Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.822
Filter
1.
Mol Med Rep ; 30(6)2024 Dec.
Article in English | MEDLINE | ID: mdl-39370806

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease whose etiology is not fully understood. Defective peripheral immune tolerance and subsequent mis­differentiation and aberrant infiltration of synovium by various immune cells, especially helper T (Th) cells, play an important role in the development of RA. There are significant sex differences in RA, but the results of studies on the effects of sex hormones on RA have been difficult to standardize and hormone replacement therapy has been limited by the potential for serious side effects. Existing research has amply demonstrated that cellular immune responses are largely determined by sex and that sex hormones play a key role in Th cell responses. Based on the aforementioned background and the plasticity of Th cells, it is reasonable to hypothesize that the action of sex hormones on Th cells will hopefully become a therapeutic target for RA. The present review discussed the role of various Th cell subsets in the pathogenesis of RA and also explored the role of sex hormones on the phenotype and function of these aberrantly regulated immune cells in RA as well as other pathologic effects on RA.


Subject(s)
Arthritis, Rheumatoid , Gonadal Steroid Hormones , T-Lymphocytes, Helper-Inducer , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Humans , Gonadal Steroid Hormones/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Female , Male
2.
Sci Rep ; 14(1): 22946, 2024 10 03.
Article in English | MEDLINE | ID: mdl-39362969

ABSTRACT

Myopia is a global public health concern, with a higher prevalence in women than that in male. As the relationship between sex hormone and myopia remains unclear, we aimed to reveal the relationship between sex hormone and myopia by removing the ovaries of mice and measuring changes in ocular parameters related to myopia. Lens-induced myopia (LIM) surgeries were performed on 3-week-old male mice and age-matched female mice to evaluate the effects of sex on myopia development, which indicated a high degree of myopia and rapid progression of axial elongation in male mice. Bilateral ovariectomy (OVX) performed on 4-week-old female mice induced myopic refraction status in ovariectomized mice. Although axial length elongation was larger in ovariectomized mice than that in sham control mice, the result was insignificant. To further reveal the relationship between female sex hormones and myopia, LIM combined with OVX was performed, which revealed a magnified myopic refraction status in ovariectomized mice. Nevertheless, elongation of the anterior chamber depth in the - 30 D lens-treated eyes significantly increased in LIM + OVX mice compared with the frame-treated eyes in LIM + OVX mice and - 30 D lens-treated eyes in LIM + Sham surgery mice. Sex hormones play a role in regulating myopia development in female mice.


Subject(s)
Myopia , Ovariectomy , Animals , Female , Myopia/etiology , Myopia/metabolism , Ovariectomy/adverse effects , Mice , Male , Disease Models, Animal , Refraction, Ocular/physiology , Gonadal Steroid Hormones/metabolism
3.
Sci Rep ; 14(1): 23891, 2024 10 12.
Article in English | MEDLINE | ID: mdl-39396092

ABSTRACT

The roles of sex hormones such as estradiol, testosterone, and sex hormone-binding globulin (SHBG) in the etiology of lung and colorectal cancers in women, among the most common cancers after breast cancer, are unclear. This Mendelian randomization (MR) study evaluated such potential causal associations in women of European ancestry. We used summary statistics data from genome-wide association studies on sex hormones and from the Trøndelag Health Study (HUNT) and large consortia on cancers. There was suggestive evidence of 1-standard deviation increase in total testosterone levels being associated with a lower risk of lung non-adenocarcinoma (hazard ratio 0.60, 95% confidence interval 0.37-0.98) in the HUNT Study. However, this was not confirmed by using data from a larger consortium. In general, we did not find convincing evidence to support a causal role of sex hormones on risk of lung and colorectal cancers in women of European ancestry.


Subject(s)
Colorectal Neoplasms , Genome-Wide Association Study , Gonadal Steroid Hormones , Lung Neoplasms , Mendelian Randomization Analysis , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/epidemiology , Female , Lung Neoplasms/genetics , Lung Neoplasms/epidemiology , Gonadal Steroid Hormones/metabolism , Sex Hormone-Binding Globulin/genetics , Sex Hormone-Binding Globulin/metabolism , Risk Factors , Testosterone/blood , Polymorphism, Single Nucleotide , Middle Aged , White People/genetics
4.
J Clin Invest ; 134(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39286970

ABSTRACT

Autoimmune diseases are a leading cause of disability worldwide. Most autoimmune diseases occur more often in women than men, with rheumatic autoimmune diseases being among those most highly expressed in women. Several key factors, identified mainly in animal models and cell culture experiments, are important in increasing autoimmune disease in females. These include sex hormones, immune genes including those found on the X chromosome, sex-specific epigenetic effects on genes by estrogen and the environment, and regulation of genes and messenger RNA by microRNAs found in extracellular vesicles. Evidence is also emerging that viruses as well as drugs or toxins that damage mitochondria may contribute to increased levels of autoantibodies against nuclear and mitochondrial antigens, which are common in many autoimmune diseases. The purpose of this Review is to summarize our current understanding of mechanisms that may determine sex differences in autoimmune disease.


Subject(s)
Autoimmune Diseases , Autoimmunity , Sex Characteristics , Humans , Female , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics , Animals , Male , Epigenesis, Genetic , Gonadal Steroid Hormones/immunology , Gonadal Steroid Hormones/metabolism , Autoantibodies/immunology , MicroRNAs/genetics , MicroRNAs/immunology , MicroRNAs/metabolism , Chromosomes, Human, X/genetics , Chromosomes, Human, X/immunology , Mitochondria/immunology , Mitochondria/metabolism , Mitochondria/genetics
5.
Skin Res Technol ; 30(10): e70086, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39349354

ABSTRACT

OBJECTIVE: This study aims to examine the expression of androgen receptor (AR) and estrogen receptor (ER) in patients with classic Kaposi's sarcoma (CKS) in Xinjiang, as well as to assess the serum levels of sex hormones in these patients. The objective is to explore potential new directions and targets for diagnosing and treating CKS in Xinjiang. METHODS: The case group comprised 35 patients diagnosed with CKS who presented at our hospital from 2014 to 2021. The control group consisted of 35 patients with pyogenic granuloma (PG) who visited the hospital during the same period, selected using propensity score matching (PSM). Immunohistochemistry was used to detect AR, human herpesvirus type 8 (HHV-8), and ER in paraffin-embedded tissue samples from patients diagnosed with CKS and PG. Additionally, enzyme-linked immunosorbent assay (ELISA) was used to quantitatively measure serum sex hormone levels in the 35 patients with CKS and 35 patients with PG. RESULTS: AR expression was relatively weak in both the CKS and PG groups, with the PG group exhibiting a slightly stronger expression than the CKS group. Conversely, the expression of ER was significantly higher in the CKS group compared to the PG group (p < 0.05). Additionally, serum testosterone (T) levels were elevated in the CKS group, while serum estradiol (E2) levels were higher in the PG group (p < 0.05). CONCLUSION: Sex hormones and their receptors are implicated in the pathogenesis of CKS in Xinjiang. The use of ER antagonists may represent a novel avenue for research and treatment of CKS.


Subject(s)
Receptors, Androgen , Receptors, Estrogen , Sarcoma, Kaposi , Humans , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/blood , Sarcoma, Kaposi/ethnology , Male , Receptors, Androgen/metabolism , Female , Middle Aged , China , Receptors, Estrogen/metabolism , Aged , Gonadal Steroid Hormones/blood , Gonadal Steroid Hormones/metabolism , Adult , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/blood , Herpesvirus 8, Human
6.
Nutrients ; 16(17)2024 Sep 08.
Article in English | MEDLINE | ID: mdl-39275346

ABSTRACT

Heat stress due to climate warming can significantly affect the synthesis of sex hormones in male adolescents, which can impair the ability of the hypothalamus to secrete gonadotropin-releasing hormone on the hypothalamic-pituitary-gonadal axis, which leads to a decrease in luteinizing hormone and follicle-stimulating hormone, which ultimately negatively affects spermatogenesis and testosterone synthesis. For optimal spermatogenesis, the testicular temperature should be 2-6 °C lower than body temperature. Heat stress directly affects the testes, damaging them and reducing testosterone synthesis. Additionally, chronic heat stress abnormally increases the level of aromatase in Leydig cells, which increases estradiol synthesis while decreasing testosterone, leading to an imbalance of sex hormones and spermatogenesis failure. Low levels of testosterone in male adolescents lead to delayed puberty and incomplete sexual maturation, negatively affect height growth and bone mineral density, and can lead to a decrease in lean body mass and an increase in fat mass. In order for male adolescents to acquire healthy reproductive capacity, it is recommended to provide sufficient nutrition and energy, avoid exposure to heat stress, and provide foods and supplements to prevent or repair testosterone reduction, germ cell damage, and sperm count reduction caused by heat stress so that they can enter a healthy adulthood.


Subject(s)
Gonadal Steroid Hormones , Heat-Shock Response , Reproduction , Male , Adolescent , Humans , Reproduction/physiology , Gonadal Steroid Hormones/metabolism , Heat-Shock Response/physiology , Testosterone/blood , Spermatogenesis , Testis/growth & development , Sexual Maturation/physiology
7.
Commun Biol ; 7(1): 1154, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39284885

ABSTRACT

Fish sex is largely influenced by steroid hormones, especially sex hormones. Here, we established a steroid hormone-free genetic model by mutation of cyp11a1 in Nile tilapia, which was confirmed by EIA assay. Gonadal phenotype and transcriptome analyses showed that the XX mutants displayed sex reversal from female to male but with defective spermatogenesis. Despite the sex reversal, the aromatase encoding gene cyp19a1a was continuously expressed in the gonads of the XX mutants, which might be caused by androgen deficiency. Whole-mount fluorescence in situ hybridization and transcriptome analysis showed that the gonads of the XX mutants firstly developed towards ovary but shifted to testis between 10 to 15 days after hatching. Detailed expression analysis of key sex differentiation pathway genes foxl3 and dmrt1 combined with apoptosis analysis revealed transdifferentiation of germ cells from female to male during sex reversal. Rescue experiments showed that both P5 and E2 treatment rescued the sex reversal of cyp11a1 mutant XX fish. Overall, our results revealed a transient ovary-like stage and transdifferentiation of germ cells from female to male in the early gonads of the steroid hormone-deprived cyp11a1 mutant XX fish.


Subject(s)
Mutation , Ovary , Sex Differentiation , Animals , Female , Ovary/metabolism , Male , Sex Differentiation/genetics , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cichlids/genetics , Cichlids/metabolism , Disorders of Sex Development/genetics , Fish Proteins/genetics , Fish Proteins/metabolism , Gonadal Steroid Hormones/metabolism , Tilapia/genetics , Tilapia/metabolism , Sex Determination Processes/genetics
9.
Arterioscler Thromb Vasc Biol ; 44(10): 2169-2190, 2024 10.
Article in English | MEDLINE | ID: mdl-39145392

ABSTRACT

Pulmonary hypertension is a rare, incurable, and progressive disease. Although there is increasing evidence that immune disorders, particularly those associated with connective tissue diseases, are a strong predisposing factor in the development of pulmonary arterial hypertension (PAH), there is currently a lack of knowledge about the detailed molecular mechanisms responsible for this phenomenon. Exploring this topic is crucial because patients with an immune disorder combined with PAH have a worse prognosis and higher mortality compared with patients with other PAH subtypes. Moreover, data recorded worldwide show that the prevalence of PAH in women is 2× to even 4× higher than in men, and the ratio of PAH associated with autoimmune diseases is even higher (9:1). Sexual dimorphism in the pathogenesis of cardiovascular disease was explained for many years by the action of female sex hormones. However, there are increasing reports of interactions between sex hormones and sex chromosomes, and differences in the pathogenesis of cardiovascular disease may be controlled not only by sex hormones but also by sex chromosome pathways that are not dependent on the gonads. This review discusses the role of estrogen and genetic factors including the role of genes located on the X chromosome, as well as the potential protective role of the Y chromosome in sexual dimorphism, which is prominent in the occurrence of PAH associated with autoimmune diseases. Moreover, an overview of animal models that could potentially play a role in further investigating the aforementioned link was also reviewed.


Subject(s)
Autoimmune Diseases , Pulmonary Arterial Hypertension , Humans , Autoimmune Diseases/genetics , Autoimmune Diseases/epidemiology , Autoimmune Diseases/immunology , Animals , Female , Male , Sex Factors , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/epidemiology , Pulmonary Arterial Hypertension/physiopathology , Sex Characteristics , Risk Factors , Genetic Predisposition to Disease , Chromosomes, Human, X/genetics , Gonadal Steroid Hormones/metabolism , Pulmonary Artery/physiopathology , Pulmonary Artery/metabolism , Chromosomes, Human, Y/genetics , Health Status Disparities
10.
Int J Biol Macromol ; 278(Pt 3): 134855, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39168222

ABSTRACT

Leptin is an important hormone in mammals, which plays a key role in regulating reproduction and energy metabolism. However, there are few studies on the function of leptin in reproductive regulation in fish, especially on tongue sole (Cynoglossus semilaevis). Thus, in this study, we firstly exploited the basic function of tongue sole leptins, the migration and growth rate of ovarian cells were reduced after knocking down lepA and lepB in ovarian cells, while increasing the apoptosis rate. Then both rlepA and rlepB were proved to be combined with lepR to further exert functions by dual luciferase assay. Transcriptome sequencing showed that differentially expressed genes (DEGs) were mainly enriched in KEGG pathways related to membrane receptors, fatty acid synthesis, growth, etc. when lepA and lepB were knocked down or additionally added in vitro. Additionally, the estradiol (E2) hormone was increased significantly after knocking down lepB. Finally, based on DEGs and the signaling pathways they participated in, we proposed a hypothesis about the signaling pathways in which leptin may be involved in ovarian cells. Taken together, these results provide new insights into the role of leptin in the regulation of physiological functions such as ovarian growth and development.


Subject(s)
Apoptosis , Cell Movement , Cell Proliferation , Flatfishes , Leptin , Ovary , Animals , Female , Ovary/metabolism , Leptin/metabolism , Leptin/genetics , Flatfishes/metabolism , Flatfishes/genetics , Cell Movement/drug effects , Signal Transduction , Fish Proteins/metabolism , Fish Proteins/genetics , Gonadal Steroid Hormones/metabolism , Gonadal Steroid Hormones/biosynthesis , Estradiol/metabolism , Receptors, Leptin/metabolism , Receptors, Leptin/genetics
12.
Poult Sci ; 103(10): 104090, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39096826

ABSTRACT

The size of the initial primordial follicle pool in the ovary depends on primordial follicle formation, which determines the female reproductive lifespan. However, the molecular regulation of primordial follicle formation in chickens remains unclear. In this study, the left ovaries of chickens were collected at 2 d posthatch (dph), 5.5 dph, and 10.5 dph to examine the formation of primordial follicles. Single-cell mRNA sequencing (scRNA-seq) and spatial transcriptomic analysis were performed to explore the ovarian microenvironment and identify regulatory pathways involved in the formation of primordial follicles in chickens. Histomorphological analysis of chicken ovary tissues revealed the presence of germ cell cysts at 1 dph, which began to disintegrate at 2 dph. Primordial follicles appeared at 5.5 dph and continued to develop into larger-diameter follicles. scRNA-seq and spatial transcriptomic analysis revealed 24 cellular clusters involved in chicken primordial follicle formation. The metabolic pathway of steroid hormone synthesis was found in pregranulosa and pretheca cells. Histological analysis showed that chicken ovaries did not form primordial follicles after the inhibition of the steroid hormone synthesis pathway by simvastatin or tamoxifen. In addition, mRNA transcriptomic and bioinformatics analyses revealed that GREB1 was a downstream gene of the steroid hormone synthesis pathway during the formation of chicken primordial follicles. This study provides a valuable foundation for investigating primordial follicle formation in avian species and optimizing their reproductive performance.


Subject(s)
Chickens , Ovarian Follicle , Single-Cell Analysis , Animals , Chickens/genetics , Chickens/growth & development , Chickens/physiology , Ovarian Follicle/physiology , Female , Sequence Analysis, RNA/veterinary , Transcriptome , Gonadal Steroid Hormones/metabolism
13.
Nat Commun ; 15(1): 7279, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39179555

ABSTRACT

Determining sex-bias in brain structure is of great societal interest to improve diagnostics and treatment of brain-related disorders. So far, studies on sex-bias in brain structure predominantly focus on macro-scale measures, and often ignore factors determining this bias. Here we study sex-bias in cortical and hippocampal microstructure in relation to sex hormones. Investigating quantitative intracortical profiling in-vivo using the T1w/T2w ratio in 1093 healthy females and males of the cross-sectional Human Connectome Project young adult sample, we find that regional cortical and hippocampal microstructure differs between males and females and that the effect size of this sex-bias varies depending on self-reported hormonal status in females. Microstructural sex-bias and expression of sex hormone genes, based on an independent post-mortem sample, are spatially coupled. Lastly, sex-bias is most pronounced in paralimbic areas, with low laminar complexity, which are predicted to be most plastic based on their cytoarchitectural properties. Albeit correlative, our study underscores the importance of incorporating sex hormone variables into the investigation of brain structure and plasticity.


Subject(s)
Cerebral Cortex , Connectome , Gonadal Steroid Hormones , Hippocampus , Humans , Female , Hippocampus/metabolism , Hippocampus/diagnostic imaging , Male , Young Adult , Adult , Gonadal Steroid Hormones/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/diagnostic imaging , Sex Characteristics , Magnetic Resonance Imaging/methods , Cross-Sectional Studies
14.
J Physiol ; 602(17): 4129-4156, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39106346

ABSTRACT

Sex as a biological variable is an underappreciated aspect of biomedical research, with its importance emerging in more recent years. This review assesses the current understanding of sex differences in human physical performance. Males outperform females in many physical capacities because they are faster, stronger and more powerful, particularly after male puberty. This review highlights key sex differences in physiological and anatomical systems (generally conferred via sex steroids and puberty) that contribute to these sex differences in human physical performance. Specifically, we address the effects of the primary sex steroids that affect human physical development, discuss insight gained from an observational study of 'real-world data' and elite athletes, and highlight the key physiological mechanisms that contribute to sex differences in several aspects of physical performance. Physiological mechanisms discussed include those for the varying magnitude of the sex differences in performance involving: (1) absolute muscular strength and power; (2) fatigability of limb muscles as a measure of relative performance; and (3) maximal aerobic power and endurance. The profound sex-based differences in human performance involving strength, power, speed and endurance, and that are largely attributable to the direct and indirect effects of sex-steroid hormones, sex chromosomes and epigenetics, provide a scientific rationale and framework for policy decisions on sex-based categories in sports during puberty and adulthood. Finally, we highlight the sex bias and problem in human performance research of insufficient studies and information on females across many areas of biology and physiology, creating knowledge gaps and opportunities for high-impact studies.


Subject(s)
Sex Characteristics , Humans , Female , Male , Gonadal Steroid Hormones/metabolism , Physical Functional Performance , Athletic Performance/physiology
15.
BMC Endocr Disord ; 24(1): 135, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090659

ABSTRACT

BACKGROUND: Prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is higher in men than in women. Hormonal and genetic causes may account for the sex differences in MASLD. Current human in vitro liver models do not sufficiently take the influence of biological sex and sex hormones into consideration. METHODS: Primary human hepatocytes (PHHs) were isolated from liver specimen of female and male donors and cultured with sex hormones (17ß-estradiol, testosterone and progesterone) for up to 72 h. mRNA expression levels of 8 hepatic lipid metabolism genes were analyzed by RT-qPCR. Sex hormones and their metabolites were determined in cell culture supernatants by LC-MS analyses. RESULTS: A sex-specific expression was observed for LDLR (low density lipoprotein receptor) with higher mRNA levels in male than female PHHs. All three sex hormones were metabolized by PHHs and the effects of hormones on gene expression levels varied depending on hepatocyte sex. Only in female PHHs, 17ß-estradiol treatment affected expression levels of PPARA (peroxisome proliferator-activated receptor alpha), LIPC (hepatic lipase) and APOL2 (apolipoprotein L2). Further changes in mRNA levels of female PHHs were observed for ABCA1 (ATP-binding cassette, sub-family A, member 1) after testosterone and for ABCA1, APOA5 (apolipoprotein A-V) and PPARA after progesterone treatment. Only the male PHHs showed changing mRNA levels for LDLR after 17ß-estradiol and for APOA5 after testosterone treatment. CONCLUSIONS: Male and female PHHs showed differences in their expression levels of hepatic lipid metabolism genes and their responsiveness towards sex hormones. Thus, cellular sex should be considered, especially when investigating the pathophysiological mechanisms of MASLD.


Subject(s)
Gonadal Steroid Hormones , Hepatocytes , Lipid Metabolism , Humans , Male , Female , Hepatocytes/metabolism , Hepatocytes/drug effects , Lipid Metabolism/genetics , Lipid Metabolism/drug effects , Gonadal Steroid Hormones/pharmacology , Gonadal Steroid Hormones/metabolism , Cells, Cultured , Middle Aged , Testosterone/pharmacology , Testosterone/metabolism , Estradiol/pharmacology , Adult , Progesterone/pharmacology , Progesterone/metabolism , Sex Factors
16.
Front Endocrinol (Lausanne) ; 15: 1458422, 2024.
Article in English | MEDLINE | ID: mdl-39188914

ABSTRACT

Despite the lack of endogenous synthesis and relevant nuclear receptors, several papers have been published over the decades claiming that the physiology of mollusks is affected by natural and synthetic sex steroids. With scant evidence for the existence of functional steroid nuclear receptors in mollusks, some scientists have speculated that the effects of steroids might be mediated via membrane receptors (i.e. via non-genomic/non-classical actions) - a mechanism that has been well-characterized in vertebrates. However, no study has yet investigated the ligand-binding ability of such receptor candidates in mollusks. The aim of the present study was to further trace the evolution of the endocrine system by investigating the presence of functional membrane sex steroid receptors in a mollusk, the great pond snail (Lymnaea stagnalis). We detected sequences homologous to the known vertebrate membrane sex steroid receptors in the Lymnaea transcriptome and genome data: G protein-coupled estrogen receptor-1 (GPER1); membrane progestin receptors (mPRs); G protein-coupled receptor family C group 6 member A (GPRC6A); and Zrt- and Irt-like protein 9 (ZIP9). Sequence analyses, including conserved domain analysis, phylogenetics, and transmembrane domain prediction, indicated that the mPR and ZIP9 candidates appeared to be homologs, while the GPER1 and GPRC6A candidates seemed to be non-orthologous receptors. All candidates transiently transfected into HEK293MSR cells were found to be localized at the plasma membrane, confirming that they function as membrane receptors. However, the signaling assays revealed that none of the candidates interacted with the main vertebrate steroid ligands. Our findings strongly suggest that functional membrane sex steroid receptors which would be homologous to the vertebrate ones are not present in Lymnaea. Although further experiments are required on other molluscan model species as well, we propose that both classical and non-classical sex steroid signaling for endocrine responses are specific to chordates, confirming that molluscan and vertebrate endocrine systems are fundamentally different.


Subject(s)
Nervous System , Animals , Nervous System/metabolism , Receptors, Steroid/metabolism , Receptors, Steroid/genetics , Lymnaea/metabolism , Lymnaea/physiology , Mollusca/metabolism , Endocrine System/metabolism , Phylogeny , Receptors, Estrogen/metabolism , Humans , Receptors, Progesterone/metabolism , Gonadal Steroid Hormones/metabolism
17.
Front Endocrinol (Lausanne) ; 15: 1400640, 2024.
Article in English | MEDLINE | ID: mdl-39109076

ABSTRACT

Sex steroid hormones (SSH) are extremely versatile molecules with a myriad of physiological functions. Next to their well-known role in sexual development and reproduction, SSH play active roles in practically every tissue in the human body, including the oral cavity. It has long been demonstrated that periodontal tissues express SSH receptors and therefore are responsive to the presence of SSH. Interestingly, SSH not only interact with the periodontal tissues but also with other tissues in the oral cavity such as dental enamel, pulp, cementum, oral mucosa, and salivary glands. Questions concerning the possible physiological functions of these receptors and their role in maintenance of oral health, remain unanswered. The purpose of this scoping review was to gather and summarize all the available evidence on the role of SSH in physiological processes in the oral cavity in humans. Two comprehensive literature searches were performed. References were screened and selected based on title, abstract and full text according to our inclusion criteria. Both searches yielded 18,992 results of which 73 were included. Results were divided into four categories: (1) Periodontium; (2) Dental structure; (3) Mucosa; and (4) Salivary glands. The interaction of these tissues with progestagens, androgens and estrogens are summarized. Sex steroid hormones are an overlooked yet fundamental factor in oral homeostasis. They play important roles in the development and function of the periodontium, dental structure, mucosa and salivary glands. Dentists and healthcare providers should consider these hormonal factors when assessing and treating oral health conditions.


Subject(s)
Gonadal Steroid Hormones , Homeostasis , Humans , Gonadal Steroid Hormones/metabolism , Homeostasis/physiology , Mouth/metabolism , Periodontium/metabolism , Oral Health
18.
Proc Natl Acad Sci U S A ; 121(36): e2404042121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39207735

ABSTRACT

The course of normal development and response to pathology are strongly influenced by biological sex. For instance, female childhood cancer survivors who have undergone cranial radiation therapy (CRT) tend to display more pronounced cognitive deficits than their male counterparts. Sex effects can be the result of sex chromosome complement (XX vs. XY) and/or gonadal hormone influence. The contributions of each can be separated using the four-core genotype mouse model (FCG), where sex chromosome complement and gonadal sex are decoupled. While studies of FCG mice have evaluated brain differences in adulthood, it is still unclear how sex chromosome and sex hormone effects emerge through development in both healthy and pathological contexts. Our study utilizes longitudinal MRI with the FCG model to investigate sex effects in healthy development and after CRT in wildtype and immune-modified Ccl2-knockout mice. Our findings in normally developing mice reveal a relatively prominent chromosome effect prepubertally, compared to sex hormone effects which largely emerge later. Spatially, sex chromosome and hormone influences were independent of one another. After CRT in Ccl2-knockout mice, both male chromosomes and male hormones similarly improved brain outcomes but did so more separately than in combination. Our findings highlight the crucial role of sex chromosomes in early development and identify roles for sex chromosomes and hormones after CRT-induced inflammation, highlighting the influences of biological sex in both normal brain development and pathology.


Subject(s)
Brain , Cranial Irradiation , Mice, Knockout , Sex Chromosomes , Animals , Male , Female , Sex Chromosomes/genetics , Brain/metabolism , Brain/radiation effects , Brain/growth & development , Mice , Cranial Irradiation/adverse effects , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Gonadal Steroid Hormones/metabolism , Magnetic Resonance Imaging
19.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125611

ABSTRACT

Sexual dimorphism among mammals includes variations in the pain threshold. These differences are influenced by hormonal fluctuations in females during the estrous and menstrual cycles of rodents and humans, respectively. These physiological conditions display various phases, including proestrus and diestrus in rodents and follicular and luteal phases in humans, distinctly characterized by varying estrogen levels. In this study, we evaluated the capsaicin responses in male and female mice at different estrous cycle phases, using two murine acute pain models. Our findings indicate that the capsaicin-induced pain threshold was lower in the proestrus phase than in the other three phases in both pain assays. We also found that male mice exhibited a higher pain threshold than females in the proestrus phase, although it was similar to females in the other cycle phases. We also assessed the mRNA and protein levels of TRPV1 in the dorsal root and trigeminal ganglia of mice. Our results showed higher TRPV1 protein levels during proestrus compared to diestrus and male mice. Unexpectedly, we observed that the diestrus phase was associated with higher TRPV1 mRNA levels than those in both proestrus and male mice. These results underscore the hormonal influence on TRPV1 expression regulation and highlight the role of sex steroids in capsaicin-induced pain.


Subject(s)
Capsaicin , Pain , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Capsaicin/pharmacology , Male , Female , Mice , Pain/metabolism , Pain/genetics , Gonadal Steroid Hormones/metabolism , Estrous Cycle/drug effects , Pain Threshold/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/drug effects , Gene Expression Regulation/drug effects , Sex Characteristics , RNA, Messenger/metabolism , RNA, Messenger/genetics
20.
Psychoneuroendocrinology ; 169: 107161, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39116520

ABSTRACT

Difficulties in fear regulation can sometimes result in maladaptive fear responses. To better understand how to improve fear regulation, it is important to determine how known factors, such as sex hormone status and stress, might interact to influence fear memory. Research has shown that women with high estradiol levels (mid-cycle) and men exhibit better extinction retention compared to women with low estradiol levels (women in the early follicular cycle or using oral contraceptives). Stress has also been demonstrated to affect both the learning and retention of extinction. Despite documented interactions between stress and sex hormones, their combined effects have not been thoroughly studied. This study aims to examine the impact of stress as a function of sex hormone status on extinction learning and retention. A total of 168 non-clinical participants were studied, including men (n = 46), women using oral contraceptives (n = 38), women in the early follicular phase (n = 40), and women in mid-cycle (n = 44). On Day 1, fear acquisition training was performed. On day 2, prior to extinction training, half of the participants were exposed to a psychosocial stressor, while the other half performed a non-stressful control task. On day 3, extinction retention was tested. Fear was quantified using skin conductance responses, while stress hormones were quantified through saliva samples. Exposure to stress prior to extinction training did not affect extinction learning, regardless of sex hormone status. In contrast, pre-extinction stress exposure had different effects on extinction retention depending on hormone status. Stressed men showed impairment in extinction retention compared to controls, while the experimental condition had no effect on naturally cycling women. Regardless of stress exposure, early follicular women exhibited a deficit in fear regulation, while mid-cycle women showed effective fear regulation. Among women using oral contraceptives, the stress group demonstrated better extinction retention compared to the control group. These results demonstrate the importance of considering sex hormone status and stress exposure during extinction learning, as both components may modulate extinction retention. These results could help identifying hormonal conditions that may enhance the effectiveness of extinction-based psychological therapies used in the treatment of fear-related disorders.


Subject(s)
Extinction, Psychological , Fear , Stress, Psychological , Humans , Extinction, Psychological/physiology , Female , Male , Adult , Fear/physiology , Fear/psychology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Young Adult , Estradiol/metabolism , Estradiol/blood , Estradiol/analysis , Sex Characteristics , Gonadal Steroid Hormones/metabolism , Gonadal Steroid Hormones/analysis , Sex Factors , Adolescent , Saliva/chemistry , Saliva/metabolism , Follicular Phase/physiology , Follicular Phase/psychology , Menstrual Cycle/physiology , Menstrual Cycle/psychology , Conditioning, Classical/physiology
SELECTION OF CITATIONS
SEARCH DETAIL