Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 413
Filtrar
1.
Front Pharmacol ; 15: 1457467, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39376609

RESUMO

Polycystic ovary syndrome (PCOS) is a complex endocrine and metabolic disorder characterized by hyperandrogenism, prolonged anovulation and polycystic ovaries. However, there are no effective interventions to treat this disorder. As previously shown, mangiferin modulated the AMPK and NLRP3 signal pathways to alleviate nonalcoholic fatty liver disease (NAFLD). In recent years, mangiferin has emerged as a promising drug candidate for treating metabolic diseases. In this study, we evaluated the effects of mangiferin on a letrozole (LET) combined with high-fat diet (HFD)-induced PCOS rat model through estrous cycle detection, serum/tissue biochemical analysis, and hematoxylin and eosin (HE) staining of ovarian tissue. The mechanisms of mangiferin's effects on PCOS rats were analyzed using 16S rRNA sequencing, RNA-seq, western blotting (WB), and immunohistochemical (IHC) staining. Our results displayed that mangiferin showed a promising effect in PCOS rats. It improved lipid metabolism, glucose tolerance, insulin resistance, hormonal imbalance, ovarian dysfunction, and adipocyte abnormalities. RNA-seq analysis indicated that mangiferin may be involved in several signal pathways, including apoptosis, necrosis, and inflammation. Furthermore, western blot and immunohistochemical staining demonstrated that mangiferin regulates Caspase-3 and Cytc, exhibiting anti-apoptotic activity in the ovaries. Additionally, mangiferin significantly altered the gut microbiota community of PCOS rats, changing the abundance of firmicutes, bacteroidota, proteobacteria, and actinobacteria at the phylum level and the abundance of Blautia, Coprococcus, Roseburia, and Pseudomonas at the genus level. In conclusion, mangiferin is a promising and novel therapeutic agent for PCOS as it ameliorates insulin resistance, gut microbiota and ovarian cell apoptosis.

2.
J Inflamm Res ; 17: 7081-7097, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39380664

RESUMO

Purpose: Acute liver injury (ALI) is characterized by inflammation and oxidative stress (OS). Although mangiferin (MGF) has antioxidant and anti-inflammatory effects, its role in ALI remains unclear. Accordingly, we investigated the MGF molecular mechanism in carbon tetrachloride (CCl4)-induced ALI in vivo and in vitro. Materials and Methods: The CCl4 was utilized to induce ALI in mice. In vivo, the therapeutic effects of MGF on CCl4-induced liver injury were evaluated through biochemical assays and histomorphological analysis. Additionally, immunohistochemistry, immunofluorescence, ELISA and Western blotting were further applied to explore the mechanism. In vitro, The CCK-8 assay and flow cytometry were employed to investigate the protective effects of MGF against CCl4-induced toxicity in HepG2 cells, while mitochondrial reactive oxygen species levels and Western blotting were used to explore the biological effects and molecular mechanisms. Results: MGF treatment resulted in a reduction in serum levels of AST and ALT, diminished concentrations of TNF-α, IL-6, and IL-1ß in liver tissue, and concurrently decreased cellular apoptosis. Furthermore, MGF pretreatment enhanced the activity of SOD and GSH while concurrently diminishing the MDA production. This study further demonstrated the upregulation of Nrf2, NQO1, and HO-1 protein expression levels, as well as the downregulation of p-p65 protein expression levels. In vitro investigations revealed that the mitigation of CCl4-induced inflammation and OS by MGF was mediated via the Nrf2- antioxidant response element (ARE) pathway, which was disrupted by ML385 in HepG2 cells. Conclusion: CCl4 can induce liver injury, while treatment with MGF mitigates ALI by inhibiting oxidative stress, inflammation, and apoptosis. The protective mechanism of MGF is mediated by the Nrf2-ARE pathway activation.

3.
Int J Mol Sci ; 25(20)2024 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-39456979

RESUMO

Inflammation in macrophages is exacerbated under hyperglycemic conditions, contributing to chronic inflammation and impaired wound healing in diabetes. This study investigates the potential of mangiferin, a natural polyphenol, to alleviate this inflammatory response by targeting a redox-sensitive transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2). Mangiferin, a known Nrf2 activator, was evaluated for its ability to counteract the hyperglycemia-induced inhibition of Nrf2 and enhance antioxidant defenses. The protective effects of mangiferin on macrophages in a hyperglycemic environment were assessed by examining the expression of Nrf2, NF-κB, NLRP3, HO-1, CAT, COX-2, IL-6, and IL-10 through gene and protein expression analyses using qPCR and immunoblotting, respectively. The mangiferin-mediated nuclear translocation of Nrf2 was evidenced, leading to a robust antioxidant response in macrophages exposed to a hyperglycemic microenvironment. This activation suppressed NF-κB signaling, reducing the expression of pro-inflammatory mediators such as COX-2 and IL-6. Additionally, mangiferin decreased NLRP3 inflammasome activation and reactive oxygen species accumulation in hyperglycemia exposed macrophages. Our findings revealed that mangiferin alleviated hyperglycemia-induced reductions in AKT phosphorylation, highlighting its potential role in modulating key signaling pathways. Furthermore, mangiferin significantly enhanced the invasiveness and migration of macrophages in a hyperglycemic environment, indicating its potential to improve wound healing. In conclusion, this study suggests that mangiferin may offer a promising therapeutic approach for managing inflammation and promoting wound healing in diabetic patients by regulating Nrf2 activity in hyperglycemia-induced macrophages.


Assuntos
Hiperglicemia , Inflamação , Macrófagos , Fator 2 Relacionado a NF-E2 , Transdução de Sinais , Xantonas , Fator 2 Relacionado a NF-E2/metabolismo , Xantonas/farmacologia , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Animais , Transdução de Sinais/efeitos dos fármacos , Camundongos , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Antioxidantes/farmacologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-39318009

RESUMO

In recent years, bioactive constituents from plants have been investigated as an alternative to synthetic approaches of therapeutics. Mangiferin (MGF) is a xanthone glycoside extracted from Mangifera indica and has shown numerous medicinal properties, such as antimicrobial, anti-diarrhoeal, antiviral, anti-inflammatory, antihypertensive, anti-tumours, and anti-diabetic effects. However, there are numerous challenges to its effective therapeutic usage, including its low water solubility, limited absorption, and poor bioavailability. Nano formulation approaches in recent years exhibited potential for the delivery of phytoconstituents with key benefits of high entrapment, sustained release, enhanced solubility, stability, improved pharmacokinetics, and site-specific drug delivery. Numerous techniques have been employed for the fabrication of MGF-loaded Nano formulations, and each technique has its advantages and limitations. The nanocarriers that have been employed to fabricate MGF nanoformulations for various therapeutic purposes include; polymeric nanoparticles, nanostructure, lipid carriers, polymeric micelles, Nano emulsions, microemulsion & self-microemulsifying drug delivery system, solid lipid nanoparticles, gold nanoparticles, carbon nanotubes, transfersomes, nanoliposomes, ethosomes & transethosomes, and glycethosomes. Different biopharmaceutical characteristics (size, shape, entrapment efficiency, zeta potential, in vitro drug release, ex vivo drug permeation,, and in vivo studies) of the mentioned MGF-loaded nanocarriers have been methodically discussed. Patent reports are also included to further strengthen the potential of MGF in the management of diseases.

5.
Int J Mol Sci ; 25(18)2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39337458

RESUMO

Myocarditis is a major cause of heart failure and death, particularly in young individuals. Current treatments are mainly symptomatic, but emerging therapies focus on targeting inflammation and fibrosis pathways. Natural bioactive compounds like flavonoids and phenolic acids show promising anti-inflammatory and antioxidant properties. Corticosteroids are frequently employed in the treatment of autoimmune myocarditis and appear to lower mortality rates compared to conventional therapies for heart failure. This study aims to explore the effects of Mangiferin on pro-inflammatory cytokine levels, nitro-oxidative stress markers, histopathological alterations, and cardiac function in experimental myosin-induced autoimmune myocarditis. The effects were compared to Prednisone, used as a reference anti-inflammatory compound, and Trolox, used as a reference antioxidant. The study involved 30 male Wistar-Bratislava rats, which were randomly divided into five groups: a negative control group (C-), a positive control group with induced myocarditis using a porcine myosin solution (C+), three groups with induced myocarditis receiving Mangiferin (M), Prednisone (P), or Trolox (T) as treatment. Cardiac function was evaluated using echocardiography. Biochemical measurements of nitro-oxidative stress and inflammatory markers were conducted. Finally, histopathological changes were assessed. At echocardiography, the evaluation of the untreated myocarditis group showed a trend toward decreased left ventricular ejection fraction (LVEF) but was not statistically significant, while all treated groups showed some improvement in LVEF and left ventricular fraction shortening (LVFS). Significant changes were seen in the Mangiferin group, with lower end-diastolic left ventricular posterior wall (LVPWd) by day 21 compared to the Trolox group (p < 0.001). In the first week of the experiment, levels of interleukins (IL)-1ß, IL-6, and tumour necrosis factor (TNF)-α were significantly higher in the myosin group compared to the negative control group (p < 0.001, p < 0.001, p < 0.01), indicating the progression of inflammation in this group. Treatment with Mangiferin, Prednisone, and Trolox caused a significant reduction in IL-1ß compared to the positive control group (p < 0.001). Notably, Mangiferin resulted in a superior reduction in IL-1ß compared to Prednisone (p < 0.05) and Trolox (p < 0.05). Furthermore, Mangiferin treatment led to a statistically significant increase in total oxidative capacity (TAC) (p < 0.001) and a significant reduction in nitric oxide (NOx) levels (p < 0.001) compared to the negative control group. Furthermore, when compared to the Prednisone-treated group, Mangiferin significantly reduced NOx levels (p < 0.001) and increased TAC levels (p < 0.001). Mangiferin treatment significantly lowered creatine kinase (CK) and aspartate aminotransferase (AST) levels on day 7 (p < 0.001 and p < 0.01, respectively) and reduced CK levels on day 21 (p < 0.01) compared to the untreated group. In the nontreated group, the histological findings at the end of the experiment were consistent with myocarditis. In the group treated with Mangiferin, only one case exhibited mild inflammatory infiltrates, represented by mononucleated leukocytes admixed with few neutrophils, with the severity graded as mild. Statistically significant correlations between the grades (0 vs. 1-2) and the study groups have been highlighted (p < 0.005). This study demonstrated Mangiferin's cardioprotective effects in autoimmune myocarditis, showing reduced oxidative stress and inflammation. Mangiferin appears promising as a treatment for acute myocarditis, but further research is needed to compare its efficacy with other treatments like Trolox and Prednisone.


Assuntos
Anti-Inflamatórios , Antioxidantes , Modelos Animais de Doenças , Miocardite , Estresse Oxidativo , Ratos Wistar , Xantonas , Animais , Miocardite/tratamento farmacológico , Miocardite/metabolismo , Miocardite/patologia , Antioxidantes/farmacologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Masculino , Xantonas/farmacologia , Xantonas/uso terapêutico , Ratos , Estresse Oxidativo/efeitos dos fármacos , Citocinas/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Cromanos
6.
Biomed Pharmacother ; 179: 117387, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39245002

RESUMO

As the global population ages, the incidence of Parkinson's Disease (PD) continues to rise, imposing significant social and economic burdens. Mangiferin (MGF), a polyphenolic, bioactive compound has been shown to play a role in the prevention and treatment of PD. This study investigates the neuroprotective effects of MGF in an MPTP-induced zebrafish model of PD through transcriptome analysis. Initially, optimal concentrations for modeling were determined using various MPTP and MGF combinations. The zebrafish were then divided into control, MPTP-treated, and MGF co-treated groups. Subsequent evaluations included hatching rates, mortality rates, growth and development conditions, spontaneous motor abilities, as well as measurements of enzymatic activities of SOD, CAT, and levels of GSH. Ultimately, the therapeutic efficacy of MGF on the PD model in zebrafish was assessed through transcriptome sequencing. The results demonstrated that MPTP treatment induced PD-associated symptoms in zebrafish, while MGF treatment significantly improved the motor abilities and survival rates of the PD model zebrafish, effectively reducing oxidative stress and ameliorating PD symptoms. Transcriptome sequencing further revealed that MGF may mitigate mitochondrial-related oxidative stress in PD zebrafish by modulating the expression of critical genes including lrrk2, vps35, atp13a, dnajc6, and uchl1. Differential gene expression analysis indicated that these genes are primarily involved in vital signaling pathways, such as neuroactive ligand-receptor interaction, and the calcium signaling pathway. In summary, our study provides robust scientific evidence supporting MGF as a potential therapeutic candidate for PD by preserving mitochondrial homeostasis and elucidating its mechanisms of action.


Assuntos
Perfilação da Expressão Gênica , Estresse Oxidativo , Xantonas , Peixe-Zebra , Animais , Xantonas/farmacologia , Perfilação da Expressão Gênica/métodos , Estresse Oxidativo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Modelos Animais de Doenças , Transcriptoma/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Antiparkinsonianos/farmacologia , Masculino
7.
J Biochem Mol Toxicol ; 38(10): e23849, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39264833

RESUMO

One of the main factors in the pathophysiology of amyotrophic lateral sclerosis is oxidative stress. Mangiferin (MF), a natural plant polyphenol, has anti-inflammatory and antioxidant effects. The aim of our study was to investigate the protective effects and mechanisms of MF in the hSOD1-G93A ALS cell model. Our result revealed that MF treatment reduced the generation of reactive oxygen species (ROS) and malondialdehyde (MDA), decreased oxidative damage, and reduced apoptosis. Additionally, it was observed that MF significantly increased the synthesis of the antioxidant genes hemeoxygenase-1 and NAD(P)H: quinone oxidoreductase 1, which are downstream of the Nrf2 signaling pathway, and increased the expression and activation of nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 knockdown greatly promoted apoptosis, which was reversed by MF treatment. To summarize, MF promoted the Nrf2 pathway and scavenged MDA and ROS to protect the ALS cell model.


Assuntos
Apoptose , Neurônios Motores , Fator 2 Relacionado a NF-E2 , Estresse Oxidativo , Transdução de Sinais , Xantonas , Xantonas/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Camundongos , Animais , Neurônios Motores/metabolismo , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/patologia , Transdução de Sinais/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/tratamento farmacológico , Humanos , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD(P)H Desidrogenase (Quinona)/genética
8.
Arch Physiol Biochem ; : 1-10, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39225043

RESUMO

The aim of this study was to investigate the protective effects of Mangiferin (MG) on glucolipotoxicity-induced pancreatic beta-cell injury. In vivo administration of MG significantly reduced the level of blood glucose in high-fat diet (HFD)-fed mice. MG treatment inhibited beta-cell apoptosis in HFD-treated mice. In vitro, MG protected INS-1 cells against apoptosis and impairment of insulin secretion following High glucose/Palmitic acid (HG/PA) treatment. MG treatment enhanced autophagy flux which was blocked by HG/PA treatment. Inhibition of autophagosome formation by 3-Methyladenine or blockade of autolysosome by Chloroquine reversed the protective effects of MG on INS-1 cells. MG treatment increased AMPK phosphorylation and reduced mTOR activation in INS-1 cells. Administration of the AMPK blocker abrogated MG-induced autophagy, and similar results were observed in INS-1 cells after cotreatment with MG and mTOR activator. In conclusion, MG ameliorated pancreatic beta-cell injury induced by glucolipotoxicity through modulation of autophagy via the AMPK-mTOR pathway.

9.
Cureus ; 16(7): e65751, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39211673

RESUMO

Background and objectives Stadice® is a proprietary herbal ingredient preparation standardized to mangiferin, developed to support cognitive wellness in healthy adults. Mangifera indica extract and its active constituent, mangiferin were known for its positive cognitive health benefits at experimental levels. This was an attempt to evaluate the clinical efficacy and safety of Stadice® on healthy subjects. Materials and methods A randomized, double-blind, placebo-controlled clinical study was designed to study the efficacy and safety of Stadice®. Sixty healthy subjects who were regularly playing virtual/mobile/computer/laptop games online or offline were asked to consume a capsule containing 300 mg of Stadice® or placebo per day for seven days. Cognitive ability tests, that were part of the NIMHANS Neuropsychological Battery and Auditory verbal learning tests were used to assess the cognitive health effects. Psychological stress response, anxiety, mood, subjective working memory, and cortisol levels were also assessed. All assessments were carried out at the baseline and at the end of the study. Results Stadice® was found to significantly improve mental speed, attention, working memory, response inhibition, and verbal learning and memory as evidenced by the results of the multiple cognitive ability tests. Additionally, Stadice ® showed beneficial responses in managing psychological stress in terms of handling nervousness, irritability, or mood swings. No safety concerns were found in the laboratory safety test parameters as they were within the normal physiological range and no adverse events were reported in this study. Conclusion The proprietary Mangifera indica extract (Stadice®) holds promise for enhancing cognitive abilities in healthy adults, particularly those engaged in esports. Improvements in learning, memory, mental speed, attention, response inhibition, and working memory among participants supplemented with Stadice® were observed with a good safety profile. Further exploration is warranted to ascertain its broader applicability as well as to elucidate the possible mechanisms.

10.
Medicina (Kaunas) ; 60(8)2024 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-39202505

RESUMO

Background and Objectives: Hyperglycemia is known to undermine the osteointegration process of implants. In this study, the effects of mangiferin (MF) on the post-implant osteointegration process in a type-II diabetes model were investigated molecularly and morphologically. Materials and Methods: Sprague Dawley male rats were divided into three groups: control, diabetes, and diabetes + MF. All animals were implanted in their tibia bones on day 0. At the end of the 3-month experimental period, the animals' blood and the implant area were isolated. Biochemical measurements were performed on blood samples and micro-CT, qRT-PCR, histological, and immunohistochemical measurements were performed on tibia samples. Results: MF significantly improved the increased glucose, triglyceride-VLDL levels, and liver enzymes due to diabetes. By administering MF to diabetic rats, the osteointegration percentage and bone volume increased while porosity decreased. DKK1 and BMP-2 mRNA expressions and OPN, OCN, and OSN mRNA-protein expressions increased by MF administration in diabetic rats. Additionally, while osteoblast and osteoid surface areas increased with MF, osteoclast and eroded surface areas decreased. Conclusions: The findings of our study indicate that MF will be beneficial to the bone-repairing process and osteointegration, which are impaired by type-II diabetes.


Assuntos
Diabetes Mellitus Experimental , Osseointegração , Ratos Sprague-Dawley , Xantonas , Animais , Xantonas/farmacologia , Xantonas/uso terapêutico , Masculino , Ratos , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Osseointegração/efeitos dos fármacos , Tíbia/efeitos dos fármacos , Microtomografia por Raio-X , Modelos Animais de Doenças , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Peptídeos e Proteínas de Sinalização Intercelular
11.
Ann Neurosci ; 31(3): 186-203, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39156628

RESUMO

Background: Parkinson's disease (PD) is typified by inflammation of dopaminergic neurons leading to the release of various inflammatory mediators. These mediators activate the transcription factor NF-κB, which in turn activates inducible nitric oxide synthase (iNOS), leading to increased inflammation. Purpose: This study was intended to study the effect of combination of mangiferin, a specific inhibitor of NF-κB with low-dose nitric oxide (NO) modulators. Methods: A total of eight Wistar rats weighing 200-250 g were used in each group. Stereotactic surgery was performed to induce 6-hydroxydopamine (6-OHDA) lesions. The treatment period extended from day 14 to day 42, during which time behavioral tests were performed to evaluate the effects of mangiferin and its combination with NO modulators. On day 42, the brains of the rats were removed for biochemical and molecular analyzes. Results: Mangiferin significantly improved locomotor activity and decreased inflammatory chemokines levels in rats with 6-OHDA lesions. Mangiferin therapy decreased myeloperoxidase (MPO) levels and reduced oxidative stress. In particular, caspase-3, caspase-9 and COX-2 activities were significantly reduced after the mangiferin treatment. A combination of 45-µg mangiferin and 10-mg/kg L-NAME showed the greatest improvement in locomotor, behavioral, biochemical, and molecular parameters impaired by 6-OHDA. Conclusion: In this study, mangiferin was found to protect rats with 6-OHDA lesions by inhibiting inflammation causing chemokines such as TNF-α and IL-6. Besides, the grouping of iNOS inhibitor L-NAME at a dose of 10 mg/kg with 45-µg mangiferin enhanced the anti-inflammatory and anti-Parkinsonian activity of mangiferin. Consequently, the combination therapy of mangiferin and L-NAME is promising for the treatment of PD. However, clinical trials will be required to evaluate the efficacy of this combination therapy in humans.

12.
Biomed Pharmacother ; 178: 117174, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39098177

RESUMO

Mangiferin(MGF) exhibits crucial biological roles, including antioxidant and anti-inflammatory functions. However, how to clearly elucidate the functioning mechanism of MGF for inhibiting cisplatin-induced hearing loss requires in-depth investigation. In this work, we aimed at gaining insight into how MGF functions as the protective agent against cisplatin-triggered ototoxicity using various assays. The variation for reactive oxygen species (ROS) concentrations was determined with MitoSOX-Red and 2',7'-Dichlorodihydrofluorescein diacetate staining (DCFH-DA). The protective function and corresponding mechanism of MGF in hair cell survival in the House Ear Institute-Organ of Corti (HEI-OC1) cell line were assessed using RNA sequencing (RNA-Seq). Our findings demonstrated that MGF significantly alleviated cisplatin-induced injury to hair cells in vitro, encompassing cell lines and cochlear explants, as well as in vivo models, including C57BL/6 J mice and zebrafish larvae. Mechanistic studies revealed that MGF reversed the increased accumulation of ROS and inhibited cell apoptosis through mitochondrial-mediated intrinsic pathway. Moreover, real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting data indicated MGF protected against cisplatin-mediated ototoxicity via the mitogen-activated protein kinase pathway (MAPK). These findings demonstrated MGF has significant potential promise in combating cisplatin-induced ototoxicity, offering a foundation for expanded investigation into therapeutic approaches for auditory protection.


Assuntos
Apoptose , Cisplatino , Células Ciliadas Auditivas , Perda Auditiva Neurossensorial , Camundongos Endogâmicos C57BL , Ototoxicidade , Espécies Reativas de Oxigênio , Xantonas , Peixe-Zebra , Cisplatino/toxicidade , Cisplatino/efeitos adversos , Animais , Ototoxicidade/prevenção & controle , Espécies Reativas de Oxigênio/metabolismo , Camundongos , Perda Auditiva Neurossensorial/induzido quimicamente , Perda Auditiva Neurossensorial/tratamento farmacológico , Perda Auditiva Neurossensorial/metabolismo , Perda Auditiva Neurossensorial/prevenção & controle , Perda Auditiva Neurossensorial/patologia , Apoptose/efeitos dos fármacos , Xantonas/farmacologia , Xantonas/uso terapêutico , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Antioxidantes/farmacologia
13.
Food Chem ; 460(Pt 3): 140712, 2024 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-39121767

RESUMO

Reactive carbonyl species (RCS), including acrolein (ACR), methylglyoxal (MGO), and glyoxal (GO), are typically generated in food processing and accumulate in the body for ages, triggering various chronic diseases. Here, we investigated the capture capability and reaction pathways of mangiferin one-to-one and one-to-many on RCS in high temperatures using UPLC-MS/MS. We found that mangiferin can capture ACR/MGO/GO to form their adducts, yet, the ability to capture RCS is arranged in different orders, with ACR > MGO > GO for a single RCS and MGO > ACR > GO for multiple RCS. After synthesizing and identifying the structures of the ACR- and MGO-adducts of MGF, our results indicated that MGF-ACR-MGO produced in the multiple-RCS-MGF system was formed by capturing MGO through MGF-ACR rather than through MGF-MGO capturing ACR, which resulting in higher inhibitory activity of MGF against MGO than against ACR. Then, the capture ability and path of MGF on RCS were verified in the coffee-leaves tea and cake.


Assuntos
Acroleína , Glioxal , Temperatura Alta , Aldeído Pirúvico , Espectrometria de Massas em Tandem , Xantonas , Xantonas/química , Aldeído Pirúvico/química , Glioxal/química , Acroleína/química , Acroleína/análogos & derivados , Cromatografia Líquida de Alta Pressão , Manipulação de Alimentos
14.
J Asian Nat Prod Res ; : 1-13, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38952150

RESUMO

Inhibition of lipid synthesis in sebocytes is essential for acne treatments. The effects of natural product-derived substances on lipid synthesis are unknown. This study investigated the effects of water extract of Mangifera indica leaves (WEML) on lipid synthesis in human sebocytes. Sebocyte differentiation in low serum conditions increased lipid accumulation and proliferator-activated receptor γ expression. WEML treatment significantly inhibited lipid accumulation and adipogenic mRNA expression in sebocytes. Mangiferin, a bioactive compound in WEML, also reduced lipid accumulation and adipogenic mRNA expression via the AKT pathway. Thus, WEML and mangiferin effectively inhibit lipid synthesis in sebocytes, showing promise for acne treatment.

15.
Future Microbiol ; 19(13): 1157-1170, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39012219

RESUMO

Aim: To evaluate the antifungal activity of mangiferin against Candida spp. resistant to fluconazole.Materials & methods: The antifungal activity of mangiferin was assessed using broth microdilution and its interaction with azoles and amphotericin B was evaluated by checkerboard. The activity of mangiferin against Candida spp. biofilms was assessed using the MTT colorimetric assay and its possible mechanism of action was evaluated using flow cytometry.Results: Mangiferin showed activity against Candida albicans, Candida tropicalis and Candida parapsilosis resistant to fluconazole and showed synergism with azoles and amphotericin B. Mangiferin increased the activity of antifungals against Candida biofilms and caused depolarization of the mitochondrial membrane and externalization of phosphatidylserine, suggesting apoptosis.Conclusion: mangiferin combined with antifungals has potential against Candida spp.


Candida is a type of fungus that can make people ill. Over time, many species of Candida have found ways to resist the drugs used to kill them. It is important to find new drugs. We decided to see if a substance called mangiferin works against Candida. We found that mangiferin works against Candida and may help other drugs to work better. We still need to do more studies to find out whether mangiferin can help prevent diseases caused by Candida in the future.


Assuntos
Anfotericina B , Antifúngicos , Biofilmes , Candida , Farmacorresistência Fúngica , Sinergismo Farmacológico , Fluconazol , Testes de Sensibilidade Microbiana , Xantonas , Antifúngicos/farmacologia , Xantonas/farmacologia , Fluconazol/farmacologia , Biofilmes/efeitos dos fármacos , Farmacorresistência Fúngica/efeitos dos fármacos , Anfotericina B/farmacologia , Candida/efeitos dos fármacos , Humanos , Apoptose/efeitos dos fármacos , Candida albicans/efeitos dos fármacos , Azóis/farmacologia
16.
Biomed Chromatogr ; 38(9): e5936, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38956791

RESUMO

Mangifera indica peels are a rich source of diverse flavonoids and xanthonoids; however, generally these are discarded. Computational studies revealed that mangiferin significantly interacts with amino acid residues of transcriptional regulators 1IK3, 3TOP, and 4f5S. The methanolic extract of Langra variety of mangoes contained the least phenol concentrations (22.6 ± 0.32 mg/gGAE [gallic acid equivalent]) compared to the chloroform (214.8 ± 0.12 mg/gGAE) and ethyl acetate fractions (195.6 ± 0.14 mg/gGAE). Similarly, the methanolic extract of Sindhri variety contained lower phenol concentrations (42.3 ± 0.13 mg/gRUE [relative utilization efficiency]) compared with the chloroform (85.6 ± 0.15 mg/gGAE) and ethyl acetate (76.1 ± 0.32 mg/gGAE) fractions. Langra extract exhibited significant α-glucosidase inhibition (IC50 0.06 mg/mL), whereas the ethyl acetate fraction was highly active (IC50 0.12 mg/mL) in Sindhri variety. Mangiferin exhibited significant inhibition (IC50 0.026 mg/mL). A moderate inhibition of 15-LOX was observed in all samples, whereas mangiferin was least active. In advanced glycation end product inhibition assay, the chloroform fraction of Langra variety exhibited significant inhibition in nonoxidative (IC50 64.4 µg/mL) and oxidative modes (IC50 54.7 µg/mL). It was concluded that both Langra and Sindhri peel extracts and fractions possess significant antidiabetic activities. The results suggest the potential use of peel waste in the management and complications of diabetes.


Assuntos
Antioxidantes , Produtos Finais de Glicação Avançada , Hipoglicemiantes , Mangifera , Extratos Vegetais , Xantonas , Xantonas/química , Xantonas/farmacologia , Xantonas/análise , Mangifera/química , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Antioxidantes/química , Antioxidantes/farmacologia , Antioxidantes/análise , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Hipoglicemiantes/análise , Produtos Finais de Glicação Avançada/química , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Simulação de Acoplamento Molecular , Frutas/química , Inibidores de Glicosídeo Hidrolases/química , Inibidores de Glicosídeo Hidrolases/farmacologia , Inibidores de Glicosídeo Hidrolases/análise , Simulação por Computador
17.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 42(4): 444-451, 2024 Aug 01.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-39049631

RESUMO

OBJECTIVES: This study aims to investigate the primary target and potential mechanism of mangiferin (MF) in treating oral submucous fibrosis (OSF) through Gene Expression Omnibus (GEO) database chip mining, network pharmacology, and molecular docking techniques. METHODS: Potential therapeutic targets for OSF were identified using GEO chip data. The potential targets of MF were predicted, and disease-related targets for OSF were collected from databases. A Venn diagram was created using the EVenn platform to identify overlapping targets. The protein-protein interaction (PPI) network was constructed using the STRING database. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed using the DAVID platform. Cytoscape 3.10.1 software was used to visualize a drug-target-pathway-disease network, while AutoDocktools 1.5.6 software was employed for molecular docking analysis. RESULTS: A total of 356 potential targets for MF and 360 disease-related targets for OSF were obtained from multiple databases. The top 15 key target proteins in the PPI network were selected as significant candidates. GO function and KEGG pathway enrichment analyses revealed that MF treatment primarily involved advanced glycation end products-receptor (AGE-RAGE), epidermal growth factor receptor (EGFR), and other signaling pathways associated with OSF pathogenesis. Molecular docking analysis demonstrated that MF exhibited a strong binding activity toward AKT serine kinase 1 (AKT1), tumor necrosis factor (TNF), and other core targets. CONCLUSIONS: These findings suggest that MF may exert its therapeutic effects on OSF through a multitarget approach involving various signaling pathways.


Assuntos
Simulação de Acoplamento Molecular , Farmacologia em Rede , Fibrose Oral Submucosa , Mapas de Interação de Proteínas , Xantonas , Xantonas/uso terapêutico , Xantonas/farmacologia , Fibrose Oral Submucosa/tratamento farmacológico , Fibrose Oral Submucosa/metabolismo , Humanos , Ontologia Genética , Mineração de Dados , Receptores ErbB/metabolismo , Software , Transdução de Sinais
18.
J Biochem Mol Toxicol ; 38(7): e23765, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38967724

RESUMO

Mangiferin is a naturally occurring glucosylxanthone that has shown promising immunomodulatory effects. It is generally isolated from the leaves, peels, bark, and kernels of Mangifera indica Linn. Mangiferin is like a miraculous natural bioactive molecule that has an immunomodulatory function that makes it a potential therapeutic candidate for the treatment of rheumatoid arthritis (RA) and cancer. The anticancer activity of mangiferin acts by blocking NF-κB, as well as regulating the ß-catenin, EMT, MMP9, MMP2, LDH, ROS, and NO, and also by the activation of macrophages. It has no cytotoxic effect on grown chondrocytes and lowers matrix metalloproteinase levels. Additionally, it has a potent proapoptotic impact on synoviocytes. The precise molecular mechanism of action of mangiferin on RA and malignancies is still unknown. This comprehensive review elaborates on the immunomodulatory effect of mangiferin and its anticancer and anti-RA activity. This also explained the total synthesis of mangiferin and its in vitro and in vivo screening models.


Assuntos
Artrite Reumatoide , Neoplasias , Xantonas , Xantonas/farmacologia , Xantonas/uso terapêutico , Xantonas/química , Humanos , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Animais , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Agentes de Imunomodulação/farmacologia , Agentes de Imunomodulação/química
19.
Curr Res Microb Sci ; 6: 100243, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38873591

RESUMO

In mango pickle industry, a significant quantity of mango seed kernels is discarded as solid wastes. These seed kernels can be an ideal source for obtaining extracts rich in bioactive polyphenolic compounds with good antioxidant properties. The potential of mango kernel phenolic extract (MKPE) was investigated as a natural and effective antimicrobial agent for controlling major postharvest fungal pathogen infections, a significant threat to global food supply chains. Fungal pathogens contribute to the deterioration of fruits, vegetables, and grains during storage and transportation, leading to economic losses and compromised food safety. MKPE was obtained from pickling variety 'Ramkela' raw mango kernels, and its phenolic composition was characterized using LC-MS. The in vitro antifungal activity of MKPE against Botrytis cinerea, Colletotrichum gloeosporoides, and Rhizopus stolonifer was evaluated in vitro. A concentration-dependent inhibition of fungal radial growth against all three pathogens was observed, exhibiting the potential of MKPE as a valuable natural resource for addressing postharvest losses caused by fungal pathogens. The extraction process yielded a total phenolic content of 2128 mg GAE/100 g. Major polyphenolic bioactive compounds present were mangiferin, quercetin, and rhamnetin. The in-vitro antimicrobial assay showed reduction in the radial growth and inhibition percent of the pathogens. EC50 values of MKPE for B. cineria, C. gloeosporoides, and R. stolonifer was found to 364.17, 963.8 and 926 ppm, respectively. Our results demonstrate an economical, sustainable, and eco-friendly approach to manage post-harvest diseases rendered by fungi using mango MKPE from pickling industry waste.

20.
Nat Prod Res ; : 1-12, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38832668

RESUMO

Mangifera indica L. (Mango), native of tropical Asia, has enormous genetic diversity. Comparative phytochemical analysis of leaves of five varieties of Mangifera indica viz. Dashahri, Chausa, Langra, Lucknow Safeda and Gola grown in North India was carried out. Mangiferin content (using HPLC) was found to vary from 0.96 g to 3.00 g per 100 g of dry leaves. Essential oil composition (through GC-MS) showed the major components of all the five varieties to be caryophyllene (4.14-46.26%), humulene (3.19-30.45%), caryophyllene oxide (2.98-17.23%) and humulene epoxide 2 (1.56-4.73%). Results indicated that there was a direct relationship between total phenolic and flavonoid contents and DPPH radical scavenging activities. Our studies indicate that M. indica leaves, which are a form of biomass waste, could be used as an economical and renewable source of antidiabetic compound mangiferin as well as other biologically active phytoconstituents having nutraceutical as well as pharmaceutical applications.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA