Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros











Intervalo de año de publicación
1.
Rep Biochem Mol Biol ; 12(4): 575-585, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39086586

RESUMEN

Background: Doxorubicin, a commonly utilized anthracycline antibiotic and chemotherapeutic agent, has been associated with hepatotoxicity as an adverse effect. This study aimed to evaluate protective effects of zingerone, a bioactive compound derived from ginger renowned for its antioxidative attributes, on oxidative stress in doxorubicin-induced rat hepatotoxicity. Methods: In this experimental study, a total of 48 male Wistar rats were allocated into six distinct groups. The first group received a control treatment of normal saline. The second group was administered an intraperitoneal dose of 20 mg/kg of doxorubicin on day 5. The third group received an oral dose of 40 mg/kg of zingerone for 8 days. The fourth, fifth, and sixth groups were administered zingerone at doses of 10, 20, and 40 mg/kg, respectively, for the same 8-day period. On day 5, all groups, except the control group, received an intraperitoneal injection of doxorubicin. Following a 72-hour interval, the animals were anesthetized, and blood samples were collected to assess serum factors. Moreover, portions of the liver tissue were subjected to histopathological analysis and assessment of oxidative stress parameters. Results: The activity levels of serum enzymes, including aspartate transaminase (AST), alanine transaminase (ALT), and liver malondialdehyde (MDA), increased in the doxorubicin group. Conversely, the levels of other parameters such as glutathione peroxidase (GPX), superoxide dismutase (SOD), and glutathione (GSH) decreased. However, the co-administration of zingerone effectively reversed these levels, restoring them back to normal. Conclusions: These findings suggest that zingerone, particularly at a high dose, exhibit a hepatoprotective effect in the doxorubicin-induced hepatotoxicity model.

2.
Artículo en Inglés | MEDLINE | ID: mdl-38985313

RESUMEN

Epicatechin (Epi) is one of the most abundant flavonoids present in different fruits and tea leaves. Emerging research illuminates the promising potential of catechins to serve as a shield against the damaging effects of arsenic (As) exposure in diverse organs.This study sought to discern whether Epi exhibits a therapeutic efficacy against arsenic-induced neurotoxicity in a murine model.The Naval Medical Research Institute (NMRI) mice were randomly partitioned into six distinct groups, which included a control group receiving normal saline, a group receiving a daily oral dose of arsenic (10 mg/kg) for 5 weeks, groups receiving As (10 mg/kg/day) orally for 5 weeks along with different doses of Epi (25-100 mg/kg) orally for the last 2 weeks, and a group receiving Epi (100 mg/kg) orally for 2 weeks. To assess the potential effects of Epi, neurobehavioral tests, various parameters of oxidative stress, and inflammation were evaluated.The findings of this investigation revealed that As-induced neurobehavioral toxicity was associated with a notable surge in lipid peroxidation and nitric oxide (NO) concentration, accompanied by a reduction in the levels of antioxidant markers. As heightened pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α) levels were observed alongside amplified nuclear factor kappa B (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2) expression. However, treatment with Epi reversed these effects.On the whole, these findings indicate that Epi may hold promise therapeutic efficacy on As-induced neurotoxicity by improving antioxidant status and mitigating oxidative stress and inflammation. Nevertheless, further research is imperative to comprehensively grasp the potential protective effects of Epi in this particular context.

3.
Artículo en Inglés | MEDLINE | ID: mdl-38953971

RESUMEN

Cardiotoxicity is one of the side effects of the anti-cancer drug doxorubicin (DOX) that limits its clinical application. Betaine (BT) is a natural agent with promising useful effects against inflammation and oxidative stress (OS). We assessed the effects of BT on DOX-induced cardiotoxicity in mice. Forty-two male NMRI mice were assigned to six groups: I: control; II: BT (200 mg/kg; orally, alone); III: DOX (2.5 mg/kg; six injections (ip)) for two weeks; IV, V, VI: BT (50 mg/kg, 100 mg/kg, and 200 mg/kg; orally, once a day for two weeks, respectively) plus DOX administration. The cardiac enzymes like cardiac troponin-I (cTn-I), lactate dehydrogenase (LDH), and creatine kinase-MB (CK-MB) were assessed in serum. Oxidative/inflammatory markers like nitric oxide (NO), malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), reduced glutathione level (GSH), and glutathione peroxidase (GPx) activities were determined in cardiac tissue. The expressions of NOD-like receptor protein 3 (NLRP3), caspase-1, interleukin (IL)-1ß, and silent information regulator 1 (SIRT1) proteins were also evaluated in cardiac tissue. The results indicated that DOX significantly increased LDH, CK-MB, cTn-I, MDA, and NO levels and also the caspase-1, NLRP3, and IL-1ß expression. Furthermore, DOX caused a significant reduction in the GSH levels and SOD, CAT, GPX activities, and the expression of SIRT1 protein in heart tissue. However, BT significantly improved all studied parameters. The findings were confirmed by histopathological assessments of the heart. BT can protect against DOX-induced cardiotoxicity by suppressing the activation of NLRP3 and OS by stimulating the SIRT1 pathway.

4.
Artículo en Inglés | MEDLINE | ID: mdl-38976045

RESUMEN

The main objective of this study was to investigate the potential efficacy of carvacrol (CAR) in mitigating bleomycin (BLM)-induced pulmonary fibrosis (PF). Sixty-six male Wistar rats were assigned into two main groups of 7 and 21 days. They were divided into the subgroups of control, BLM, CAR 80 (only for the 21-day group), and CAR treatment groups. The CAR treatment groups received CAR (20, 40, and 80 mg/kg, orally) for 7 or 21 days after an instillation of BLM (5 mg/kg, intratracheally). Results indicated that BLM significantly increased total cell count in bronchoalveolar lavage fluid and the percentages of neutrophils and lymphocytes, and reduced the percentage of macrophages. CAR dose-dependently decreased total cell count and the percentage of neutrophils and lymphocytes. CAR significantly reduced thiobarbituric acid reactive substances and hydroxyproline levels and elevated the total thiol level and catalase, superoxide dismutase, and glutathione peroxidase activities in BLM-exposed rats. Furthermore, CAR decreased the transforming growth factor-ß1, connective transforming growth factor, and tumor necrosis factor-α on days 7 and 21. BLM increased interferon-γ on day 7 but decreased its level on day 21. However, CAR reversed interferon-γ levels on days 7 and 21. Based on histopathological findings, BLM induced inflammation on days 7 and 21, but for induction of fibrosis, 21-day study showed more fibrotic injuries than the 7-day group. CAR showed the improvement of fibrotic injuries. The effect of CAR against BLM-induced pulmonary fibrosis is possibly due to its antioxidant, anti-inflammatory, and antifibrotic activity.

5.
Arch Biochem Biophys ; 758: 110076, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38942108

RESUMEN

Arsenic, an environmental pollutant and poisonous metalloid, has adverse effects on different body organs, including the kidneys. Betaine is a natural nutrient that has many beneficial health effects. This research was conducted to examine the impact of betaine on nephrotoxicity caused by inorganic arsenic (NaAsO2) in mice. Mice were separated into following groups: control, NaAsO2 (50 ppm), NaAsO2 (50 ppm) + betaine (500 mg/kg), and betaine (500 mg/kg). Mice were received NaAsO2 via drinking water for 8 consecutive weeks and betaine was given to the animals via gavage once daily in the 7th and 8th weeks of the study. Upon completion of the study, the mice were euthanized and samples of serum and kidney were obtained for further evaluations. Administration of NaAsO2 increased the levels of blood urea nitrogen and creatinine in the serum. It enhanced the amounts of renal malondialdehyde and decreased the total thiol levels, as well as the activity of antioxidant enzymes (catalase, superoxide dismutase, and glutathione peroxidase). Furthermore, it enhanced the levels of renal inflammatory indicators (tumor necrosis factor-alpha and nitric oxide). Western blot results exhibited an increase in the protein expression of nuclear factor kappa B (NF-κB), and phosphorylated NF-κB in NaAsO2-treated mice. Histopathological results also confirmed kidney damage caused by NaAsO2. However, treatment with betaine improved NaAsO2-related kidney injuries in mice. The results of this work indicated that betaine can attenuate kidney damage caused by NaAsO2 by inhibiting oxidative stress and inflammation.


Asunto(s)
Betaína , Inflamación , Riñón , Estrés Oxidativo , Animales , Betaína/farmacología , Estrés Oxidativo/efectos de los fármacos , Ratones , Riñón/efectos de los fármacos , Riñón/patología , Riñón/metabolismo , Masculino , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Arsénico/toxicidad , FN-kappa B/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo
6.
Cardiovasc Toxicol ; 24(6): 539-549, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38703273

RESUMEN

NaAsO2 is known as a harmful pollutant all over the world, and many chronic heart diseases can be attributed to its prolonged exposure in NaAsO2-contaminated water. Therefore, considering the anti-inflammatory and antioxidant effects of betaine (BET), in this study, our team investigated the cardioprotective effects of this phytochemical agent on sodium arsenite (NaAsO2)-induced cardiotoxicity. Forty male mice were randomly divided into 4 groups: (I) Control; (II) BET (500 mg/kg); (III) NaAsO2 (50 ppm); and (IV) NaAsO2 + BET. NaAsO2 was given to the animals for 8 weeks, but BET was given in the last two weeks. After decapitation, inflammatory factors and biochemical parameters were measured, and Western blot analyses were performed. BET decrease the activity level of alanine aspartate aminotransferase, creatine kinase MB, thiobarbituric acid reactive substances level, inflammatory factors (tumor necrosis factor-α) content, and nuclear factor kappa B expression. Furthermore, BET increased cardiac total thiol and activity levels of catalase, superoxide dismutase, and glutathione peroxidase and nuclear factor erythroid-2 expression. Hence, the administration of BET ameliorated the deleterious effects stemming from the imbalance of oxidative and antioxidant pathways and histopathological alterations observed in NaAsO2-intoxicated mice, thereby attenuating oxidative stress-induced damage and inflammation.


Asunto(s)
Antiinflamatorios , Antioxidantes , Arsenitos , Betaína , Cardiotoxicidad , Modelos Animales de Enfermedad , Cardiopatías , Mediadores de Inflamación , Estrés Oxidativo , Transducción de Señal , Compuestos de Sodio , Animales , Arsenitos/toxicidad , Compuestos de Sodio/toxicidad , Masculino , Antioxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Antiinflamatorios/farmacología , Ratones , Betaína/farmacología , Cardiopatías/prevención & control , Cardiopatías/inducido químicamente , Cardiopatías/patología , Cardiopatías/metabolismo , Mediadores de Inflamación/metabolismo , Transducción de Señal/efectos de los fármacos , Biomarcadores/metabolismo , Biomarcadores/sangre , Citoprotección , Miocardio/patología , Miocardio/metabolismo
7.
Int J Biol Macromol ; 271(Pt 1): 132670, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38806083

RESUMEN

This study focuses on creating new forms of biomimetic nanofiber composites by combining copolymerizing and electrospinning approaches in the field of nanomedicine. The process involved utilizing the melt polymerization of proline (Pr) and hydroxyl proline (Hyp) to synthesize polymers based on Pr (PPE) and Hyp (PHPE). These polymers were then used in a grafting copolymerization process with chitosan (CS) to produce PHPC (1560 ± 81.08 KDa). A novel electrospun nanofiber scaffold was then produced using PHPC and/or CS, hyaluronic acid, polyvinyl alcohol, and naringenin (NR) as a loading drug. Finally, Mouse Dermal Fibroblast (MDF) cells were introduced to the wound dressing and assessed their therapeutic potential for wound healing in rats. The scaffolds were characterized by FTIR, NMR, DSC, and SEM analysis, which confirmed the amino acid grafting, loading drug, and porous and nanofibrous structures (>225 nm). The results showed that the PHPC-based scaffolds were more effective for swelling/absorption of wound secretions, had more elasticity/elongation, faster drug release, more MDF-cytocompatibility, and antibacterial activity against multidrug-resistant S. aureus compared to CS-based scaffolds. The in vivo studies showed that NR in combination with MDF can accelerate cell migration/proliferation, and remodeling phases of wound healing in both PHPC/CS-based scaffolds. Moreover, PHPC-based scaffolds promote collagen content, and better wound contraction, epithelialization, and neovascularization than CS-based, showing potential as wound-dressing.


Asunto(s)
Quitosano , Citrus , Flavonoides , Nanofibras , Cicatrización de Heridas , Quitosano/química , Quitosano/farmacología , Cicatrización de Heridas/efectos de los fármacos , Animales , Citrus/química , Ratas , Nanofibras/química , Ratones , Flavonoides/farmacología , Flavonoides/química , Flavonoides/administración & dosificación , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/administración & dosificación , Sistemas de Liberación de Medicamentos , Staphylococcus aureus/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Piel/efectos de los fármacos , Liberación de Fármacos , Masculino , Portadores de Fármacos/química , Flavanonas/farmacología , Flavanonas/química , Flavanonas/administración & dosificación
8.
Biomed Pharmacother ; 175: 116797, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38776675

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda , Caspasa 1 , Caspasa 3 , Cisplatino , Cetotifen , FN-kappa B , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal , Proteína X Asociada a bcl-2 , Animales , Cisplatino/toxicidad , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal/efectos de los fármacos , Ratones , FN-kappa B/metabolismo , Caspasa 1/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Caspasa 3/metabolismo , Humanos , Cetotifen/farmacología , Proteína X Asociada a bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Apoptosis/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/patología , Riñón/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Células HeLa , Estrés Oxidativo/efectos de los fármacos
9.
JBRA Assist Reprod ; 28(3): 464-470, 2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-38801315

RESUMEN

OBJECTIVE: Methotrexate (MTX) is widely administered for the treatment of various cancers. However, MTX induces male reproductive toxicity. In the current study, the effect of ozone therapy (OT) on reducing the toxic effects of MTX in the mouse testicles has been investigated. METHODS: Twenty-four mice were divided into four groups: control, OT (4 mg/kg ozone), MTX (20 mg/kg), and MTX + OT. Testosterone levels, histological changes, and oxidative stress biomarkers were assessed to evaluate the protective effects of OT. RESULTS: The results demonstrated that MTX disrupted germinal epithelium, reduced serum testosterone levels, and enhanced oxidative stress in testicular tissue. However, treatment with OT attenuated these adverse effects. OT effectively restored the levels of antioxidant enzymes, such as catalase (CAT), glutathione (GSH), and superoxide dismutase (SOD). OT reduced lipid peroxidation, as indicated by decreased malondialdehyde (MDA) levels. OT preserved normal spermatogenesis, improved morphometric parameters, and reduced histological changes by MTX. Moreover, OT effectively restored testosterone levels. CONCLUSIONS: OT protects against MTX-induced testicular damage by suppressing oxidative stress.


Asunto(s)
Metotrexato , Estrés Oxidativo , Ozono , Testículo , Testosterona , Animales , Masculino , Metotrexato/efectos adversos , Metotrexato/toxicidad , Ratones , Testículo/efectos de los fármacos , Testículo/patología , Estrés Oxidativo/efectos de los fármacos , Testosterona/sangre , Peroxidación de Lípido/efectos de los fármacos , Antioxidantes/farmacología , Malondialdehído/metabolismo
10.
J Assist Reprod Genet ; 41(7): 1881-1891, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38568464

RESUMEN

PURPOSE: In this study, the effect of thymoquinone (TQ) on CP-induced spermatogenesis defects in mice has been investigated. METHODS: Sperm parameters, serum testosterone concentration, histology, Bax/Bcl-2 ratio, and expression of autophagy-related biomarkers have been assessed. Total antioxidant capacity (TAC), total oxidant status (TOS), and oxidative stress index (OSI) in testicular tissue were examined for the evaluation of oxidative stress levels. RESULTS: CP has induced histological changes and significantly increased the Bax/Bcl-2 ratio, decreased testosterone concentration, testicular weight, and sperm quality. CP induced oxidative stress by elevating OSI in the testicular tissue (p < 0.05). Expression of the autophagy-inducer genes (ATG7, ATG5, and Beclin-1) and ratio of LC3B/LC3A proteins were significantly decreased, while mTOR expression was increased in the CP group. TQ pretreatment dose-dependently decreased the Bax/Bcl-2 ratio and mTOR gene expression while increasing the expression of ATG5 and ATG7 genes, LC3B/LC3A ratio, and Beclin-1 proteins. TQ could also dose-dependently reverse the histology, testosterone level, and sperm quality of the CP-intoxicated mice. CONCLUSIONS: These findings show that TQ pretreatment can enhance sperm production by inducing autophagy and reducing apoptosis and oxidative stress in the CP-intoxicated mouse testicles.


Asunto(s)
Apoptosis , Autofagia , Benzoquinonas , Cisplatino , Estrés Oxidativo , Espermatozoides , Testículo , Masculino , Animales , Estrés Oxidativo/efectos de los fármacos , Ratones , Autofagia/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/metabolismo , Testículo/patología , Apoptosis/efectos de los fármacos , Benzoquinonas/farmacología , Cisplatino/efectos adversos , Cisplatino/farmacología , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Espermatozoides/patología , Testosterona/sangre , Espermatogénesis/efectos de los fármacos , Antioxidantes/farmacología , Antioxidantes/metabolismo , Beclina-1/genética , Beclina-1/metabolismo , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo
11.
Tissue Cell ; 88: 102384, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38626526

RESUMEN

Autophagy disruption suppresses insulin production and induces diabetes. The role of autophagy in the differentiation of Wharton's jelly (WJ)-derived mesenchymal stem cells (WJSCs) into insulin-producing cells (IPCs) was investigated in this experimental study. The WJSCs were incubated in a differentiation medium (DM) with or without an autophagy inhibitor (3-methyladenine: 3MA). The differentiation of IPCs was confirmed by flow cytometry analysis of PDX-1 and insulin-positive cells, insulin secretion, and the high expression of ß cell-specific genes, Glucose transporter 2 (GLUT-2), and INSULIN. Autophagy has been assessed by calculating the percentage of Acridine orange (AO)-positive cells, expression of autophagy-related genes, and the LC3B/LC3A ratio. ß cell-specific genes were up-regulated in the DM group, and 3MA decreased their expression. In the DM+3MA-treated cells, the expression of GLUT-2 and INSULIN genes and insulin secretion decreased compared to the DM group. In cells treated with 3MA, there was a significant decrease in the percentage of PDX-1 and insulin-positive cells compared to 3MA-untreated cells. Additionally, in the group receiving both DM and 3MA treatment, the expression of autophagy-related genes, the LC3B/LC3A protein ratio, and the percentage of AO-stained cells were significantly reduced compared to the group receiving only DM treatment. These findings suggest autophagy is essential for ß cell differentiation and insulin secretion.


Asunto(s)
Autofagia , Diferenciación Celular , Células Secretoras de Insulina , Insulina , Células Madre Mesenquimatosas , Gelatina de Wharton , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Gelatina de Wharton/citología , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citología , Insulina/metabolismo , Adenina/farmacología , Adenina/análogos & derivados
12.
Mol Neurobiol ; 2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38430353

RESUMEN

Bisphenol A (BPA), an endocrine disruptor, is commonly used to produce epoxy resins and polycarbonate plastics. Continuous exposure to BPA may contribute to the development of diseases in humans and seriously affect their health. Previous research suggests a significant relationship between the increased incidence of neurological diseases and the level of BPA in the living environment. Syringic acid (SA), a natural derivative of gallic acid, has recently considered much attention due to neuromodulator activity and its anti-oxidant, anti-apoptotic, and anti-inflammatory effects. Therefore, in this study, we aimed to investigate the effects of SA on oxidative stress, apoptosis, memory and locomotor disorders, and mitochondrial function, and to identify the mechanisms related to Alzheimer's disease (AD) in the brain of rats receiving high doses of BPA. For this purpose, male Wistar rats received BPA (50, 100, and 200 mg/kg) and SA (50 mg/kg) for 21 days. The results showed that BPA exposure significantly altered the rats' neurobehavioral responses. Additionally, BPA, by increasing the level of ROS, and MDA level, increased the level of oxidative stress while reducing the level of antioxidant enzymes, such as SOD, CAT, GPx, and mitochondrial GSH. The administration of BPA at 200 mg/kg significantly decreased the expression of ERRα, TFAM, irisin, PGC-1α, Bcl-2, and FNDC5, while it increased the expression of TrkB, cytochrome C, caspase 3, and Bax. Moreover, the Western blotting results showed that BPA increased the levels of P-AMPK, GSK3b, p-tau, and Aß, while it decreased the levels of PKA, P-PKA, Akt, BDNF, CREB, P-CREB, and PI3K. Meanwhile, SA at 50 mg/kg reversed the behavioral, biochemical, and molecular changes induced by high doses of BPA. Overall, BPA could lead to the development of AD by affecting the mitochondria-dependent apoptosis pathway, as well as AMPK/PGC-1α/FNDC5 and CREB/BDNF/TrkB signaling pathways, and finally, by increasing the expression of tau and Aß proteins. In conclusion, SA, as an antioxidant, significantly reduced the toxicity of BPA.

13.
Iran J Med Sci ; 49(2): 121-129, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38356483

RESUMEN

Background: Stem cell-derived secretome (SE) released into the extracellular space contributes to tissue repair. The present study aimed to investigate the impact of isolated secretome (SE) from adipose-derived mesenchymal stem cells (ASCs) on Leishmania major (L. major) lesions in BALB/c mice. Methods: This experimental study was conducted at Ahvaz University of Medical Sciences (Ahvaz, Iran) in 2021. Forty female BALB/c mice were infected with stationary phase promastigotes through intradermal injection in the bottom of their tail and randomly divided into four groups (n=10 per group). The mice were given SE (20 mg/mL), either alone or in combination with Glucantime (GC, 20 mg/mL/Kg), meglumine antimoniate (20 mg/mL/Kg) for the GC group, and phosphate-buffered saline (PBS) for the control group. After eight weeks, the lesion size, histopathology, the levels of Interleukin 10 (IL-10), and Interleukin 12 (IL-12) were assessed. For the comparison of values between groups, the parametric one-way ANOVA was used to assess statistical significance. Results: At the end of the experiment, the mice that received SE had smaller lesions (4.56±0.83 mm versus 3.62±0.59 mm, P=0.092), lower levels of IL-10 (66.5±9.7 pg/mL versus 285.4±25.2 pg/mL, P<0.001), and higher levels of IL-12 (152.2±14.2 pg/mL versus 24.2±4.4 pg/mL, P<0.001) than the control. Histopathology findings revealed that mice treated with SE had a lower parasite burden in lesions and spleen than the control group. Conclusion: The current study demonstrated that ADSC-derived SE could protect mice infected with L. major against leishmaniasis.


Asunto(s)
Leishmania major , Leishmaniasis Cutánea , Parásitos , Femenino , Animales , Ratones , Leishmaniasis Cutánea/terapia , Leishmaniasis Cutánea/parasitología , Interleucina-10 , Secretoma , Antimoniato de Meglumina , Interleucina-12
14.
Mol Biol Rep ; 50(11): 9417-9430, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37831347

RESUMEN

BACKGROUND: Quercetin (QC) is a naturally occurring flavonoid found in abundance in fruits and vegetables. Its anti-cancer and anti-inflammatory properties have been previously demonstrated, but its low bioavailability hampers its clinical use. Triple-negative breast cancer is a subtype of breast cancer with a poor response to chemotherapy. This study investigates the anti-cancer effects of quercetin-solid lipid nanoparticles (QC-SLN) on the triple-negative breast cancer cell line MDA-MB231. MATERIALS AND METHODS: MCF-7 and MDA-MB231 cells were treated with 18.9 µM of QC and QC-SLN for 48 h. Cell viability, apoptosis, colony formation assay, and the anti-angiogenic effects of the treatment were evaluated. RESULTS: QC-SLN displayed optimal properties (particle size of 154 nm, zeta potential of -27.7 mV, encapsulation efficiency of 99.6%, and drug loading of 1.81%) and exhibited sustained release of QC over 72 h. Compared to the QC group, the QC-SLN group showed a significant decrease in cell viability, colony formation, angiogenesis, and a substantial increase in apoptosis through the modulation of Bax and Bcl-2 at both gene and protein levels. The augmentation in the proportion of cleaved-to-pro caspases 3 and 9, as well as poly (ADP-ribose) polymerase (PARP), under the influence of QC-SLN, was conspicuously observed in both cancer cell lines. CONCLUSIONS: This study showcases quercetin-solid lipid nanoparticles (QC-SLN) as a promising therapy for triple-negative breast cancer. The optimized QC-SLN formulation improved physicochemical properties and sustained quercetin release, resulting in reduced cell viability, colony formation, angiogenesis, and increased apoptosis in the MDA-MB231 cell line. These effects were driven by modulating Bax and Bcl-2 expression, activating caspases 3 and 9, and poly (ADP-ribose) polymerase (PARP). Further in vivo studies are needed to confirm QC-SLN's efficacy and safety.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Nanopartículas , Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Neoplasias de la Mama/metabolismo , Quercetina , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Proteína X Asociada a bcl-2 , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Ribosa , Línea Celular Tumoral , Nanopartículas/química , Proliferación Celular , Caspasas , Antineoplásicos/química , Apoptosis
15.
Brain Res Bull ; 204: 110779, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37827266

RESUMEN

Traumatic brain injury (TBI) is recognized as an important risk factor for cognitive deficits. The present study was designed to determine the potential neuroprotective effects of chrysin, a natural flavonoid compound, against TBI-induced spatial cognitive decline and the possible mechanisms involved. Oral administration of chrysin (25, 50, or 100 mg/kg/day) was initiated in rats immediately following the induction of the diffuse TBI model using the weight-dropping Marmarou model. Spatial cognitive ability, hippocampal synaptic plasticity, blood-brain barrier (BBB) permeability, brain water content, and histological changes were assessed at scheduled time points. The animals subjected to TBI exhibited spatial cognitive decline in the Morris water maze (MWM) test, which was accompanied by inhibition of hippocampal long-term potentiation (LTP) induction at the perforant path-dentate gyrus (PP-DG) synapses. Additionally, TBI caused BBB disruption, brain edema, and neuronal loss. Interestingly, treatment with chrysin (especially in the dose of 100 mg/kg) alleviated all the above-mentioned neuropathological changes related to TBI. The results provide evidence that chrysin improves TBI-induced spatial cognitive decline, which may be partly related to the amelioration of hippocampal synaptic dysfunction, alleviation of BBB disruption, reduction of brain edema, and prevention of neuronal loss.


Asunto(s)
Conmoción Encefálica , Edema Encefálico , Lesiones Traumáticas del Encéfalo , Disfunción Cognitiva , Fármacos Neuroprotectores , Ratas , Animales , Conmoción Encefálica/complicaciones , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Edema Encefálico/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Flavonoides/farmacología , Flavonoides/uso terapéutico , Disfunción Cognitiva/etiología , Disfunción Cognitiva/complicaciones , Hipocampo , Aprendizaje por Laberinto
17.
Food Chem Toxicol ; 178: 113924, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37393015

RESUMEN

Arsenic is a toxic metalloid that increases the risk of hepatotoxicity and hyperglycemia. The objective of the present study was to assess the effect of ferulic acid (FA) in mitigating glucose intolerance and hepatotoxicity caused by sodium arsenite (SA). A total of six groups including control, FA 100 mg/kg, SA 10 mg/kg, and groups that received different doses of FA (10, 30, and 100 mg/kg), respectively just before SA (10 mg/kg) for 28 days were examined. Fasting blood sugar (FBS) and glucose tolerance tests were conducted on the 29th day. On day 30, mice were sacrificed and blood and tissues (liver and pancreas) were collected for further investigations. FA reduced FBS and improved glucose intolerance. Liver function and histopathological studies confirmed that FA preserved the structure of the liver in groups received SA. Furthermore, FA increased antioxidant defense and decreased lipid peroxidation and tumor necrosis factor-alpha level in SA-treated mice. FA, at the doses of 30 and 100 mg/kg, prevented the decrease in the expression of PPAR-γ and GLUT2 proteins in the liver of mice exposed to SA. In conclusion, FA prevented SA-induced glucose intolerance and hepatotoxicity by reducing oxidative stress, inflammation, and hepatic overexpression of PPAR-γ and GLUT2 proteins.


Asunto(s)
Arsénico , Enfermedad Hepática Inducida por Sustancias y Drogas , Intolerancia a la Glucosa , Ratones , Animales , Arsénico/toxicidad , Arsénico/metabolismo , Intolerancia a la Glucosa/inducido químicamente , Intolerancia a la Glucosa/tratamiento farmacológico , Intolerancia a la Glucosa/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Antioxidantes/farmacología , Hígado , Estrés Oxidativo , Hipoglucemiantes/farmacología , Hipoglucemiantes/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo
20.
Biochem Biophys Res Commun ; 664: 69-76, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37141640

RESUMEN

BACKGROUND: The presence of cancer stem cells (CSCs) is a major cause of resistance to cancer therapy and recurrence. Triple-negative breast cancer (TNBC) is a subtype that responds poorly to therapy, making it a significant global health issue. Quercetin (QC) has been shown to affect CSC viability, but its low bioavailability limits its clinical use. This study aims to increase the effectiveness of QC in inhibiting CSC generation by using solid lipid nanoparticles (SLNs) in MDA-MB231 cells. MATERIALS AND METHODS: After treating MCF-7 and MDA-MB231 cells with 18.9 µM and 13.4 µM of QC and QC-SLN for 48 h, respectively, cell viability, migration, sphere formation, protein expression of ß-catenin, p-Smad 2 and 3, and gene expression of EMT and CSC markers were evaluated. RESULTS: The QC-SLN with particle size of 154 nm, zeta potential of -27.7 mV, and encapsulation efficacy of 99.6% was found to be the most effective. Compared to QC, QC-SLN significantly reduced cell viability, migration, sphere formation, protein expression of ß-catenin and p-Smad 2 and 3, and gene expression of CD44, zinc finger E-box binding homeobox 1 (ZEB1), vimentin, while increasing the gene expression of E-cadherin. CONCLUSIONS: Our findings demonstrate that SLNs improve the cytotoxic effect of QC in MDA-MB231 cells by increasing its bioavailability and inhibiting epithelial-mesenchymal transition (EMT), thereby effectively inhibiting CSC generation. Therefore, SLNs could be a promising new treatment for TNBC, but more in vivo studies are needed to confirm their efficacy.


Asunto(s)
Neoplasias de la Mama Triple Negativas , beta Catenina , Humanos , beta Catenina/metabolismo , Quercetina/farmacología , Quercetina/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Línea Celular Tumoral , Fosforilación , Transducción de Señal , Células Madre Neoplásicas/metabolismo , Transición Epitelial-Mesenquimal , Movimiento Celular , Proteína smad3/metabolismo , Proteína Smad2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA