Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Biomed Pharmacother ; 169: 115783, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37944439

ABSTRACT

Nuclear receptors (NRs) represent intracellular proteins that function as a signaling network of transcriptional factors to control genes in response to a variety of environmental, dietary, and hormonal stimulations or serve as orphan receptors lacking a recognized ligand. They also play an essential role in normal development, metabolism, cell growth, cell division, physiology, reproduction, and homeostasis and function as biological markers for tumor subclassification and as targets for hormone therapy. NRs, including steroid hormone receptors (SHRs), have been studied as tools to examine the fundamentals of transcriptional regulation within the development of mammals and human physiology, in addition to their links to disturbances. In this regard, it is widely recognized that aberrant NR signaling is responsible for the pathological growth of hormone-dependent tumors in response to SHRs dysregulation and consequently represents a potential therapeutic candidate in a range of diseases, as in the case of prostate cancer and breast cancer. On the other hand, phytosterols are a group of plant-derived compounds that act directly as ligands for NRs and have proven their efficacy in the management of diabetes, heart diseases, and cancers. However, these plants are not suggested in cases of hormone-dependent cancer since a certain group of plants contains molecules with a chemical structure similar to that of estrogens, which are known as phytoestrogens or estrogen-like compounds, such as lignans, coumestans, and isoflavones. Therefore, it remains an open and controversial debate regarding whether consuming a phytosterol-rich diet and adopting a vegetarian lifestyle like the Mediterranean diet may increase the risk of developing steroid hormone-dependent cancers by constitutively activating SHRs and thereby leading to tumor transformation. Overall, the purpose of this review is to better understand the relevant mechanistic pathways and explore epidemiological investigations in order to establish that phytosterols may contribute to the activation of NRs as cancer drivers in hormone-dependent cancers.


Subject(s)
Breast Neoplasms , Phytosterols , Receptors, Steroid , Animals , Humans , Male , Estrogens/metabolism , Mammals , Phytoestrogens , Receptors, Cytoplasmic and Nuclear , Receptors, Steroid/chemistry , Receptors, Steroid/physiology , Steroids
2.
Int J Mol Sci ; 24(17)2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37686439

ABSTRACT

This study investigated the interplay between transforming growth factor beta (TGF-ß1/T1 and TGF-ß3/T3), and sex hormone receptors using our 3D in vitro cornea stroma model. Primary human corneal fibroblasts (HCFs) from healthy donors were plated in transwells at 106 cells/well and cultured for four weeks. HCFs were supplemented with stable vitamin C (VitC) and stimulated with T1 or T3. 3D construct proteins were analyzed for the androgen receptor (AR), progesterone receptor (PR), estrogen receptor alpha (ERα) and beta (ERß), luteinizing hormone receptor (LHR), follicle-stimulating hormone receptor (FSHR), gonadotropin-releasing hormone receptor (GnRHR), KiSS1-derived peptide receptor (KiSS1R/GPR54), and follicle-stimulating hormone subunit beta (FSH-B). In female constructs, T1 significantly upregulated AR, PR, ERα, FSHR, GnRHR, and KiSS1R. In male constructs, T1 significantly downregulated FSHR and FSH-B and significantly upregulated ERα, ERß, and GnRHR. T3 caused significant upregulation in expressions PR, ERα, ERß, LHR, FSHR, and GNRHR in female constructs, and significant downregulation of AR, ERα, and FSHR in male constructs. Semi-quantitative Western blot findings present the interplay between sex hormone receptors and TGF-ß isoforms in the corneal stroma, which is influenced by sex as a biological variable (SABV). Additional studies are warranted to fully delineate their interactions and signaling mechanisms.


Subject(s)
Corneal Stroma , Transforming Growth Factor beta3 , Humans , Female , Male , Estrogen Receptor alpha , Receptors, Kisspeptin-1 , Estrogen Receptor beta/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta , Follicle Stimulating Hormone
3.
J Mol Endocrinol ; 69(4): T25-T36, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36069789

ABSTRACT

The retinoic acid receptors (RARα, ß, and γ) are multi-domain polypeptides that heterodimerize with retinoid X receptors (RXRα, ß, and γ) to form functional transcription factors. Understanding the three-dimensional molecular organization of these nuclear receptors (NRs) began with RAR and RXR DNA-binding domains (DBDs), and were followed with studies on isolated ligand-binding domains (LBDs). The more complete picture emerged in 2017 with the multi-domain crystal structure of RXRα-RARß on its response element with retinoic acid molecules and coactivator segments on both proteins. The analysis of that structure and its complementary studies have clarified the direct communication pathways within RXR-RAR polypeptides, through which DNA binding, protein-ligand, and protein-protein interactions are integrated for overall functional responses. Understanding the molecular connections in the RXR-RAR complex has benefited from direct observations of the multi-domain structures of RXRα-PPARγ, RXRα-LXRß, HNF-4α homodimer, and androgen receptor homodimer, each bound to its response element. These comprehensive NR structures show unique quaternary architectures, yet all have DBD-DBD, LBD-LBD, and DBD-LBD domain-domain contacts within them. These convergence zones allow signals from discrete domains of their polypeptides to be propagated and integrated across their entire complex, shaping their overall responses in an allosteric fashion.


Subject(s)
PPAR gamma , Receptors, Androgen , DNA , DNA-Binding Proteins/metabolism , Ligands , Receptors, Retinoic Acid/genetics , Retinoid X Receptors , Tretinoin
4.
BMC Complement Med Ther ; 22(1): 18, 2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35057779

ABSTRACT

BACKGROUND: Breast Cancer (BC) is the most common cancer in women worldwide and, although 70% of patients are responsive to selective Estrogen Receptor (ER) modulators such as Tamoxifen (Tam), patients' survival is comprised by resistance to endocrine therapy. Brazilian flora, especially the Amazon biome, is one of the richest global sources of native species with potentially bioactive compounds. Arrabidaea chica is a plant native to the Amazon that has been used in the treatment of different diseases. However, its action on BC remains unclear. METHODS: Herein the biological effects of the chloroform extract of A. chica (CEAC) were evaluated on BC cells and in in vivo model. After confirmation of CEAC antioxidant capacity, cells were treated with CEAC and Tam, alone and with CEAC+Tam. The cell viability was evaluated by MTT and hormone receptor transcripts levels were assessed (ESR1, ESR2 and AR). Finally, anticarcinogenicity of CEAC was recorded in Drosophila melanogaster through Epithelial Tumor Test (ETT). RESULTS: The study confirmed the antioxidant activity of CEAC. CEAC was selective for MCF-7, downregulating ESR2 and AR transcripts and upregulating ESR2 expression. The modulatory effects of CEAC on ERs did not differ between cells treated with Tam and with CEAC+Tam. Interestingly, previous treatment with CEAC, followed by treatment with Tam promoted a significant decrease in cell viability. The extract also presented anticarcinogenic effect in in vivo assay. CONCLUSION: The bioassays on breast tumor cells demonstrated the antiproliferative activity of the extract, which modulated the expression of hormone receptors and sensitized luminal tumor cells to Tam. These results suggest that CEAC could be a complementary treatment for BC.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antioxidants/pharmacology , Bignoniaceae , Breast Neoplasms/drug therapy , Phytotherapy , Plant Extracts/pharmacology , Animals , Biological Assay , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drosophila melanogaster , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Humans , MCF-7 Cells/drug effects , Plants, Medicinal , Receptors, Androgen/metabolism
5.
J Clin Med ; 10(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34884213

ABSTRACT

Background Rheumatoid Arthritis (RA) patients show a higher risk of heart failure. The present study investigated possible causes of cardiac dysfunction related to thyroid hormone (TH) signaling in a RA mouse model. Methods A TNF-driven mouse model of RA[TghuTNF (Tg197)] was used. Cardiac function was evaluated by echocardiography. SERCA2a and phospholamban protein levels in left ventricle (LV) tissue, thyroid hormone levels in serum, TH receptors in LV and TH-related kinase signaling pathways were measured. T3 hormone was administered in female Tg197 mice. Results We show LV and atrial dilatation with systolic dysfunction in Tg197 animals, accompanied by downregulated SERCA2a. We suggest an interaction of pro-inflammatory and thyroid hormone signaling indicated by increased p38 MAPK and downregulation of TRß1 receptor in Tg197 hearts. Interestingly, female Tg197 mice showed a worse cardiac phenotype related to reduced T3 levels and Akt activation. T3 supplementation increased Akt activation, restored SERCA2a expression and improved cardiac function in female Tg197 mice. Conclusions TNF overexpression of Tg197 mice results in cardiac dysfunction via p38 MAPK activation and downregulation of TRß1. Gender-specific reduction in T3 levels could cause the worse cardiac phenotype observed in female mice, while T3 administration improves cardiac function and calcium handling via modified Akt activation.

6.
Cells ; 10(6)2021 06 09.
Article in English | MEDLINE | ID: mdl-34207527

ABSTRACT

The relationship between endocrine disrupting chemicals (EDCs) and the pathogenesis of acne vulgaris has yet to be explored in the literature. Acne vulgaris is a chronic inflammatory skin disease of the pilosebaceous unit. The pathogenesis of acne involves several hormonal pathways, including androgens, insulin-like growth factor 1(IGF-1), estrogens, and corticosteroids. EDCs influence these pathways primarily through two mechanisms: altering endogenous hormone levels and interfering with hormone receptor function. This review article describes the mechanistic links between EDCs and the development of acne lesions. Highlighted is the contributory role of androgen receptor ligands, such as bisphenol A (BPA) and mono-2-ethylhexyl Phthalate (MEHP), via upregulation of lipogenic genes and resultant exacerbation of cholesterol synthesis. Additionally discussed is the protective role of phytoestrogen EDCs in counteracting androgen-induced sebocyte maturation through attenuation of PPARy transcriptional activity (i.e., resveratrol) and restoration of estrogen-regulated TGF-B expression in skin cells (i.e., genistein). Examination of the relationship between EDCs and acne vulgaris may inform adjunctive avenues of treatment such as limiting environmental exposures, and increasing low-glycemic, plant-rich foods in the diet. With a better understanding of the cumulative role that EDCs play in acne, clinicians can be better equipped to treat and ultimately improve the lives of their patients.


Subject(s)
Acne Vulgaris , Endocrine Disruptors/toxicity , Phytoestrogens/pharmacology , Acne Vulgaris/chemically induced , Acne Vulgaris/drug therapy , Adolescent , Adult , Animals , Female , Humans , Young Adult
7.
Nutr Res Rev ; 34(2): 276-302, 2021 12.
Article in English | MEDLINE | ID: mdl-34057057

ABSTRACT

Dietary intake and tissue levels of carotenoids have been associated with a reduced risk of several chronic diseases, including cardiovascular diseases, type 2 diabetes, obesity, brain-related diseases and some types of cancer. However, intervention trials with isolated carotenoid supplements have mostly failed to confirm the postulated health benefits. It has thereby been speculated that dosing, matrix and synergistic effects, as well as underlying health and the individual nutritional status plus genetic background do play a role. It appears that our knowledge on carotenoid-mediated health benefits may still be incomplete, as the underlying mechanisms of action are poorly understood in relation to human relevance. Antioxidant mechanisms - direct or via transcription factors such as NRF2 and NF-κB - and activation of nuclear hormone receptor pathways such as of RAR, RXR or also PPARs, via carotenoid metabolites, are the basic principles which we try to connect with carotenoid-transmitted health benefits as exemplified with described common diseases including obesity/diabetes and cancer. Depending on the targeted diseases, single or multiple mechanisms of actions may play a role. In this review and position paper, we try to highlight our present knowledge on carotenoid metabolism and mechanisms translatable into health benefits related to several chronic diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Antioxidants , Carotenoids , Dietary Supplements , Humans , Nutritional Status
8.
Nutr Res ; 85: 84-98, 2021 01.
Article in English | MEDLINE | ID: mdl-33453499

ABSTRACT

L-carnitine is an indispensable metabolite facilitating the transport of fatty acids into the mitochondrial matrix and has been previously postulated to exert a nutrigenomic effect. However, the underlying molecular mechanisms remain mostly unclear. We hypothesized that L-carnitine interacts with nuclear receptors involved in metabolic regulation, thereby modulating downstream targets of cellular metabolism. Therefore, we investigated the effect of L-carnitine supplementation on protein activity, mRNA expression, and binding affinities of nuclear receptors as well as mRNA expression of downstream targets in skeletal muscle cells, hepatocytes, and differentiated adipocytes. L-carnitine supplementation to hepatocytes increased the protein activity of multiple nuclear receptors (RAR, RXR, VDR, PPAR, HNF4, ER, LXR). Diverging effects on the mRNA expression of PPAR-α, PPAR-δ, PPAR-γ, RAR-ß, LXR-α, and RXR-α were observed in adipocytes, hepatocytes, and skeletal muscle cells. mRNA levels of PPAR-α, a key regulator of lipolysis and ß-oxidation, were significantly upregulated, emphasizing a role of L-carnitine as a promoter of lipid catabolism. L-carnitine administration to hepatocytes modulated the transcription of key nuclear receptor target genes, including ALDH1A1, a promoter of adipogenesis, and OGT, a contributor to insulin resistance. Electrophoretic mobility shift assays proved L-carnitine to increase binding affinities of nuclear receptors to their promoter target sequences, suggesting a molecular mechanism for the observed transcriptional modulation. Overall, these findings indicate that L-carnitine modulates the activity and expression of nuclear receptors, thereby promoting lipolytic gene expression and decreasing transcription of target genes linked to adipogenesis and insulin resistance.


Subject(s)
Adipocytes/metabolism , Carnitine/metabolism , Carnitine/pharmacology , Cell Nucleus/metabolism , Hepatocytes/metabolism , Muscle Fibers, Skeletal/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , 3T3-L1 Cells , Animals , Binding Sites , Carnitine O-Acetyltransferase/genetics , Carnitine O-Acetyltransferase/metabolism , Cells, Cultured , Culture Media , Humans , Liver X Receptors/genetics , Mice , Nutrigenomics , PPAR alpha/genetics , PPAR alpha/metabolism , Promoter Regions, Genetic , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Signal Transduction , Transcription, Genetic
9.
Int J Mol Sci ; 21(22)2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33198405

ABSTRACT

Gonadotropin-releasing hormones (GnRHs) play pivotal roles in reproduction via the hypothalamus-pituitary-gonad axis (HPG axis) in vertebrates. GnRHs and their receptors (GnRHRs) are also conserved in invertebrates lacking the HPG axis, indicating that invertebrate GnRHs do not serve as "gonadotropin-releasing factors" but, rather, function as neuropeptides that directly regulate target tissues. All vertebrate and urochordate GnRHs comprise 10 amino acids, whereas amphioxus, echinoderm, and protostome GnRH-like peptides are 11- or 12-residue peptides. Intracellular calcium mobilization is the major second messenger for GnRH signaling in cephalochordates, echinoderms, and protostomes, while urochordate GnRHRs also stimulate cAMP production pathways. Moreover, the ligand-specific modulation of signal transduction via heterodimerization between GnRHR paralogs indicates species-specific evolution in Ciona intestinalis. The characterization of authentic or putative invertebrate GnRHRs in various tissues and their in vitro and in vivo activities indicate that invertebrate GnRHs are responsible for the regulation of both reproductive and nonreproductive functions. In this review, we examine our current understanding of and perspectives on the primary sequences, tissue distribution of mRNA expression, signal transduction, and biological functions of invertebrate GnRHs and their receptors.


Subject(s)
Hypothalamus/metabolism , Invertebrates/metabolism , Receptors, LHRH/metabolism , Animals , Biological Evolution , COS Cells , Calcium/metabolism , Chlorocebus aethiops , Ciona intestinalis , Cyclic AMP/metabolism , Echinodermata , Female , Gonadotropin-Releasing Hormone/metabolism , HEK293 Cells , Humans , Ligands , Male , Markov Chains , Mollusca , Signal Transduction , Tissue Distribution , Urochordata
10.
Brain Res ; 1748: 147096, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32898507

ABSTRACT

Estrogen replacement therapy decreases some risk factors of the metabolic syndrome but increases the risk of some types of cancer. Tibolone (TIB) has shown similar neuroprotective effects as estrogens. This study aimed to evaluate the effects of TIB on metabolic parameters and the expression of sex hormone receptors in the CNS in ovariectomised rats fed with a hypercaloric diet. Sprague-Dawley female rats were ovariectomised and fed for 30 days with a standard diet (SD) or high-fat high-fructose diet (HFFD) and treated with TIB (1 mg/kg) or vehicle. At the end of the treatments, HFFD increased body weight, glucose tolerance, triglycerides and cholesterol levels, while TIB treatment decreased these parameters. Subsequently, the hippocampus, the hypothalamus and the frontal cortex were dissected. RT-PCR was performed for progesterone receptor (PR), androgen receptor (AR), estrogen receptors alpha and beta (ERα, ERß), insulin receptor (IR) and insulin-like growth factor 1 (IGF-1). HFFD altered the expression of sex hormone receptors in specific brain structures involved in the regulation of homeostasis and cognition, which highlights the importance of a healthy diet. In turn, TIB modulated the expression of these receptors, particularly in the hypothalamus.


Subject(s)
Diet, High-Fat , Dietary Carbohydrates , Estrogen Receptor Modulators/pharmacology , Frontal Lobe/metabolism , Hippocampus/metabolism , Hypothalamus/metabolism , Norpregnenes/pharmacology , Animals , Female , Frontal Lobe/drug effects , Fructose , Hippocampus/drug effects , Hypothalamus/drug effects , Ovariectomy , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
11.
Gen Comp Endocrinol ; 285: 113250, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31445009

ABSTRACT

Seasonally breeding animals initiate gonadal recrudescence when mechanisms that suppress reproduction give way to mechanisms that stimulate it. However, knowledge of mechanistic changes in hormonal regulation during this transition is limited. Further, most studies of reproductive timing have focused on males, despite the critical role of females in determining breeding phenology. Closely related populations that live in the same environment but differ in reproductive timing provide an opportunity to examine differences in mechanisms during the transition from the pre-reproductive to reproductive state. We studied closely related migrant and resident populations of dark-eyed juncos (Junco hyemalis) that reside in the same environment in spring but differ in breeding phenology. Residents initiate breeding earlier than migrants, which do not breed until after they have migrated. To directly study differences in the hypothalamic mechanisms of reproduction, we captured 16 migrant and 13 resident females from the field on March 25-April 11. We quantified expression of mRNA transcripts and show that resident females had higher abundance of gonadotropin-releasing hormone transcripts than migrant females, indicating greater reproductive development in resident than migrant females living in the same environment. We also found higher transcript abundance of estrogen receptor and androgen receptor in migrant than resident females, suggesting that negative feedback may delay reproductive development in migrant females until after they migrate. These differences in hypothalamic mechanisms may help to explain differences in reproductive timing in populations that differ in migratory strategy.


Subject(s)
Animal Migration/physiology , Neurosecretory Systems/metabolism , Seasons , Songbirds/physiology , Sympatry/physiology , Animals , Female , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Linear Models , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Am J Clin Nutr ; 109(3): 597-605, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30831601

ABSTRACT

BACKGROUND: Soy-based dietary supplements have been promoted as natural alternatives to menopausal hormone therapy, but their potential effect on breast cancer development is controversial. OBJECTIVES: We examined the relation between the consumption of soy supplements and the risk of breast cancer, overall and by tumor hormone receptor status, among women aged >50 y. METHODS: In total, 76,442 women from the Etude Epidemiologique aupres de Femmes de la Mutuelle Generale de l'Education Nationale (E3N) cohort, born between 1925 and 1950, were followed from 2000 to 2011 (11.2 y on average, starting at a mean age of 59.5 y; 3608 incident breast cancers), with soy supplement use assessed every 2-3 y. HRs of breast cancer were estimated with the use of multivariable Cox models. RESULTS: Compared with never using soy supplements, the HRs associated with current use of soy supplements were 0.92 (95% CI: 0.76, 1.11) for all, 0.78 (95% CI: 0.60, 0.99) for estrogen receptor (ER)-positive, and 2.01 (95% CI: 1.41, 2.86) for ER-negative breast cancers. There was no association between past use of soy supplements and breast cancer. HRs for current use were 1.36 (95% CI: 0.95, 1.93) and 0.82 (95% CI: 0.65, 1.02) among women with and without a family history of breast cancer, respectively (P-interaction = 0.03) and 1.06 (95% CI: 0.87, 1.30) ≥5 y after menopause compared with 0.50 (95% CI: 0.31, 0.81) in premenopause or ≤5 y postmenopause (P-interaction = 0.04). CONCLUSIONS: In this cohort of women aged >50 y, we report opposing associations of soy supplements with ER-positive and ER-negative breast cancer risk. Our results also caution against the use of these supplements in women with a family history of breast cancer. Whether the risk profile of soy supplements could be more favorable among premenopausal or recently postmenopausal women deserves further investigation.


Subject(s)
Breast Neoplasms/etiology , Dietary Supplements/analysis , Isoflavones/administration & dosage , Menopause/drug effects , Aged , Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cohort Studies , Female , Follow-Up Studies , Humans , Isoflavones/adverse effects , Menopause/genetics , Menopause/metabolism , Middle Aged , Prospective Studies , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Risk Factors
13.
J Trace Elem Med Biol ; 47: 89-97, 2018 May.
Article in English | MEDLINE | ID: mdl-29544812

ABSTRACT

OBJECTIVE: Our aim was to investigate thyroid function alterations attributed to high iodide supplementation in maternal rats and their offspring. METHODS: Depending on their iodide intake, the pregnant rats were randomly divided into three groups: normal iodide intake (NI), 10 times high iodide intake (10 HI) and 100 times high iodide intake (100 HI) groups. Iodine concentration in the urine and maternal milk; iodine content and mitochondrial superoxide production; expression of TRα1, TRß1, NIS and Dio1 in both the thyroid and mammary glands were all measured. The offspring were exposed to different iodide-containing water (NI, 10 HI and 100 HI) from weaning to postnatal day 180 (PN180). Serum thyroid hormone levels were measured in both maternal rats and their offspring. RESULTS: Iodine concentration in the urine and maternal milk, as well as iodine content in the thyroid and mammary glands was significantly increased in both the 10 HI and 100 HI groups (p < .05). In the 100 HI group of maternal rats, low FT3 levels, high FT4, TPOAb and TgAb levels were detected. In addition, an increased mitochondrial superoxide production and decreased expression of TRα1, TRß1, NIS and Dio1 in the thyroid and mammary glands was found (p < .05). A positive staining of CD4+ that co-localized with TRß1 in the infiltrated cells within the thyroid follicles was observed. At PN180 in the offspring, the FT3 and FT4 levels showed a significant decrease, while the levels of serum TSH, TPOAb and TgAb were significantly increased in both 10 HI and 100 HI groups (p < .05). CONCLUSION: In maternal rats, although normal thyroid function can be maintained following 10 HI, thyroiditis can be induced following 100 HI on lactation days 7, 14, and 21. In the offspring at PN180, hypothyroidism complicated with thyroiditis can occur in both the 10 HI and 100 HI groups.


Subject(s)
Iodine/administration & dosage , Mammary Glands, Animal/drug effects , Thyroid Gland/drug effects , Animals , Animals, Newborn , Female , Hypothyroidism/chemically induced , Iodide Peroxidase/genetics , Iodine/analysis , Iodine/urine , Male , Mammary Glands, Animal/metabolism , Milk/chemistry , Rats, Wistar , Receptors, Thyroid Hormone/genetics , Superoxides/metabolism , Thyroid Function Tests , Thyroid Gland/physiology , Thyroxine/analysis , Triiodothyronine/analysis
14.
Am J Chin Med ; 45(6): 1147-1156, 2017.
Article in English | MEDLINE | ID: mdl-28830207

ABSTRACT

The first record of ginseng use dates back over two millennia, and ginseng is now popular in more than 35 countries. Ginsenosides are the pharmacological constituents responsible for the beneficial effects of ginseng. There is increasing evidence that ginseng and its bioactive ingredients are involved in the regulation of nuclear receptors, molecules that act in response to the specific binding of hormones, which link to a diverse array of signaling pathways, such as the ERK and PI3K/Akt pathways. Knowledge of the mechanism of how ginseng mediates these complexes is essential for the development of multi-target phytomedicine as possible therapy for different diseases. Here, we discuss the literature on the effects of ginseng and its constituents on estrogen, glucocorticoid, peroxisome proliferator-activated, and androgen nuclear hormone receptors, as well as how ginseng and its constituents exert their biological function in the treatment of cancer, obesity, and cardiovascular and neurological disorders. The accumulated results definitely show that the nuclear receptors are cellular targets of ginsenosides, but more rigorous data are required to establish and provide a scientific basis to confirm the suggested efficacy of ginseng or products with ginsenosides.


Subject(s)
Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Panax/chemistry , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Receptors, Cytoplasmic and Nuclear/drug effects , Animals , Cardiovascular Diseases/drug therapy , Female , Ginsenosides/isolation & purification , Humans , MAP Kinase Signaling System , Male , Neoplasms/drug therapy , Nervous System Diseases/drug therapy , Obesity/drug therapy , Peroxisome Proliferator-Activated Receptors/drug effects , Peroxisome Proliferator-Activated Receptors/physiology , Plant Extracts/isolation & purification , Receptors, Androgen/drug effects , Receptors, Androgen/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Estrogen/drug effects , Receptors, Estrogen/physiology , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/physiology
15.
Mol Cell Endocrinol ; 450: 113-125, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28479374

ABSTRACT

We evaluated the effect of cafeteria diet (CAF) on the mRNA levels and DNA methylation state of feeding-related neuropeptides, and neurosteroidogenic enzymes in discrete hypothalamic nuclei. Besides, the expression of steroid hormone receptors was analyzed. Female rats fed with CAF from weaning increased their energy intake, body weight, and fat depots, but did not develop metabolic syndrome. The increase in energy intake was related to an orexigenic signal of paraventricular (PVN) and ventromedial (VMN) nuclei, given principally by upregulation of AgRP and NPY. This was mildly counteracted by the arcuate nucleus, with decreased AgRP expression and increased POMC and kisspeptin expression. CAF altered the transcription of neurosteroidogenic enzymes in PVN and VMN, and epigenetic mechanisms associated with differential promoter methylation were involved. The changes observed in the hypothalamic nuclei studied could add information about their differential role in food intake control and how their action is disrupted in obesity.


Subject(s)
DNA Methylation/genetics , Diet , Eating/genetics , Gene Expression Regulation , Hypothalamus/metabolism , Adipose Tissue/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Binding Sites , Body Weight , Computer Simulation , Energy Intake/genetics , Female , Glucose Tolerance Test , Neuropeptides/genetics , Neuropeptides/metabolism , Organ Size/genetics , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Steroids/metabolism , Transcription, Genetic , Ventromedial Hypothalamic Nucleus/metabolism
16.
Cell Rep ; 19(4): 760-773, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28445727

ABSTRACT

Environmental enrichment (EE) replicates mind-body therapy by providing complex housing to laboratory animals to improve their activity levels, behavior, and social interactions. Using a Tcf4Het/+ApcMin/+-mediated model of colon tumorigenesis, we found that EE vastly improved the survival of tumor-bearing animals, with differential effect on tumor load in male compared to female animals. Analysis of Tcf4Het/+ApcMin/+ males showed drastically reduced expression of circulating inflammatory cytokines and induced nuclear hormone receptor (NHR) signaling, both of which are common in the wound repair process. Interestingly, EE provoked tumor wound repair resolution through revascularization, plasma cell recruitment and IgA secretion, replacement of glandular tumor structures with pericytes in a process reminiscent of scarring, and normalization of microbiota. These EE-dependent changes likely underlie the profound improvement in survival of colon-tumor-bearing Tcf4Het/+ApcMin/+ males. Our studies highlight the exciting promise of EE in the design of future therapeutic strategies for colon cancer patients.


Subject(s)
Colonic Neoplasms/pathology , Environment , Immunoglobulin A/metabolism , Wound Healing/physiology , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Alphaproteobacteria/isolation & purification , Alphaproteobacteria/physiology , Animals , Colon/microbiology , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Cytokines/blood , Disease Models, Animal , Female , Male , Mice , Microbiota , Neovascularization, Physiologic , Pericytes/cytology , Pericytes/metabolism , Proteobacteria/isolation & purification , Proteobacteria/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Survival Rate , Transcription Factor 4/genetics , Transcription Factor 4/metabolism
17.
Fitoterapia ; 110: 150-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26976217

ABSTRACT

Pumpkin seeds have been known in folk medicine as remedy for kidney, bladder and prostate disorders since centuries. Nevertheless, pumpkin research provides insufficient data to back up traditional beliefs of ethnomedical practice. The bioactivity of a hydro-ethanolic extract of pumpkin seeds from the Styrian pumpkin, Cucurbita pepo L. subsp. pepo var. styriaca, was investigated. As pumpkin seed extracts are standardized to cucurbitin, this compound was also tested. Transactivational activity was evaluated for human androgen receptor, estrogen receptor and progesterone receptor with in vitro yeast assays. Cell viability tests with prostate cancer cells, breast cancer cells, colorectal adenocarcinoma cells and a hyperplastic cell line from benign prostate hyperplasia tissue were performed. As model for non-hyperplastic cells, effects on cell viability were tested with a human dermal fibroblast cell line (HDF-5). No transactivational activity was found for human androgen receptor, estrogen receptor and progesterone receptor, for both, extract and cucurbitin. A cell growth inhibition of ~40-50% was observed for all cell lines, with the exception of HDF-5, which showed with ~20% much lower cell growth inhibition. Given the receptor status of some cell lines, a steroid-hormone receptor independent growth inhibiting effect can be assumed. The cell growth inhibition for fast growing cells together with the cell growth inhibition of prostate-, breast- and colon cancer cells corroborates the ethnomedical use of pumpkin seeds for a treatment of benign prostate hyperplasia. Moreover, due to the lack of androgenic activity, pumpkin seed applications can be regarded as safe for the prostate.


Subject(s)
Cucurbita/chemistry , Growth Inhibitors/pharmacology , Plant Extracts/pharmacology , Plant Proteins/pharmacokinetics , Seeds/chemistry , Cell Line, Tumor/drug effects , Fibroblasts/drug effects , Growth Inhibitors/isolation & purification , Humans , Male , Plant Proteins/isolation & purification , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Saccharomyces cerevisiae/drug effects
18.
Chinese Journal of Immunology ; (12): 1145-1149, 2016.
Article in Chinese | WPRIM | ID: wpr-495095

ABSTRACT

Objective:To explore the effect on sex hormone functional of extracts of Herba Epimedii /Fructus Ligustri Lucidi in osteoporosis rats.Methods:50 cases of 3 months Wistar rats were rando mly divided into normal group ,model group,extract low dose group(extract-L),extract middle dose group(extract-M),extract high dose group(extract-H),10 cases in each group,in addition to the normal group,other 4 groups were bulided OP model by filling and feeding retinoic acid for 15 d,then normal group and model group were filling physiological saline;extract-L,extract-M,extract-H were respectively feeding extracts of Herba Epimedii /Fructus Ligustri Lucidi at 50 mg/kg,100 mg/kg,200 mg/kg,both for 3 weeks.Measured the bone mineral density (BMD),observed the femoral pathology change of left femur with hematoxylin and eosin ( HE ) staining, measured trabecular bone tissue morphology related parameters ,analyzed the hormone receptor protein expression with immunohistochemistry .Results:The BMD of the whole femur ,femur head,and femur neck in normal group were all significantly higher than that of model group ,the BMD of the whole femur ,femur head, and femur neck in extract-M and extract-H group were all significantly higher than that of model group ,the BMD of the whole femur in extract-L group was significantly higher than that of model group ,the differences were statistically significant (P<0.05 or P<0.01).The bone trabecular number had significantly increased ,integrity,continuity improved significantly ,bone histomorphometry such as Tb.Ar, Tb.Th,and Tb.N in model group were decreased ,and Tb.Sp was increased,extract-M and extract-H group had remarkable effect on each index ,while extract-L group only had a remarkable effect on Tb .Ar and Tb.Th ( P<0.05 or P<0.01 ) , compared with model group.The expression of AR , ER in normal groups were significantly higher than that of model group , the expression of AR , ER in extract-H group was significantly higher than that of model group , the differences were statistically significant ( P<0.05 or P<0.01 ) . Conclusion:This study demonstrated that the combination with TFE and TIFL had potential protective effects on osteoporosis rats induced by retinoic acid .The combination with TFE and TIFL is able to increase BMD ,reverse pathological changes of bone tissue ,co-ordinate hormones of the hypothalamic-pituitary-gonadal axis ,and upregulate the expression of sex hormone receptors .

19.
J Ethnopharmacol ; 155(1): 721-30, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24945396

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Prostate cancer is one of the most diagnosed forms of cancer among men in western regions. Many traditional applications or phytotherapeutic concepts propose to inhibit the proliferation of prostate cancer cells. In order to detect influences of plant or fungal extracts and derived fractions on androgen receptor signaling pathways, a differentiating cell proliferation assay was established, which enables the simultaneous detection of hormonal and cytotoxic effects. MATERIAL AND METHODS: The well characterized prostate cancer cell lines LNCaP and PC-3 were used in a multiple readout assay. In all, 186 fractions of 23 traditionally used organisms were screened regarding their effects on proliferation of the two prostate cancer cell lines. The fractions were prepared by accelerated solvent extraction followed by gradient extrography. Extracts of the potential hormonally active plants Cibotium barometz, Heteropterys chrysophylla, and Sideroxylon obtusifolium (= Bumelia sartorum) were phytochemically investigated. RESULTS: Fractions from Cibotium barometz, Cortinarius rubellus, Cyrtomium falcatum, Heteropterys chrysophylla, Nephrolepis exaltata, Salvia miltiorrhiza, Sideroxylon obtusifolium, Trichilia emetica, and Trimeria grandifolia exhibited hormonal influences on prostate cancer cells. Cytotoxic activity towards human cell lines was detected for the first time for fractions from Aglaia spectabilis (A. gigantea), Nephrolepis exaltata and Cortinarius brunneus. CONCLUSIONS: The differential behavior of the two prostate cancer cell lines allows the discrimination between potential androgenic or antiandrogenic activities and effects on the estrogen or glucocorticoid receptor as well as cytotoxic activities. The combined cell lines assay can help to assess the biological activities of material used in traditional medicine.


Subject(s)
Fungi/chemistry , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Prostatic Neoplasms/drug therapy , Androgen Antagonists/isolation & purification , Androgen Antagonists/pharmacology , Androgens/isolation & purification , Androgens/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Prostatic Neoplasms/pathology , Solvents/chemistry
20.
Hepatol Res ; 44(10): E218-28, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24033844

ABSTRACT

AIM: Bile acid synthesis is regulated by nuclear receptors including farnesoid X receptor (FXR) and small heterodimer partner (SHP), and by fibroblast growth factor 15/19 (FGF15/19). We hypothesized that hepatic cysteine sulfinic acid decarboxylase (CSAD) (a key enzyme in taurine synthesis) is regulated by bile acids (BA). The aim of this study was to investigate CSAD regulation by BA dependent regulatory mechanisms. METHODS: Mice were fed a control diet or a diet supplemented with either 0.5% cholate or 2% cholestyramine. To study BA dependent pathways, we utilized GW4064 (FXR agonist), FGF19 or T-0901317 (liver X receptor [LXR] agonist) and Shp-/- mice. Tissue mRNA was determined by quantitative reverse transcription polymerase chain reaction. Amino acids were measured by high-performance liquid chromatography. RESULTS: Mice supplemented with dietary cholate exhibited reduced hepatic CSAD mRNA while those receiving cholestyramine exhibited increased mRNA. Activation of FXR suppressed CSAD mRNA expression whereas CSAD expression was increased in Shp-/- mice. Hepatic hypotaurine concentration (the product of CSAD) was higher in Shp-/- mice with a corresponding increase in serum taurine conjugated BA. FGF19 administration suppressed hepatic cholesterol 7-α-hydroxylase (CYP7A1) mRNA but did not change CSAD mRNA expression. LXR activation induced CYP7A1 mRNA yet failed to induce CSAD mRNA expression. CONCLUSION: BA regulate CSAD mRNA expression in a feedback fashion via mechanisms involving SHP and FXR but not FGF15/19 or LXR. These findings implicate BA as regulators of CSAD mRNA via mechanisms shared with CYP7A1.

SELECTION OF CITATIONS
SEARCH DETAIL