected and classified into control group, ECH group, ECH + Ov-NC group, and ECH + Ov-AKR1B10 group. Then Western blot was employed to analyze the expression of AKR1B10/ERK pathway-associated proteins. CCK-8 and 5-ethynyl-2'-deoxyuridine(EdU) assay were used to examine cell proliferation. Cell migration was appraised with scratch assay, Transwell assay, and Western blot. Eventually, MCF-7 cells were treated with ADR for 48 h to induce ADR resistance. Cell viability was tested by CCK-8 assay and cell apoptosis was estimated based on terminal-deoxynucleoitidyl transferase mediated nick end labeling(TUNEL) assay and Western blot. Based on Protein Data Bank(PDB) and molecular docking, the binding affinity of ECH to AKR1B10 was assessed. Various doses of ECH decreased the expression of AKR1B10/ERK pathway-associated proteins in a dose-dependent manner and declined cell viability compared with the control group. Compared with the control group, 40 µg·mL~(-1) ECH blocked the AKR1B10/ERK pathway in MCF-7 cells and inhibited the proliferation, metastasis and ADR resistance of the cells. Compared with the ECH + Ov-NC group, ECH + Ov-AKR1B10 group showed the recovery of some biological behaviors of MCF-7 cells. ECH also targeted AKR1B10. ECH can inhibit the proliferation, metastasis, and ADR resistance of BC cells by blocking AKR1B10/ERK pathway.


Assuntos
Neoplasias , Humanos , Células MCF-7 , Simulação de Acoplamento Molecular , Transdução de Sinais , Aldo-Ceto Redutases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA