Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.272
Filter
1.
Appl Microbiol Biotechnol ; 108(1): 399, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951177

ABSTRACT

Dehydroepiandrosterone (DHEA) has a promising market due to its capacity to regulate human hormone levels as well as preventing and treating various diseases. We have established a chemical esterification coupled biocatalytic-based scheme by lipase-catalyzed 4-androstene-3,17-dione (4-AD) hydrolysis to obtain the intermediate product 5-androstene-3,17-dione (5-AD), which was then asymmetrically reduced by a ketoreductase from Sphingomonas wittichii (SwiKR). Co-enzyme required for KR is regenerated by a glucose dehydrogenase (GDH) from Bacillus subtilis. This scheme is more environmentally friendly and more efficient than the current DHEA synthesis pathway. However, a significant amount of 4-AD as by-product was detected during the catalytic process. Focused on the control of by-products, we investigated the source of 4-AD and identified that it is mainly derived from the isomerization activity of SwiKR and GDH. Increasing the proportion of glucose in the catalytic system as well as optimizing the catalytic conditions drastically reduced 4-AD from 24.7 to 6.5% of total substrate amount, and the final yield of DHEA achieved 40.1 g/L. Furthermore, this is the first time that both SwiKR and GDH have been proved to be promiscuous enzymes with dehydrogenase and ketosteroid isomerase (KSI) activities, expanding knowledge of the substrate diversity of the short-chain dehydrogenase family enzymes. KEY POINTS: • A strategy of coupling lipase, ketoreductase, and glucose dehydrogenase in producing DHEA from 4-AD • Both SwiKR and GDH are identified with ketosteroid isomerase activity. • Development of catalytic strategy to control by-product and achieve highly selective DHEA production.


Subject(s)
Dehydroepiandrosterone , Lipase , Sphingomonas , Dehydroepiandrosterone/metabolism , Lipase/metabolism , Sphingomonas/enzymology , Sphingomonas/metabolism , Biocatalysis , Bacillus subtilis/enzymology , Bacillus subtilis/metabolism , Bacillus subtilis/genetics , Glucose 1-Dehydrogenase/metabolism , Glucose 1-Dehydrogenase/genetics , Androstenedione/metabolism , Androstenedione/biosynthesis , Hydrolysis
2.
Stress ; 27(1): 2330009, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38952223

ABSTRACT

Dehydroepiandrosterone (DHEA) and cortisol release appear to have contrasting effects on stress perception during stressful tasks. This study aimed to investigate anticipatory examination stress in college students by considering DHEA, cortisol, psycho-emotional aspects and examination performance. Seventy-six students (66 females, 10 males; age range 18-25 years) provided saliva samples and completed questionnaires in two sessions 48 hours apart. During the second session, the students performed the examination. The questionnaires used were the State-Trait Anxiety Inventory, the Positive and Negative Affect Scale, and the Brief-Coping Orientation to Problems Experienced Inventory. DHEA, cortisol, anxiety and negative affect showed an anticipatory rise before the examination (all ps < 0.001). This rise of DHEA and cortisol was associated with lower positive affect (p = 0.001 and p = 0.043, respectively). However, only the DHEA anticipatory levels were linked to poorer examination marks (p = 0.020). Higher levels of the DHEA/cortisol ratio in anticipation of the examination were related to lower scores on the support-seeking strategy (p = 0.022). There was no association between DHEA and cortisol levels and anxiety, negative affect, active and avoidant coping strategies, or academic record. These results suggest that how DHEA and cortisol respond in anticipation of examination stress significantly impacts students' emotional well-being during examination periods and how they cope with stress. They also suggest that levels of DHEA in anticipation of an academic stressor have detrimental effects on stress management.


Subject(s)
Adaptation, Psychological , Affect , Anxiety , Dehydroepiandrosterone , Hydrocortisone , Saliva , Stress, Psychological , Students , Humans , Male , Female , Hydrocortisone/metabolism , Hydrocortisone/analysis , Dehydroepiandrosterone/analysis , Dehydroepiandrosterone/metabolism , Young Adult , Students/psychology , Adult , Adolescent , Saliva/chemistry , Stress, Psychological/metabolism , Stress, Psychological/psychology , Affect/physiology , Anxiety/psychology , Surveys and Questionnaires , Anticipation, Psychological/physiology , Universities
3.
Psychoneuroendocrinology ; 166: 107072, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38733756

ABSTRACT

BACKGROUND: Frequent or prolonged exposure to stressors may jeopardize young children's health. The onset of the COVID-19 pandemic, coupled with disruptions in daily routines and social isolation resulting from public health preventive measures, have raised concerns about its potential impact on children' experienced stress, particularly for young children and vulnerable families. However, whether the pandemic was accompanied by changes in physiological stress remains unknown as perceived stress is not a good proxy of physiological stress. This study examined if preschoolers showed increasing hair steroid concentrations following the onset of the COVID-19 pandemic and whether family characteristics may have exacerbated or buffered these changes. METHODS: 136 preschoolers (2-4 years) provided hair for steroid measurement (cortisol, dehydroepiandrosterone (DHEA), cortisone, cortisol-to-DHEA ratio, cortisol-to-cortisone ratio) in October-November 2019 (T0) and in July-August 2020 (T1). A 2-centimeter hair segment was analyzed, reflecting steroid production over the two months leading up to collection. Family income, conflict resolution and lack of cohesion, as well as parents' COVID-19 stress were reported by parents. Linear mixed models for repeated measures and Bayes factors were used. RESULTS: No significant changes were noted from before to after the onset of the COVID-19 pandemic for most hair steroids. However, a moderating role of family conflict resolution was noted. Children living with parents with a better ability to resolve conflicts had lower levels of DHEA compared to those who had more difficulty managing conflicts. Additionally, lower levels of family cohesion and income were linked to some steroids, especially DHEA, suggesting that these factors may relate to children's physiological stress. Finally, boys had higher DHEA levels than girls. CONCLUSION: Our findings suggest that stress biomarkers were comparable from before to during the COVID-19 pandemic. This observation holds true despite the pandemic being perceived by many as a novel, unpredictable, and potentially threatening event. Findings further suggest that family characteristics are associated with hair steroid, especially DHEA, which deserves further investigation.


Subject(s)
COVID-19 , Dehydroepiandrosterone , Family Characteristics , Hair , Hydrocortisone , SARS-CoV-2 , Stress, Psychological , Humans , Child, Preschool , COVID-19/metabolism , COVID-19/psychology , Male , Hair/chemistry , Hair/metabolism , Female , Hydrocortisone/analysis , Hydrocortisone/metabolism , Dehydroepiandrosterone/analysis , Dehydroepiandrosterone/metabolism , Stress, Psychological/metabolism , Cortisone/analysis , Cortisone/metabolism , Stress, Physiological/physiology
4.
J Inorg Biochem ; 257: 112582, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38723329

ABSTRACT

When subjected to γ-irradiation at cryogenic temperatures the oxygenated complexes of Cytochrome P450 CYP17A1 (CYP17A1) bound with either of the lyase substrates, 17α-Hydroxypregnenolone (17-OH PREG) or 17α-Hydroxyprogesterone (17-OH PROG) are shown to generate the corresponding lyase products, dehydroepiandrosterone (DHEA) and androstenedione (AD) respectively. The current study uses gas chromatography-mass spectrometry (GC/MS) to document the presence of the initial substrates and products in extracts of the processed samples. A rapid and efficient method for the simultaneous determination of residual substrate and products by GC/MS is described without derivatization of the products. It is also shown that no lyase products were detected for similarly treated control samples containing no nanodisc associated CYP17 enzyme, demonstrating that the product is formed during the enzymatic reaction and not by GC/MS conditions, nor the conditions produced by the cryoradiolysis process.


Subject(s)
Gas Chromatography-Mass Spectrometry , Steroid 17-alpha-Hydroxylase , Steroid 17-alpha-Hydroxylase/metabolism , Dehydroepiandrosterone/chemistry , Dehydroepiandrosterone/metabolism , 17-alpha-Hydroxyprogesterone/chemistry , 17-alpha-Hydroxyprogesterone/metabolism , 17-alpha-Hydroxypregnenolone/chemistry , 17-alpha-Hydroxypregnenolone/metabolism , Androstenedione/chemistry , Androstenedione/metabolism , Humans , Lyases/metabolism , Lyases/chemistry , Gamma Rays , Substrate Specificity , Oxygen/chemistry
5.
Psychoneuroendocrinology ; 165: 107049, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38657340

ABSTRACT

BACKGROUND: Past studies on schizophrenia (SCZ) and the stress-sensitive neuroendocrine systems have mostly focused on a single system and traditionally utilized acute biomarkers (e.g., biomarkers from blood, urine and saliva) that poorly match the chronic course of schizophrenia in time span. Using eight biomarkers in hair, this study aimed to explore the functional characteristics of SCZ patients in the hypothalamic-pituitary-adrenocortical (HPA) and hypothalamic-pituitary-gonadal (HPG) axes and the interaction between the two axes. METHODS: Hair samples were taken from 137 SCZ patients and 73 controls. The SCZ patients were diagnosed by their attending physician according to the Diagnostic and Statistical Manual of Mental Disorders IV and were clinically stable after treatment. Gender, age, BMI, frequency of hair washing, marital status, education level, family history of mental illness and clozapine dosage were concurrently collected as covariates. The 10-item perceived stress scale (PSS-10) and the social readjustment rating scale were used to assess chronic stress status in SCZ patients. Eight hair biomarkers, cortisol, cortisone, dehydroepiandrosterone (DHEA), testosterone, progesterone, cortisol/cortisone, cortisol/DHEA and cortisol/testosterone, were measured by high performance liquid chromatography tandem mass spectrometer. Among them, cortisol, cortisone, DHEA and cortisol/DHEA reflected the functional activity of the HPA axis, and testosterone and progesterone reflected the functional activity of the HPG axis, and cortisol/cortisone reflected the activity of 11ß-hydroxysteroid dehydrogenase types 2 (11ß-HSD 2), and cortisol/testosterone reflected the HPA-HPG interaction. RESULTS: SCZ patients showed significantly higher cortisone and cortisol/testosterone than controls (p<0.001, η²p=0.180 and p=0.015, η²p=0.031), lower testosterone (p=0.009, η²p=0.034), progesterone (p<0.001, η²p=0.069) and cortisol/cortisone (p=0.001, η²p=0.054). There were significant intergroup differences in male and female progesterone (p=0.003, η²p=0.088 and p=0.030, η²p=0.049) and female testosterone (p=0.028, η²p=0.051). In SCZ patients, cortisol, cortisol/cortisone, cortisol/DHEA and cortisol/testosterone were positively associated with PSS-10 score (ps<0.05, 0.212

Subject(s)
Biomarkers , Cortisone , Dehydroepiandrosterone , Hair , Hydrocortisone , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Schizophrenia , Stress, Psychological , Testosterone , Humans , Female , Male , Hypothalamo-Hypophyseal System/metabolism , Schizophrenia/metabolism , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Hair/chemistry , Hair/metabolism , Biomarkers/metabolism , Adult , Hydrocortisone/metabolism , Hydrocortisone/analysis , Cortisone/metabolism , Cortisone/analysis , Testosterone/metabolism , Testosterone/analysis , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone/analysis , Stress, Psychological/metabolism , Middle Aged , Progesterone/metabolism , Progesterone/analysis , Case-Control Studies
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167182, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653359

ABSTRACT

OBJECTIVE: This work aimed to investigate the role of rhythm gene PER1 in mediating granulosa cell ferroptosis and lipid metabolism of polycystic ovary syndrome (PCOS). METHODS: We injected dehydroepiandrosterone and Ferrostatin-1 (Fer-1) into mice to explore the mechanism of ferroptosis in PCOS. The effect of PER1 on ferroptosis-like changes in granulosa cells was explored by overexpression of PER1 plasmid transfection and Fer-1 treatment. RESULTS: We found that Fer-1 ameliorated the characteristic polycystic ovary morphology, suppressed ferroptosis in the PCOS mice. PER1 and ALOX15 were highly expressed in PCOS, whereas SREBF2 was lowly expressed. Overexpression of PER1 decreased granulosa cell viability and inhibited proliferation. Meanwhile, overexpression of PER1 increased lipid reactive oxygen species, 4-Hydroxynonenal (4-HNE), Malondialdehyde (MDA), total Fe, and Fe2+ levels in granulosa cells and decreased Glutathione (GSH) content. Fer-1, SREBF2 overexpression, or ALOX15 silencing treatment reversed the effects of PER1 overexpression on granulosa cells. PER1 binds to the SREBF2 promoter and represses SREBF2 transcription. SREBF2 binds to the ALOX15 promoter and represses ALOX15 transcription. Correlation analysis of clinical trials showed that PER1 was positively correlated with total cholesterol, low-density lipoprotein cholesterol, luteinizing hormone, testosterone, 4-HNE, MDA, total Fe, Fe2+, and ALOX15. In contrast, PER1 was negatively correlated with SREBF2, high-density lipoprotein cholesterol, follicle-stimulating hormone, progesterone, and GSH. CONCLUSION: This study demonstrates that the rhythm gene PER1 promotes ferroptosis and dysfunctional lipid metabolism in granulosa cells in PCOS by inhibiting SREBF2/ALOX15 signaling.


Subject(s)
Ferroptosis , Granulosa Cells , Lipid Metabolism , Polycystic Ovary Syndrome , Animals , Female , Humans , Mice , Arachidonate 12-Lipoxygenase , Arachidonate 15-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Cyclohexylamines/pharmacology , Dehydroepiandrosterone/metabolism , Ferroptosis/genetics , Granulosa Cells/metabolism , Granulosa Cells/pathology , Lipid Metabolism/genetics , Phenylenediamines/pharmacology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/pathology , Reactive Oxygen Species/metabolism
7.
Front Cell Infect Microbiol ; 14: 1328741, 2024.
Article in English | MEDLINE | ID: mdl-38665877

ABSTRACT

Polycystic ovary syndrome (PCOS) is a common systemic disorder related to endocrine disorders, affecting the fertility of women of childbearing age. It is associated with glucose and lipid metabolism disorders, altered gut microbiota, and insulin resistance. Modern treatments like pioglitazone, metformin, and spironolactone target specific symptoms of PCOS, while in Chinese medicine, moxibustion is a common treatment. This study explores moxibustion's impact on PCOS by establishing a dehydroepiandrosterone (DHEA)-induced PCOS rat model. Thirty-six specific pathogen-free female Sprague-Dawley rats were divided into four groups: a normal control group (CTRL), a PCOS model group (PCOS), a moxibustion treatment group (MBT), and a metformin treatment group (MET). The MBT rats received moxibustion, and the MET rats underwent metformin gavage for two weeks. We evaluated ovarian tissue changes, serum testosterone, fasting blood glucose (FBG), and fasting insulin levels. Additionally, we calculated the insulin sensitivity index (ISI) and the homeostasis model assessment of insulin resistance index (HOMA-IR). We used 16S rDNA sequencing for assessing the gut microbiota, 1H NMR spectroscopy for evaluating metabolic changes, and Spearman correlation analysis for investigating the associations between metabolites and gut microbiota composition. The results indicate that moxibustion therapy significantly ameliorated ovarian dysfunction and insulin resistance in DHEA-induced PCOS rats. We observed marked differences in the composition of gut microbiota and the spectrum of fecal metabolic products between CTRL and PCOS rats. Intriguingly, following moxibustion intervention, these differences were largely diminished, demonstrating the regulatory effect of moxibustion on gut microbiota. Specifically, moxibustion altered the gut microbiota by increasing the abundance of UCG-005 and Turicibacter, as well as decreasing the abundance of Desulfovibrio. Concurrently, we also noted that moxibustion promoted an increase in levels of short-chain fatty acids (including acetate, propionate, and butyrate) associated with the gut microbiota of PCOS rats, further emphasizing its positive impact on gut microbes. Additionally, moxibustion also exhibited effects in lowering FBG, testosterone, and fasting insulin levels, which are key biochemical indicators associated with PCOS and insulin resistance. Therefore, these findings suggest that moxibustion could alleviate DHEA-induced PCOS by regulating metabolic levels, restoring balance in gut microbiota, and modulating interactions between gut microbiota and host metabolites.


Subject(s)
Disease Models, Animal , Gastrointestinal Microbiome , Insulin Resistance , Moxibustion , Polycystic Ovary Syndrome , Rats, Sprague-Dawley , Animals , Polycystic Ovary Syndrome/therapy , Polycystic Ovary Syndrome/metabolism , Female , Moxibustion/methods , Rats , Dehydroepiandrosterone/metabolism , Blood Glucose/metabolism , Insulin/blood , Insulin/metabolism , Metformin/pharmacology , Testosterone/blood , Ovary/metabolism , Ovary/microbiology
8.
J Mol Histol ; 55(2): 187-199, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38478190

ABSTRACT

Polycystic ovary syndrome (PCOS) is an endocrine disease, and its pathogenesis and treatment are still unclear. Hexokinase domain component 1 (HKDC1) participates in regulating mitochondrial function and glycolysis. However, its role in PCOS development remains unrevealed. Here, female C57BL/6 mice were intraperitoneally injected with dehydroepiandrosterone (DHEA; 60 mg/kg body weight) to establish an in vivo model of PCOS. In vitro, KGN cells, a human ovarian granular cell line, were used to explore the potential mechanisms. DHEA-treated mice exhibited a disrupted estrus cycle, abnormal hormone levels, and insulin resistance. Dysfunction in mitochondria and glycolysis is the main reason for PCOS-related growth inhibition of ovarian granular cells. Here, we found that the structure of mitochondria was impaired, less ATP was generated and more mitochondrial Reactive Oxygen Species were produced in HKDC1-silenced KGN cells. Moreover, HKDC1 knockdown inhibited glucose consumption and decreased the production of glucose-6-phosphate and lactic acid. Conclusively, HKDC1 protects ovarian granulocyte cells from DHEA-related damage at least partly by preserving mitochondrial function and maintaining glycolysis.


Subject(s)
Polycystic Ovary Syndrome , Female , Mice , Humans , Animals , Polycystic Ovary Syndrome/metabolism , Hexokinase/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/metabolism , Granulocytes/metabolism , Granulocytes/pathology
9.
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
10.
Psychoneuroendocrinology ; 165: 107034, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38554595

ABSTRACT

BACKGROUND: Despite evidence that early life stress (ELS) can influence the functioning of the hypothalamic-pituitary-adrenal (HPA) axis and increase maladaptive behaviors in adolescence, less attention has been paid to the role of the coordinated effects of the two primary adrenal hormones, cortisol and dehydroepiandrosterone (DHEA), in these associations. METHODS: 138 typically developing adolescents (76 females) reported the stressful events experienced during childhood and early adolescence across 30 domains. Two years later we assessed levels of externalizing problems and obtained salivary levels of cortisol and DHEA. Using causal moderated mediation analyses, we examined whether the ratio of cortisol to DHEA (CD ratio) mediates the association between ELS and subsequent externalizing problems. RESULTS: We found that ELS is associated with both a lower CD ratio and more externalizing problems. Importantly, a lower CD ratio mediated the association between ELS and externalizing problems in boys. CONCLUSIONS: An imbalance in adrenal hormones may be a mechanism through which ELS leads to an increase in externalizing problems in adolescent boys. These findings underscore the utility of using the CD ratio to index HPA-axis functioning.


Subject(s)
Adverse Childhood Experiences , Dehydroepiandrosterone , Hydrocortisone , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Saliva , Stress, Psychological , Humans , Male , Hydrocortisone/metabolism , Hydrocortisone/analysis , Adolescent , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone/analysis , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Saliva/chemistry , Saliva/metabolism , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Female , Child , Adolescent Behavior/physiology , Adolescent Behavior/psychology , Problem Behavior/psychology
11.
Environ Pollut ; 345: 123329, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38281572

ABSTRACT

The biological pathways linking lead exposure to adverse outcomes are beginning to be understood. Rodent models suggest lead exposure induces dysfunction within the hypothalamic-pituitary-adrenal (HPA) axis and glucocorticoid regulation, a primary physiological stress response system. Over time, HPA axis and glucocorticoid dysfunction has been associated with adverse neurocognitive and cardiometabolic health, much like lead exposure. This systematic review utilized PRISMA guidelines to synthesize the literature regarding associations between lead exposure and downstream effector hormones of the HPA axis, including cortisol, a glucocorticoid, and dehydroepiandrosterone (DHEA), a glucocorticoid antagonist. We additionally determined the state of the evidence regarding lead exposure and allostatic load, a measure of cumulative body burden resultant of HPA axis and glucocorticoid dysfunction. A total of 18 articles were included in the review: 16 assessed cortisol or DHEA and 3 assessed allostatic load. Generally, the few available child studies suggest a significant association between early life lead exposure and altered cortisol, potentially suggesting the impact of developmental exposure. In adulthood, only cross sectional studies were available. These reported significant associations between lead and reduced cortisol awakening response and increased cortisol reactivity, but few associations with fasting serum cortisol. Two studies reported significant associations between increasing lead exposure and allostatic load in adults and another between early life lead exposure and adolescent allostatic load. The paucity of studies examining associations between lead exposure and allostatic load or DHEA and overall heterogeneity of allostatic load measurements limit conclusions. However, these findings cautiously suggest associations between lead and dysregulation of physiological stress pathways (i.e., glucocorticoids) as seen through cortisol measurement in children and adults. Future research would help to elucidate these associations and could further examine the physiological stress pathway as a mediator between lead exposure and detrimental health outcomes.


Subject(s)
Glucocorticoids , Hydrocortisone , Adult , Child , Adolescent , Humans , Glucocorticoids/toxicity , Glucocorticoids/metabolism , Hydrocortisone/metabolism , Lead/toxicity , Lead/metabolism , Hypothalamo-Hypophyseal System , Cross-Sectional Studies , Pituitary-Adrenal System/metabolism , Stress, Physiological , Dehydroepiandrosterone/metabolism , Stress, Psychological
12.
Psychoneuroendocrinology ; 159: 106668, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37944209

ABSTRACT

The COVID-19 pandemic generated significant life stress and increases in internalizing disorders. Moreover, COVID-related stressors disproportionately impacted women, consistent with outcomes showing a gender gap in stress-related disorders. Gender-related stress vulnerability emerges in adolescence alongside gender-specific changes in neuroendocrine signaling. Most research on the neuroendocrinology of stress-related disorders has focused on differences in the hypothalamic-pituitary-adrenal (HPA) axis effector hormone cortisol. More recent studies, however, emphasize dehydroepiandrosterone (DHEA), a neuroprotective and neuroactive hormone released concurrently with cortisol that balances its biobehavioral actions during stress. Notably, women show lower cortisol responses and higher DHEA responses to stress. However, lower cortisol and higher DHEA are associated with internalizing disorders in women, while those associations are opposite in men. Thus, gender-specific factors perhaps result in a neuroendocrine profile that places women at greater risk for stress-related disorders. The current study prospectively examined socially evaluated cold-pressor task (SECPT) induced neuroendocrine responses at age 15 and internalizing symptoms during the COVID-19 pandemic at age 21 in a cohort of 175 primarily Black low-socioeconomic status participants, while controlling for internalizing symptoms at age 15. The association between COVID-related stress and internalizing symptoms was not stronger in women. Lower DHEA-cortisol ratios were associated with a weaker relationship between COVID-related stress and internalizing symptoms in women, while higher ratios were associated with a weaker relationship in men. These findings suggest gender differences in the relationship between DHEA and cortisol and internalizing outcomes during a stressful period, and support differential neuroendocrine protective and risk pathways for young men and women.


Subject(s)
COVID-19 , Hydrocortisone , Male , Adolescent , Humans , Female , Young Adult , Adult , Hydrocortisone/metabolism , Pandemics , Stress, Psychological/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Psychophysiologic Disorders/metabolism , Saliva/metabolism , Dehydroepiandrosterone/metabolism
13.
J Steroid Biochem Mol Biol ; 238: 106452, 2024 04.
Article in English | MEDLINE | ID: mdl-38160767

ABSTRACT

CYP68JX, a P450 hydroxylase, derived from Colletotrichum lini ST-1 is capable of biotransforming dehydroepiandrosterone (DHEA) to 3ß,7α,15α-trihydroxy-5-androstene-17-one (7α,15α-diOH-DHEA). Redox partners and cofactor supply are important factors affecting the catalytic activity of CYP68JX. In this study, the heterologous expression of CYP68JX in Saccharomyces cerevisiae BY4741 was realized resulting in a 17.1% target product yield. In order to increase the catalytic efficiency of CYP68JX in S. cerevisiae BY4741, a complete cytochrome P450 redox system was constructed. Through the combination of CYP68JX and heterologous CPRs, the yield of the target product 7α,15α-diOH-DHEA in CYP68JX recombinant system was increased to 37.8%. Furthermore, by adding NADPH coenzyme precursor tryptophan of 40 mmol/L and co-substrate fructose of 20 g/L during the conversion process, the catalytic efficiency of CYP68JX was further improved, the target product yield reached 57.9% which was 3.39-fold higher than initial yield. Overall, this study provides a reference for improving the catalytic activity of P450s.


Subject(s)
Dehydroepiandrosterone , Saccharomyces cerevisiae , Dehydroepiandrosterone/metabolism , Hydroxylation , NADP/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Oxidation-Reduction , Steroids
14.
Methods Enzymol ; 689: 67-86, 2023.
Article in English | MEDLINE | ID: mdl-37802583

ABSTRACT

Cytochrome P450 aromatase (AROM) and steroid (estrone (E1)/dehydroepiandrosterone (DHEA)) sulfatase (STS) are the two key enzymes responsible for the biosynthesis of estrogens in human, and maintenance of the critical balance between androgens and estrogens. Human AROM, an integral membrane protein of the endoplasmic reticulum, is a member of the Fe-heme containing cytochrome P450 superfamily having a cysteine thiolate as the fifth Fe-coordinating ligand. It is the only enzyme known to catalyze the conversion of androgens with non-aromatic A-rings to estrogens characterized by the aromatic A-ring. Human STS, also an integral membrane protein of the endoplasmic reticulum, is a Ca2+-dependent enzyme that catalyzes the hydrolysis of sulfate esters of E1 and DHEA to yield the respective unconjugated steroids, the precursors of the most potent forms of estrogens and androgens, namely, 17ß-estradiol (E2), 16α,17ß-estriol (E3), testosterone (TST) and dihydrotestosterone (DHT). Expression of these steroidogenic enzymes locally within various organs and tissues of the endocrine, reproductive, and central nervous systems is the key for maintaining high levels of the reproductive steroids. Thus, the enzymes have been drug targets for the prevention and treatment of diseases associated with steroid hormone excesses, especially in breast and prostate malignancies and endometriosis. Both AROM and STS have been the subjects of vigorous research for the past six decades. In this article, we review the procedures of their extraction and purification from human term placenta are described in detail, along with the activity assays.


Subject(s)
Aromatase , Steryl-Sulfatase , Female , Humans , Pregnancy , Androgens/metabolism , Aromatase/metabolism , Dehydroepiandrosterone/metabolism , Estrogens/metabolism , Estrone/metabolism , Membrane Proteins/metabolism , Placenta/metabolism , Steryl-Sulfatase/metabolism
15.
Methods Enzymol ; 689: 89-119, 2023.
Article in English | MEDLINE | ID: mdl-37802584

ABSTRACT

The enzyme 3ß-hydroxysteroid dehydrogenase-1 (3ßHSD1), encoded by the gene HSD3B1, plays an essential role in the peripheral conversion of 3ß-OH, Δ5-steroids to 3-keto, Δ4-steroids. In human physiology, the adrenal produces dehydroepiandrosterone (DHEA) and DHEA-sulfate, which are major precursors for the biosynthesis of potent androgens and estrogens. DHEA is converted by 3ßHSD1 and subsequently is converted by steroid-5α-reductase to potent androgens or by aromatase to estrogens. Assessment of 3ßHSD1 is therefore critical under various conditions. In this chapter, we detail several approaches to assessing 3ßHSD1. First, we describe a genotyping protocol for the identification of a common missense-encoding variation that regulates 3ßHSD1 cellular metabolic activity. This protocol distinguishes between the HSD3B1(1245A) and the HSD3B1(1245C) allele which have lower and higher metabolic activity, respectively. Second, we detail mass spectrometry approaches to determining 3ßHSD1 activity using stable isotope dilution. Third, we describe methods for using tritiated DHEA and high performance liquid chromatography coupled with a beta-RAM to also determine 3ßHSD1 activity. Together, we provide multiple methods of directly assessing 3ßHSD1 activity or anticipated 3ßHSD1 activity.


Subject(s)
Androgens , Estrogens , Humans , Androgens/metabolism , Multienzyme Complexes/metabolism , Dehydroepiandrosterone/metabolism , Steroids
16.
ACS Appl Mater Interfaces ; 15(43): 50034-50046, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37851924

ABSTRACT

Measuring bioactive stress hormones, including cortisol and dehydroepiandrosterone (DHEA), allows for evaluating the hypothalamic-pituitary-adrenal (HPA) axis functioning, offering valuable insights into an individual's stress response through adrenocortex stress profiles (ASPs). Conventional methods for detecting steroid hormones involve sample collections and competitive immunoassays, which suffer from drawbacks such as time-consuming labeling and binding procedures, reliance on unstable biological receptors, and the need for sophisticated instruments. Here, we report a label-free and external redox reagent-free amperometric assay directly detecting sweat cortisol and DHEA levels on the skin. The approach utilizes multitarget sensors based on redox-active molecularly imprinted polymers (redox MIPs) capable of selectively binding cortisol and DHEA, inducing changes in electrochemical redox features. The redox MIP consists of imprinted cavities for specific capture of cortisol or DHEA in a poly(pyrrole-co-(dimethylamino)pyrrole) copolymer containing hydrophobic moieties to enhance affinity toward steroid hormones. The polymer matrix also incorporates covalently linked interpenetrating redox-active polyvinylferrocene, offering a stable electrochemical redox feature that enables sensitive current change in response to the target capture in the vicinity. The multiplexed sensor detects cortisol and DHEA within 5 min, with detection limits of 115 and 390 pM, respectively. Through the integration of redox MIP sensors into a wireless wearable sensing system, we successfully achieved ambulatory detection of these two steroid hormones in sweat directly on the skin. The new sensing method facilitates rapid, robust determination of the cortisol-DHEA ratio, providing a promising avenue for point-of-care assessment of an individual's physiological state.


Subject(s)
Molecular Imprinting , Wearable Electronic Devices , Dehydroepiandrosterone/metabolism , Hydrocortisone , Polymers , Pyrroles
17.
Prostate ; 83(13): 1306-1309, 2023 09.
Article in English | MEDLINE | ID: mdl-37321973

ABSTRACT

BACKGROUND: Conversion of adrenally produced dehydroepiandrosterone (DHEA) to the potent androgen dihydrotestosterone (DHT) is an important mechanism by which prostate cancer reaches castration resistance. At the start of this pathway is a branch point at which DHEA can be converted to Δ4 -androstenedione by the enzyme 3ß-hydroxysteroid dehydrogenase (3ßHSD) or to Δ5 -androstenediol by 17ßHSD. To better understand this process, we studied the kinetics of these reactions in cells. METHODS: Prostate cancer cells (LNCaP cell line) were incubated with steroids (DHEA and Δ5 -androstenediol) over a range of concentrations and the steroid metabolism reaction products were measured by mass spectrometry or by high-performance liquid chromatography to determine reaction kinetics. To confirm the generalizability of results, experiments were also performed in JEG-3 placental choriocarcinoma cells. RESULTS: The two reactions displayed very different saturation profiles, with only the 3ßHSD-catalyzed reaction beginning to saturate within a physiological substrate concentration range. Strikingly, incubating LNCaP cells with low (in the ~10 nM range) concentrations of DHEA resulted in a large majority of the DHEA undergoing 3ßHSD-catalyzed conversion to Δ4 -androstenedione, whereas high concentrations of DHEA (in the 100s of nM range) resulted in most of the DHEA undergoing 17ßHSD-catalyzed conversion to Δ5 -androstenediol. CONCLUSION: Contrary to expectations from previous studies that used purified enzyme, cellular metabolism of DHEA by 3ßHSD begins to saturate in the physiological concentration range, suggesting that fluctuations in DHEA concentrations could be buffered at the downstream active androgen level.


Subject(s)
Androgens , Prostatic Neoplasms , Humans , Male , Androstenediols , Androstenedione/metabolism , Cell Line, Tumor , Dehydroepiandrosterone/metabolism , Prostatic Neoplasms/pathology
18.
J Steroid Biochem Mol Biol ; 232: 106359, 2023 09.
Article in English | MEDLINE | ID: mdl-37390976

ABSTRACT

Estetrol (E4) has emerged as a novel and highly promising estrogen for therapeutic use. E4 is a weak natural estrogen produced only in pregnancy. Because of its novelty, there is considerable interest by clinicians in how it is produced in pregnancy. Although the fetal liver plays a key role in its production, the placenta is also involved. A current view is that estradiol (E2) formed in the placenta enters the fetal compartment and is then rapidly sulfated. E2 sulfate then undergoes 15α-/16α-hydroxylation in the fetal liver thereby forming E4 sulfate (phenolic pathway). However, another pathway involving 15α,16α-dihydroxy-DHEAS formed in the fetal liver and converted to E4 in the placenta also plays a significant role (neutral pathway). It is not known which pathway predominates, but both pathways appear to be important in E4 biosynthesis. In this commentary, we summarize the well-established pathways in the formation of estrogens in the nonpregnant and pregnant female. We then review what is known about the biosynthesis of E4 and describe the 2 proposed pathways involving the fetus and placenta.


Subject(s)
Estetrol , Pregnancy , Humans , Female , Estetrol/metabolism , Estrogens/metabolism , Estradiol/metabolism , Dehydroepiandrosterone/metabolism , Placenta/metabolism
19.
Iran J Med Sci ; 48(2): 187-197, 2023 03.
Article in English | MEDLINE | ID: mdl-36895462

ABSTRACT

Background: Polycystic ovary syndrome (PCOS) is the most common reproductive dysfunction in premenopausal women. PCOS is associated with oxidative stress (OS), which is the main risk factor for renal diseases. This study aimed to investigate the mechanisms responsible for renal injury in a hyperandrogenemic female rat model. Methods: This study was conducted from December 2019 to September 2021 at Shiraz Nephro-Urology Research Centre, Shiraz University of Medical Sciences (Shiraz, Iran). Thirty female Sprague-Dawley rats were randomly divided into three groups (n=10), namely control, sham, and dehydroepiandrosterone (DHEA). Plasma total testosterone, plasma creatinine (Cr), and blood urea nitrogen (BUN) levels were measured. In addition, total oxidant status (TOS), total antioxidant capacity (TAC), oxidative stress index (OSI), and histopathological changes in the ovaries and kidneys were determined. Data were analyzed using the GraphPad Prism software, and P<0.05 was considered statistically significant. Results: Plasma total testosterone levels increased by nine-fold in DHEA-treated rats compared to controls (P=0.0001). Administration of DHEA increased Cr and BUN levels and caused severe renal tubular cell injury. In addition, plasma and tissue (kidney and ovary) TAC levels decreased significantly, but TOS levels and OSI values were significantly increased (P=0.019). Significant damage to both glomerular and tubular parts of the kidney and ovarian follicular structure was observed in the DHEA group. Conclusion: Hyperandrogenemia caused systemic abnormalities through OS-related mechanisms and damaged renal and ovarian tissues. DHEA treatment in rat models is recommended to study the mechanisms that mediate PCOS-associated renal injury.


Subject(s)
Hyperandrogenism , Kidney Diseases , Polycystic Ovary Syndrome , Humans , Rats , Female , Animals , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Rats, Sprague-Dawley , Hyperandrogenism/complications , Hyperandrogenism/metabolism , Hyperandrogenism/pathology , Oxidative Stress , Kidney , Antioxidants/metabolism , Kidney Diseases/pathology , Testosterone/metabolism , Dehydroepiandrosterone/metabolism
20.
Org Biomol Chem ; 21(15): 3172-3176, 2023 04 12.
Article in English | MEDLINE | ID: mdl-36950968

ABSTRACT

Menarandroside A, which bears a 12α-hydroxypregnenolone steroid backbone, was isolated from the plant, Cynanchum menarandrense. Treatment of extracts from this plant containing menarandroside A against secretin tumor cell line (STC-1) intestinal cells, resulted in an increased secretion of glucagon-like peptide 1 (GLP-1), a peptide that plays a role in the regulation of blood sugar levels. Increase in GLP-1 is beneficial for the treatment of type 2 diabetes. We disclose the synthesis of menarandroside A from dehydroepiandrosterone (DHEA). Key features of this synthesis include: (i) Wittig reaction of the C17-ketone of a 12-oxygenated DHEA derivative to introduce the C17-acetyl moiety, and (ii) the stereoselective reduction of a C12-keto intermediate bearing an sp2-center at C17 to yield the C12α-hydroxy group. In addition, an oxidation of a methyl enol ether derivative to an α-hydroxy methyl ester using tetrapropylammonium perruthenate (TPAP) and N-methyl-morpholine-N-oxide (NMO) was discovered.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Steroids , Glucagon-Like Peptide 1/metabolism , Oxidation-Reduction , Dehydroepiandrosterone/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...