Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.961
Filter
1.
Dev Psychobiol ; 66(7): e22550, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39378306

ABSTRACT

Meconium, the first stool produced by neonates, has been used as an analyte for exogenous fetal exposures. However, few studies have investigated the relationship between meconium and androgen exposure in utero. Here, we examine the associations of testosterone and dehydroepiandrosterone (DHEA) across maternal antenatal salivary testosterone, cord blood, meconium, and infant salivary testosterone. A total of 47 women with singleton, uncomplicated pregnancies, and their infants were included in this study. Participants were recruited from an academic obstetric clinic. Maternal saliva was collected at 36-weeks' gestation. Cord blood and meconium were collected at birth. Infant salivary testosterone was collected at 1 and 4 weeks of age. Multivariate model results showed that meconium testosterone was associated with neonatal testosterone at 1 (F = 5.62, p = 0.029) and 4 weeks (F = 4.28, p = 0.048) postnatal age; no sex differences were detected. This study suggests meconium is a valuable tool for evaluating endogenous androgen exposure and should be used in future studies to investigate the fetal hormonal milieu.


Subject(s)
Biomarkers , Dehydroepiandrosterone , Fetal Blood , Meconium , Saliva , Testosterone , Humans , Meconium/chemistry , Meconium/metabolism , Female , Pregnancy , Infant, Newborn , Adult , Testosterone/analysis , Testosterone/metabolism , Dehydroepiandrosterone/analysis , Dehydroepiandrosterone/metabolism , Saliva/chemistry , Fetal Blood/chemistry , Male , Androgens/analysis
2.
F S Sci ; 5(1): 16-23, 2024 Feb.
Article in English | MEDLINE | ID: mdl-39382269

ABSTRACT

OBJECTIVE: To investigate the direct effect of growth differentiation factor 9 (GDF9) on androgen production in human theca cells. DESIGN: Experimental study. SETTING: Tertiary hospital-based research laboratory. PATIENT(S): Women who underwent in vitro fertilization and intracytoplasmic sperm injections at our clinic were included in this study. INTERVENTION(S): Primary cultured human theca cells from women undergoing in vitro fertilization and intracytoplasmic sperm injection treatment were treated with GDF9, an activin receptor-like kinase 5 (ALK5) inhibitor, and a SMAD4 agonist. MAIN OUTCOME MEASURE(S): The expression of androgen synthesis-related genes StAR, CYP17A1, and LHCGR, levels of androstenedione and testosterone, phosphorylation of SMAD2/3, and the interaction between bone morphogenic protein-activated type II receptor and ALK5 were evaluated using reverse transcription-quantitative polymerase chain reaction, Western blot, enzyme-linked immunosorbent assays, and coimmunoprecipitation assays, respectively. RESULT(S): Growth differentiation factor 9 decreased StAR, CYP17A1, and LHCGR expression levels in human theca cells, which was prevented by treatment with the ALK5 inhibitor, and suppressed production of androgen in human theca cells. Growth differentiation factor 9 increased SMAD2/3 phosphorylation, and the ALK5 inhibitor also suppressed this effect. Bone morphogenic protein-activated type II receptor and ALK5 bound to each other after GDF9 stimulation. The SMAD4 agonist kartogenin also decreased messenger RNA levels of StAR and CYP17A1 and protein levels of StAR in human theca cells. CONCLUSION(S): Growth differentiation factor 9 can activate the bone morphogenic protein-activated type II receptor-ALK5-SMAD2/3 signaling pathway, suppress CYP17A1 expression, and decrease androgen production in human theca cells.


Subject(s)
Growth Differentiation Factor 9 , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta , Smad2 Protein , Smad3 Protein , Steroid 17-alpha-Hydroxylase , Theca Cells , Humans , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Theca Cells/metabolism , Theca Cells/drug effects , Female , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Transforming Growth Factor beta/genetics , Growth Differentiation Factor 9/metabolism , Growth Differentiation Factor 9/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Androgens/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , Smad4 Protein/metabolism , Smad4 Protein/genetics , Phosphorylation/drug effects , Cells, Cultured , Oocytes/metabolism , Oocytes/drug effects , Androstenedione/metabolism , Testosterone/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction/drug effects
3.
Mol Biol Rep ; 51(1): 1047, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39388074

ABSTRACT

BACKGROUND: Furan is an organic compound that occurs as a result of heat treatment during the processing and cooking of many food products. Furthermore, the environment contains furan in tobacco smoke and vehicle exhaust gases, and it serves as an intermediate molecule in the synthesis of various pharmaceutical and chemical agents, pesticides, and stabilizers. Studies on the male reproductive system have not been able to elucidate the pathway through which furan exerts its negative effects. METHODS AND RESULTS: In this study, the TM3 Leydig cell line was exposed to various furan concentrations (0.03, 0.3, and 3 mM) for 24 h. In order to assess the cytotoxic effects of furan on Leydig cells, we examined cell viability, cell proliferation, and lactate dehydrogenase enzyme levels. To investigate the detrimental effects of furan on testosterone biosynthesis, quantitative analyses were conducted on cAMP and testosterone levels, as well as the expression levels of key genes and transcription factors implicated in the steroidogenic pathway. The results indicate that furan inhibited the viability and proliferation of Leydig cells and enhanced the activity of lactate dehydrogenase. Leydig cells administered to furan exhibited notable reductions in cAMP and testosterone levels. Additionally, while the expression levels of steroidogenic genes were downregulated, significant changes were detected in the expression levels of the transcription factors responsible for the regulation of these genes. CONCLUSIONS: Consequently, our findings suggest that furan exerts inhibitory effects on steroidogenesis in Leydig cells through multiple mechanisms, ultimately leading to infertility by inducing dysfunction in Leydig cells.


Subject(s)
Cell Proliferation , Cell Survival , Furans , Leydig Cells , Testosterone , Leydig Cells/metabolism , Leydig Cells/drug effects , Male , Animals , Testosterone/biosynthesis , Testosterone/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Mice , Cell Line , Cyclic AMP/metabolism , L-Lactate Dehydrogenase/metabolism , Steroids/biosynthesis
4.
Front Endocrinol (Lausanne) ; 15: 1418056, 2024.
Article in English | MEDLINE | ID: mdl-39403584

ABSTRACT

Introduction: Mammalian reproductive and somatic development is regulated by steroid hormones, growth hormone (GH), and insulin-like growth factor-1 (IGF-1). Based largely on information from humans, model organisms, and domesticated animals, testosterone (T) and the GH/IGF-1 system activate sexually differentiated development, promoting male-biased growth, often at a cost to health and survivorship. To test if augmented prenatal androgen exposure in females produces similar developmental patterns and trade-offs, we examine maternal effects in wild meerkats (Suricata suricatta), a non-model species in which adult females naturally, albeit differentially by status, express exceptionally high androgen concentrations, particularly during pregnancy. In this cooperative breeder, the early growth of daughters predicts future breeding status and reproductive success. Methods: We examine effects of normative and experimentally induced variation in maternal androgens on the ontogenetic patterns in offspring reproductive hormones (androstenedione, A4; T; estradiol, E2), IGF-1, growth from pup emergence at 1 month to puberty at 1 year, and survivorship. Specifically, we compare the male and female offspring of dominant control (DC or high-T), subordinate control (SC or lower-T), and dominant treated (DT or blocked-T) dams, the latter having experienced antiandrogen treatment in late gestation. Results: Meerkat offspring showed sex differences in absolute T and IGF-1 concentrations, developmental rates of A4 and E2 expression, and survivorship - effects that were sometimes socially or environmentally modulated. Atypical for mammals were the early male bias in T that disappeared by puberty, the absence of sex differences in A4 and E2, and the female bias in IGF-1. Food availability was linked to steroid concentrations in females and to IGF-1, potentially growth, and survival in both sexes. Maternal treatment significantly affected rates of T, E2, and IGF-1 expression, and weight, with marginal effects on survivorship; offspring of DT dams showed peak IGF-1 concentrations and the best survivorship. Discussion: Maternal effects thus impact offspring development in meerkats, with associated trade-offs: Whereas prenatal androgens modify postnatal reproductive and somatic physiology, benefits associated with enhanced competitiveness in DC lineages may have initial costs of reduced IGF-1, delay in weight gain, and decreased survivorship. These novel data further confirm the different evolutionary and mechanistic pathways to cooperative breeding and call for greater consideration of natural endocrine variation in both sexes.


Subject(s)
Androgens , Herpestidae , Animals , Female , Male , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Insulin-Like Growth Factor I/metabolism , Testosterone/metabolism , Estradiol , Reproduction/drug effects , Reproduction/physiology
5.
Commun Biol ; 7(1): 1250, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39363102

ABSTRACT

Friedreich's ataxia (FA) is an autosomal recessive disorder caused by reduced frataxin (FXN) expression in mitochondria, where the lethal component is cardiomyopathy. Using the conditional Fxnflox/null::MCK-Cre knock-out (Fxn-cKO) mouse model, we discovered significant sex differences in the progression towards heart failure, with Fxn-cKO males exhibiting a worse cardiac phenotype, low survival rate, kidney and reproductive organ deficiencies. These differences are likely due to a decline in testosterone in Fxn-cKO males. The decrease in testosterone was related to decreased expression of proteins involved in cholesterol transfer into the mitochondria: StAR and TSPO on the outer mitochondrial membrane, and the cholesterol side-chain cleavage enzyme P450scc and ferredoxin on the inner mitochondrial membrane. Expression of excitation-contraction coupling proteins (L-type calcium channel, RyR2, SERCA2, phospholamban and CaMKIIδ) was decreased significantly more in Fxn-cKO males. This is the first study that extensively investigates the sexual dimorphism in FA mouse model with cardiac calcium signaling impairment.


Subject(s)
Cardiomyopathies , Disease Models, Animal , Frataxin , Friedreich Ataxia , Iron-Binding Proteins , Mice, Knockout , Sex Characteristics , Animals , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/etiology , Male , Female , Mice , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Testosterone/metabolism , Testosterone/blood , Receptors, GABA
6.
Sci Rep ; 14(1): 23931, 2024 10 13.
Article in English | MEDLINE | ID: mdl-39397165

ABSTRACT

Testosterone (T) is a critical predictor of polycystic ovary syndrome (PCOS) but the genetic overlap between T and PCOS has not been established. Here by leveraging genetic datasets from large-scale genome-wide association studies, we assessed the genetic correlation and polygenic overlap between PCOS and three T-related traits using linkage disequilibrium score regression and the bivariate causal mixture model methods. The conjunctional false discovery rate (conjFDR) method was employed to identify shared causal variants. Functional annotation of variants was conducted using FUMA. Total T and bioavailable T exhibited positive correlations with PCOS, while sex hormone-binding globulin (SHBG) showed a negative correlation. All three traits demonstrated extensive genetic overlap with PCOS, with a minimum of 68% of T-related variants influencing PCOS. The conjFDR revealed 4 to 6 causal variants within joint genomic loci shared between PCOS and T-related traits. Functional annotations suggested that these variants might impact PCOS by modulating nearby genes, such as FSHB. Our findings support the hypothesis that PCOS is significantly influenced by androgen abnormalities. Additionally, this study identified several causal variants potentially involved in shared biological mechanisms between PCOS and T regulation.


Subject(s)
Genome-Wide Association Study , Linkage Disequilibrium , Polycystic Ovary Syndrome , Polymorphism, Single Nucleotide , Testosterone , Polycystic Ovary Syndrome/genetics , Humans , Female , Testosterone/metabolism , Genetic Predisposition to Disease , Sex Hormone-Binding Globulin/genetics , Sex Hormone-Binding Globulin/metabolism , Multifactorial Inheritance/genetics
7.
J Comp Neurol ; 532(10): e25675, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39387367

ABSTRACT

Highly sensitive in situ hybridization procedures (RNAScope) were used to quantify the expression of three dopamine receptors (Drd1, Drd2, and Drd3) in two song control nuclei (HVC and the Area X of the basal ganglia) that are known to receive dopaminergic inputs and in the periaqueductal gray (PAG) of male and female canaries. Both sexes were treated with testosterone to ensure they would sing actively. We also determined the excitatory versus inhibitory phenotype of the cells expressing these receptors as well as their activation following a period of song production. The three receptor types were identified in each brain area, with the exception of Drd3 in Area X. The density of cells expressing each receptor varied as a function of receptor type and brain area. Surprisingly few sex differences were detected; they do not seem to explain the sex differences in testosterone-induced song. Overall, the density of Drd-positive cells was much lower in PAG than in the two song control nuclei. In HVC, the majority of cells expressing the three receptor subtypes were VGlut2-positive, whereas colocalization with Vglut2 occurred in few cells in Area X and in an intermediate proportion of cells in PAG. The number of inhibitory cells expressing dopamine receptors was limited. Most dopaminoceptive cells in Area X did not express either excitatory or inhibitory markers. Finally, cellular activation during singing behavior, as measured by the expression of Egr1, was observed in cells expressing each of the three dopamine receptor subtypes, except Drd3 in the PAG.


Subject(s)
Canaries , In Situ Hybridization, Fluorescence , Vocalization, Animal , Animals , Male , Female , Vocalization, Animal/physiology , Vocalization, Animal/drug effects , Canaries/physiology , In Situ Hybridization, Fluorescence/methods , Receptors, Dopamine/metabolism , Sex Characteristics , Testosterone/metabolism , Periaqueductal Gray/metabolism
8.
Sci Rep ; 14(1): 21029, 2024 09 09.
Article in English | MEDLINE | ID: mdl-39251671

ABSTRACT

Benign prostatic hyperplasia (BPH) is a prevalent age-related condition often characterized by debilitating urinary symptoms. Its etiology is believed to stem from hormonal imbalance, particularly an elevated estradiol-to-testosterone ratio and chronic inflammation. Our previous studies using a mouse steroid hormone imbalance model identified a specific increase in macrophages that migrated and accumulated in the prostate lumen where they differentiated into lipid-laden foam cells in mice implanted with testosterone and estradiol pellets, but not in sham animals. The current study focused on further characterizing the cellular heterogeneity of the prostate in this model as well as identifying the specific transcriptomic signature of the recruited foam cells. Moreover, we aimed to identify epithelia-derived signals that drive macrophage infiltration and luminal translocation. Male C57BL/6J mice were implanted with slow-release testosterone and estradiol pellets (T + E2) or sham surgery was performed and the ventral prostates were harvested two weeks later for scRNA-seq analysis. We identified Ear2 + and Cd72 + macrophages that were elevated in response to steroid hormone imbalance, whereas a Mrc1 + resident macrophage population did not change. In addition, an Spp1 + foam cell cluster was almost exclusively found in T + E2 mice. Further markers of foam cells were also identified, including Gpnmb and Trem2, and GPNMB was confirmed as a novel histological marker with immunohistochemistry. Foam cells were also shown to express known pathological factors Vegf, Tgfb1, Ccl6, Cxcl16 and Mmp12. Intriguingly, a screen for chemokines identified the upregulation of epithelia-derived Cxcl17, a known monocyte attractant, in T + E2 prostates suggesting that it might be responsible for the elevated macrophage number as well as their translocation to the lumen. Our study identified macrophage subsets that responded to steroid hormone imbalance as well as further confirmed a potential pathological role of luminal foam cells in the prostate. These results underscore a potential pathological role of the identified prostate foam cells and suggests CXCL17-mediated macrophage migration as a critical initiating event.


Subject(s)
Estradiol , Foam Cells , Macrophages , Mice, Inbred C57BL , Prostate , Testosterone , Animals , Male , Mice , Testosterone/metabolism , Macrophages/metabolism , Prostate/metabolism , Prostate/pathology , Estradiol/pharmacology , Foam Cells/metabolism , Disease Models, Animal , Chemokines, CXC/metabolism , Chemokines, CXC/genetics , Biomarkers/metabolism , Up-Regulation
9.
Sci Rep ; 14(1): 21864, 2024 09 19.
Article in English | MEDLINE | ID: mdl-39300146

ABSTRACT

The decrease in sperm count and infertility is a global issue that remains unresolved. By screening environmental bacterial isolates, we have found that a novel lactic acid bacterium, Lactiplantibacillus plantarum SNI3, increased testis size, testosterone levels, sperm count, sexual activity and fertility in mice that have consumed the bacteria for four weeks. The abundance of L. plantarum in the colon microbiome was positively associated with sperm count. Fecal microbiota transplantation (FMT) from L. plantarum SNI3-dosed mice improved testicular functions in microbiome-attenuated recipient animals. To identify mediators that confer pro-reproductive effects on the host, untargeted in situ mass spectrometry metabolomics was performed on testis samples of L. plantarum SNI3-treated and control mice. Enrichment pathway analysis revealed several perturbed metabolic pathways in the testis of treated mice. Within the testis, a dipeptide, glutamyl-glutamate (GluGlu) was the most upregulated metabolite following L. plantarum SNI3 administration. To validate the pro-reproductive feature of GluGlu, systemic and local injections of the dipeptide have been performed. γ-GluGlu increased sperm count but had no effect on testosterone. These findings highlight the role of γ-GluGlu in mediating spermatogenetic effects of L. plantarum on the male mouse host and -following relevant human clinical trials- may provide future tools for treating certain forms of male infertility.


Subject(s)
Dipeptides , Spermatogenesis , Testis , Animals , Male , Mice , Dipeptides/metabolism , Testis/metabolism , Testis/microbiology , Sperm Count , Fecal Microbiota Transplantation , Testosterone/metabolism , Host Microbial Interactions , Metabolomics/methods , Gastrointestinal Microbiome , Fertility
10.
Int J Mol Sci ; 25(18)2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39337617

ABSTRACT

The differences in muscle development potential between male and female ducks lead to variations in body weight, significantly affecting the growth of the Muscovy duck meat industry. The aim of this study is to explore the regulatory mechanisms for the muscle development differences between genders. Muscovy ducks of both sexes were selected for measurements of body weight, growth traits, hormone levels, and muscle gene expression. The results show that male ducks compared to females had greater weight and growth traits (p < 0.05). Compared to male ducks, the level of serum testosterone in female ducks was decreased, and the estradiol levels were increased (p < 0.05). The RNA-seq analysis identified 102 upregulated and 49 downregulated differentially expressed genes. KEGG analysis revealed that among the top 10 differentially enriched pathways, the AMPK signaling pathway is closely related to muscle growth and development. Additionally, the mRNA and protein levels of CD36, CPT1A, LPL, and SREBP1 were increased and the P-AMPK protein level decreased in the female ducks compared to the male ducks (p < 0.05). In conclusion, muscle development potential difference between male and female ducks is regulated by sex hormones. This process is likely mediated through the activation of the AMPK pathway.


Subject(s)
AMP-Activated Protein Kinases , Ducks , Muscle Development , Signal Transduction , Animals , Ducks/genetics , Ducks/growth & development , Ducks/metabolism , Male , Female , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Muscle Development/genetics , Estradiol/blood , Estradiol/metabolism , Body Weight , Testosterone/blood , Testosterone/metabolism , Sex Characteristics , Muscle, Skeletal/metabolism , Muscle, Skeletal/growth & development , Sex Factors
11.
J Clin Invest ; 134(17)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39225098

ABSTRACT

Testosterone (T) and 17ß-estradiol (E2) are produced in male and female humans and are potent metabolic regulators in both sexes. When E2 and T production stops or decreases during aging, metabolic dysfunction develops and promotes degenerative metabolic and vascular disease. Here, we discuss the shared benefits afforded by E2 and T for metabolic function human females and males. In females, E2 is central to bone and vascular health, subcutaneous adipose tissue distribution, skeletal muscle insulin sensitivity, antiinflammatory immune function, and mitochondrial health. However, T also plays a role in female skeletal, vascular, and metabolic health. In males, T's conversion to E2 is fundamental to bone and vascular health, as well as prevention of excess visceral adiposity and the promotion of insulin sensitivity via activation of the estrogen receptors. However, T and its metabolite dihydrotestosterone also prevent excess visceral adiposity and promote skeletal muscle growth and insulin sensitivity via activation of the androgen receptor. In conclusion, T and E2 are produced in both sexes at sex-specific concentrations and provide similar and potent metabolic benefits. Optimizing levels of both hormones may be beneficial to protect patients from cardiometabolic disease and frailty during aging, which requires further study.


Subject(s)
Estradiol , Testosterone , Humans , Testosterone/metabolism , Male , Female , Estradiol/metabolism , Insulin Resistance , Sex Characteristics , Aging/metabolism , Muscle, Skeletal/metabolism , Animals
12.
Arch Microbiol ; 206(10): 401, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261350

ABSTRACT

Staphylococcus aureus is a notorious pathogen predominantly involved in skin and soft tissue infections, exhibiting a distinct innate sex bias. This study explores the influence of testosterone on the virulence of S. aureus and elucidates its underlying mechanisms. Utilizing a skin abscess model in intact and castrated male mice, we assessed the effects of testosterone on S. aureus pathogenicity. Compared to controls, castrated mice showed significantly reduced abscess sizes and decreased bacterial loads, highlighting the role of testosterone in modulating the severity of S. aureus infections. In vitro experiments revealed that testosterone enhances the hemolytic activity, cytotoxicity, and oxidative stress resistance of S. aureus. Real-time quantitative PCR analysis showed a significant upregulation of the genes encoding α-hemolysin (hla) and phenol-soluble modulin (psmα). Importantly, testosterone treatment significantly enhanced the expression of the accessory gene regulator (Agr) quorum-sensing system components (agrC, agrA, agrB, agrD), while the SaeRS system (saeR, saeS, and sbi) exhibited only slight changes. Gene knockout experiments revealed that deletion of agrC, rather than saeRS and agrBD, abolishes the testosterone-induced enhancement of hemolysis and gene expression, underscoring the key role of AgrC. Molecular docking simulations indicated a direct interaction between testosterone and AgrC protein, with a strong binding affinity at the active site residue SER201. This study provides new insights into the mechanistic basis of how testosterone enhances the pathogenicity of S. aureus, potentially contributing to increased male susceptibility to S. aureus infections and offering a targeted approach for therapeutic interventions.


Subject(s)
Bacterial Proteins , Staphylococcal Infections , Staphylococcus aureus , Testosterone , Male , Testosterone/pharmacology , Testosterone/metabolism , Animals , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Mice , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Virulence , Staphylococcal Infections/microbiology , Trans-Activators/genetics , Trans-Activators/metabolism , Gene Expression Regulation, Bacterial , Quorum Sensing , Molecular Docking Simulation , Bacterial Toxins/metabolism , Bacterial Toxins/genetics , Abscess/microbiology , Hemolysis , Hemolysin Proteins/metabolism , Hemolysin Proteins/genetics
13.
Bull Exp Biol Med ; 177(5): 630-634, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39343843

ABSTRACT

The mechanisms of regulation of the organic anion transporting polypeptide OATP1B3 by sex hormones were studied using HepG2 cells. Estradiol, progesterone, and testosterone were added to cells at concentrations of 1, 10, 100 µM for 24 h. The relative content of OATP1B3 was evaluated by Western blotting. Estradiol at concentrations of 10 and 100 µM increased the level of OATP1B3 acting through the farnesoid X-receptor, testosterone at concentrations of 1, 10, and 100 µM increased the expression of the transporter protein due to its effect on the liver X-receptor subtype α (LXRα), and progesterone did not affect the expression of OATP1B3.


Subject(s)
Estradiol , Progesterone , Solute Carrier Organic Anion Transporter Family Member 1B3 , Testosterone , Humans , Estradiol/metabolism , Estradiol/pharmacology , Progesterone/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Testosterone/metabolism , Hep G2 Cells , Liver X Receptors/metabolism , Liver X Receptors/genetics , Gene Expression Regulation/drug effects
14.
Int J Mol Sci ; 25(18)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39337689

ABSTRACT

This study aims to investigate the effect of a supraphysiological dose of testosterone on the levels of sex steroid hormones and the expression and distribution of sex steroid receptors in the uterus during the endometrial receptivity development period. In this study, adult female Sprague-Dawley rats (n = 24) were subcutaneously administered 1 mg/kg/day of testosterone alone or in combination with the inhibitors (finasteride or anastrozole or both) from day 1 to day 3 post-coitus, while a group of six untreated rats served as a control group. The rats were sacrificed on the evening of post-coital day 4 of to measure sex steroid hormone levels by ELISA. Meanwhile, gene expression and protein distribution of sex steroid receptors were analysed by quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC), respectively. In this study, treatment with a supraphysiological dose of testosterone led to a significant reduction in oestrogen and progesterone levels compared to the control. The mRNA expression of the androgen receptor increased significantly in all treatment groups, while the mRNA expression of both the progesterone receptor and the oestrogen receptor-α decreased significantly in all treatment groups. The IHC findings of all sex steroid receptors were coherent with all mRNAs involved. This study shows that a supraphysiological dose of testosterone was able to interrupt the short period of the implantation window. This finding could serve as a basis for understanding the role of testosterone in endometrial receptivity in order to develop further therapeutic approaches targeting androgen-mediated disorders of endometrial receptivity.


Subject(s)
Endometrium , Rats, Sprague-Dawley , Testosterone , Animals , Female , Testosterone/metabolism , Endometrium/metabolism , Endometrium/drug effects , Rats , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , Embryo Implantation/drug effects , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Receptors, Steroid/metabolism , Receptors, Steroid/genetics , Progesterone/metabolism
15.
PLoS One ; 19(9): e0310036, 2024.
Article in English | MEDLINE | ID: mdl-39255308

ABSTRACT

The main purpose of this longitudinal study was to investigate football players' recovery status, through hormonal response, in relation to accumulated workload at two comparable time points of the first (T1) and second half (T2) of the competitive season. Moreover, this study investigated athletes' hormonal response to a typical weekly conditioning session (5 days before match: MD-5), at T1 and T2, to detect changes in players' recovery capability over time. Salivary cortisol (sC) and testosterone (sT) of 24 professional players (27.8 ± 4.1 years of age) were collected before, after, and 24 hours following MD-5 in two comparable microcycles of T1 and T2. GPS training data (total and high-intensity distance) of the 7 and 28 days before sampling were used to obtain athletes' acute and chronic workloads. Results showed a pre-training significant decrease of sT and an increase of sC (p<0.05) in T2, compared to T1. Moreover, athletes showed high sC and low sT levels before, after and 24 hours following MD-5 in T2. Workload analysis revealed significant correlations of chronic load with sC (r = 0.45, p = 0.056) and T/C ratio (r = -0.59; p = 0.007). These results suggested that, in professional football, chronic workload has a greater impact on players' recovery time than acute workload over the sport season. Moreover, athletes' hormonal response to the weekly conditioning session at T2 revealed an altered anabolic/catabolic balance, highlighting the key role of continuous internal and external workload monitoring during the season.


Subject(s)
Hydrocortisone , Testosterone , Workload , Humans , Longitudinal Studies , Male , Adult , Hydrocortisone/metabolism , Hydrocortisone/analysis , Testosterone/metabolism , Young Adult , Soccer/physiology , Saliva/metabolism , Saliva/chemistry , Athletes , Physical Conditioning, Human/physiology , Physical Conditioning, Human/methods , Athletic Performance/physiology
16.
PLoS One ; 19(9): e0309965, 2024.
Article in English | MEDLINE | ID: mdl-39240904

ABSTRACT

The implementation of biocatalytic steroid hydroxylation processes plays a crucial role in the pharmaceutical industry due to a plethora of medicative effects of hydroxylated steroid derivatives and their crucial role in drug approval processes. Cytochrome P450 monooxygenases (CYP450s) typically constitute the key enzymes catalyzing these reactions, but commonly entail drawbacks such as poor catalytic rates and the dependency on additional redox proteins for electron transfer from NAD(P)H to the active site. Recently, these bottlenecks were overcome by equipping Escherichia coli cells with highly active variants of the self-sufficient single-component CYP450 BM3 together with hydrophobic outer membrane proteins facilitating cellular steroid uptake. The combination of the BM3 variant KSA14m and the outer membrane pore AlkL enabled exceptionally high testosterone hydroxylation rates of up to 45 U gCDW-1 for resting (i.e., living but non-growing) cells. However, a rapid loss of specific activity heavily compromised final product titers and overall space-time yields. In this study, several stabilization strategies were evaluated on enzyme-, cell-, and reaction level. However, neither changes in biocatalyst configuration nor variation of cultivation media, expression systems, or inducer concentrations led to considerable improvement. This qualified the so-far used genetic construct pETM11-ksa14m-alkL, M9 medium, and the resting-cell state as the best options enabling comparatively efficient activity along with fast growth prior to biotransformation. In summary, we report several approaches not enabling a stabilization of the high testosterone hydroxylation rates, providing vital guidance for researchers tackling similar CYP450 stability issues. A comparison with more stable natively steroid-hydroxylating CYP106A2 and CYP154C5 in equivalent setups further highlighted the high potential of the investigated CYP450 BM3-based whole-cell biocatalysts. The immense and continuously developing repertoire of enzyme engineering strategies provides promising options to stabilize the highly active biocatalysts.


Subject(s)
Biocatalysis , Cytochrome P-450 Enzyme System , Escherichia coli , Hydroxylation , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Testosterone/metabolism , Steroids/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , NADPH-Ferrihemoprotein Reductase/metabolism , NADPH-Ferrihemoprotein Reductase/genetics , Enzyme Stability
17.
Biol Res ; 57(1): 64, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252136

ABSTRACT

BACKGROUND: Testicular macrophages (TM) have long been recognized for their role in immune response within the testicular environment. However, their involvement in steroid hormone synthesis, particularly testosterone, has not been fully elucidated. This study aims to explore the capability of TM to synthesize and secrete testosterone de novo and to investigate the regulatory mechanisms involved. RESULTS: Transcriptomic analysis revealed significant expression of Cyp11a1, Cyp17a1, Hsd3b1, and Hsd17b3 in TM, which are key enzymes in the testosterone synthesis pathway. qPCR analysis and immunofluorescence validation confirmed the autonomous capability of TM to synthesize testosterone. Ablation of TM in mice resulted in decreased physiological testosterone levels, underscoring the significance of TM in maintaining testicular testosterone levels. Additionally, the study also demonstrated that Cebpb regulates the expression of these crucial genes, thereby modulating testosterone synthesis. CONCLUSIONS: This research establishes that TM possess the autonomous capacity to synthesize and secrete testosterone, contributing significantly to testicular testosterone levels. The transcription factor Cebpb plays a crucial role in this process by regulating the expression of key genes involved in testosterone synthesis.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta , Macrophages , Testis , Testosterone , Animals , Male , Testosterone/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Testis/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Gene Expression Profiling
18.
Reproduction ; 168(4)2024 10 01.
Article in English | MEDLINE | ID: mdl-39102452

ABSTRACT

In brief: Male reproductive problems under psychological stress were widely studied. Using chronically unpredictable mild stress-treated mice, we found that reduced serum testosterone levels were related to the low level of cholesterol in the Leydig cells. Abstract: Testosterone deficiency in humans can be caused by depressive symptoms; however, the causes of this deficiency are incompletely understood. This study demonstrates that male mice with depression-like symptoms due to chronic unpredictable mild stress (CUMS) show reduced serum testosterone levels and disrupted sexual behaviors. However, the observed testosterone reductions were not caused by apoptosis of Leydig cells. Oil red O staining revealed that lipid droplets were dramatically decreased in Leydig cells, suggesting that defects in cholesterol uptake might be related to testosterone deficiency in depression-like mice. To investigate the potential mechanism, lipid homeostasis was examined by liquid chromatography-tandem mass spectrometry. The results revealed that higher levels of sphingomyelins (SM 8:0;2O/28:1, 18:0;2O/22:2, 33:0;3O, 33:1;2O) were linked to decreased cholesterol levels. Further investigation indicated that testosterone biosynthesis from cholesterol in Leydig cells was impaired by the downregulation of Ldlr, Srb1, Lhr, and P450scc. Elevated levels of interferon signaling-associated pathways in depression-like mice testes may also contribute to decreased testosterone levels. Taken together, these findings provide a novel understanding of male reproductive problems under psychological stress and suggest that cholesterol uptake might be a causal factor in reduced testosterone production in depression-like mice.


Subject(s)
Cholesterol , Depression , Leydig Cells , Stress, Psychological , Testosterone , Animals , Male , Leydig Cells/metabolism , Testosterone/blood , Testosterone/metabolism , Mice , Cholesterol/metabolism , Cholesterol/blood , Depression/metabolism , Depression/etiology , Stress, Psychological/metabolism , Sexual Behavior, Animal , Mice, Inbred C57BL
19.
Horm Behav ; 165: 105611, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089160

ABSTRACT

During breeding when testosterone concentrations are high, male songbirds that are open-ended vocal learners like canaries (Serinus canaria) tend to produce a stable, stereotyped song that facilitates mate attraction or territory defense. Outside breeding contexts, song becomes more variable. The neuroendocrine mechanisms controlling this vocal variability across seasons are not entirely clear. We tested whether androgen signaling within the lateral magnocellular nucleus of the anterior nidopallium (LMAN), a cortical-like brain region of the vocal control system known as a vocal variability generator, plays a role in seasonal vocal variability. We first characterized song in birds housed alone on a short day (SD) photoperiod, which simulates non-breeding conditions. Then, cannulae filled with the androgen receptor (AR) blocker flutamide or left empty as control were implanted bilaterally in LMAN. Birds were then transferred to long days (LD) to simulate the breeding season and song was analyzed again. Blocking AR in LMAN increased acoustic variability of song and the acoustic variability of syllables. However, blocking AR in LMAN did not impact the variability of syllable usage nor their sequencing in LD birds, song features that are controlled by androgen signaling in a somatosensory brain region of the vocal control system called HVC. These findings highlight the multifactorial, non-redundant actions of steroid hormones in controlling complex social behaviors such as birdsong. They also support the hypothesis that LMAN is a key brain area for the effects of testosterone on song plasticity both seasonally in adults and during the song crystallization process at sexual maturity.


Subject(s)
Androgens , Canaries , Vocalization, Animal , Animals , Male , Vocalization, Animal/physiology , Vocalization, Animal/drug effects , Canaries/physiology , Androgens/pharmacology , Stereotyped Behavior/drug effects , Stereotyped Behavior/physiology , Receptors, Androgen/metabolism , Receptors, Androgen/physiology , Flutamide/pharmacology , Photoperiod , Seasons , Signal Transduction/physiology , Signal Transduction/drug effects , Testosterone/metabolism , Testosterone/pharmacology , Androgen Antagonists/pharmacology
20.
Int J Mol Sci ; 25(16)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39201373

ABSTRACT

Low or insufficient testosterone levels caused by caponization promote fat deposition in animals. However, the molecular mechanism of fat deposition in caponized animals remains unclear. This study aimed to investigate the metabolomics and transcriptomic profiles of adipose tissues and study the effect of testosterone and leptin on the proliferation of adipocytes. We observed a significant enlargement in the areas of adipocytes in the abdominal fat tissues in capon, as well as increased luciferase activity of the serum leptin and a sharp decrease in the serum testosterone in caponized gander. Metabolomics and transcriptomic results revealed differentially expressed genes and differentially expressed metabolites with enhanced PARR signal pathway. The mRNA levels of peroxisome proliferators-activated receptor γ, fatty acid synthase, and suppressor of cytokine signaling 3 in goose primary pre-adipocytes were significantly upregulated with high leptin treatment and decreased significantly with increasing testosterone dose. Hence, reduced testosterone and increased leptin levels after caponization possibly promoted adipocytes proliferation and abdominal fat deposition by altering the expression of PPAR pathway related genes in caponized ganders. This study provides a new direction for the mechanism through which testosterone regulates the biological function of leptin and fat deposition in male animals.


Subject(s)
Adipogenesis , Leptin , Signal Transduction , Testosterone , Animals , Leptin/metabolism , Testosterone/pharmacology , Testosterone/metabolism , Adipogenesis/drug effects , Male , Signal Transduction/drug effects , Adipocytes/metabolism , Adipocytes/drug effects , Geese/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Peroxisome Proliferator-Activated Receptors/genetics , Cell Proliferation/drug effects , Adipose Tissue/metabolism , Adipose Tissue/drug effects , PPAR gamma/metabolism , PPAR gamma/genetics , Orchiectomy
SELECTION OF CITATIONS
SEARCH DETAIL