Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
1.
Cells ; 12(13)2023 07 07.
Article in English | MEDLINE | ID: mdl-37443838

ABSTRACT

Sertoli cells are essential for germ cell development and function. Their disruption by endocrine disrupting chemicals (EDCs) or drugs could jeopardize spermatogenesis, contributing to male infertility. Perinatal exposure to EDCs and acetaminophen (APAP) disrupts male reproductive functions in animals and humans. Infants can be exposed simultaneously to the dietary soy phytoestrogen genistein (GEN) and APAP used for fever or pain relief. Our goal was to determine the effects of 10-100 µM APAP and GEN, alone or mixed, on immature Sertoli cells using mouse TM4 Sertoli cell line and postnatal-day 8 rat Sertoli cells, by measuring cell viability, proliferation, prostaglandins, genes and protein expression, and functional pathways. A value of 50 µM APAP decreased the viability, while 100 µM APAP and GEN decreased the proliferation. Sertoli cell and eicosanoid pathway genes were affected by GEN and mixtures, with downregulation of Sox9, Cox1, Cox2, and genes relevant for Sertoli cell function, while genes involved in inflammation were increased. RNA-seq analysis identified p53 and TNF signaling pathways as common targets of GEN and GEN mixture in both cell types. These results suggest that APAP and GEN dysregulate immature Sertoli cell function and may aid in elucidating novel EDC and drug targets contributing to the etiology of male infertility.


Subject(s)
Genistein , Infertility, Male , Animals , Female , Male , Mice , Pregnancy , Rats , Acetaminophen/adverse effects , Genistein/adverse effects , Infertility, Male/chemically induced , Infertility, Male/metabolism , Rodentia , Sertoli Cells/metabolism
2.
J Epidemiol ; 33(7): 342-349, 2023 07 05.
Article in English | MEDLINE | ID: mdl-34924453

ABSTRACT

BACKGROUND: The number of people with cognitive impairment, including dementia, in the world is steadily increasing. Although the consumption of isoflavones and soy is associated with a reduced risk of cardiovascular disease, it might also be associated with cognitive impairment. The low number of studies investigating the association between soy/isoflavone intake and cognitive function warrant additional research. METHODS: The Japan Public Health Center-based prospective (JPHC) Study is a large population-based cohort. Midlife dietary intake of soy and the isoflavone genistein was assessed on two occasions: in the years 1995 and 2000. In 2014-2015, 1,299 participants from Nagano prefecture completed a mental health screening. Of these, a total of 1,036 participants were included in analyses. Logistic regression was used to determine Odds Ratios (OR) and 95% Confidence Intervals (CI) for the association between midlife energy-adjusted genistein and soy food intake and cognitive impairment. RESULTS: There were 392 cases of cognitive impairment (346 cases of MCI and 46 cases of dementia). Compared to the lowest dietary quartile of energy-adjusted genistein intake, the highest quartile was significantly associated with cognitive impairment (OR = 1.51; 95% CI, 1.02-2.24; P for trend = 0.03) in the final multivariable analysis. CONCLUSION: High midlife intake of the isoflavone genistein is associated with late-life cognitive impairment.


Subject(s)
Cognitive Dysfunction , Dementia , Isoflavones , Soy Foods , Humans , Genistein/adverse effects , Isoflavones/adverse effects , Prospective Studies , Public Health , Japan/epidemiology , Mental Health , Risk Factors , Surveys and Questionnaires , Cognitive Dysfunction/epidemiology , Dementia/epidemiology
3.
Clin Pharmacol Drug Dev ; 12(2): 190-201, 2023 02.
Article in English | MEDLINE | ID: mdl-36301689

ABSTRACT

A pharmaceutical formulation of genistein, produced as an amorphous solid dispersion by hot melt extrusion (genistein HME), has been developed that can be administered prophylactically to improve outcomes and survival following radiation exposure. Here, genistein HME was evaluated in a phase 1, open-label, single ascending dose (SAD) and multiple single dose (MSD) study enrolling 34 healthy volunteers. In the SAD study, participants were administered a single dose (500, 1000, 2000, or 3000 mg) and in the MSD study, participants were administered a single daily dose for six consecutive days (3000 mg/day). The overall adverse event profile and pharmacokinetics of genistein HME were determined. Additionally, biomarkers of genistein HME were evaluated by profiling whole blood for changes in gene expression by RNA sequencing. Genistein HME was found to be safe at doses up to 3000 mg. Most toxicities were mild to moderate gastrointestinal events, and no dose-limiting toxicities were reported. The maximum tolerated dose was not determined and the no observable adverse effect level was 500 mg. Genistein HME bioavailability greatly increased between the 2000 mg and 3000 mg doses. RNA sequencing analysis revealed that the majority of drug-related changes in gene expression occurred 8-12 hours after the sixth dose in the MSD study. Based on these results, the putative effective dose in humans is 3000 mg.


Subject(s)
Genistein , Radiation-Protective Agents , Humans , Biological Availability , Biomarkers/blood , Drug Compounding/methods , Genistein/adverse effects , Genistein/blood , Genistein/pharmacokinetics , Healthy Volunteers , Radiation-Protective Agents/adverse effects , Radiation-Protective Agents/pharmacokinetics
4.
J Egypt Natl Canc Inst ; 34(1): 37, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36058937

ABSTRACT

BACKGROUND: About 7 million people die from various types of cancer every year representing nearly 12.5% of deaths worldwide. This fact raises the demand to develop new, effective anticancer, onco-suppressive, and chemoprotective agents for the future fighting of cancers. Genistein exhibits pleiotropic functions in cancer, metabolism, and inflammation. It functions as an antineoplastic agent through its effect on the cell cycle, apoptotic processes, angiogenesis, invasion, and metastasis. AIM OF THE STUDY: The current study aimed to study the genistein onco-suppressive effects during 7,12-dimethylbenz[a]anthracene (DMBA)-induced oral carcinogenesis in hamsters' buccal pouch utilizing flow cytometry analysis (FMA), as a fast-diagnosing tool, in addition to the histopathology. MATERIAL AND METHODS: The buccal mucosa of adult male Syrian hamsters was painted with paraffin oil only (group 1), DMBA mixed in mineral oil (group 2), or orally administrated genistein along with painting DMBA (group 2B). The buccal mucosa was utilized for flow cytometric analysis and histopathological examination. RESULTS: Grossly, DMBA-induced carcinogenesis started at the 9th week. Progressive signs appeared in the following weeks reaching to large ulcerative oral masses and exophytic nodules at the 21st week. Histologically, invasive well-differentiated oral squamous cell carcinoma (OSCC) appeared in the underlying tissues from the 12th week, showing malignant criteria. Genistein had delayed clinicopathological change, which started 6 weeks later, than the DMBA-painted hamsters, as mild epithelial dysplastic changes. This became moderate during the last 6 weeks, without dysplastic changes. Flow cytometry revealed that DMBA led to considerable variation in DNA proliferation activity, aneuploid DNA pattern, in 47.22% of hamsters and significantly raised the S-phase fragment (SPF) values, which drastically reduced after genistein treatment. CONCLUSION: Taken together, genistein could be employed as an onco-suppressive agent for carcinogenesis. Moreover, FMA could be used as an aiding fast tool for diagnosis of cancer.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Carcinogenesis/pathology , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cricetinae , Genistein/adverse effects , Humans , Male , Mesocricetus , Mouth Neoplasms/chemically induced , Mouth Neoplasms/drug therapy , Squamous Cell Carcinoma of Head and Neck
5.
Nutrients ; 14(9)2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35565830

ABSTRACT

Skeletal muscle atrophy is a complex degenerative disease characterized by decreased skeletal muscle mass, skeletal muscle strength, and function. MicroRNAs (miRNAs) are a potential therapeutic target, and natural products that regulate miRNA expression may be a safe and effective treatment strategy for muscle atrophy. Previous studies have shown beneficial effects of genistein treatment on muscle mass and muscle atrophy, but the mechanism is not fully understood. Differential co-expression network analysis revealed that miR-222 was upregulated in multiple skeletal muscle atrophy models. Subsequent in vitro (C2C12 myoblasts) and in vivo (C57BL/6 mice) experiments showed that genistein could alleviate dexamethasone-induced muscle atrophy and downregulate the expression of miR-222 in muscle tissue and C2C12 myotubes. The dual-luciferase reporter assay system confirmed that IGF1 is a target gene of miR-222 and is regulated by genistein. In C2C12 myotubes, both dexamethasone and miR-222 overexpression promoted muscle atrophy, however, this function was significantly reduced after genistein treatment. Furthermore, we also observed that both genistein and miR-222 antagomiR could significantly inhibit dexamethasone-induced muscle atrophy in vivo. These results suggest that miR-222 may be involved in the regulation of genistein on muscle atrophy, and genistein and miR-222 may be used to improve muscle health.


Subject(s)
Genistein , MicroRNAs , Animals , Cell Line , Dexamethasone/adverse effects , Genistein/adverse effects , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle Fibers, Skeletal , Muscle, Skeletal/metabolism , Muscular Atrophy/chemically induced , Muscular Atrophy/drug therapy , Muscular Atrophy/genetics
6.
Nutrients ; 14(8)2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35458112

ABSTRACT

Genistein is an isoflavone phytoestrogen that has been shown to improve obesity; however, the underlying molecular mechanisms involved therein have not been clearly elucidated. In this study, we administered genistein to high-fat diet-induced obese mice to investigate its effect on hepatic gluconeogenesis. The results showed that genistein treatment significantly inhibited body weight gain, hyperglycemia, and adipose and hepatic lipid deposition in high-fat diet-induced obese mice. Glucose tolerance test (GTT), insulin tolerance test (ITT) and pyruvate tolerance test (PTT) showed that genistein treatment significantly inhibited gluconeogenesis and improved insulin resistance in obese mice. In addition, this study also found that genistein could promote the expression of miR-451 in vitro and in vivo, and the dual-luciferase reporter system showed that G6pc (glucose-6-phosphatase) may be a target gene of miR-451. Both genistein treatment and in vivo injection of miR-451 agomir significantly inhibited gluconeogenesis and inhibited the expression of G6pc and Gk (glycerol kinase, a known target gene of miR-451). In conclusion, genistein may inhibit gluconeogenesis in obese mice by regulating the expression of Gk and G6pc through miR-451. These results may provide insights into the functions of miR-451 and food-derived phytoestrogens in ameliorating and preventing gluconeogenesis-related diseases.


Subject(s)
Insulin Resistance , MicroRNAs , Animals , Diet, High-Fat/adverse effects , Genistein/adverse effects , Gluconeogenesis , Insulin Resistance/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , MicroRNAs/genetics , MicroRNAs/metabolism , Obesity/chemically induced , Obesity/etiology , Phytoestrogens/adverse effects
7.
Probiotics Antimicrob Proteins ; 14(3): 560-572, 2022 06.
Article in English | MEDLINE | ID: mdl-35411448

ABSTRACT

Soy isoflavone (SIF), a natural phytoestrogen, is used in the condition of hormonal imbalance. These isoflavones generally have low solubility resulting in low bioavailability and bioactivity. It is reported that trans-glycosylation by cyclodextrin glycosyltransferase (CGTase) is widely utilized for increasing the solubility and bioavailability of isoflavones. Present investigation was aimed to study the effect of Bacillus coagulans (a probiotic) in potentiating the bioactivity of soy isoflavones in letrozole-induced PCOS. Initial consideration was focused on proving CGTase assay of B. coagulans. After that, animal study was performed to check the enhancement of bioactivity of SIF along with B. coagulans. A total of 36 rats, separated into six groups (6 rats in each), were used. Group I received vehicles, group II received letrozole (1 mg/kg) for 21 days, and group III animals were administered with soy isoflavones (SIF-100 mg/kg). In the case of group IV, V, and VI, animals received SIF (100 mg/kg) along with B. coagulans 0.65, 3.25, and 6.50 mg/kg, respectively. Treatment was given for 2 weeks after induction of disease. All the animals were sacrificed at the end of the study and endpoint parameters were performed. Present investigation revealed that combination of SIF with B. coagulans showed hormone restoration, reduce oxidative stress, recovery in the menstrual cycle, and improvement in ovarian physiology. SIF (genistein & daidzein) together with B. coagulans exhibits a beneficial role in the enhancement of the bioactivity of soy isoflavones. Further, it showed that a higher dose of B. coagulans (6.50 mg/kg) is more effective in ameliorating the PCOS symptoms.


Subject(s)
Bacillus coagulans , Isoflavones , Polycystic Ovary Syndrome , Animals , Female , Genistein/adverse effects , Hormones , Humans , Isoflavones/adverse effects , Letrozole/adverse effects , Rats
8.
Sci Rep ; 11(1): 18297, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521933

ABSTRACT

Genistein is applied worldwide as an alternative medicament for psoriasis (Ps) because of its anti-inflammatory activity and perceived beneficial impact on the skin. Hereby, we report our in vivo and in vitro investigations to supplement scientific research in this area. The reduction of clinical and biochemical scores in mild to moderate Ps patients taking genistein, its safety, good tolerability with no serious adverse events or discontinuations of treatment, no dose-limiting toxicities, negligible changes in pharmacodynamic parameters and remarkable serum interleukin level alterations were documented in this study. A certain regression of the Ps phenotype was visible, based on photo-documented Ps lesion evaluation. Through in vitro experiments, we found that genistein reduced IL-17A and TNF-α induced MAPK, NF-κB, and PI3K activation in normal human epidermal keratinocytes. Moreover, at the mRNA level of genes associated with the early inflammatory response characteristic for Ps (CAMP, CCL20, DEFB4A, PIK3CA, S100A7, and S100A9) and key cellular signalling (MTORC1 and TFEB), we showed that this isoflavone attenuated the increased response of IL-17A- and TNF-α-related pathways. This allows us to conclude that genistein is a good candidate for Ps treatment, being attractive for co-pharmacotherapy with other drugs.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Genistein/therapeutic use , Psoriasis/drug therapy , Adult , Anti-Inflammatory Agents/adverse effects , Cell Line , Cytokines/blood , Female , Fluorescent Antibody Technique , Genistein/adverse effects , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Male , Middle Aged , Psoriasis/blood , Real-Time Polymerase Chain Reaction
9.
Expert Opin Investig Drugs ; 29(5): 429-441, 2020 May.
Article in English | MEDLINE | ID: mdl-32450051

ABSTRACT

INTRODUCTION: There are no radioprotectors currently approved by the United States Food and Drug Administration (US FDA) for either the hematopoietic acute radiation syndrome (H-ARS) or for the acute radiation gastrointestinal syndrome (GI-ARS). There are currently, however, three US FDA-approved medicinals that serve to mitigate acute irradiation-associated hematopoietic injury. AREA COVERED: We present the current status of a promising radiation countermeasure, BIO 300 (a genistein-based agent), that has been extensively investigated in murine models of H-ARS and models of the delayed effects of acute radiation exposure (DEARE) and is currently being evaluated in large animal models. It is also being developed for the prevention of radiation-induced toxicities associated with solid tumor radiotherapy and is the subject of two active Investigational New Drug (IND) applications. We have included a listing and brief review of significant investigations of this promising medical countermeasure. EXPERT OPINION: BIO 300 is a leading radioprotector under advanced development for H-ARS and DEARE, as well as for select oncologic indication(s). Efficacy following oral administration (po), lack of clinical side effects, storage at ambient temperature, and intended dual use makes BIO 300 an ideal candidate for military and civilian use as well as for storage in the Strategic National Stockpile.


Subject(s)
Acute Radiation Syndrome/prevention & control , Genistein/pharmacology , Radiation-Protective Agents/pharmacology , Animals , Drug Approval , Genistein/administration & dosage , Genistein/adverse effects , Humans , Mice , Neoplasms/radiotherapy , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/adverse effects , United States , United States Food and Drug Administration
10.
Adv Biosyst ; 4(4): e1900187, 2020 04.
Article in English | MEDLINE | ID: mdl-32293160

ABSTRACT

Isoflavones are a class of flavonoids present in legumes and are called phytoestrogens because of their estrogen-like activity. Endogenous estrogen is well known to regulate mammary gland morphogenesis during pregnancy. Each isoflavone also has different physiological activities. However, it is difficult to investigate the direct effect of each isoflavone in mammary morphogenesis in vivo because isoflavones are metabolized into different isoflavones by enteric bacteria. In this study, investigated are the direct influences of coumestrol, daidzein, and genistein on mammary structure development and future milk production ability of mammary epithelial cells (MECs) using in vitro culture models. Mouse MECs are cultured in Matrigel with basic fibroblast growth factor and epidermal growth factor to induce ductal branching and alveolar formation, respectively. Coumestrol and genistein inhibit ductal branching and alveolar formation by affecting the proliferation and migration of MECs with the induction of apoptosis. Daidzein hardly influences mammary structure development. Furthermore, pretreatment with coumestrol adversely affects the induction of milk production ability of MECs. These results suggest that each isoflavone differentially influences mammary morphogenesis and future milk production by affecting MEC behaviors. These results also suggest that the culture models are effective to study mammary epithelial morphogenesis in vitro.


Subject(s)
Apoptosis/drug effects , Coumestrol/adverse effects , Epithelial Cells/metabolism , Genistein/adverse effects , Lactation/drug effects , Mammary Glands, Animal/metabolism , Animals , Coumestrol/pharmacology , Epithelial Cells/pathology , Female , Genistein/pharmacology , Mammary Glands, Animal/pathology , Mice , Mice, Inbred ICR
11.
Environ Health Perspect ; 128(3): 37001, 2020 03.
Article in English | MEDLINE | ID: mdl-32186404

ABSTRACT

BACKGROUND: Embryo implantation relies on precise hormonal regulation, associated gene expression changes, and appropriate female reproductive tract tissue architecture. Female mice exposed neonatally to the phytoestrogen genistein (GEN) at doses similar to those in infants consuming soy-based infant formulas are infertile due in part to uterine implantation defects. OBJECTIVES: Our goal was to determine the mechanisms by which neonatal GEN exposure causes implantation defects. METHODS: Female mice were exposed to GEN on postnatal days (PND)1-5 and uterine tissues collected on PND5, PND22-26, and during pregnancy. Analysis of tissue weights, morphology, and gene expression was performed using standard histology, confocal imaging with three-dimensional analysis, real-time reverse transcription polymerase chain reaction (real-time RT-PCR), and microarrays. The response of ovariectomized adults to 17ß-estradiol (E2) and artificial decidualization were measured. Leukemia inhibitory factor (LIF) injections were given intraperitoneally and implantation sites visualized. Gene expression patterns were compared with curated data sets to identify upstream regulators. RESULTS: GEN-exposed mice exhibited reduced uterine weight gain in response to E2 treatment or artificial decidualization compared with controls; however, expression of select hormone responsive genes remained similar between the two groups. Uteri from pregnant GEN-exposed mice were posteriorized and had reduced glandular epithelium. Implantation failure was not rescued by LIF administration. Microarray analysis of GEN-exposed uteri during early pregnancy revealed significant overlap with several conditional uterine knockout mouse models, including Foxa2, Wnt4, and Sox17. These models exhibit reduced endometrial glands, features of posteriorization and implantation failure. Expression of Foxa2, Wnt4, and Sox17, as well as genes important for neonatal uterine differentiation (Wnt7a, Hoxa10, and Msx2), were severely disrupted on PND5 in GEN-exposed mice. DISCUSSION: Our findings suggest that neonatal GEN exposure in mice disrupts expression of genes important for uterine development, causing posteriorization and diminished gland function during pregnancy that contribute to implantation failure. These findings could have implications for women who consumed soy-based formulas as infants. https://doi.org/10.1289/EHP6336.


Subject(s)
Embryo Implantation/drug effects , Genistein/adverse effects , Phytoestrogens/adverse effects , Uterus/drug effects , Animals , Female , Mice , Pregnancy , Uterus/growth & development , Uterus/physiopathology
12.
Menopause ; 27(1): 57-65, 2020 01.
Article in English | MEDLINE | ID: mdl-31567873

ABSTRACT

OBJECTIVE: PhytoSERM is a selective estrogen receptor beta (ERß) modulator comprised of three phytoestrogens: genistein, daidzein, and S-equol. The PhytoSERM formulation promotes estrogenic action in the brain while largely inactive or inhibitory in reproductive tissue. A phase Ib/IIa clinical trial (ClinicalTrial.gov ID: NCT01723917) of PhytoSERM demonstrated safety and pharmacokinetics profile of PhytoSERM. While this study was not powered for efficacy analysis, we conducted a pilot, retrospective analysis to identify potential responders to PhytoSERM treatment, and to determine the optimal populations to pursue in a phase II clinical trial of efficacy of the PhytoSERM formulation. METHODS: In this retrospective analysis involving 46 participants (n = 16, placebo; n = 18, 50 mg/d PhytoSERM; and n = 12, 100 mg/d PhytoSERM), the therapeutic effect of PhytoSERM was stratified by 2 genetic risk modulators for Alzheimer's disease: mitochondrial haplogroup and APOE genotype. RESULTS: Our retrospective responder analysis indicated that participants on 50 mg of daily PhytoSERM (PS50) for 12 weeks significantly reduced hot flash frequency compared with their baseline (mean [95% CI])-1.61, [-2.79, -0.42], P = 0.007). Participants on 50 mg of PhytoSERM also had significantly greater reduction in hot flash frequency at 12 weeks compared with the placebo group (-1.38, -0.17 [median PS50, median placebo], P = 0.04). Fifty milligrams of daily PhytoSERM also preserved cognitive function in certain aspects of verbal learning and executive function. Our analysis further suggests that mitochondrial haplogroup and APOE genotype can modify PhytoSERM response. CONCLUSION: Our data support a precision medicine approach for further development of PhytoSERM as a safe and effective alternative to hormone therapy for menopause-associated hot flash and cognitive decline. While definitive determination of PhytoSERM efficacy is limited by the small sample size, these data provide a reasonable rationale to extend analyses to a larger study set powered to address statistical significance.


Subject(s)
Apolipoproteins E/genetics , Cognitive Dysfunction/drug therapy , Equol/administration & dosage , Genistein/administration & dosage , Haplotypes , Hot Flashes/drug therapy , Isoflavones/administration & dosage , Menopause , Mitochondria/genetics , Phytoestrogens/administration & dosage , Selective Estrogen Receptor Modulators/administration & dosage , Cognition/drug effects , Cognitive Dysfunction/genetics , Double-Blind Method , Equol/adverse effects , Feasibility Studies , Female , Genistein/adverse effects , Hot Flashes/genetics , Humans , Isoflavones/adverse effects , Middle Aged , Phytoestrogens/adverse effects , Pilot Projects , Retrospective Studies , Selective Estrogen Receptor Modulators/adverse effects , Treatment Outcome
13.
Food Chem Toxicol ; 135: 110982, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31747621

ABSTRACT

With epidemic of obesity, it affects aspects of female reproduction. Genistein could ameliorate obesity in people and animals, but might exert adverse effects on the female reproductive system. To evaluate the effects of fetal and neonatal genistein exposure on the ovarian health of F1 obese female mice with obesity induced by high-fat diet after weaning, we simulated a diet-induced obesity model to observe and determine biological effects of genistein exposure on the ovarian follicle of overfed female mice. Results showed that F1 female mice with obesity induced by high-fat diet significantly prolonged the estrus cycle, disrupted sex hormonal balance and ovarian follicle development after they were exposed to 25 mg/kg b.w./day of genistein during the fetal and neonatal stages. Genistein significantly up-regulated the ovarian mRNA expression of estrogen receptor beta in F1 obese female mice, and high-fat diet influenced the ovarian mRNA expression of estrogen receptor alpha, luteinizing hormone receptor and follicle-stimulating hormone receptor. Hence, genistein exposure from the fetal stage might increase the risk of reproductive diseases in obese females in later life. Thus, the long-term risks of genistein to obese females should be thoroughly assessed.


Subject(s)
Diet, High-Fat , Genistein/adverse effects , Obesity/drug therapy , Ovarian Follicle/drug effects , Animals , Animals, Newborn , Estradiol/metabolism , Estrogen Receptor beta/genetics , Estrous Cycle/drug effects , Female , Fetus/drug effects , Follicle Stimulating Hormone/metabolism , Gene Expression/drug effects , Luteinizing Hormone/metabolism , Mice, Inbred ICR , Obesity/metabolism , Ovarian Follicle/embryology , Ovarian Follicle/pathology , Pregnancy , RNA, Messenger/metabolism
14.
Cancer Epidemiol Biomarkers Prev ; 29(2): 500-508, 2020 02.
Article in English | MEDLINE | ID: mdl-31826911

ABSTRACT

BACKGROUND: Very few previous studies have examined the relationship between thyroid cancer risk and intake of phytoestrogens (PE); furthermore, these studies have reached inconsistent results. METHODS: We analyzed data from a population-based case-control study in Connecticut from 2010 to 2011, including 387 histologically confirmed thyroid cancer cases and 433 population-based controls, with compound data available concerning specific PEs. Multivariate unconditional logistic regression models were used to estimate the associations between specific PEs and the risk of thyroid cancer, adjusting for potential confounders. RESULTS: An elevated risk of thyroid cancer was associated with moderate to high levels of coumestrol intake [OR = 2.48, 95% confidence interval (CI), 1.39-4.43 for 40-80 µg/day; OR = 2.41, 95% CI, 1.32-4.40 for 80-130 µg/day; and OR = 2.38, 95% CI, 1.26-4.50 for >200 µg/day compared with <40 µg/day], and the main elevation in risk appeared among microcarcinomas (≤1 cm). A decreased risk of papillary macrocarcinomas (>1 cm; OR = 0.26, 95% CI, 0.08-0.85 for 1,860-3,110 µg/day compared with <760 µg/day) was associated with moderate genistein intake among women. CONCLUSIONS: Our study suggests that high coumestrol intake increases the risk of thyroid cancer, especially microcarcinomas, whereas moderate amounts of genistein intake appear to be protective for females with thyroid macrocarcinomas. IMPACT: The study highlights the importance of distinguishing between microcarcinomas and macrocarcinomas in future research on the etiology of thyroid cancer.


Subject(s)
Diet Surveys/statistics & numerical data , Feeding Behavior , Phytoestrogens/administration & dosage , Thyroid Neoplasms/epidemiology , Adult , Aged , Aged, 80 and over , Case-Control Studies , Connecticut/epidemiology , Coumestrol/administration & dosage , Coumestrol/adverse effects , Female , Genistein/administration & dosage , Genistein/adverse effects , Humans , Male , Middle Aged , Phytoestrogens/adverse effects , Protective Factors , Risk Factors , Thyroid Neoplasms/etiology , Thyroid Neoplasms/prevention & control , Young Adult
15.
J Agric Food Chem ; 67(49): 13737-13750, 2019 Dec 11.
Article in English | MEDLINE | ID: mdl-31789024

ABSTRACT

Genistein is abundant in animal feed. In this study, the side effects of high-dose genistein on intestinal health and hypothalamic RNA profile were evaluated. Chicks exposed to high-dose genistein by intraperitoneal injection (416 ± 21, 34.5 ± 2.5) and feed supplementation (308 ± 19, 27.2 ± 2.1) both showed a reduced body weight gain and feed intake in comparison with the control group (261 ± 16, 22.7 ± 1.6, P < 0.01). In comparison with the control (22.4 ± 0.5, 33.3 ± 2.4), serum levels of albumin and total protein were decreased after high-dose genistein injection (21.6 ± 0.5, 31.8 ± 1.6) and diet supplementation (20.6 ± 0.9, 29.9 ± 2.5, P < 0.001). Interestingly, the genistein diet presented the chick hypothalamus with downregulated expression of bitter receptors (TAS1R3, P < 0.05). Meanwhile, it upregulated the expressions of TAS2R1 (P < 0.05) and downstream genes (PLCB2 and IP3R3) in the ileum (P < 0.05). Accordingly, high-dose dietary genistein reduced villus height and the abundance of Lactobacillus, along with the increased abundance of pathogenic bacteria in the ileum (P < 0.05). Furthermore, transcriptomic analysis identified 348 differently expressed genes (168 upregulated and 224 downregulated) in the high-dose dietary genistein treated group in comparison with the control (P < 0.05, |log2FoldChange| > 0.585). Therefore, high-dose dietary genistein altered the hypothalamic RNA profile and signal processing. Cluster analysis further revealed that high-dose dietary genistein significantly influenced apoptosis, the immune process, and the whole synthesis of steroid hormones in the hypothalamus (P < 0.05). In conclusion, high-dose dietary genistein altered the hypothalamic RNA profile and intestinal health of female chicks.


Subject(s)
Chickens/metabolism , Dietary Supplements/adverse effects , Genistein/adverse effects , Hypothalamus/metabolism , RNA/genetics , Animal Feed/adverse effects , Animal Feed/analysis , Animals , Body Weight/drug effects , Chickens/genetics , Chickens/growth & development , Chickens/immunology , Eating/drug effects , Female , Genistein/analysis , Hypothalamus/drug effects , Ileum/drug effects , Ileum/metabolism , RNA/metabolism , Steroids/metabolism
16.
Brain Res ; 1724: 146434, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31491419

ABSTRACT

As demonstrated in previous studies, early postnatal genistein (GEN) administration to mice pups of both sexes, at doses similar to that of infant soy-based formulas, may affect the development of some steroid-sensitive neuronal circuits (i.e. nitrergic and vasopressinergic systems), causing irreversible alterations in adults. Here, we investigated the hypothalamic and mesencephalic dopaminergic system (identified with tyrosine hydroxylase immunohistochemistry). GEN administration (50 mg/kg) to mice of both sexes during the first week of postnatal life specifically affected tyrosine hydroxylase immunohistochemistry in the hypothalamic subpopulation of neurons, abolishing their sexual dimorphism. On the contrary, we did not observe any effects in the mesencephalic groups. Due to the large involvement of dopamine in circuits controlling rodent sexual behavior and food intake, these results clearly indicate that the early postnatal administration of GEN may irreversibly alter the control of reproduction, of energetic metabolism, and other behaviors. These results suggest the need for a careful evaluation of the use of soy products in both human and animal newborns.


Subject(s)
Genistein/pharmacology , Sex Differentiation/drug effects , Animals , Animals, Newborn , Dopamine/physiology , Female , Genistein/adverse effects , Genistein/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/physiology , Male , Mesencephalon/metabolism , Mice , Neurons/physiology , Phytoestrogens , Sex Characteristics , Glycine max , Tyrosine 3-Monooxygenase
17.
Int J Mol Sci ; 20(5)2019 Mar 12.
Article in English | MEDLINE | ID: mdl-30870965

ABSTRACT

Genistein is a naturally occurring phytoestrogen isoflavone and is the active drug ingredient in BIO 300, a radiation countermeasure under advanced development for acute radiation syndrome (H-ARS) and for the delayed effects of acute radiation exposure (DEARE). Here we have assessed the pharmacokinetics (PK) and safety of BIO 300 in the nonhuman primate (NHP). In addition, we analyzed serum samples from animals receiving a single dose of BIO 300 for global metabolomic changes using ultra-performance liquid chromatography (UPLC) quadrupole time-of-flight mass spectrometry (QTOF-MS). We present a comparison of how either intramuscularly (im) or orally (po) administered BIO 300 changed the metabolomic profile. We observed transient alterations in phenylalanine, tyrosine, glycerophosphocholine, and glycerophosphoserine which reverted back to near-normal levels 7 days after drug administration. We found a significant overlap in the metabolite profile changes induced by each route of administration; with the po route showing fewer metabolic alterations. Taken together, our results suggest that the administration of BIO 300 results in metabolic shifts that could provide an overall advantage to combat radiation injury. This initial assessment also highlights the utility of metabolomics and lipidomics to determine the underlying physiological mechanisms involved in the radioprotective efficacy of BIO 300.


Subject(s)
Genistein/administration & dosage , Genistein/pharmacokinetics , Metabolome/drug effects , Nanoparticles/administration & dosage , Suspensions/administration & dosage , Suspensions/pharmacokinetics , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/metabolism , Animals , Chromatography, High Pressure Liquid/methods , Female , Genistein/adverse effects , Macaca mulatta , Male , Metabolomics/methods , Nanoparticles/adverse effects , Nanoparticles/metabolism , Primates , Suspensions/adverse effects
18.
Biol Pharm Bull ; 41(10): 1581-1585, 2018.
Article in English | MEDLINE | ID: mdl-30270327

ABSTRACT

The objective of this study was to confirm the effect of maternal genistein exposure on body weight of male offspring and the metabolic alterations associated with maternal genistein-induced obesity. Pregnant female Sprague-Dawley (SD) rats were supplemented with 300 mg/kg diet of genistein (GEN) or no genistein (CON) throughout pregnancy and lactation. The growth of male offspring was investigated until 12 week age and the mechanism of obesity was studied using metabonomics by ultra performance liquid chromatography and quadrupole time-of-flight (UPLC Q-TOF) MS with electrospray ionization in positive ESI mode (ESI+). Compared with the CON group, body weight, fat pad and food intake of male offspring in GEN group were increased significantly at the age of weeks 10 to 12 (p<0.05). Ten urine principal metabolites contributing to the clusters were identified, including increased 8-Isoprostaglandin F2a, and decreased L-Proline, Betaine, L-Acetylcarnitine, Norsalsolinol, Indoleacrylic acid, L-Tryptophan, Lysophosphatidylcholines (LysoPC) (20 : 4), Lysophosphatidylethanolamines (LysoPE) (18 : 1) and LysoPC (O-18 : 0). Our results confirmed weight-increasing effects of maternal genistein exposure, accompanied by favorable changes in metabolites in the male offspring' urine. Therefore, this research enables us to better understand obesity and predict risk of obesity-related disease by studying metabolites present in the urine.


Subject(s)
Genistein/adverse effects , Lactation , Maternal Nutritional Physiological Phenomena , Metabolome , Obesity/etiology , Phytoestrogens/adverse effects , Prenatal Exposure Delayed Effects , Adipose Tissue/metabolism , Animals , Biomarkers/metabolism , Chromatography, High Pressure Liquid , Diet , Dietary Supplements , Eating , Female , Male , Metabolomics/methods , Obesity/urine , Pregnancy , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization , Weight Gain
19.
Article in English | MEDLINE | ID: mdl-29870789

ABSTRACT

Based on the assumed oestrogenic and apoptotic properties of soya isoflavones (genistein, daidzein), and following the current OECD test-guidelines and principle of 3Rs, we have studied the potential toxicity of phytochemicals on the zebrafish embryos test (ZFET). For this purpose, zebrafish embryos at 2-3 h post-fertilisation (hpf) were exposed to both soya isoflavones (from 1.25 mg/L to 20 mg/L) and assayed until 96 hpf. Lethal and sub-lethal endpoints (mortality, hatching rates and malformations) were estimated in the ZFET, which was expanded to potential gene expression markers, determining the lowest observed effect (and transcriptional) concentrations (LOEC, LOTEC), and the no-observable effect (and transcriptional) concentrations (NOEC, NOTEC). The results revealed that genistein is more toxic (LC50-96 hpf: 4.41 mg/L) than daidzein (over 65.15 mg/L). Both isoflavones up-regulated the oestrogen (esrrb) and death receptors (fas) and cyp1a transcript levels. Most thyroid transcript signals were up-regulated by genistein (except for thyroid peroxidase/tpo), and the hatching enzyme (he1a1) was exclusively up-regulated by daidzein (from 1.25 mg/L onwards). The ZFET proved suitable for assessing toxicant effects of both isoflavones and potential disruptions (i.e. oestrogenic, apoptotic, thyroid, enzymatic) during the embryogenesis and the endotrophic larval period.


Subject(s)
Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Genistein/adverse effects , Isoflavones/adverse effects , Phytoestrogens/adverse effects , Thyroid Gland/metabolism , Animals , Apoptosis , Cytochrome P-450 CYP1A1/chemistry , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Dietary Supplements/adverse effects , Ectogenesis , Embryo, Nonmammalian/enzymology , Endocrine Disruptors/adverse effects , Endocrine Disruptors/metabolism , Genistein/metabolism , Isoflavones/metabolism , Larva/enzymology , Larva/growth & development , Larva/metabolism , Lethal Dose 50 , Receptors, Estrogen/chemistry , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Seeds/chemistry , Signal Transduction , Glycine max/chemistry , Thyroid Gland/embryology , Thyroid Gland/enzymology , Toxicity Tests, Acute , Zebrafish , fas Receptor/agonists , fas Receptor/chemistry , fas Receptor/metabolism
20.
Environ Health Perspect ; 126(4): 047002, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29624291

ABSTRACT

BACKGROUND: Female reproductive tract development is sensitive to the endocrine-disrupting potential of environmental estrogens. Early-life exposure to the dietary phytoestrogen genistein impairs fertility and persistently alters the transcriptome in the oviduct and uterus of rodents. Glucocorticoid signaling, which has recently been shown to be essential for normal fertility in the female mouse uterus, is antagonized by genistein. OBJECTIVE: Our goal was to determine whether early-life exposure to genistein disrupts glucocorticoid signaling in the mouse uterus, which may contribute to infertility. METHODS: Female C57Bl/6 mice were exposed to either 50 mg/kg per day genistein, 10 µg/kg per day estradiol, or vehicle (corn oil) on postnatal days 1-5 (PND1-5), and then treated with the synthetic glucocorticoid dexamethasone (Dex: 1 mg/kg) or vehicle (saline) on PND5, at weaning on PND21, or as adults on PND56 following adrenalectomy and ovariectomy to evaluate glucocorticoid responsiveness. Uteri were isolated following treatment for gene expression or chromatin immunoprecipitation. RESULTS: Neonatal exposure to genistein altered the uterine transcriptome of adult mice and caused substantial changes to the transcriptional response to glucocorticoids. Although expression of the glucocorticoid receptor was not affected, genistein exposure disrupted glucocorticoid receptor recruitment to specific regulatory sites in target genes. Many genes involved in chromatin remodeling were dysregulated in genistein-exposed mice, suggesting that epigenetic reprograming may contribute to the altered glucocorticoid response of the uterus following early-life exposure to genistein. These changes affected the biological activity of glucocorticoids within the uterus, as glucocorticoids antagonized the proliferative effects of estradiol in the uterus of control mice but not genistein-exposed mice. CONCLUSIONS: Our findings suggest that disruption of glucocorticoid signaling due to early-life exposure to environmental estrogens may in part render the uterus unable to support implantation. https://doi.org/10.1289/EHP1575.


Subject(s)
Dexamethasone/metabolism , Genistein/adverse effects , Glucocorticoids/metabolism , Phytoestrogens/adverse effects , Signal Transduction/drug effects , Uterus/drug effects , Animals , Animals, Newborn , Female , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...