Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 577
Filtrar
1.
Int J Mol Sci ; 25(4)2024 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-38397051

RESUMO

Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disorder caused by α-L-iduronidase deficiency. The standard treatment, enzyme replacement therapy with laronidase, has limited effectiveness in treating neurological symptoms due to poor blood-brain barrier penetration. An alternative is substrate reduction therapy using molecules, such as genistein, which crosses this barrier. This study evaluated the effectiveness of a combination of laronidase and genistein in a mouse model of MPS I. Over 12 weeks, MPS I and wild-type mice received laronidase, genistein, or both. Glycosaminoglycan (GAG) storage in visceral organs and the brain, its excretion in urine, and the serum level of the heparin cofactor II-thrombin (HCII-T) complex, along with behavior, were assessed. The combination therapy resulted in reduced GAG storage in the heart and liver, whereas genistein alone reduced the brain GAG storage. Laronidase and combination therapy decreased liver and spleen weights and significantly reduced GAG excretion in the urine. However, this therapy negated some laronidase benefits in the HCII-T levels. Importantly, the combination therapy improved the behavior of female mice with MPS I. These findings offer valuable insights for future research to optimize MPS I treatments.


Assuntos
Mucopolissacaridose I , Feminino , Camundongos , Animais , Mucopolissacaridose I/tratamento farmacológico , Iduronidase/uso terapêutico , Genisteína/farmacologia , Genisteína/uso terapêutico , Encéfalo , Barreira Hematoencefálica , Glicosaminoglicanos/uso terapêutico , Trombina/uso terapêutico , Modelos Animais de Doenças , Terapia de Reposição de Enzimas/métodos
2.
ScientificWorldJournal ; 2024: 5338212, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38304042

RESUMO

Endometriosis (EM) is a gynecological disorder that causes morbidity in women and is characterized by endometrial tissue in the uterus cavity. This study investigated the mechanism of genistein in the VEGF-A and ER-α expression through in vivo and in silico approaches. An in vivo study was conducted by thirty-six mice that were divided into six groups including control, EM, and EM treatment with genistein with the doses of 1.3, 1.95, 2.6, and 3.25 mg/day for 14 days. Peritoneal tissues with lesions were collected and analyzed by immunohistochemistry to measure the VEGF-A and ER-α expression. The data were analyzed using a statistical approach using one-way ANOVA followed by Tukey HSD test with a significant value p < 0.05. In silico study was conducted for investigating the inhibition mechanism of genistein in VEGF-A and ER-α protein. Genistein significantly reduced the VEGF-A and ER-α expression with the optimum dose of 3.25 mg/day. Molecular docking showed that genistein inhibited VEGF-A in several active site residues of VEGF-A, also blocked the ER-α protein in estradiol binding sites. This study concluded that genistein prevented endometriosis by performing the antiangiogenic activity and showed a similar function to estradiol.


Assuntos
Endometriose , Fator A de Crescimento do Endotélio Vascular , Humanos , Feminino , Camundongos , Animais , Genisteína/farmacologia , Genisteína/uso terapêutico , Endometriose/tratamento farmacológico , Endometriose/patologia , Simulação de Acoplamento Molecular , Receptores de Estrogênio/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estradiol/farmacologia , Modelos Animais de Doenças
3.
Kidney Blood Press Res ; 49(1): 137-143, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38266504

RESUMO

INTRODUCTION: The process of vascular calcification has severe clinical consequences in a number of diseases, including diabetes, atherosclerosis, and end-stage renal disease. In the present study, we investigated the effect of policosanol (Poli), genistein (Gen), and vitamin D (VitD) separately and in association to evaluate the possible synergistic action on inorganic phosphate (Pi)-induced calcification of vascular smooth muscle cells (VSMCs). METHODS: Primary human VSMCs were cultured with either growth medium or growth medium supplemented with calcium and phosphorus (calcification medium) in combination with Poli, Gen, and VitD. Alizarin Red staining, mineralization, and the protein expression of RUNX2 and superoxide dismutase-2 (SOD2) were investigated. RESULTS: All three substances tested were effective at reducing osteogenic differentiation of VSMCs in a dose-dependent manner. Poli+Gen, Poli+VitD, Gen+VitD treatment induced a greater inhibition of calcification and RUNX2 expression compared to single compounds treatments. Moreover, the association of Poli+Gen+VitD (Reduplaxin®) was more effective at inhibiting VSMCs mineralization and preventing the increase in RUNX2 expression induced by calcification medium but not modified SOD2 expression. CONCLUSIONS: The association of Pol, Gen, and VitD (Reduplaxin®) has an additive inhibitory effect on the calcification process of VSMCs induced in vitro by a pro-calcifying medium.


Assuntos
Álcoois Graxos , Genisteína , Músculo Liso Vascular , Calcificação Vascular , Vitamina D , Humanos , Vitamina D/farmacologia , Álcoois Graxos/farmacologia , Células Cultivadas , Calcificação Vascular/prevenção & controle , Calcificação Vascular/induzido quimicamente , Calcificação Vascular/tratamento farmacológico , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citologia , Genisteína/farmacologia , Genisteína/uso terapêutico , Superóxido Dismutase/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo
4.
Toxicol In Vitro ; 96: 105785, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38266663

RESUMO

Secondary metabolites from medicinal plants have a well-established therapeutic potential, with many of these chemicals having specialized medical uses. Isoflavonoids, a type of secondary metabolite, have little cytotoxicity against healthy human cells, making them interesting candidates for cancer treatment. Extensive research has been conducted to investigate the chemo-preventive benefits of flavonoids in treating various cancers. Biochanin A (BA), an isoflavonoid abundant in plants such as red clover, soy, peanuts, and chickpeas, was the subject of our present study. This study aimed to determine how BA affected glucose-6-phosphate dehydrogenase (G6PD) in human lung cancer cells. The study provides meaningful insight and a significant impact of BA on the association between metastasis, inflammation, and G6PD inhibition in A549 cells. Comprehensive in vitro tests revealed that BA has anti-inflammatory effects. Molecular docking experiments shed light on BA's high binding affinity for the G6PD receptor. BA substantially decreased the expression of G6PD and other inflammatory and metastasis-related markers. In conclusion, our findings highlight the potential of BA as a therapeutic agent in cancer treatment, specifically by targeting G6PD and related pathways. BA's varied effects, which range from anti-inflammatory capabilities to metastasis reduction, make it an appealing option for future investigation in the development of new cancer therapeutics.


Assuntos
Anti-Inflamatórios , Carcinoma Pulmonar de Células não Pequenas , Genisteína , Neoplasias Pulmonares , Humanos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Genisteína/farmacologia , Genisteína/uso terapêutico , Glucosefosfato Desidrogenase , Neoplasias Pulmonares/tratamento farmacológico , Simulação de Acoplamento Molecular
5.
Drug Resist Updat ; 73: 101056, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38277755

RESUMO

BACKGROUND: The treatment of dopamine agonists (DA) resistant prolactinomas remains a formidable challenge, as the mechanism of resistance is still unclear, and there are currently no viable alternative drug therapies available. This study seeks to investigate the mechanism of DA resistance in prolactinomas and identify new potentially effective drugs. METHODS: To explore the mechanism of DA resistance in prolactinomas, this study conducted transcriptome sequencing analysis on 27 cases of DA-resistant prolactinomas and 10 cases of sensitive prolactinomas. In addition, single-cell sequencing analysis was performed on 3 cases of DA-resistant prolactinomas and 3 cases of sensitive prolactinomas. Furthermore, to screen for potential therapeutic drugs, the study successfully established an organoids model for DA-resistant prolactinomas and screened 180 small molecule compounds using 8 organoids. The efficacy of the identified drugs was verified through various assays, including CCK-8, colony formation, CTG, and flow cytometry, and their mechanisms of action were confirmed through WB and IHC. The effectiveness of the identified drugs was evaluated both in vitro and in vivo. RESULTS: The results of transcriptome sequencing and single-cell sequencing analyses showed that DA resistance in prolactinomas is associated with the upregulation of the Focal Adhesion (FA) signaling pathway. Additionally, immunohistochemical validation revealed that FAK and Paxillin were significantly upregulated in DA-resistant prolactinomas. Screening of 180 small molecule compounds using 8 organoids identified Genistein as a potentially effective drug for DA-resistant prolactinomas. Experimental validation demonstrated that Genistein inhibited the proliferation of pituitary tumor cell lines and organoids and promoted apoptosis in pituitary tumor cells. Moreover, both the cell sequencing results and WB validation results of the drug-treated cells indicated that Genistein exerts its anti-tumor effect by inhibiting the FA pathway. In vivo, experiments also showed that Genistein can inhibit subcutaneous tumor formation. CONCLUSION: DA resistance in prolactinomas is associated with upregulation of the Focal Adhesion (FA) signaling pathway, and Genistein can exert its anti-tumor effect by inhibiting the expression of the FA pathway.


Assuntos
Tumores Neuroendócrinos , Neoplasias Hipofisárias , Prolactinoma , Humanos , Neoplasias Hipofisárias/tratamento farmacológico , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Agonistas de Dopamina/farmacologia , Agonistas de Dopamina/uso terapêutico , Prolactinoma/tratamento farmacológico , Prolactinoma/genética , Prolactinoma/metabolismo , Prolactina/metabolismo , Prolactina/uso terapêutico , Genisteína/uso terapêutico , Tumores Neuroendócrinos/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética
6.
Phytother Res ; 38(2): 489-506, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37905329

RESUMO

Cancer is considered a leading cause of mortality. This rising cancer death rate and several existing limitations like side effects, poor efficacies, and high cost of the present chemotherapeutic agents have increased the demand for more potent and alternative cancer treatments. This review elucidated a brief overview of Biochanin A (BCA) and its potentiality on various cancers with details of anticancer mechanism. According to our review, a number of studies including in silico, in vitro, pre-clinical, and clinical trials have tested to evaluate the efficacy of BCA. This compound is effective against 15 types of cancer, including breast, cervical, colorectal, gastric, glioblastoma, liver, lung, melanoma, oral, osteosarcoma, ovarian, pancreatic, pharynx, prostate, and umbilical vein cancer. The general anticancer activities of this compound are mediated via several molecular processes, including regulation of apoptosis, cell proliferation, metastasis and angiogenesis, signaling, enzymatic pathways, and other mechanisms. Targeting both therapeutic and oncogenic proteins, as well as different pathways, makes up the molecular mechanism underlying the anticancer action. Many signaling networks and their components, such as EFGR, PI3K/Akt/mTOR, MAPK, MMP-2, MMP-9, PARP, Caspase-3/8/9, Bax, Bcl2, PDL-1, NF-κB, TNF-α, IL-6, JAK, STAT3, VEGFR, VEGF, c-MY, Cyclin B1, D1, E1 and CDKs, Snail, and E-cadherin proteins, can be regulated in cancer cells by BCA. Such kind of anticancer properties of BCA could be a result of its correct structural chemistry. The use of BCA-based therapies as nano-carriers for the delivery of chemotherapeutic medicines has the potential to be very effective. This natural compound synergises with other natural compounds and standard drugs, including sorafenib, 5-fluorouracil, temozolomide, doxorubicin, apigenin, and genistein. Moreover, proper use of this compound can reverse multidrug resistance through numerous mechanisms. BCA has better drug-likeness and pharmacokinetic properties and is nontoxic (eye, liver, kidney, skin, cardio) in human bodies. As having a wide range of cancer-fighting mechanisms, synergistic effects, and good pharmacokinetic properties, BCA can be used as a supplementary food until standard drugs are available at pharma markets.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Masculino , Humanos , Genisteína/farmacologia , Genisteína/uso terapêutico , Fosfatidilinositol 3-Quinases/metabolismo , Apoptose
7.
Osteoporos Int ; 35(3): 413-430, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37875614

RESUMO

Due to estrogen deficiency, postmenopausal women may suffer from an imbalance in bone metabolism that leads to bone fractures. Isoflavones, a type of phytoestrogen, have been suggested to improve bone metabolism and increase bone mass. Therefore, isoflavones are increasingly recognized as a promising natural alternative to hormone replacement therapy for postmenopausal women who face a heightened risk of osteoporosis and are susceptible to bone fractures. PURPOSE: This study aimed to evaluate the efficacy of isoflavone interventions on bone mineral density (BMD) in postmenopausal women by means of systematic review and meta-analysis. METHODS: The electronic database searches were performed on PubMed, Embase, Scopus, and Cochrane Library databases, covering literature up to April 20, 2023. A random-effects model was used to obtain the main effect estimates, with a mean difference (MD) and its 95% confidence interval (CI) as the effect size summary. The risk of bias assessment was conducted using the Risk of Bias 2 (RoB2) tool. RESULTS: A total of 63 randomized controlled trials comparing isoflavone interventions (n = 4,754) and placebo (n = 4,272) were included. The results indicated that isoflavone interventions significantly improved BMD at the lumbar spine (MD = 0.0175 g/cm2; 95% CI, 0.0088 to 0.0263, P < 0.0001), femoral neck (MD = 0.0172 g/cm2; 95% CI, 0.0046 to 0.0298, P = 0.0073), and distal radius (MD = 0.0138 g/cm2; 95% CI, 0.0077 to 0.0198, P < 0.0001) in postmenopausal women. Subgroup analysis showed that the isoflavone intervention was effective for improving BMD when the duration was ≥ 12 months and when the intervention contained genistein of at least 50 mg/day. CONCLUSION: This systematic review and meta-analysis suggests that isoflavone interventions, especially those containing genistein of at least 50 mg/day, can effectively enhance BMD in postmenopausal women.


Assuntos
Fraturas Ósseas , Isoflavonas , Osteoporose Pós-Menopausa , Feminino , Humanos , Densidade Óssea , Isoflavonas/farmacologia , Isoflavonas/uso terapêutico , Genisteína/farmacologia , Genisteína/uso terapêutico , Osteoporose Pós-Menopausa/tratamento farmacológico , Osteoporose Pós-Menopausa/prevenção & controle , Pós-Menopausa , Ensaios Clínicos Controlados Aleatórios como Assunto , Fraturas Ósseas/tratamento farmacológico
8.
J Mater Chem B ; 11(43): 10404-10417, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37877170

RESUMO

Ulcerative colitis (UC) is an inflammatory disease involving ulcers in the colon and rectum. The conventional treatments for UC still have many limitations, such as non-specific release, adverse effects and low absorption, resulting in the poor bioavailability of therapeutic agents. To address these challenges, targeting delivery systems are required to specifically deliver drugs to the colonic site with controlled release. Herein, we present a novel microgel oral delivery system, loaded with liposome nanoparticles (Li NPs) containing a natural anti-inflammatory compound genistein (Gen) into alginate microgels, thereby achieving the targeted release of Gen in the colonic region and ameliorating UC symptoms. Initially, Gen was loaded into phosphatidylserine (PS)-functionalized Li NPs to form Gen@Li NPs with an average size of 245.9 ± 9.6 nm. In vitro assessments confirmed that Gen@Li NPs efficiently targeted macrophages and facilitated the internalization of Gen into cells. To prevent rapid degradation in the harsh gastrointestinal tract, Gen@Li NPs were further encapsulated into alginate microgels through electric spraying technology, forming Gen@Li microgels. In vivo distribution tests demonstrated that Gen@Li microgels possessed long-term retention in the colon and gradual release characteristics compared to Gen@Li NPs. Furthermore, in vivo experiments confirmed that Gen@Li microgels significantly alleviated UC symptoms in mice induced by dextran sulfate sodium salt (DSS) mainly through reducing the expression levels of pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6) and promoting colonic mucosal barrier repair through upregulation of mucosal protein expression. This study shed light on the potential of utilizing oral administration of natural compounds for UC treatment.


Assuntos
Colite Ulcerativa , Microgéis , Animais , Camundongos , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/metabolismo , Lipossomos/uso terapêutico , Fosfatidilserinas/efeitos adversos , Genisteína/farmacologia , Genisteína/uso terapêutico , Alginatos/uso terapêutico
9.
Molecules ; 28(13)2023 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-37446555

RESUMO

INTRODUCTION: The third-generation of aromatase inhibitors (AIs)-Exemestane (Exe), Letrozole (Let), and Anastrozole (Ana)-is the main therapeutic approach applied for estrogen receptor-positive (ER+) breast cancer (BC), the most common neoplasm in women worldwide. Despite their success, the development of resistance limits their efficacy. Genistein (G), a phytoestrogen present in soybean, has promising anticancer properties in ER+ BC cells, even when combined with anticancer drugs. Thus, the potential beneficial effects of combining G with AIs were investigated in sensitive (MCF7-aro) and resistant (LTEDaro) BC cells. METHODS: The effects on cell proliferation and expression of aromatase, ERα/ERß, and AR receptors were evaluated. RESULTS: Unlike the combination of G with Ana or Let, which negatively affects the Ais' therapeutic efficacy, G enhanced the anticancer properties of the steroidal AI Exe, increasing the antiproliferative effect and apoptosis relative to Exe. The hormone targets studied were not affected by this combination when compared with Exe. CONCLUSIONS: This is the first in vitro study that highlights the potential benefit of G as an adjuvant therapy with Exe, emphasizing, however, that soy derivatives widely used in the diet or applied as auxiliary medicines may increase the risk of adverse interactions with nonsteroidal AIs used in therapy.


Assuntos
Antineoplásicos , Neoplasias da Mama , Feminino , Humanos , Inibidores da Aromatase/farmacologia , Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Genisteína/farmacologia , Genisteína/uso terapêutico , Letrozol , Antineoplásicos/uso terapêutico , Nitrilas/uso terapêutico
10.
Sci Rep ; 13(1): 11225, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37433869

RESUMO

Depression is a common mental disease, with some patients exhibiting ideas and behaviors such as self-harm and suicide. The drugs currently used to treat depression have not achieved good results. It has been reported that metabolites produced by intestinal microbiota affect the development of depression. In this study, core targets and core compounds were screened by specific algorithms in the database, and three-dimensional structures of these compounds and proteins were simulated by molecular docking and molecular dynamics software to further study the influence of intestinal microbiota metabolites on the pathogenesis of depression. By analyzing the RMSD gyration radius and RMSF, it was finally determined that NR1H4 had the best binding effect with genistein. Finally, according to Lipinski's five rules, equol, genistein, quercetin and glycocholic acid were identified as effective drugs for the treatment of depression. In conclusion, the intestinal microbiota can affect the development of depression through the metabolites equol, genistein and quercetin, which act on the critical targets of DPP4, CYP3A4, EP300, MGAM and NR1H4.


Assuntos
Microbioma Gastrointestinal , Biologia de Sistemas , Humanos , Depressão/tratamento farmacológico , Equol , Genisteína/farmacologia , Genisteína/uso terapêutico , Simulação de Acoplamento Molecular , Quercetina/farmacologia , Quercetina/uso terapêutico
11.
Redox Rep ; 28(1): 2218679, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37260037

RESUMO

OBJECTIVES: Gastric ulcer (GU) is a prevalent chronic digestive disease affecting about 10% of the world's population leading to gastrointestinal perforation and bleeding. Genistein is a legume flavonoid with antioxidants, anti-inflammatory and antibacterial activities. Therefore, we aimed to investigate the ability of genistein to reduce experimentally induced GU in rats by affecting gastric tissue fibrosis Wnt/ß-catenin/TGF-ß/SMAD4 pathway. METHODS: Thirty rats were used. Ten rats served as control, and GU was induced in twenty rats using a single dose of indomethacin (80 mg/kg) orally. Following induction of GU, ten were treated with genistein 25 mg/kg orally. The gastric tissues were isolated to investigate markers of gastric fibrosis, Wnt, ß-catenin, transforming growth factor (TGF)-ß, SMAD4, and Protein kinase B (PKB). In addition, gastric sections were stained with PAS and anti-TGF-ß antibodies. RESULTS: Investigation GU micro-images revealed degeneration in both surface cells and glandular epithelial cells, which was improved by genistein. In addition, treatment with genistein significantly reduced the expression of Wnt, ß-catenin, TGF-ß, SMAD4, and PKB. CONCLUSION: Besides antioxidant activity, genistein improves experimentally induced GU in rats, at least in part, via reduction of gastric tissue fibrosis as indicated by reduction in expression of Wnt, ß-catenin, TGF-ß, SMAD4, and PKB.


Assuntos
Genisteína , Úlcera Gástrica , Fator de Crescimento Transformador beta , Animais , Ratos , beta Catenina/metabolismo , Cateninas , Fibrose , Genisteína/uso terapêutico , Úlcera Gástrica/induzido quimicamente , Úlcera Gástrica/tratamento farmacológico , Fator de Crescimento Transformador beta/metabolismo
12.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 2893-2910, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37300702

RESUMO

Genistein, a commonly occurring isoflavone, has recently gained popularity owing to its ever-expanding spectrum of pharmacological benefits. In addition to health benefits such as improved bone health and reduced postmenopausal complications owing to its phytoestrogen properties, it has been widely evaluated for its anti-cancer potential. Several studies have established the potential for its usage in the management of breast, lung, and prostate cancers, and its usage has significantly evolved from early applications in traditional systems of medicine. This review offers an insight into its current status of usage, the chemistry, and pharmacokinetics of the molecule, an exploration of its apoptotic mechanisms in cancer management, and opportunities for synergism to improve therapeutic outcomes. In addition to this, the authors have presented an overview of recent clinical trials, to offer an understanding of contemporary studies and explore prospects for a greater number of focused trials, moving forward. Advancements in the application of nanotechnology as a strategy to improve safety and efficacy have also been highlighted, with a brief discussion of results from safety and toxicology studies.


Assuntos
Isoflavonas , Neoplasias da Próstata , Masculino , Humanos , Genisteína/farmacologia , Genisteína/uso terapêutico , Isoflavonas/farmacologia , Fitoestrógenos/farmacologia , Fitoestrógenos/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Apoptose
13.
Aging (Albany NY) ; 15(9): 3678-3689, 2023 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-37155147

RESUMO

Colorectal cancer (CRC) is presently a health challenge in China. Although clinical chemotherapy is prescribed availably, the negative effects and poor prognoses still occur. Genistein has antitumor properties in our previous studies. However, the molecular mechanisms underlying the anti-CRC effects of genistein remain unclear. Increasing evidences have indicated that the induction of autophagy, one of cell death models, is closely associated with the formation and development of human cancer. In the current study, a systematic bioinformatics approach using network pharmacology and molecular docking imitation was aimed at identifying the pharmacological targets and anti-CRC mechanisms of genistein, characterized by autophagy-related processes and pathways. Moreover, experimental validation was conducted by using clinical and cell culture samples. All 48 potential targets of genistein-anti-CRC-associated autophagy were screened accordingly. Further bioinformatics analyses identified 10 core genistein-anti-CRC targets related to autophagy, and enrichment-assayed results revealed that the biological processes of these core targets might regulate multiple molecular pathways, including the estrogen signaling pathway. Additionally, molecular docking data demonstrated that genistein has a high affinity for epidermal growth factor receptor (EGFR) and estrogen receptor 1 (ESR1). Both EGFR and ESR1 proteins were highly expressed in clinical CRC samples. Preliminary in vitro data showed that genistein effectively reduced cellular proliferation, activated apoptosis, and suppressed EGFR and ESR1 protein expressions in CRC cells. Our research findings uncovered the molecular mechanisms of genistein against CRC, and the potential drug targets associated with autophagy in genistein treatment of CRC were identified and validated experimentally, including EGFR and ESR1.


Assuntos
Neoplasias Colorretais , Genisteína , Humanos , Genisteína/farmacologia , Genisteína/uso terapêutico , Simulação de Acoplamento Molecular , Neoplasias Colorretais/patologia , Receptores ErbB/metabolismo , Transdução de Sinais
14.
Biomed Pharmacother ; 163: 114821, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37167726

RESUMO

Glucocorticoid-induced osteoporosis (GIO) complicates the clinical management of patients subjected to long-term glucocorticoid use. This study explored the effects of genistein on bone loss in a randomized double-blind alendronate-controlled trial in postmenopausal women with GIO. 200 postmenopausal women (taking at least 5 mg of prednisone equivalents) since 3 months, or more, and expected to continue for at least other 12 months, were randomized to receive genistein (54 mg/day daily) or alendronate (70 mg once a week) for 24 months. Both groups received also Calcium and Vitamin D3 supplementation. Median bone mineral density (BMD) at the antero-posterior lumbar spine significantly increased from 0.75 g/cm2 at baseline to 0.77 g/cm2 at 1 year and 0.79 g/cm2 at 2 years in alendronate-treated patients and from 0.77 g/cm2 at baseline to 0.79 g/cm2 at 12 months and to 0.80 g/cm2 at 24 months in genistein recipients. No difference was observed between the two treatments. Median BMD at the femoral neck increased from 0.67 g/cm2 at baseline to 0.68 g/cm2 at 1 year and 0.69 g/cm2 at 2 years in alendronate-treated patients and from 0.68 g/cm2 at baseline to 0.70 g/cm2 at 12 months and to 0.71 g/cm2 at 24 months in genistein recipients. No difference was observed between alendronate and genistein groups in BMD. Regarding bone markers genistein and alendronate statistically decreased c-terminal telopeptide, while osteocalcin, bone-ALP, and sclerostin showed greater changes in genistein treated patients. This randomized clinical trial suggests that genistein aglycone represents an additional therapeutic option for patients with GIO.


Assuntos
Conservadores da Densidade Óssea , Osteoporose Pós-Menopausa , Osteoporose , Humanos , Feminino , Alendronato/uso terapêutico , Glucocorticoides/farmacologia , Genisteína/farmacologia , Genisteína/uso terapêutico , Conservadores da Densidade Óssea/farmacologia , Conservadores da Densidade Óssea/uso terapêutico , Osteoporose/induzido quimicamente , Osteoporose/tratamento farmacológico , Densidade Óssea , Osteoporose Pós-Menopausa/induzido quimicamente , Osteoporose Pós-Menopausa/tratamento farmacológico , Método Duplo-Cego
15.
Int J Mol Sci ; 24(8)2023 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-37108207

RESUMO

Glycine max Merr. (GM) is a functional food that provides many beneficial phytochemicals. However, scientific evidence of its antidepressive and sedative activities is scarce. The present study was designed to investigate the antidepressive and calmative effects of GM and its biologically active compound, genistein (GE), using electroencephalography (EEG) analysis in an electric foot shock (EFS)-stressed rat. The underlying neural mechanisms of their beneficial effects were determined by assessing corticotropin-releasing factor (CRF), serotonin (5-HT), and c-Fos immunoreactivity in the brain using immunohistochemical methods. In addition, the 5-HT2C receptor binding assay was performed because it is considered a major target of antidepressants and sleep aids. In the binding assay, GM displayed binding affinity to the 5-HT2C receptor (IC50 value of 14.25 ± 11.02 µg/mL). GE exhibited concentration-dependent binding affinity, resulting in the binding of GE to the 5-HT2C receptor (IC50, 77.28 ± 26.57 mg/mL). Administration of GM (400 mg/kg) increased non-rapid eye movement (NREM) sleep time. Administration of GE (30 mg/kg) decreased wake time and increased rapid eye movement (REM) and NREM sleep in EPS-stressed rats. In addition, treatment with GM and GE significantly decreased c-Fos and CRF expression in the paraventricular nucleus (PVN) and increased 5-HT levels in the dorsal raphe in the brain. Overall, these results suggest that GM and GE have antidepressant-like effects and are effective in sleep maintenance. These results will benefit researchers in developing alternatives to decrease depression and prevent sleep disorders.


Assuntos
Hormônio Liberador da Corticotropina , Transtornos do Sono-Vigília , Ratos , Animais , Hormônio Liberador da Corticotropina/farmacologia , Genisteína/farmacologia , Genisteína/uso terapêutico , Serotonina/metabolismo , Receptor 5-HT2C de Serotonina , Sono , Hipnóticos e Sedativos/farmacologia , Hipnóticos e Sedativos/uso terapêutico , Eletroencefalografia , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Transtornos do Sono-Vigília/tratamento farmacológico , Transtornos do Sono-Vigília/etiologia
16.
Naunyn Schmiedebergs Arch Pharmacol ; 396(10): 2481-2500, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37083712

RESUMO

Neuroblastoma is one of the most common solid tumors in children younger than 1 year of age, with poor prognosis and survival rates. Therefore, novel molecular targets and therapeutic strategies are needed to prolong patient survival. For this purpose, we investigated the effects of rottlerin and genistein separately and in combination on neuroblastoma cells (SH-SY5Y, Kelly). First, the effects of rottlerin and genistein were investigated on cell proliferation. Different rottlerin (1-50 µM) and genistein (5-150 µM) doses were used as experimental groups compared to the control (DMSO/vehicle). The IC50 dose was found to be 5 µM for rottlerin and 30 µM for genistein (P < 0.0001). Other analyses, such as colony formation assays, annexin V/propidium iodide staining, matrigel invasion assays, and Western blot analysis, were performed with these doses and their combinations. To assess statistical significance, statistical analysis was conducted using the one-way ANOVA with the post hoc Tukey test. Our results showed that IC50 doses of rottlerin and genistein induced a significant reduction in cell proliferation, colony formation, and invasion in neuroblastoma cells (P < 0.0001). The combination of these doses increased the levels of inhibition of cell proliferation and invasion while decreasing the level of apoptosis (P 0.0001). Furthermore, these agents caused G1-cell cycle arrest in these cells. Our western blot data showed that rottlerin and genistein treatments markedly inhibit elongation factor 2 kinase (EF2K) and other pro-tumorigenic, metastatic proteins in neuroblastoma cells. These agents probably showed their anti-proliferative, anti-metastatic, and pro-apoptotic effects through EF2K downregulation. Our results suggested that rottlerin and genistein have inhibitory effects on cancer cell proliferation, invasion, and cell cycle and induce apoptosis in both cell lines. Combined treatment with rottlerin and genistein may be a viable approach and beneficial to neuroblastoma patients as the combined effect significantly suppresses the above-mentioned pathways.


Assuntos
Genisteína , Neuroblastoma , Criança , Humanos , Genisteína/farmacologia , Genisteína/uso terapêutico , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Quinase do Fator 2 de Elongação , Proliferação de Células , Apoptose , Pontos de Checagem da Fase G1 do Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular
17.
Hum Exp Toxicol ; 42: 9603271231164913, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36932924

RESUMO

BACKGROUND: Studies have shown oxidative stress and apoptosis are the main pathogenic mechanisms of renal ischemia/reperfusion (IR) injury (IRI). Genistein, a polyphenolic non-steroidal compound, has been extensively explored in oxidative stress, inflammation and apoptosis. Our research aims to reveal the potential role of genistein on renal IRI and its potential molecular mechanism both in vivo and in vitro. METHODS: In vivo experiments, mice were pretreated with or without genistein. Renal pathological changes and function, cell proliferation, oxidative stress and apoptosis were measured. In vitro experiments, overexpression of ADORA2A and knockout of ADORA2A cells were constructed. Cells proliferation, oxidative stress and apoptosis were analyzed. RESULTS: Our results in vivo showed that the renal damage induced by IR was ameliorated by genistein pretreatment. Moreover, ADORA2A was activated by genistein, along with inhibition of oxidative stress and apoptosis. The results in vitro showed that genistein pretreatment and ADORA2A overexpression reversed the increase of apoptosis and oxidative stress in NRK-52E cells induced by H/R, while the knockdown of ADORA2A partially weakened this reversal from genistein treatment. CONCLUSIONS: Our results demonstrated that genistein have a protective effect against renal IRI by inhibiting oxidative stress and apoptosis via activating ADORA2A, presenting its potential use for the treatment of renal IRI.


Assuntos
Genisteína , Traumatismo por Reperfusão , Camundongos , Animais , Genisteína/farmacologia , Genisteína/uso terapêutico , Genisteína/metabolismo , Rim , Traumatismo por Reperfusão/metabolismo , Estresse Oxidativo , Antioxidantes/farmacologia , Apoptose
18.
Clinics (Sao Paulo) ; 78: 100141, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36905879

RESUMO

OBJECTIVE: The tyrosine-protein kinase inhibitor, genistein, can inhibit cell malignant transformation and has an antitumor effect on various types of cancer. It has been shown that both genistein and KNCK9 can inhibit colon cancer. This research aimed to investigate the suppressive effects of genistein on colon cancer cells and the association between the application of genistein and KCNK9 expression level. METHODS: The Cancer Genome Atlas (TCGA) database was used to study the correlation between the KCNK9 expression level and the prognosis of colon cancer patients. HT29 and SW480 colon cancer cell lines were cultured to examine the inhibitory effects of KCNK9 and genistein on colon cancer in vitro, and a mouse model of colon cancer with liver metastasis was established to verify the inhibitory effect of genistein in vivo. RESULTS: KCNK9 was overexpressed in colon cancer cells and was associated with a shorter Overall Survival (OS), a shorter Disease-Specific Survival (DFS), and a shorter Progression-Free Interval (PFI) of colon cancer patients. In vitro experiments showed that downregulation of KCNK9 or genistein application could suppress cell proliferation, migration, and invasion abilities, induce cell cycle quiescence, promote cell apoptosis, and reduce epithelial-mesenchymal transition of the colon cancer cell line. In vivo experiments revealed that silencing of KCNK9 or application of genistein could inhibit hepatic metastasis from colon cancer. Additionally, genistein could inhibit KCNK9 expression, thereby attenuating Wnt/ß-catenin signaling pathway. CONCLUSION: Genistein inhibited the occurrence and progression of colon cancer through Wnt/ß-catenin signaling pathway that could be mediated by KCNK9.


Assuntos
Neoplasias do Colo , Neoplasias Hepáticas , Animais , Camundongos , Genisteína/farmacologia , Genisteína/uso terapêutico , Proliferação de Células , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Apoptose , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Via de Sinalização Wnt , Linhagem Celular Tumoral , Movimento Celular , Regulação Neoplásica da Expressão Gênica , beta Catenina/genética , beta Catenina/metabolismo , beta Catenina/farmacologia
19.
J Biochem Mol Toxicol ; 37(4): e23298, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36727417

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA) is a highly infectious Gram-positive pathogen known to cause severe diseases such as endocarditis, food poisoning, pneumonia, osteomyelitis, and septicemia. MRSA is a major public health issue. Among these, osteomyelitis is inflammation of the bone caused by the invasion of the bacterial pathogen in the bones. Its prominent symptoms include fever, pain, and redness of bones. In the case of children, it affects the long bones of arms and legs, whereas in the case of adults it affects the hip, feet, and spine. Bacterial osteomyelitis can trigger pathological remodeling of bones and hence causes substantial morbidity and mortality. The present study aims to evaluate the isoflavone genistein's (5,7-dihydroxy-3-(4-hydroxyphenyl)-4H-1-benzopyran-4-one,4',5,7 trihydroxyisoflavone) antimicrobial and anti-inflammatory effects against osteomyelitis induced by MRSA in male Wistar rats. Classification of the animals was into the following: sham (Group I), osteomyelitis (Group II, control), genistein (25 mg/kg body weight, Group III), and genistein (50 mg/kg body weight, Group IV). The rats did not receive any treatment for 4 weeks after bacterial inoculation. Genistein was then administered twice daily for 2 weeks. Bacterial growth, mean body weight bone infection status, and side effects of genistein treatment were assessed. Furthermore, lipid peroxidation, superoxide dismutase, glutathione (GSH) peroxidase, catalase, reduced GSH, tumor necrosis factor-α (TNF-α), and interleukin (IL)-6 were also determined. Two days after treatment, it was found that genistein significantly suppressed bacterial growth and reduced serum pro-inflammatory cytokines TNF-α and IL-6. Therefore, the study suggests that genistein could be a promising lead against MRSA-induced osteomyelitis.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Osteomielite , Infecções Estafilocócicas , Ratos , Masculino , Animais , Staphylococcus aureus , Genisteína/farmacologia , Genisteína/uso terapêutico , Fator de Necrose Tumoral alfa , Ratos Wistar , Antibacterianos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Osteomielite/etiologia , Osteomielite/microbiologia , Glutationa , Interleucina-6/farmacologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico
20.
Biogerontology ; 24(3): 329-346, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36828983

RESUMO

Genistein (GEN) is a non-steroidal phytoestrogen that belongs to the isoflavone class. It is abundantly found in soy. Soy and its products are used as food components in many countries including India. The present review is focused to address roles of GEN in brain functions in the context of learning and memory as a function of aging and neurological disorders. Memory decline is one of the most disabling features observed during normal aging and age-associated neurodegenerative disorders namely Alzheimer's disease (AD) and Parkinson's disease (PD), etc. Anatomical, physiological, biochemical and molecular changes in the brain with advancement of age and pathological conditions lead to decline of cognitive functions. GEN is chemically comparable to estradiol and binds to estrogen receptors (ERs). GEN acts through ERs and mimics estrogen action. After binding to ERs, GEN regulates a plethora of brain functions including learning and memory; however detailed study still remains elusive. Due to the neuroprotective, anti-oxidative and anti-inflammatory properties, GEN is used to restore or improve memory functions in different animal models and humans. The present review may be helpful to understand roles of GEN in learning and memory during aging and neurological disorders, its direction of research and therapeutic perspectives.


Assuntos
Genisteína , Doenças do Sistema Nervoso , Animais , Humanos , Genisteína/farmacologia , Genisteína/uso terapêutico , Fitoestrógenos/farmacologia , Estradiol , Doenças do Sistema Nervoso/tratamento farmacológico , Envelhecimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...