genistein treatment
Recently Published Documents


TOTAL DOCUMENTS

71
(FIVE YEARS 19)

H-INDEX

18
(FIVE YEARS 2)

Author(s):  
Takahiro Ikeda ◽  
Shun Watanabe ◽  
Takakazu Mitani

Abstract Genistein exerts anti-adipogenic effects, but its target molecules remain unclear. Here, we delineated the molecular mechanism underlying the anti-adipogenic effect of genistein. A pulldown assay using genistein-immobilized beads identified adenine nucleotide translocase-2 as a genistein-binding protein in adipocytes. Adenine nucleotide translocase-2 exchanges ADP/ATP through the mitochondrial inner membrane. Similar to the knockdown of adenine nucleotide translocase-2, genistein treatment decreased ADP uptake into the mitochondria and ATP synthesis. Genistein treatment and adenine nucleotide translocase-2 knockdown suppressed adipogenesis and increased phosphorylation of AMP-activated protein kinase. Adenine nucleotide translocase-2 knockdown reduced the transcriptional activity of CCAAT/enhancer-binding protein β, whereas AMP-activated protein kinase inhibition restored the suppression of adipogenesis by adenine nucleotide translocase-2 knockdown. These results indicate that genistein interacts directly with adenine nucleotide translocase-2 to suppress its function. The downregulation of adenine nucleotide translocase-2 reduces the transcriptional activity of CCAAT/enhancer-binding protein β via activation of AMP-activated protein kinase, which consequently represses adipogenesis.


2021 ◽  
Vol 43 (1) ◽  
pp. 127-141
Author(s):  
Matúš Čoma ◽  
Veronika Lachová ◽  
Petra Mitrengová ◽  
Peter Gál

Estrogen deprivation is one of the major factors responsible for many age-related processes including poor wound healing in postmenopausal women. However, the reported side-effects of estrogen replacement therapy (ERT) have precluded broad clinical administration. Therefore, selective estrogen receptor modulators (SERMs) have been developed to overcome the detrimental side effects of ERT on breast and/or uterine tissues. The use of natural products isolated from plants (e.g., soy) may represent a promising source of biologically active compounds (e.g., genistein) as efficient alternatives to conventional treatment. Genistein as natural SERM has the unique ability to selectively act as agonist or antagonist in a tissue-specific manner, i.e., it improves skin repair and simultaneously exerts anti-cancer and chemopreventive properties. Hence, we present here a wound healing phases-based review of the most studied naturally occurring SERM.


Author(s):  
Xiangjun Cheng ◽  
Peilian Xu

The study aimed to investigate the preservative effects of genistein on articular cartilage in an experimental model of knee osteoarthritis in rats. Thirty male Wistar rats were assigned to three equal groups: the sham group (SG), osteoarthritis control group (OAG), and genistein-treated osteoarthritis group (GTG). Intra-articular injections of monosodium iodoacetate were used for osteoarthritis induction. After two weeks of rest for the induction of the inflammatory process, genistein (30 mg/kg/day) vs. saline gavage was administered for eight weeks. The expression of matrix metalloproteinase (MMP) 8 and 13, Sox5/Sox6, Indian hedgehog (IHH), and Col2 were evaluated in medial femoral condyle sections by immunohistochemical staining. The number of chondrocytes and cartilage thicknesses were also measured and compared among the groups. No significant change in cartilage thickness was observed in GTG compared with OAG (p=0.188). Chondrocyte count was significantly higher in the articular cartilage of GTG compared with OAG (p=0.006). Induction of OA significantly increased the expression of MMP-8, MMP-13, and IHH, but decreased Col2, Sox5, and Sox6 expression (p<0.001); these were partially prevented in the GTG. Our findings support the effectiveness of genistein treatment in the prevention of articular cartilage damage in the experimental model of knee osteoarthritis. The proposed mechanism of action is through the suppression of the MMP, IHH, Col2 pathways, besides the induction of Sox5 and Sox6 expression. Novelty: -Genistein prevent articular cartilage damage in the experimental model of knee osteoarthritis. -The osteoprotective effect is trough modulation of expression of MMP, Sox, IHH, and Col2 proteins.


2021 ◽  
Vol 10 (5) ◽  
pp. e36410515061
Author(s):  
Naara Gabriela Monteiro ◽  
Fábio Roberto De Souza Batista ◽  
Maria Isabela Lopes Gandolfo ◽  
Leonardo Perez Faverani ◽  
Letícia Pitol Palin ◽  
...  

The present study aimed to evaluate the peri-implantar bone healing in the presence of genistein treatment in ovariectomized rats. Thirty female rats with 4 months old were divided into 3 groups according to the experimental condition and the drug treatment: SHAM (rats submitted to the fictional surgery and gavage with 0.9% saline solution); OVX (rats submitted to bilateral ovariectomy and gavage with 0.9% saline solution); OVX GEN (rats submitted to bilateral ovariectomy and gavage with 1mg/day of genistein). 60 implants were installed, with two implants in each animal. The calcified group was subjected microcomputerized tomography and the parameters analysed was bone volume per tissue volume (BV/TV) and connective density (Cnn.Dn). The decalcified samples were evaluated through immunolabeling analysis, in order to detect the presence of RUNX2, Alkaline Phosphatase, Osteocalcin, Osteopontin and TRAP. All the quantitative data were submitted to the normality curve to determine the most adequate test. The significance level of p<0.05 was considered for all tests. The morphometric analysis of the OVX GEN group showed higher percentage of bone volume and lower connective density when compared with OVX. Immunohistochemical analysis favors expression. For the markers that positively label osteoblastic activity. This study shows that genistein therapy improves peri-implant bone healing in ovariectomized rats.


2021 ◽  
Vol 22 (8) ◽  
pp. 4220
Author(s):  
Graciela Argüello ◽  
Elisa Balboa ◽  
Pablo J. Tapia ◽  
Juan Castro ◽  
María José Yañez ◽  
...  

Niemann–Pick type C disease (NPCD) is a lysosomal storage disease (LSD) characterized by abnormal cholesterol accumulation in lysosomes, impaired autophagy flux, and lysosomal dysfunction. The activation of transcription factor EB (TFEB), a master lysosomal function regulator, reduces the accumulation of lysosomal substrates in LSDs where the degradative capacity of the cells is compromised. Genistein can pass the blood–brain barrier and activate TFEB. Hence, we investigated the effect of TFEB activation by genistein toward correcting the NPC phenotype. We show that genistein promotes TFEB translocation to the nucleus in HeLa TFEB-GFP, Huh7, and SHSY-5Y cells treated with U18666A and NPC1 patient fibroblasts. Genistein treatment improved lysosomal protein expression and autophagic flux, decreasing p62 levels and increasing those of the LC3-II in NPC1 patient fibroblasts. Genistein induced an increase in β-hexosaminidase activity in the culture media of NPC1 patient fibroblasts, suggesting an increase in lysosomal exocytosis, which correlated with a decrease in cholesterol accumulation after filipin staining, including cells treated with U18666A and NPC1 patient fibroblasts. These results support that genistein-mediated TFEB activation corrects pathological phenotypes in NPC models and substantiates the need for further studies on this isoflavonoid as a potential therapeutic agent to treat NPCD and other LSDs with neurological compromise.


2021 ◽  
Author(s):  
Faeze Daghigh ◽  
Masoumeh Majidi Zolbin ◽  
Pouran Karimi ◽  
Alireza Alihemmati ◽  
Nasser Ahmadiasl

Abstract Background Phytoestrogens are suggested to have estrogenic effects in the pulmonary system and have been revealed with a few adverse side effects. In this study, we tried to investigate the effect of genistein treatment on estrogen deficiency-induced lung injury and demonstrating whether genistein supplementation could replace estrogen hormone in postmenopausal women. Methods Forty adult female rats were divided into four groups; sham: rats that underwent surgery without ovariectomy, OVX: rats that underwent ovariectomies, OVX.E: ovariectomized rats with eight weeks period of estrogen treatment (20µg/kg/day), OVX.Gen: ovariectomized rats with eight week period of genistein treatment (1mg/kg/day). At the end of the experiment, lung tissue was removed and inflammatory and fibrotic biomarkers were evaluated with western blotting technique. Hematoxylin-eosin and immunohistochemical staining were used to evaluate histomorphological changes in the lung tissue. Results Genistein treatment restored ERK1/2, TGFβ1, MMP2, and IL1β, Bcl-2, and caspase3 expression levels, implying the effectiveness of genistein supplementation in targeting estrogen deficiency symptoms. Conclusions Genistein supplementation exerted protective effects against ovariectomy-induced lung injury with reducing inflammation and fibrosis, moreover, it can be recommended as a natural alternative to postmenopausal hormone therapy.


Molecules ◽  
2021 ◽  
Vol 26 (4) ◽  
pp. 1156
Author(s):  
Lena Seidemann ◽  
Anne Krüger ◽  
Victoria Kegel-Hübner ◽  
Daniel Seehofer ◽  
Georg Damm

Nonalcoholic fatty liver disease (NAFLD) is among the leading causes of end-stage liver disease. The impaired hepatic lipid metabolism in NAFLD is exhibited by dysregulated PPARα and SREBP-1c signaling pathways, which are central transcription factors associated with lipid degradation and de novo lipogenesis. Despite the growing prevalence of this disease, current pharmacological treatment options are unsatisfactory. Genistein, a soy isoflavone, has beneficial effects on lipid metabolism and may be a candidate for NAFLD treatment. In an in vitro model of hepatic steatosis, primary human hepatocytes (PHHs) were incubated with free fatty acids (FFAs) and different doses of genistein. Lipid accumulation and the cytotoxic effects of FFAs and genistein treatment were evaluated by colorimetric and enzymatic assays. Changes in lipid homeostasis were examined by RT-qPCR and Western blot analyses. PPARα protein expression was induced in steatotic PHHs, accompanied by an increase in CPT1L and ACSL1 mRNA. Genistein treatment increased PPARα protein expression only in control PHHs, while CPTL1 and ACSL1 were unchanged and PPARα mRNA was reduced. In steatotic PHHs, genistein reversed the increase in activated SREBP-1c protein. The model realistically reflected the molecular changes in hepatic steatosis. Genistein suppressed the activation of SREBP-1c in steatotic hepatocytes, but the genistein-mediated effects on PPARα were abolished by high hepatic lipid levels.


2021 ◽  
Author(s):  
Qing-peng Hu ◽  
Xiang-yi Huang ◽  
Wei Feng ◽  
Fen-fang Chen ◽  
Hong-xia Yan ◽  
...  

Abstract Background: Epilepsy is a common chronic neurological disease caused by the over-synchronization of neurons that lead to brain dysfunction. Recurrent seizures or status epilepticus can cause irreversible brain damage. The JAK2-STAT3 signal transduction pathway is stimulated by cytokines and involved in various pathological processes including inflammation, apoptosis and immune regulation in central system diseases. Keap1/Nrf2 is an important anti-oxidative stress pathway, which can reduce the toxic effects of oxygen free radicals and endogenous toxins on neurons. Genistein (Gen) can modulate inflammation and neuronal apoptosis, and may thereby have antiepileptic effects. This study aimed to explore the regulation of Genistein on JAK2/STAT3 and Keap1/Nrf2 signaling pathway and the protective effects on brain injury after epilepsy. Methods: Pentylenetetrazole (PTZ) was used to induce epilepsy in developing rats and Genistein was used for pretreatment of epilepsy. The seizure latency, grade scores and duration of the first generalized tonic-clonic seizure (GTCs) were recorded. Hippocampus tissue was sampled at 24 hours post-epilepsy. Immunofluorescence staining was used to observe the number of mature neurons, activated microglia and astrocytes in the hippocampal CA1 region. Western blot and qRT-PCR were used to determine the protein and mRNA levels of p-JAK2, p-STAT3, TNF-α, IL-1β, Keap1, Nrf2, HO-1, NQO1, caspase3, Bax and Bcl2 in the hippocampus. Results: Immunofluorescence showed that the number of neurons significantly decreased, and activated microglia and astrocytes significantly increased after epilepsy; Western blot and q-PCR showed that the expressions of p-JAK2, p-STAT3, TNF-α, IL-1β, Keap1, caspase3 and Bax significantly increased, while Nrf2, HO-1, NQO1 and Bcl-2 were significantly reduced after epilepsy. These effects were reversed by Genistein treatment. Moreover, Genistein was found to prolong seizure latency and reduce seizure intensity score and duration of generalized tonic-clonic seizures(GTCs). Conclusions: Genistein can activate the Keap1/Nrf2 antioxidant stress pathway and attenuate the activation of microglia and astrocytes. Genistein also inhibits the JAK2-STAT3 inflammation pathway and expression of apoptotic proteins, and increases the number of surviving neurons, thus having a protective effect on epilepsy-induced brain damage.


Nutrients ◽  
2020 ◽  
Vol 12 (11) ◽  
pp. 3410
Author(s):  
Carmen P. Ortega-Santos ◽  
Layla Al-Nakkash ◽  
Corrie M. Whisner

Genistein (Gen) and exercise (Exe) have been postulated as potential strategies to ameliorate obesity, inflammation, and gut microbiota (GM) with promising results. However, the impact of the combination of both Exe and Gen is yet to be investigated. We aimed to analyze the impacts of Exe, Gen, and their combined effects on GM and inflammation in mice after a 12-week high-fat, high-sugar diet (HFD). Eighty-three C57BL/6 mice were randomized to control, HFD, HFD + Exe, HFD + Gen, or HFD + Exe + Gen. The V4 region of the 16S rRNA gene was analyzed with Illumina MiSeq. Serum samples were used to analyze interleukin (Il)-6 and Tumor Necrosis Factor alpha (TNF-alpha). The HFD + Exe and HFD + Exe + Gen treatments resulted in significantly greater microbial richness compared to HFD. All the treatments had a significantly different impact on the GM community structure. Ruminococcus was significantly more abundant after the HFD + Exe + Gen treatment when compared to all the other HFD groups. Exe + Gen resulted in serum Il-6 concentrations similar to that of controls. TNF-alpha concentrations did not differ by treatment. Overall, Exe had a positive impact on microbial richness, and Ruminococcus might be the driving bacteria for the GM structure differences. Exe + Gen may be an effective treatment for preventing HFD-induced inflammation.


2020 ◽  
Vol 2020 ◽  
pp. 1-15
Author(s):  
Qiuchen Cheng ◽  
Wen Qin ◽  
Yanhong Yu ◽  
Guojian Li ◽  
Jizhou Wu ◽  
...  

The objective of this study is to improve the bioavailability of genistein by encapsulation with polyethylene glycol-polylactic acid (PEG-PLA) copolymers. Genistein micelles (GMs) prepared using a modified emulsion-evaporation method were more stable than those made with the original method. The effect of polyvinyl alcohol, Tween 80, sonication time, PEG-PLA/genistein ratio, and organic phase (acetone)/H2O ratio on the size, polydispersity index, encapsulation efficiency, and drug loading efficiency of GMs was investigated. GMs were obtained and characterized under optimal experimental conditions. For long-term storage, GMs were lyophilized by freeze drying with trehalose to produce genistein lyophilized powder (GLP). The analysis of GLP by Fourier-transform infrared spectroscopy and differential scanning calorimetry showed that genistein was successfully incorporated into the micellar structure. In vitro release experiments revealed that the incorporation of genistein into PEG-PLA copolymers significantly improved its solubility and bioavailability. GLP was more potent in inhibiting the proliferation of HSC-T6 cells than genistein. Treatment with GLP at 10–20 μg/mL for 48 h significantly inhibited the protein expression of α-smooth muscle actin and collagen I in HSC-T6 cells compared with the control. These data demonstrated that the improved solubility and bioavailability of genistein in the form of GLP enhanced its antifibrotic effect in vitro.


Sign in / Sign up

Export Citation Format

Share Document