Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 441
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Drug Discov Ther ; 18(2): 117-129, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38644207

ABSTRACT

Polycystic ovary syndrome (PCOS) is a common gynecological endocrine disorder characterized by a complex pathogenesis and limited treatment options. Yishen Huatan and Huoxue decoction (YHHD), as a traditional Chinese Medicine formula, has shown effectiveness in treating PCOS. However, the specific mechanisms by which YHHD exerts its therapeutic effects remain unclear. In this study, we performed to investigate the therapeutic effects of YHHD and quercetin on dehydroepiandrosterone-induced PCOS mice, and examine the effect of quercetin on the decidualization of T-HESCs under hyperinsulinemic conditions. The results showed that YHHD could reduce early miscarriage rates in PCOS patients and significantly improved glucose metabolism disorders, sex hormone levels, and the estrous cycles in PCOS mice. Quercetin could alleviate effect of high insulin levels and restore the low expression of insulin receptor substrate1/2 (IRS1/2) and glucose transporte 4 (GLUT4) in T-HESCs, demonstrating its potential to mitigate hyperinsulin-induced decidualization dysfunction via the GLUT4 signaling pathway mediated by IRS1/2. This study provides valuable molecular insights of YHHD and highlight the therapeutic potential of quercetin in treating decidualization dysfunction in PCOS.


Subject(s)
Drugs, Chinese Herbal , Polycystic Ovary Syndrome , Quercetin , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , Female , Quercetin/pharmacology , Quercetin/therapeutic use , Animals , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Mice , Humans , Disease Models, Animal , Glucose Transporter Type 4/metabolism , Insulin Receptor Substrate Proteins/metabolism , Signal Transduction/drug effects , Adult , Abortion, Spontaneous/drug therapy , Insulin/blood , Insulin/metabolism , Dehydroepiandrosterone/pharmacology , Decidua/drug effects , Decidua/metabolism , Estrous Cycle/drug effects , Pregnancy
2.
Phytomedicine ; 128: 155456, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38537446

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a heterogeneous metabolic and endocrine disorder that causes anovulatory infertility and abnormal folliculogenesis in women of reproductive age. Several studies have revealed inflammation in PCOS follicles, and recent evidence suggests that Berberine (BBR) effectively reduces inflammatory responses in PCOS, however, the underlying mechanisms remain unclear. PURPOSE: To determine the underlying mechanisms by which BBR alleviates inflammation in PCOS. STUDY DESIGN: Primary human GCs from healthy women and women with PCOS, and KGN cells were used for in vitro studies. ICR mice were used for in vivo studies. METHODS: Gene expression was measured using RT-qPCR. HAS2, inflammatory cytokines, and serum hormones were assayed by ELISA. Protein expression profiles were assayed by Western blot. Chronic low-grade inflammatory mouse models were developed by intraperitoneal injection with LPS, and PCOS mouse models were established by subcutaneous intraperitoneal injection of DHEA. BBR and 4-MU were administered by gavage. Ovarian morphologic changes were evaluated using H&E staining. HAS2 expression in the ovary was assayed using Western blot and immunohistochemistry. RESULTS: Our results confirmed that HAS2 expression and hyaluronan (HA) accumulation are closely associated with inflammatory responses in PCOS. Data obtained from in vitro studies showed that HAS2 and inflammatory genes (e.g., MCP-1, IL-1ß, and IL-6) are significantly upregulated in PCOS samples and LPS-induced KGN cells compared to their control groups. In addition, these effects were reversed by blocking HAS2 expression or HA synthesis using BBR or 4-MU, respectively. Furthermore, HAS2 overexpression induces the expression of inflammatory genes in PCOS. These results were further confirmed in LPS- and DHEA-induced mouse models, where inflammatory genes were reduced by BBR or 4-MU, and ovarian morphology was restored. CONCLUSIONS: Our results define previously unknown links between HAS2 and chronic low-grade inflammation in the follicles of women with PCOS. BBR exerts its anti-inflammatory effects by down-regulating HAS2. This study provides a novel therapeutic target for alleviating ovarian inflammation in women with PCOS.


Subject(s)
Berberine , Disease Models, Animal , Hyaluronan Synthases , Inflammation , Mice, Inbred ICR , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/drug therapy , Berberine/pharmacology , Female , Animals , Humans , Hyaluronan Synthases/metabolism , Inflammation/drug therapy , Mice , Hyaluronic Acid , Adult , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Dehydroepiandrosterone/pharmacology , Ovary/drug effects , Lipopolysaccharides , Cytokines/metabolism
3.
Exp Anim ; 73(3): 319-335, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38494723

ABSTRACT

Dehydroepiandrosterone (DHEA) is frequently integrated as an adjuvant in over a quarter of controlled ovarian hyperstimulation (COH) protocols, despite the ongoing debate regarding its impact. This study aimed to evaluate the efficacy and mechanism of action of DHEA on ovarian follicular development and ovarian response in rats with varying ovarian reserves. The study involved 75 rats categorized into 15 distinct groups. The ovarian tissues of rats in both the normal ovarian reserve group and the premature ovarian insufficiency (POI) group, induced by 4-vinylcyclohexene diepoxide (VCD) injection, were subjected to histomorphological and biochemical analyses following the administration of DHEA, either alone or in combination with COH. Follicle counting was performed on histological sections obtained from various tissues. Serum concentrations of anti-Müllerian hormone (AMH) and the quantification of specific proteins in ovarian tissue, including phosphatase and tensin homolog of chromosome 10 (PTEN), phosphoinositide 3-kinase (PI3K), phosphorylated protein kinase B (pAKT), cyclooxygenase 2 (COX-2), caspase-3, as well as assessments of total antioxidant status and total oxidant status, were conducted employing the ELISA method. The impact of DHEA exhibited variability based on ovarian reserve. In the POI model, DHEA augmented follicular development and ovarian response to the COH protocol by upregulating the PTEN/PI3K/AKT signaling pathway, mitigating apoptosis, inflammation, and oxidative stress, contrary to its effects in the normal ovarian reserve group. In conclusion, it has been determined that DHEA may exert beneficial effects on ovarian stimulation response by enhancing the initiation of primordial follicles and supporting antral follicle populations.


Subject(s)
Cyclohexenes , Dehydroepiandrosterone , PTEN Phosphohydrolase , Phosphatidylinositol 3-Kinases , Primary Ovarian Insufficiency , Proto-Oncogene Proteins c-akt , Signal Transduction , Vinyl Compounds , Animals , Female , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/metabolism , PTEN Phosphohydrolase/metabolism , Cyclohexenes/pharmacology , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/administration & dosage , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Sprague-Dawley , Ovary/drug effects , Ovary/metabolism , Ovarian Reserve/drug effects , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism
4.
Eur J Med Chem ; 269: 116296, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38467086

ABSTRACT

Steroid hybrids have emerged as a type of advantageous compound as they could offer improved pharmacological and pharmaceutical properties. Here, we report a series of novel peptide-dehydroepiandrosterone hybrids, which would effectively induce endoplasmic reticulum stress (ERS) and lead to apoptosis with outstanding in vitro and in vivo anti-melanoma effects. The lead compound IId among various steroids conjugated with peptides and pyridines showed effective in vivo activity in B16 xenograft mice: in medium- and high-dose treatment groups (60 and 80 mg/kg), compound IId would significantly inhibit the growth of tumours by 98%-99% compared to the control group, with the highest survival rate as well. Further mechanism studies showed that compound IId would damage the endoplasmic reticulum and upregulate the ERS markers C/EBP homologous protein (CHOP) and glucose-regulated protein 78 (GRP78), which could further regulate caspase and Bcl-2 family proteins and lead to cell apoptosis. The compound IId was also proven to be effective in inhibiting B16 cell migration and invasion.


Subject(s)
Apoptosis , Endoplasmic Reticulum , Humans , Mice , Animals , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Peptides/pharmacology , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone/pharmacology
5.
Menopause ; 30(11): 1167-1173, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37788418

ABSTRACT

IMPORTANCE: Whether dehydroepiandrosterone (DHEA) supplementation improves cognitive performance in older women is uncertain. Nonetheless, DHEA supplements are readily available over the counter in several countries and are potentially being used to prevent cognitive decline and dementia. OBJECTIVE: This systematic review was conducted to evaluate the effect of exogenous DHEA on cognitive performance in postmenopausal women. EVIDENCE REVIEW: Ovid MEDLINE, EMBASE, PsycINFO, Web of Science Core Collection, and the Cochrane Central Register of Controlled Trials databases were searched for studies of postmenopausal women until November 30, 2022. Eligible studies were required to be randomized clinical trials, be at least single blind, have a placebo or comparator arm and published in English. Risk of bias of the included studies was assessed by the revised Cochrane risk-of-bias tool. FINDINGS: Of the 15 articles retrieved for full-text review, four met the inclusion criteria. In all studies DHEA was administered as a 50-mg oral daily dose and all were double blind with an identical placebo. Three were placebo-controlled, crossover studies and one was a parallel-group clinical trial. The only positive outcome was limited to a 4-wk cross-over study in which DHEA statistically significantly enhanced five of six tests of visual-spatial performance compared with placebo in 24 cognitively normal postmenopausal women. Improvement in cognitive performance with DHEA treatment over placebo group was not seen in any other study. Heterogeneity of design and use of multiple measures of cognitive performance was a barrier to meta-analysis and between study comparisons. The studies were limited by high risk of bias in multiple domains. CONCLUSION AND RELEVANCE: Overall, this systematic review does not support a beneficial effect of DHEA therapy on cognitive performance in postmenopausal women.


Subject(s)
Cognition Disorders , Dehydroepiandrosterone , Aged , Female , Humans , Cognition , Cognition Disorders/prevention & control , Cross-Over Studies , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Postmenopause , Randomized Controlled Trials as Topic , Single-Blind Method
6.
Neurobiol Aging ; 132: 246-249, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37866084

ABSTRACT

Old male rhesus macaques often show cognitive impairment, and also have attenuated circulating levels of testosterone and dehydroepiandrosterone sulfate (DHEAS). However, it is unclear if these age-associated decreases in circulating androgen levels are casually related to mechanisms that support cognition. To test this possibility, old male rhesus macaques were given daily supplements of testosterone and DHEA for ∼7 months, using a paradigm designed to mimic the 24-hour circulating hormone patterns of young adults. Animals completed the Delayed Match-to-Sample (DMS) task to assess recognition, and the Delayed Response (DR) task to assess working memory. The animals all showed significant delay-dependent performance, with longer delays resulting in lower accuracy; and timepoint-dependent performance, showing improvement with the repeated opportunities for practice. However, there were no differences between the androgen supplemented animals and age-matched controls. These data indicate that the specific short-term supplementation paradigm employed here offers no obvious benefits for DMS or DR task performance.


Subject(s)
Androgens , Dehydroepiandrosterone , Animals , Male , Androgens/pharmacology , Macaca mulatta/physiology , Dehydroepiandrosterone/pharmacology , Testosterone , Cognition/physiology , Dietary Supplements
7.
Reprod Biol ; 23(4): 100797, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37633225

ABSTRACT

The present study aimed to build a DHEA-induced polycystic ovary syndrome (PCOS) rat model to evaluate the potential mechanism of DHEA-induced AMH rise in these rat ovarian tissues. A total of 36 female 3-week-old rats were allocated into two groups at random. The control group received merely the same amount of sesame oil for 20 days while the experimental group received 0.2 mL of sesame oil Plus DHEA 6 mg/100 g daily. Both groups' vaginal opening times were noted, and vaginal smears were taken. By using RT-qPCR and Western blot, the mRNA and protein expression of AMH, GATA4, SF1, and SOX9 in the ovarian tissues of the two groups was investigated.The rats in the experimental group appeared to have obvious disorders of the estrus cycle, as evidenced by the ratio of estrus being significantly higher than that in the control group (P < 0.05); HE staining revealed that the ovarian volume, follicular vacuoles, and follicular lumen of the rats in the experimental group increased significantly.The ELISA results revealed that T and AMH in the experimental group were higher than those in the control group at day 15 and 20. AMH、GATA4 and SF1 mRNA and protein expression were higher in the experimental group than in the control group on day 15 and 20 (P < 0.05). On day 20, the experimental group outperformed the control group (P < 0.05). In the DHEA-induced PCOS rat model, androgen may have enhanced AMH expression via increasing the expression of genes associated to the AMH promoter binding site (GATA4, SF1, SOX9).


Subject(s)
Polycystic Ovary Syndrome , Humans , Rats , Female , Animals , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Sesame Oil/adverse effects , Dehydroepiandrosterone/pharmacology , RNA, Messenger , Anti-Mullerian Hormone/genetics
8.
Drug Discov Ther ; 17(2): 87-94, 2023 May 15.
Article in English | MEDLINE | ID: mdl-37019659

ABSTRACT

Diminished ovarian reserve (DOR) refers to the decline in fertility caused by the loss of normal ovarian function. DOR is associated with adverse reactions to ovarian stimulation during in vitro fertilization and embryo transfer (IVF-ET), increasing cycle cancellation rates and reducing pregnancy rates. Although it is well known that dehydroepiandrosterone (DHEA) can be used as a dietary supplement for age-related diseases, its potential has gradually been shown for many diseases. In this review, we focus on the effects of DHEA on DOR, briefly analysing its clinical benefits and limitations and describing the mechanism of function and the clinical trials conducted. Therefore, we summarize the mechanisms and indications of DHEA for DOR.


Subject(s)
Dehydroepiandrosterone , Ovarian Reserve , Pregnancy , Female , Humans , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Fertilization in Vitro , Pregnancy Rate , Ovary/physiology
9.
Mol Immunol ; 155: 110-123, 2023 03.
Article in English | MEDLINE | ID: mdl-36773597

ABSTRACT

Fatty liver hemorrhage syndrome (FLHS) seriously threatens the health and performance of laying hens, and the occurrence and development of FLHS are closely related to oxidative damage and inflammation; thus, diets supplemental with activated substances to relive the oxidative stress and inflammation maybe effectively control the occurrences of FLHS. Dehydroepiandrosterone (DHEA) has beneficial effects in fat-reduction, anti-oxidation and anti-inflammation, and it was widely applied to alleviate multiple metabolic-related diseases; however, there are few reports on whether DHEA can prevent against metabolic-related diseases by modulating oxidative stress and inflammation, especially FLHS in laying hens. Herein, present study aimed to investigate the regulatory actions and potential molecular mechanism of DHEA on inflammation and oxidative stress triggered by oleic acid (OA)-stimulation in primary chicken hepatocytes and chicken hepatocellular carcinoma cell line (LMH). The results showed that DHEA significantly alleviated oxidative stress challenged by OA-stimulation via activation of AMP-activated protein kinase (AMPK)-nuclear factor-erythroid 2-related factor 2 (Nrf2) signaling pathway in hepatocytes, which led to relieving effect of DHEA on inflammatory by inhibiting mitogen-activated protein kinases (MAPKs) and nuclear factor κB (NF-κB) signaling pathways. Mechanistically, we found that the activation of AMPK-Nrf2 signaling pathway by DHEA treatment was mediated by G-protein coupled estrogen receptor (GPR30/GPER) in OA-stimulated hepatocytes. Further investigation found that DHEA activated the GPR30-mediated AMPK-Nrf2 signaling pathways to increase antioxidant capacity and inhibit mitochondrial reactive oxygen species (ROS) overproduction, which thereby inhibiting the activation of ROS-induced MAPK and NF-κB signaling pathways in OA-stimulated hepatocytes. Overall, these data demonstrated that DHEA attenuates the oxidative stress and inflammation triggered by OA-stimulation, and these beneficial effects of DHEA are achieved by activating the GPR30-mediated AMPK-Nrf2 signaling to prevent the impairment of mitochondrial function, and thereby inhibiting the activation of ROS-induced MAPK and NF-κB signaling pathways in hepatocytes. These results revealed the effects and mechanisms of DHEA on oxidative stress and inflammation, and also provide substantial information to support it as a potential nutritional supplement in preventing the occurrences of FLHS in laying hens and other metabolic-related diseases in animals and humans.


Subject(s)
AMP-Activated Protein Kinases , Oleic Acid , Humans , Animals , Female , Reactive Oxygen Species/metabolism , AMP-Activated Protein Kinases/metabolism , Oleic Acid/adverse effects , Oleic Acid/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Chickens , Oxidative Stress , Hepatocytes/metabolism , Inflammation/metabolism , Receptors, G-Protein-Coupled/metabolism , Dehydroepiandrosterone/pharmacology
10.
Zygote ; 31(2): 111-122, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36617989

ABSTRACT

Although the role of myo-inositol (MYO) in promoting the oocyte quality of PCOS patients has been documented in human studies; the cellular effects of this supplement on oocytes have not been directly examined due to ethical limitations. In the first phase of this study, MYO dosimetry was carried out simultaneously with the PCOS model development. An effective dose was obtained following the assessment of fasting insulin and testosterone levels using ELISA and ovarian morphology appraisal by histopathology. In the second phase, following the continuous administration of the effective dose of MYO and dehydroepiandrosterone (DHEA), cellular evaluation was performed. The quality of oocytes from superovulation was analyzed by examining maturity and normal morphology percentage using a stereomicroscope, intracellular reactive oxygen species (ROS) and glutathione (GSH) levels using fluorometry, and ATP count evaluation using ELISA. The results revealed that, among the four different MYO concentrations, the 0.36 mg/g dose compared with the DHEA group reduced testosterone levels and large atretic antral follicles (LAtAnF) diameter. This dose also increased the corpus luteum count and the granulosa:theca (G/T)layer thickness ratio in antral follicles. Furthermore, this dose increased mature oocytes and normal morphology percentage, ATP count, and GSH levels; however, it decreased ROS levels in mature oocytes. Our findings provide the grounds for further cellular and molecular studies on the PCOS mouse model, suggesting that the improvement in mitochondrial function and its antioxidant properties is probably one of the mechanisms by which MYO increases oocyte quality.


Subject(s)
Folic Acid , Polycystic Ovary Syndrome , Female , Animals , Mice , Humans , Folic Acid/pharmacology , Reactive Oxygen Species , Inositol/pharmacology , Oocytes , Glutathione , Testosterone/pharmacology , Dehydroepiandrosterone/pharmacology , Adenosine Triphosphate/pharmacology
11.
Poult Sci ; 102(2): 102385, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36565630

ABSTRACT

The incident of lipid metabolism disorders has obviously increased under the undue pursuit of efficiency, which had seriously threatened to the health development of poultry industry. As an important cholesterol-derived intermediate, though dehydroepiandrosterone (DHEA) has the fat-reduction effect in animals and humans, but the underlying mechanism still poorly understood. Herein, the present study aimed to investigate the regulatory effects and its molecular mechanism of DHEA on disturbance of lipid metabolism induced by oleic acid (OA) in primary chicken hepatocytes. The hepatocytes were treated with 0, 0.1, 1, 10 µM DHEA for 4 h, and then supplemented with 0 or 0.5 mM OA stimulation for another 24 h. Our findings demonstrated that DHEA treatment effectively reduced TG content and alleviated lipid droplet deposition in OA-induced hepatocytes. DHEA inhibited the lipogenesis related factors (ACC, FAS, SREBP-1c, and ACLY) mRNA level and increased the lipolysis key factors (CPT-1 and PPARα) mRNA levels. In addition, DHEA obviously elevated the protein levels of CPT-1A, p-ACC, and ECHS1; whereas decreased the protein levels of FAS and SREBP-1 in hepatocytes stimulated by OA. Furthermore, DHEA promoted the phosphorylation of AMP-activated protein kinase (AMPK) and inhibited the phosphorylation of mammalian target of rapamycin (mTOR). Mechanistically, the hepatocytes were pre-treated with AMPK inhibitor compound C or AMPK activator AICAR before addition of DHEA treatment, and the results certified that DHEA activated cAMP/AMPK pathway and which subsequently led the inhibition of mTOR signal, which finally reduced the fat excessive accumulation in OA-stimulated hepatocytes. Collectively, our study unveiled that DHEA protects against the lipid metabolism disorders triggered by OA stimulation through activation of AMPK-mTOR signaling pathway, which prompts the value of DHEA as a potential nutritional supplement in regulating the lipid metabolism and its related disease in poultry.


Subject(s)
AMP-Activated Protein Kinases , Lipid Metabolism Disorders , Animals , AMP-Activated Protein Kinases/metabolism , Chickens/genetics , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/metabolism , Hepatocytes , Lipid Metabolism , Lipid Metabolism Disorders/metabolism , Lipid Metabolism Disorders/veterinary , Mammals/genetics , Oleic Acid/pharmacology , Oleic Acid/metabolism , RNA, Messenger/genetics , Signal Transduction , Sirolimus , TOR Serine-Threonine Kinases/metabolism
12.
Mini Rev Med Chem ; 23(8): 941-952, 2023.
Article in English | MEDLINE | ID: mdl-36121077

ABSTRACT

Dehydroepiandrosterone (DHEA) is the most abundant steroid hormone in primates, which is predominantly synthesized in the adrenal cortex. A characteristic curve of growth and decline of its synthesis during life was observed, together with the corresponding formation of its sulphate ester (DHEAS). High levels of plasma circulating DHEA are suggested as a marker of human longevity, and various pathophysiological conditions lead to a decreased DHEA level, including adrenal insufficiency, severe systemic diseases, acute stress, and anorexia. More recent studies have established the importance of DHEA in the central nervous system (CNS). A specific intranuclear receptor for DHEA has not yet been identified; however, highly specific membrane receptors have been detected in endothelial cells, the heart, kidney, liver, and the brain. Research shows that DHEA and DHEAS, as well as their metabolites, have a wide range of effects on numerous organs and organ systems, which places them in the group of potential pharmacological agents useful in various clinical entities. Their action as neurosteroids is especially interesting due to potential neuroprotective, pro-cognitive, anxiolytic, and antidepressant effects. Evidence from clinical studies supports the use of DHEA in hypoadrenal individuals and in treating depression and associated cognitive disorders. However, there is also an increasing trend of recreational DHEA misuse in healthy people, as it is classified as a dietary supplement in some countries. This article aims to provide a critical review regarding the biological and pharmacological effects of DHEA, its mechanism of action, and potential therapeutic use, especially in CNS disorders.


Subject(s)
Dehydroepiandrosterone , Endothelial Cells , Animals , Humans , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Endothelial Cells/metabolism , Dehydroepiandrosterone Sulfate/metabolism , Dehydroepiandrosterone Sulfate/pharmacology , Brain/metabolism , Steroids
13.
Comb Chem High Throughput Screen ; 26(5): 916-927, 2023.
Article in English | MEDLINE | ID: mdl-35996249

ABSTRACT

BACKGROUND AND OBJECTIVE: Although many trials have evaluated the use of dehydroepiandrosterone to improve outcomes in poor responders undergoing assisted reproductive technology treatment, evidence supporting this approach is controversial. We aimed to conduct a systematic review and meta-analysis of existing published data to further elucidate and supplement the use of Dehydroepiandrosterone (DHEA) to improve the effectiveness of vitro fertilization in patients with diminished ovarian reserve or adverse ovarian reactions. METHODS: PubMed, Embase, Cochrane Library, and the Web of Science databases were searched through December 2020. Oocyte yield, metaphase II oocytes, fertilized oocytes, top-quality embryos, clinical pregnancy rate, ongoing pregnancy rate, and live birth rate were analyzed as relative outcomes. Meta-analysis was performed and fitted to both fixed-effects models and random-effects models. RESULTS: Eight prospective randomized controlled studies, five prospective case-control studies, and three retrospective cohort studies were conducted with a total of 1998 participants. Meta-analyses of these studies showed a significantly higher number of oocytes retrieved (WMD 1.09, 95% CI 0.38 to 1.80), metaphase II oocytes (WMD 0.78, 95% CI 0.16 to 1.40), fertilized oocytes (WMD 0.84, 95% CI 0.42 to 1.26), top-quality embryos (WMD 0.60, 95% CI 0.34 to 0.86), clinical pregnancy rate (RR 1.35, 95% CI 1.13 to 1.61), and ongoing pregnancy rate (RR 1.82, 95% CI 1.34 to 2.46), although there was no difference in live birth rate (RR 1.35, 95% CI 0.94 to 1.94) in the DHEA supplementation groups compared with that in the control groups. CONCLUSION: Oral DHEA supplementation appears to improve some IVF outcomes. On the basis of this limited evidence, we conclude that further studies are required to provide sufficient data.


Subject(s)
Dehydroepiandrosterone , Fertilization in Vitro , Pregnancy , Female , Humans , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Retrospective Studies , Pregnancy Rate , Ovary
14.
Endocrinology ; 163(12)2022 10 23.
Article in English | MEDLINE | ID: mdl-36201601

ABSTRACT

Dehydroepiandrosterone (DHEA), an adrenal androgen precursor, can be metabolized in target tissues into active sex steroids. It has been proposed that DHEA supplementation might result in restoration of physiological local sex steroid levels, but knowledge on the effect of DHEA treatment on local sex steroid levels in multiple tissues is lacking. To determine the effects of DHEA on tissue-specific levels of sex steroids, we treated orchiectomized (ORX) male mice with DHEA for 3 weeks and compared them with vehicle-treated ORX mice and gonadal intact mice. Intra-tissue levels of sex steroids were analyzed in reproductive organs (seminal vesicles, prostate, m. levator ani), major body compartments (white adipose tissue, skeletal muscle, and brain), adrenals, liver, and serum using a sensitive and validated gas chromatography-mass spectrometry method. DHEA treatment restored levels of both testosterone (T) and dihydrotestosterone (DHT) to approximately physiological levels in male reproductive organs. In contrast, this treatment did not increase DHT levels in skeletal muscle or brain. In the liver, DHEA treatment substantially increased levels of T (at least 4-fold) and DHT (+536%, P < 0.01) compared with vehicle-treated ORX mice. In conclusion, we provide a comprehensive map of the effect of DHEA treatment on intra-tissue sex steroid levels in ORX mice with a restoration of physiological levels of androgens in male reproductive organs while DHT levels were not restored in the skeletal muscle or brain. This, and the unexpected supraphysiological androgen levels in the liver, may be a cause for concern considering the uncontrolled use of DHEA.


Subject(s)
Androgens , Dihydrotestosterone , Male , Mice , Animals , Dihydrotestosterone/pharmacology , Androgens/pharmacology , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/metabolism , Testosterone , Dietary Supplements
15.
Life Sci ; 308: 120926, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36058264

ABSTRACT

Fatty liver hemorrhagic syndrome (FLHS) seriously threatens the layer industry due to it can cause a sudden decline in egg production and acute death, and dietary supplement with bioactive substance is considered an effective way to prevent the FLHS occurrence. Dehydroepiandrosterone (DHEA) is a popular dietary supplement and it possesses anti-oxidative and anti-inflammatory functions; however, the effect and underlying mechanism about DHEA in protecting against the occurrence and development of FLHS remain elucidated. The current results showed that DHEA relieved HELP-induced decrease of egg productivity and liver injury in laying hens. Meanwhile, DHEA markedly enhanced the antioxidant capacity and then alleviated oxidative stress via activation of nuclear factor (erythroid-derived 2)-like 2 (NRF-2) signal in laying hens fed with HELP diets. In addition, DHEA significantly alleviated HELP-stimulated systemic inflammatory response by suppressing the overproduction of hepatic pro-inflammatory factors in laying hens, and further found this beneficial effect was achieved by blocking the activation of NF-κB pathway. Furthermore, we found that DHEA promoted the AMP-activated protein kinase α (AMPKα) activation and increased the G-protein-coupled estrogen receptor (GPER) expression level in laying hens fed with HELP diets. In summary, our data demonstrated that DHEA attenuates oxidative stress and inflammation through the activation of GPER-AMPK signal axis in laying hens fed with HELP diets. These results might facilitate an understanding of the benefits and mechanism of DHEA on the development of FLHS, and provide sufficient data to support it as a dietary supplement to control the FLHS-related metabolic diseases in chickens.


Subject(s)
Fatty Liver , Poultry Diseases , AMP-Activated Protein Kinases/metabolism , Abnormalities, Multiple , Animal Feed/analysis , Animals , Antioxidants/metabolism , Chickens/metabolism , Craniofacial Abnormalities , Dehydroepiandrosterone/pharmacology , Diet , Diet, Protein-Restricted , Estrogens , Fatty Liver/metabolism , Female , GTP-Binding Proteins/metabolism , Growth Disorders , Heart Septal Defects, Ventricular , Hemorrhage/etiology , NF-kappa B/metabolism , Oxidative Stress , Poultry Diseases/etiology , Poultry Diseases/metabolism , Receptors, Estrogen/metabolism , Signal Transduction
16.
Pak J Pharm Sci ; 35(3): 701-710, 2022 May.
Article in English | MEDLINE | ID: mdl-35791467

ABSTRACT

Androgen exposure may be an important factor in promoting the development of polycystic ovary syndrome (PCOS) and disease progression. Bushen Huoxue Formula (BHF), a traditional Chinese medicine, is prescribed in clinical settings as a PCOS remedy, albeit with unclear pharmacological effects on granulosa cells. The present research explores potentially advantageous BHF impacts and whereby BHF alleviates dehydroepiandrosterone (DHEA)-induced inflammation and endocrine disruption. Six chemical components in BHF were identified and fingerprint analysis showed good reproducibility. Using a human granulosa cell line (KGN), BHF effects on cell viability, secretion of steroidogenic and inflammatory factors were evaluated and TLR4/NF-κB pathway expression was examined. Our results demonstrate that BHF treatment of KGN cells in a DHEA-induced inflammatory state led to increased cell viability, decreased testosterone and estradiol production, and decreased CYP19A1 and HSD3B2 mRNA expression. Further experiments revealed that BHF inhibited the expression of pro-inflammatory cytokines and considerably hindered up-regulation in protein levels of TLR4, MyD88, and TRAF6, while inhibiting the activation of NF-κB and phosphorylation of IκBα. Collectively, BHF administration protected granulosa cells from DHEA-induced injuries through down-regulating pro-inflammatory cytokines and blocking the pathway of TLR4/NF-κB. Therefore, BHF hold promise as a therapeutic formulation for preventing androgen induced PCOS.


Subject(s)
NF-kappa B , Polycystic Ovary Syndrome , Androgens/metabolism , Androgens/pharmacology , Androgens/therapeutic use , Cytokines/metabolism , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Drugs, Chinese Herbal , Female , Granulosa Cells/metabolism , Humans , Inflammation/metabolism , NF-kappa B/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , Reproducibility of Results , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
17.
Oxid Med Cell Longev ; 2022: 2606880, 2022.
Article in English | MEDLINE | ID: mdl-35585881

ABSTRACT

Cerebral oxygenation disturbances contribute to the pathogenesis of brain lesions in preterm infants with white matter damage. These children are at risk of developing long-term neurodevelopmental disabilities. Preterm birth is associated with sudden hormonal changes along with an untimely increase in oxygen tissue tension. There is a persistent high postnatal production of fetal zone steroids (FZS), which serve in the fetoplacental unit as precursors for placental estrogen synthesis during pregnancy. The role of FZS in events associated with oxygenation differences and their impact on the developing white matter is not well understood. Therefore, we investigated the effect of hyperoxia (80% O2) and subsequent administration of FZS on the protein composition and migration capabilities of immature oligodendrocytes using the OLN93 (rat-derived OPC) cell line as an experimental model. We tested the effect of the FZS, dehydroepiandrosterone (DHEA), 16α-OH-DHEA, and adiol (5-androstene-3ß, 17ß-diol). After 24-hour exposure to hyperoxia, we monitored the changes in the proteome profile following treatment and observed significant alterations in pathways regulating cytoskeletal remodelling, cell migration, and cell survival. Additionally, hyperoxia leads to impaired migration of the OLN93 cells in culture. Administration of the FZS showed positive effects on the migration process under normoxic conditions in general. However, under hyperoxic conditions, the trend was less prominent. The observed effects could be related to changes in levels of cofilin/LIMK pathway-associated proteins. Adiol had a negative effect when administered together with estradiol, and the proteomic data reveal the activation of ephrin receptor signalling that might be responsible for the attenuation of migration. The results suggest that FZS can differentially regulate pathways involved in the migration of OLN93 cells. A deeper insight into the precise role of endogenous FZS would be an essential prerequisite for developing new treatment strategies including supplementation of estradiol and other steroids in preterm infants.


Subject(s)
Hyperoxia , Oligodendrocyte Precursor Cells , Premature Birth , Animals , Dehydroepiandrosterone/pharmacology , Estradiol/pharmacology , Female , Humans , Hyperoxia/metabolism , Infant, Newborn , Infant, Premature/metabolism , Oligodendrocyte Precursor Cells/metabolism , Placenta/metabolism , Pregnancy , Proteomics , Rats , Steroids/pharmacology
18.
Int J Eat Disord ; 55(6): 733-746, 2022 06.
Article in English | MEDLINE | ID: mdl-35460091

ABSTRACT

OBJECTIVE: This study was designed to determine the status of dehydroepiandrosterone (DHEA) in women with anorexia nervosa (AN) and to assess the efficacy of DHEA supplementation as a treatment for bone health in women with AN. METHOD: Studies were retrieved from the PubMed, Embase, Cochrane Library, MEDLINE, and Scopus databases from inception to February 14, 2022. Observational studies that compared serum DHEA levels between women with AN and healthy controls were included for meta-analysis, and randomized controlled trials (RCTs) that evaluated the effects of DHEA supplementation on bone mass were reviewed. RESULTS: Meta-analysis of 15 cross-sectional studies revealed that patients with AN had significantly elevated serum DHEA levels (mean difference (MD) = 311.63 ng/dl; 95% confidence interval (CI), 78.01-545.25) and reduced DHEAS levels (MD = -24.90 µg/dl; 95% CI, -41.72 to -8.07) compared with healthy controls. A systematic review of seven RCTs found that DHEA monotherapy does not improve bone mineral density (BMD) compared with placebo after adjusting for weight gain. While the combination of DHEA and conjugated oral contraceptives has led to increased bone strength and decreased bone loss, the beneficial effect appears to be limited to older adolescents and adults with closed physes. Potential detrimental effects on BMD were identified in younger adolescents with open physes in one study. DISCUSSION: Due to the lack of apparent benefit of DHEA in women with AN and its potential detrimental effect on BMD in young patients with AN, current evidence does not support the use of DHEA. PUBLIC SIGNIFICANCE: This study demonstrates that women with anorexia nervosa have abnormal levels of dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEAS), which have been suggested by previous studies to play a role in the development of low bone density in this condition. However, current evidence does not support the use of DHEA as a treatment to preserve bone health in patients with anorexia nervosa given the lack of clear benefit following its use and also because of a potential detrimental effect on bone mineral density in young patients with anorexia nervosa.


OBJETIVO: Este estudio fue diseñado para determinar el estado de la dehidroepiandrosterona (DHEA) en mujeres con anorexia nerviosa (AN) y para evaluar la eficacia de la suplementación con DHEA como tratamiento para la salud ósea en mujeres con AN. MÉTODO: Los estudios se obtuvieron de las bases de datos PubMed, Embase, Cochrane library, MEDLINE y Scopus desde su inicio hasta el 14 de febrero de 2022. Se incluyeron estudios observacionales que compararon los niveles séricos de DHEA entre mujeres que padecen AN y controles sanos para el metanálisis, y se revisaron los ensayos controlados aleatorios (ECA) que evaluaron los efectos de la suplementación con DHEA sobre la masa ósea. RESULTADOS: El metanálisis de 15 estudios transversales reveló que los pacientes que padecen AN tenían niveles séricos significativamente elevados de DHEA (diferencia de medias [DM] = 311,63 ng/dL; intervalo de confianza [IC] del 95%, 78,01-545,25) y niveles reducidos de DHEAS (DM = -24,90 µg/dL; IC del 95%, -41,72 a -8,07) en comparación con los controles sanos. La revisión sistemática de siete ECA encontró que la monoterapia con DHEA no mejora la densidad mineral ósea (DMO) en comparación con placebo después de ajustar el aumento de peso. Si bien la combinación de DHEA y anticonceptivos orales conjugados ha llevado a un aumento de la fuerza ósea y una disminución de la pérdida ósea, el efecto beneficioso parece limitarse a adolescentes mayores y adultos con placas de crecimiento cerradas. En un estudio se identificaron posibles efectos perjudiciales sobre la DMO en adolescentes más jóvenes con placas de crecimiento abiertas. DISCUSIÓN: Debido a la falta de beneficio aparente de la DHEA en mujeres que padecen AN y su posible efecto perjudicial sobre la DMO en pacientes jóvenes que padecen AN, la evidencia actual no apoya el uso de la DHEA.


Subject(s)
Anorexia Nervosa , Bone Density , Adolescent , Adult , Anorexia Nervosa/chemically induced , Anorexia Nervosa/drug therapy , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Dietary Supplements , Female , Humans
19.
Cell Signal ; 95: 110336, 2022 07.
Article in English | MEDLINE | ID: mdl-35452821

ABSTRACT

The intratumoral androgen synthesis is one of the mechanisms by which androgen receptor (AR) is aberrantly re-activated in castration-resistant prostate cancer (CRPC) after androgen ablation. However, pathways controlling steroidogenic enzyme expression and de novo androgen synthesis in prostate cancer (PCa) cells are largely unknown. In this study, we explored the potential roles of DAB2IP in testosterone synthesis and CRPC tumor growth. Indeed, DAB2IP loss could maintain AR transcriptional activity, PSA re-expression and tumor growth under castrated condition in vitro and in vivo, and reprogram the expression profiles of steroidogenic enzymes, including AKR1C3. Mechanistically, DAB2IP could dramatically inhibit the AKR1C3 promoter activity and the conversion from androgen precursors (i.e., DHEA) to testosterone through PI3K/AKT/mTOR/ETS1 signaling. Consistently, there was a high co-expression of ETS1 and AKR1C3 in PCa tissues and xenografts, and their expression in prostate tissues could also restore AR nuclear staining in castrated DAB2IP-/- mice after DHEA supplement. Together, this study reveals a novel regulation of intratumoral de novo androgen synthesis in CRPC, and provides the DAB2IP/ETS1/AKR1C3 signaling as a potential therapeutic target.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3 , Androgens , Prostatic Neoplasms, Castration-Resistant , Proto-Oncogene Protein c-ets-1 , Testosterone , ras GTPase-Activating Proteins , Aldo-Keto Reductase Family 1 Member C3/metabolism , Androgens/metabolism , Animals , Cell Line, Tumor , Dehydroepiandrosterone/pharmacology , Humans , Male , Mice , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Proto-Oncogene Protein c-ets-1/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Testosterone/biosynthesis , Testosterone/metabolism , ras GTPase-Activating Proteins/metabolism
20.
Cell Death Dis ; 13(4): 372, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440074

ABSTRACT

As a widely acknowledged FDA-approved dietary supplement or over-the-counter medicines, dehydroepiandrosterone (DHEA) exerts anti-inflammatory and immunomodulatory function. Pyroptosis is an important form of programmed cell death (PCD), and which acts a key role in the body's anti-infection and inflammatory responses. But the effects and mechanisms of DHEA on pyroptosis remain unclear. Here, we found that DHEA inhibited the NLRP3 inflammasome components expression by blocking inflammatory signals in lipopolysaccharide (LPS)-primed macrophages, and prevented the bacterial toxin nigericin (Nig)-induced NLRP3 inflammasome assembly. However, DHEA exacerbated NLRP3-independent cell death in Nig-treated inflammatory macrophages. During this process, DHEA induced the abnormal autophagy, which reflected as the blocking of autophagic flux and the accumulation of autophagy receptor p62 (SQSTM1) protein. In addition, DHEA caused a burst of reactive oxygen species (ROS) and activated extracellular signal-regulated kinase (ERK) phosphorylation in LPS plus Nig-stimulated macrophages but not in LPS-treated macrophages. Mechanistically, the present study certified that the activation of G protein-coupled estrogen receptor (GPER) signal mediated the cell death induced by DHEA in Nig-stimulated inflammatory macrophages, as GPER specific inhibitor G15 alleviated the abnormal autophagy and ultimately prevented the gasdermin D (GSDMD)-mediated pyroptosis induced by DHEA. Collectively, DHEA can exacerbate Nig-induced abnormal autophagy and pyroptosis via activation of GPER in LPS-primed macrophages, which prompts us the potential application value of DHEA in anti-infection or anti-tumor immunity.


Subject(s)
Lipopolysaccharides , Pyroptosis , Autophagy , Dehydroepiandrosterone/pharmacology , Inflammasomes/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nigericin/pharmacology , Receptors, G-Protein-Coupled/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL