Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biology (Basel) ; 11(9)2022 Aug 24.
Article in English | MEDLINE | ID: mdl-36138736

ABSTRACT

Obesity and type II diabetes mellitus (T2DM) are prominent risk factors for secondary osteoporosis due to the negative impacts of hyperglycemia and excessive body fat on bone metabolism. While the armamentarium of anti-diabetic drugs is expanding, their negative or unknown impacts on bone metabolism limits effectiveness. The inactivation of inositol hexakisphosphate kinase 1 (IP6K1) protects mice from high-fat-diet (HFD)-induced obesity (DIO) and insulin resistance by enhancing thermogenic energy expenditure, but the role of this kinase and the consequences of its inhibition on bone metabolism are unknown. To determine if IP6K1 inhibition in obese mice affords protection against obesity-induced metabolic derangements and bone loss, we maintained 2-month-old mice on a normal chow control diet or HFD under thermal neutral conditions for 100 d. Beginning on day 40, HFD-fed mice were divided into two groups and administered daily injections of vehicle or the pan-IP6K inhibitor TNP [N2-(m-Trifluorobenzyl), N6-(p-nitrobenzyl) purine]. HFD-fed mice developed obesity, hyperglycemia, hyperlipidemia, and secondary osteoporosis, while TNP administration protected mice against HFD-induced metabolic and lipid derangements and preserved bone mass, mineral density, and trabecular microarchitecture, which correlated with reduced serum leptin levels, reduced marrow adiposity, and preservation of marrow resident skeletal stem/progenitor cells (SSPCs). TNP also exhibited hypotensive activity, an unrealized benefit of the drug, and its prolonged administration had no adverse impacts on spermatogenesis. Together, these data indicate that the inhibition of IP6K1 using selective inhibitors, such as TNP, may provide an effective strategy to manage obesity and T2DM due to its bone sparing effects.

2.
Am J Pathol ; 191(9): 1592-1609, 2021 09.
Article in English | MEDLINE | ID: mdl-34126085

ABSTRACT

Male factors, including those of autoimmune origin, contribute to approximately 50% of infertility cases in humans. However, the mechanisms underlying autoimmune male infertility are poorly understood. Deficiency in autoimmune regulator (AIRE) impairs central immune tolerance because of diminished expression of self-antigens in the thymus. Humans with AIRE mutations and mice with engineered ablation of Aire develop multiorgan autoimmunity and infertility. To determine the immune targets contributing to infertility in male Aire-deficient (-/-) mice, Aire-/- or wild-type (WT) males were paired with WT females. Aire-/- males exhibited dramatically reduced mating frequency and fertility, hypogonadism, and reduced serum testosterone. Approximately 15% of mice exhibited lymphocytic infiltration into the testis, accompanied by atrophy, azoospermia, and reduced numbers of mitotically active germ cells; the remaining mice showed normal testicular morphology, sperm counts, and motility. However, spermatozoa from all Aire-/- mice were defective in their ability to fertilize WT oocytes in vitro. Lymphocytic infiltration into the epididymis, seminal vesicle, and prostate gland was evident. Aire-/- male mice generated autoreactive antibodies in an age-dependent manner against sperm, testis, epididymis, prostate gland, and seminal vesicle. Finally, expression of Aire was evident in the seminiferous epithelium in an age-dependent manner, as well as in the prostate gland. These findings suggest that Aire-dependent central tolerance plays a critical role in maintaining male fertility by stemming autoimmunity against multiple reproductive targets.


Subject(s)
Infertility, Male/immunology , Polyendocrinopathies, Autoimmune/pathology , Transcription Factors/metabolism , Animals , Female , Infertility, Male/genetics , Male , Mice , Mice, Knockout , Polyendocrinopathies, Autoimmune/genetics , Transcription Factors/genetics , AIRE Protein
3.
Physiol Genomics ; 52(2): 81-95, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31841397

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that suppresses gene expression. Previously, we developed a conditional null model where EZH2 is knocked out in uterus. Deletion of uterine EZH2 increased proliferation of luminal and glandular epithelial cells. Herein, we used RNA-Seq in wild-type (WT) and EZH2 conditional knockout (Ezh2cKO) uteri to obtain mechanistic insights into the gene expression changes that underpin the pathogenesis observed in these mice. Ovariectomized adult Ezh2cKO mice were treated with vehicle (V) or 17ß-estradiol (E2; 1 ng/g). Uteri were collected at postnatal day (PND) 75 for RNA-Seq or immunostaining for epithelial proliferation. Weighted gene coexpression network analysis was used to link uterine gene expression patterns and epithelial proliferation. In V-treated mice, 88 transcripts were differentially expressed (DEG) in Ezh2cKO mice, and Bmp5, Crabp2, Lgr5, and Sprr2f were upregulated. E2 treatment resulted in 40 DEG with Krt5, Krt15, Olig3, Crabp1, and Serpinb7 upregulated in Ezh2cKO compared with control mice. Transcript analysis relative to proliferation rates revealed two module eigengenes correlated with epithelial proliferation in WT V vs. Ezh2cKO V and WT E2 vs. Ezh2cKO E2 mice, with a positive relationship in the former and inverse in the latter. Notably, the ESR1, Wnt, and Hippo signaling pathways were among those functionally enriched in Ezh2cKO females. Current results reveal unique gene expression patterns in Ezh2cKO uterus and provide insight into how loss of this critical epigenetic regulator assumingly contributes to uterine abnormalities.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Transcriptome , Uterus/metabolism , Animals , Cell Proliferation , Cluster Analysis , Computational Biology , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Estradiol/pharmacology , Estrogens/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation , Genotype , Heterozygote , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , RNA-Seq , Signal Transduction , Up-Regulation , Uterus/abnormalities , Wnt Proteins/metabolism
4.
Biol Reprod ; 101(2): 306-317, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31201420

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is a rate-limiting catalytic subunit of a histone methyltransferase, polycomb repressive complex, which silences gene activity through the repressive histone mark H3K27me3. EZH2 is critical for epigenetic effects of early estrogen treatment, and may be involved in uterine development and pathologies. We investigated EZH2 expression, regulation, and its role in uterine development/function. Uterine epithelial EZH2 expression was associated with proliferation and was high neonatally then declined by weaning. Pre-weaning uterine EZH2 expression was comparable in wild-type and estrogen receptor 1 knockout mice, showing neonatal EZH2 expression is ESR1 independent. Epithelial EZH2 was upregulated by 17ß-estradiol (E2) and inhibited by progesterone in adult uteri from ovariectomized mice. To investigate the uterine role of EZH2, we developed a EZH2 conditional knockout (Ezh2cKO) mouse using a cre recombinase driven by the progesterone receptor (Pgr) promoter that produced Ezh2cKO mice lacking EZH2 in Pgr-expressing tissues (e.g. uterus, mammary glands). In Ezh2cKO uteri, EZH2 was deleted neonatally. These uteri had reduced H3K27me3, were larger than WT, and showed adult cystic endometrial hyperplasia. Ovary-independent uterine epithelial proliferation and increased numbers of highly proliferative uterine glands were seen in adult Ezh2cKO mice. Female Ezh2cKO mice were initially subfertile, and then became infertile by 9 months. Mammary gland development in Ezh2cKO mice was inhibited. In summary, uterine EZH2 expression is developmentally and hormonally regulated, and its loss causes aberrant uterine epithelial proliferation, uterine hypertrophy, and cystic endometrial hyperplasia, indicating a critical role in uterine development and function.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Uterus/enzymology , Uterus/growth & development , Animals , Enhancer of Zeste Homolog 2 Protein/genetics , Epithelial Cells/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Female , Histones/metabolism , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Pregnancy , Progesterone/metabolism
5.
Biol Reprod ; 101(2): 392-404, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31141131

ABSTRACT

Both membrane and nuclear fractions of estrogen receptor 1 (ESR1) mediate 17ß-estradiol (E2) actions. Mice expressing nuclear (n)ESR1 but lacking membrane (m)ESR1 (nuclear-only estrogen receptor 1 [NOER] mice) show reduced E2 responsivity and reproductive abnormalities culminating in adult male and female infertility. Using this model, we investigated whether reproductive pathologies caused by the synthetic estrogen diethylstilbestrol (DES) are mitigated by mESR1 ablation. Homozygous and heterozygous wild-type (WT and HET, respectively) and NOER male and female mice were subcutaneously injected with DES (1 mg/kg body weight [BW]) or vehicle daily from postnatal day (PND) 1-5. Uterine histology was assessed in select DES-treated females at PND 5, whereas others were ovariectomized at PND 60 and treated with E2 (10 µg/kg BW) or vehicle 2 weeks later. Neonatal DES exposure resulted in ovary-independent epithelial proliferation in the vagina and uterus of WT but not NOER females. Neonatal DES treatment also induced ovary-independent adult expression of classical E2-induced transcripts (e.g., lactoferrin [Ltf] and enhancer of zeste homolog 2 [Ezh2]) in WT but not NOER mice. At PND 90, DES-treated WT and HET males showed smaller testes and a high incidence of bacterial pyogranulomatous inflammation encompassing the testes, epididymis and occasionally the ductus deferens with spread to lumbar lymph nodes; such changes were largely absent in NOER males. Results indicate that male and female NOER mice are protected from deleterious effects of neonatal DES, and thus mESR1 signaling is required for adult manifestation of DES-induced reproductive pathologies in both sexes.


Subject(s)
Diethylstilbestrol/toxicity , Estrogen Receptor alpha/genetics , Estrogens, Non-Steroidal/toxicity , Prenatal Exposure Delayed Effects , Animals , Female , Gene Expression Regulation/drug effects , Genital Diseases, Male/chemically induced , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Uterus/metabolism
6.
Physiol Rev ; 97(3): 995-1043, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28539434

ABSTRACT

Estrogens have historically been associated with female reproduction, but work over the last two decades established that estrogens and their main nuclear receptors (ESR1 and ESR2) and G protein-coupled estrogen receptor (GPER) also regulate male reproductive and nonreproductive organs. 17ß-Estradiol (E2) is measureable in blood of men and males of other species, but in rete testis fluids, E2 reaches concentrations normally found only in females and in some species nanomolar concentrations of estrone sulfate are found in semen. Aromatase, which converts androgens to estrogens, is expressed in Leydig cells, seminiferous epithelium, and other male organs. Early studies showed E2 binding in numerous male tissues, and ESR1 and ESR2 each show unique distributions and actions in males. Exogenous estrogen treatment produced male reproductive pathologies in laboratory animals and men, especially during development, and studies with transgenic mice with compromised estrogen signaling demonstrated an E2 role in normal male physiology. Efferent ductules and epididymal functions are dependent on estrogen signaling through ESR1, whose loss impaired ion transport and water reabsorption, resulting in abnormal sperm. Loss of ESR1 or aromatase also produces effects on nonreproductive targets such as brain, adipose, skeletal muscle, bone, cardiovascular, and immune tissues. Expression of GPER is extensive in male tracts, suggesting a possible role for E2 signaling through this receptor in male reproduction. Recent evidence also indicates that membrane ESR1 has critical roles in male reproduction. Thus estrogens are important physiological regulators in males, and future studies may reveal additional roles for estrogen signaling in various target tissues.


Subject(s)
Estrogens/metabolism , Genitalia, Male/metabolism , Receptors, Estrogen/metabolism , Reproduction , Animals , Aromatase/genetics , Aromatase/metabolism , Genitalia, Male/pathology , Genitalia, Male/physiopathology , Genotype , Humans , Male , Mice, Knockout , Mutation , Phenotype , Prostate/metabolism , Prostate/pathology , Prostate/physiopathology , Prostatic Diseases/metabolism , Prostatic Diseases/pathology , Prostatic Diseases/physiopathology , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , Signal Transduction
7.
J Endocrinol Reprod ; 21(1): 1-14, 2017 Jun.
Article in English | MEDLINE | ID: mdl-34321782

ABSTRACT

Steroid hormones, acting through their cognate nuclear receptors, are critical for many reproductive and non-reproductive functions. Over the past two decades, it has become increasingly clear that in addition to cytoplasmic/nuclear steroid receptors that alter gene transcription when liganded, a small fraction of cellular steroid receptors are localized to the cell membranes, where they mediate rapid steroid hormone effects. 17ß-Estradiol (E2), a key steroid hormone for both male and female reproduction, acts predominately through its main receptor, estrogen receptor 1 (ESR1). Most ESR1 is nuclear; however, 5-10% of ESR1 is localized to the cell membrane after being palmitoylated at cysteine 451 in mice. This review discusses reproductive phenotypes of a newly-developed mouse model with a C451A point mutation that precludes membrane targeting of ESR1. This transgenic mouse, termed the nuclear-only ESR1 (NOER) mouse, shows extensive male and female reproductive abnormalities and infertility despite normally functional nuclear ESR1 (nESR1). These results provide the first in vivo evidence that membrane-initiated E2/ESR1 signaling is required for normal male and female reproductive functions and fertility. Signaling mechanisms for membrane ESR1 (mESR1), as well as how mESR1 works with nESR1 to mediate estrogen effects, are still being established. We discuss some possible mechanisms by which mESR1 might facilitate nESR1 signaling, as well as the emerging evidence that mESR1 might be a major mediator of epigenetic effects of estrogens, which are potentially linked to various adult-onset pathologies.

8.
Asian J Androl ; 19(1): 3-4, 2017.
Article in English | MEDLINE | ID: mdl-27633909

Subject(s)
Meiosis , Humans
9.
Endocrinology ; 157(7): 2909-19, 2016 07.
Article in English | MEDLINE | ID: mdl-27145009

ABSTRACT

Estrogen receptor 1 (ESR1) mediates major reproductive functions of 17ß-estradiol (E2). Male Esr1 knockout (Esr1KO) mice are infertile due to efferent ductule and epididymal abnormalities. The majority of ESR1 is nuclear/cytoplasmic; however, a small fraction is palmitoylated at cysteine 451 in mice and localized to cell membranes, in which it mediates rapid E2 actions. This study used an Esr1 knock-in mouse containing an altered palmitoylation site (C451A) in ESR1 that prevented cell membrane localization, although nuclear ESR1 was expressed. These nuclear-only estrogen receptor 1 (NOER) mice were used to determine the roles of membrane ESR1 in males. Epididymal sperm motility was reduced 85% in 8-month-old NOER mice compared with wild-type controls. The NOER mice had decreased epididymal sperm viability and greater than 95% of sperm had abnormalities, including coiled midpieces and tails, absent heads, and folded tails; this was comparable to 4-month Esr1KO males. At 8 months, daily sperm production in NOER males was reduced 62% compared with controls. The NOER mice had histological changes in the rete testes, efferent ductules, and seminiferous tubules that were comparable with those previously observed in Esr1KO males. Serum T was increased in NOER males, but FSH, LH, and E2 were unchanged. Critically, NOER males were initially subfertile, becoming infertile with advancing age. These findings identify a previously unknown role for membrane ESR1 in the development of normal sperm and providing an adequate environment for spermatogenesis.


Subject(s)
Estrogen Receptor alpha/metabolism , Infertility, Male/metabolism , Reproduction/physiology , Sperm Motility/genetics , Spermatogenesis/physiology , Animals , Epididymis/metabolism , Estradiol/blood , Estrogen Receptor alpha/genetics , Follicle Stimulating Hormone/blood , Infertility, Male/genetics , Luteinizing Hormone/blood , Male , Mice , Mice, Knockout , Testosterone/blood
10.
Biol Reprod ; 92(5): 122, 2015 May.
Article in English | MEDLINE | ID: mdl-25882702

ABSTRACT

Progesterone (P4) and the synthetic glucocorticoid dexamethasone (Dex) inhibit luminal epithelial (LE) proliferation in neonatal mouse uteri. This study determined the roles of progesterone receptor and estrogen receptor 1 (PR and ESR1, respectively) in P4- and Dex-induced inhibition of LE proliferation using PR knockout (PRKO) and Esr1 knockout (Esr1KO) mice. Wild-type (WT), heterozygous, and homozygous PRKO female pups were injected with vehicle, P4 (40 µg/g body weight), or Dex (4 or 40 µg/g body weight) on Postnatal Day 5, then 24 h later immunostained for markers of cell proliferation. In WT and heterozygous mice, P4 sharply reduced LE proliferation, and Dex produced dose-responsive decreases equaling those of P4 at the higher dose. Critically, although both doses of Dex similarly decreased proliferation compared to vehicle-treated PRKOs, treatment of PRKO pups with the high Dex dose (40 µg/g) did not inhibit LE as much as treatments of WT mice with this Dex dose or with P4. Stromal proliferation was stimulated by P4 in WT but not PRKO mice, and Dex did not alter stromal proliferation. Uteri of all genotypes strongly expressed glucocorticoid receptor (GR), demonstrating that impaired Dex effects in PRKOs did not reflect GR deficiency. Furthermore, inhibition of LE proliferation by Dex (40 µg/g body weight) in Esr1KO mice was normal, so this process does not involve ESR1. In summary, inhibitory Dex effects on LE proliferation occur partially through non-PR-mediated mechanisms, presumably GR, as indicated by Dex inhibition of LE proliferation in PRKOs. However, maximal inhibitory Dex effects on uterine LE proliferation are not seen in PRKO mice with even high Dex, indicating that maximal Dex effects in WT mice also involve PR.


Subject(s)
Dexamethasone/pharmacology , Epithelial Cells/drug effects , Progesterone/pharmacology , Receptors, Progesterone/metabolism , Uterus/physiology , Animals , Animals, Newborn , Epithelial Cells/physiology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation , Mice , Mice, Knockout , Receptors, Progesterone/genetics , Uterus/cytology , Uterus/drug effects
11.
Asian J Androl ; 17(3): 441-2, 2015.
Article in English | MEDLINE | ID: mdl-25791734

ABSTRACT

How the primordial germ cell (PGC) lineage, which eventually gives rise to spermatozoa in males and oocytes in females, is established in the developing mammalian embryo has been a critical topic in both developmental and reproductive biology for many years. There have been significant breakthroughs over the past two decades in establishing both the source of PGCs and the factors that regulate the specification of this lineage in mice, [1] but our understanding of the factors that control PGC development in the human is rudimentary. The SRY-related HMG-box (SOX) family of transcription factors consists of 20 genes in humans and mice that are involved in the maintenance of pluripotency, male sexual development, and other processes. A recent paper in Cell has identified one member of this family, SOX17, as an essential factor for inducing the PGC lineage in humans. [2] Surprisingly, this protein does not appear to have a role in PGC specification in mice. This work not only introduces a new and important player to the field of germ cell specification, but also emphasizes the uniqueness of human PGC development compared to more extensively studied mouse models.


Subject(s)
Cell Differentiation , Germ Cells/cytology , SOXF Transcription Factors/metabolism , Animals , Humans
12.
Biol Reprod ; 92(3): 78, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25653281

ABSTRACT

Neonatal uterus and vagina express estrogen receptor 1 (ESR1) and respond mitogenically to exogenous estrogens. However, neonatal ovariectomy does not inhibit preweaning uterine cell proliferation, indicating that this process is estrogen independent. Extensive literature suggests that ESR1 can be activated by growth factors in a ligand-independent manner and drive uterine cell proliferation. Alternatively, neonatal uterine cell proliferation could be ESR1 independent despite its obligatory role in adult luminal epithelial proliferation. To determine ESR1's role in uterine and vaginal development, we analyzed cell proliferation, apoptosis, and uterine gland development (adenogenesis) in wild-type (WT) and Esr1 knockout (Esr1KO) mice from Postnatal Day 2 to Postnatal Day 60. Uterine and vaginal cell proliferation, apoptosis, and uterine adenogenesis were comparable in WT and Esr1KO mice before weaning. By Days 29-60, glands had regressed, and uterine cell proliferation was reduced in Esr1KO mice in contrast to continued adenogenesis and proliferation in WT. Apoptosis in Esr1KO uterine epithelium was not increased compared to WT at any age, indicating that differences in cell proliferation, rather than apoptosis, cause divergence of uterine size in these two groups at puberty. Similarly, vaginal epithelial proliferation was reduced, and the epithelium became atrophic in Esr1KO mice by 29 days of age and later in Esr1KO mice. These results indicate that preweaning uterine and vaginal development is ESR1 independent but becomes dependent on ESR1 by Day 29 on. It is not yet clear what mechanisms drive preweaning vaginal and uterine development, but ligand-independent activation of ESR1 is not involved.


Subject(s)
Animals, Newborn/physiology , Cell Proliferation/physiology , Estrogen Receptor alpha/physiology , Uterus/cytology , Uterus/growth & development , Vagina/cytology , Vagina/growth & development , Animals , Apoptosis/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Female , Genotype , Heterozygote , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Progesterone/physiology , Sexual Maturation/genetics , Sexual Maturation/physiology , Time Factors , Uterus/physiology , Vagina/physiology
13.
Asian J Androl ; 17(3): 355-9, 2015.
Article in English | MEDLINE | ID: mdl-25677134

ABSTRACT

There have been significant breakthroughs over the past decade in the development and use of pluripotent stem cells as a potential source of cells for applications in regenerative medicine. It is likely that this methodology will begin to play an important role in human clinical medicine in the years to come. This review describes the plasticity of one type of pluripotent cell, spermatogonial stem cells (SSCs), and their potential therapeutic applications in regenerative medicine and male infertility. Normally, SSCs give rise to sperm when in the testis. However, both human and murine SSCs can give rise to cells with embryonic stem (ES) cell-like characteristics that can be directed to differentiate into tissues of all three embryonic germ layers when placed in an appropriate inductive microenvironment, which is in contrast to other postnatal stem cells. Previous studies have reported that SSCs expressed an intermediate pluripotent phenotype before differentiating into a specific cell type and that extended culture was necessary for this to occur. However, recent studies from our group using a tissue recombination model demonstrated that SSCs differentiated rapidly into another tissue, in this case, prostatic epithelium, without expression of pluripotent ES cell markers before differentiation. These results suggest that SSCs are capable of directly differentiating into other cell types without going through an intermediate ES cell-like stage. Because SSCs do not require reprogramming to achieve a pluripotent state, they are an attractive source of pluripotent cells for use in regenerative medicine.


Subject(s)
Adult Stem Cells/physiology , Cell Plasticity/physiology , Pluripotent Stem Cells/physiology , Adult Stem Cells/cytology , Animals , Cell Differentiation/physiology , Humans , Infertility, Male/therapy , Male , Mice , Pluripotent Stem Cells/cytology , Regenerative Medicine/trends
14.
Toxicol Lett ; 225(3): 479-87, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24472609

ABSTRACT

There is concern that early-life exposure to bisphenol A (BPA) may alter developmental programming and predispose individuals to obesity and reproductive anomalies. The present study was designed to determine if a high fat diet at sexual maturation moderates testicular toxicity occasioned by exposure to BPA during reproductive development. Therefore, male rats were exposed to BPA by maternal gavage (0, 2.5 or 25 µg/kg body weight/day) from gestational day 12 to postnatal day 21. At weaning, control and BPA-exposed animals were placed on a regular normal fat diet (NFD) until 70 days of age when they were continued on the NFD or were maintained on a high fat diet (HFD) until euthanasia at 98 days. Adult male rats maintained on HFD were generally heavier than NFD animals due to greater energy intake but energy intake per unit body weight gain was similar in all animals. However, perinatal exposure to BPA decreased (P<0.05) serum adiponectin as well as adiponectin and AdipoR2 protein expression levels in Leydig cells. Importantly, the combination of BPA exposure and HFD consumption promoted lipid peroxidation evidenced by elevated serum thiobarbituric acid reactive substances and glutathione concentrations. These findings imply that interaction between BPA and HFD potentially causes testicular dysfunction to a greater degree than would be due to BPA exposure or HFD consumption. Given the relationship that exists between energy homeostasis and reproductive activity, additional studies are warranted to investigate the consequences of BPA-diet interactions on testicular function.


Subject(s)
Benzhydryl Compounds/toxicity , Diet, High-Fat/adverse effects , Estrogens, Non-Steroidal/toxicity , Phenols/toxicity , Prenatal Exposure Delayed Effects , Sexual Maturation/drug effects , Testis/drug effects , Adiponectin/blood , Animals , Blotting, Western , Body Weight/physiology , Estradiol/blood , Female , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Pregnancy , Rats , Rats, Long-Evans , Receptors, Adiponectin/blood , Sexual Maturation/physiology , Testis/cytology , Testis/metabolism , Testosterone/blood
15.
Food Chem Toxicol ; 65: 252-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24373827

ABSTRACT

The present study investigated if Aflatoxin B1 (AFB1), a potent and naturally occurring mycotoxin, interferes with the steroidogenic pathway in rat Leydig cells. Testicular Leydig cells are the predominant source of the male sex steroid hormone testosterone (T) that maintains the male phenotype and support fertility. Leydig cells, isolated from 35-day-old male Long-Evans rats (Rattus norvegicus), were incubated with AFB1 at 0, 0.01, 0.1, 1, 10µM followed by measurement of T secretion by radioimmunoassay and analysis of protein expression in western blots. Results demonstrated that AFB1 suppressed testosterone secretion in a dose-dependent manner and inhibited expression of cholesterol transporter steroidogenic acute regulatory protein (StAR) and steroidogenic enzymes [(3ß-hydroxysteroid dehydrogenase (3ß-HSD) and 17ß-hydroxysteroid dehydrogenase enzyme (HSD17B3)]. Protein expression analysis showed that AFB1 treatment increased ERK phosphorylation but suppressed p38 MAPK and JNK activation in Leydig cells. AFB1-induced inhibition of Leydig cells was alleviated by co-treatment with the ERK inhibitor UO 126, implying that ERK mediates, at least in part, the inhibitory effects of AFB1 in Leydig cells. The findings highlight potential extra-hepatic effects of aflatoxin exposure and indicate that exposure to AFB1 has significant reproductive health implications for consumers of contaminated products even under conditions of low dietary toxin levels.


Subject(s)
Aflatoxin B1/toxicity , Androgens/biosynthesis , Leydig Cells/drug effects , Animals , Cells, Cultured , Leydig Cells/metabolism , Male , Radioimmunoassay , Rats , Rats, Long-Evans
16.
Front Neurosci ; 7: 108, 2013.
Article in English | MEDLINE | ID: mdl-23781171

ABSTRACT

Traumatic brain injury (TBI) is an important and costly medical problem for which no clinically proven treatment currently exists. Studies in rodents and humans have shown beneficial effects of progesterone (P4) on both mortality and functional outcomes following TBI. Neuroprotective effects of P4 in TBI likely involve the classical nuclear progesterone receptors (Pgr) that are widely distributed in both glial cells and neurons of the brain. However, P4 may have critical effects not mediated through Pgr. In the brain, P4 is converted to a metabolite, allopregnanolone (ALLO), whose beneficial effects equal or exceed those of P4 in TBI. ALLO does not bind Pgr, suggesting it acts through non-classical pathways. ALLO has effects on GABAA and pregnane X receptors, as well as on the mitochondrial permeability transition pore. In addition, ALLO is metabolized to another compound, 5alpha-dihydroprogesterone, which binds Pgr, suggesting ALLO actions may involve signaling through Pgr as well as the aforementioned mechanisms of action. P4 and ALLO also signal through a number of membrane receptors (progesterone receptor membrane component 1, and membrane progesterone receptors (mPRs) alpha, beta, gamma, delta, and epsilon) in the brain that are distinct from Pgr, although the role of these receptors in the normal brain and in the therapeutic response to P4 and ALLO following TBI is unclear. In summary, P4 has the potential to become the first clinically effective treatment for TBI, and the effects of P4 and its metabolite ALLO in TBI may involve Pgr, mPRs, and other signaling pathways. Elucidating these mechanisms will more clearly reveal the potential of classical and non-classical pathways to mediate important effects of P4 and its metabolites, and potentially offer new therapeutic approaches to TBI.

17.
Biol Reprod ; 86(5): 135, 1-12, 2012 May.
Article in English | MEDLINE | ID: mdl-22302688

ABSTRACT

The presence of bisphenol A (BPA) in consumer products has raised concerns about potential adverse effects on reproductive health. Testicular Leydig cells are the predominant source of the male sex steroid hormone testosterone, which supports the male phenotype. The present report describes the effects of developmental exposure of male rats to BPA by gavage of pregnant and lactating Long-Evans dams at 2.5 and 25 µg/kg body weight from Gestational Day 12 to Day 21 postpartum. This exposure paradigm stimulated Leydig cell division in the prepubertal period and increased Leydig cell numbers in the testes of adult male rats at 90 days. Observations from in vitro experiments confirmed that BPA acts directly as a mitogen in Leydig cells. However, BPA-induced proliferative activity in vivo is possibly mediated by several factors, such as 1) protein kinases (e.g., mitogen-activated protein kinases or MAPK), 2) growth factor receptors (e.g., insulin-like growth factor 1 receptor-beta and epidermal growth factor receptors), and 3) the Sertoli cell-secreted anti-Mullerian hormone (also called Mullerian inhibiting substance). On the other hand, BPA suppressed protein expression of the luteinizing hormone receptor (LHCGR) and the 17beta-hydroxysteroid dehydrogenase enzyme (HSD17B3), thereby decreasing androgen secretion by Leydig cells. We interpret these findings to mean that the likely impact of deficits in androgen secretion on serum androgen levels following developmental exposure to BPA is alleviated by increased Leydig cell numbers. Nevertheless, the present results reinforce the view that BPA causes biological effects at environmentally relevant exposure levels and its presence in consumer products potentially has implication for public health.


Subject(s)
Cell Proliferation/drug effects , Estrogens, Non-Steroidal/toxicity , Leydig Cells/drug effects , Phenols/toxicity , 17-Hydroxysteroid Dehydrogenases/biosynthesis , Androgens/biosynthesis , Androgens/blood , Animals , Benzhydryl Compounds , Female , Leydig Cells/metabolism , Male , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Long-Evans , Receptors, LH/biosynthesis , Steroids/biosynthesis
18.
Toxicol Lett ; 209(1): 78-85, 2012 Feb 25.
Article in English | MEDLINE | ID: mdl-22155228

ABSTRACT

Testicular Leydig cells are the predominant source of the male sex steroid hormone testosterone (T), which is required to maintain male fertility. There is now growing evidence that environmental stressors, including chemicals present in food, air and water, may affect energy balance. A relationship between energy balance and reproductive capacity has been proposed for a long time. In the present study, developmental exposures of male rats to soy isoflavones in the maternal diet from gestational day 12 to day 21 post-partum enhanced adiponectin expression in adipose tissue and increased serum adiponectin concentrations in adulthood. However, exposure to soy isoflavones caused a decrease in T production and expression of adiponectin and its receptor (adipoR2) in Leydig cells. In separate experiments, incubation of Leydig cells with recombinant adiponectin in the absence of isoflavones caused a decrease in T biosynthesis associated with diminished expression of the cholesterol transporter steroidogenic acute regulatory protein (StAR). Thus, chemical-induced alterations in serum adiponectin concentrations have implication for steroid hormone secretion. The results also imply that changes in adipose tissue metabolism occasioned by exposure to dietary estrogens, and perhaps other estrogenic agents, possibly contribute to deficiencies in reproductive capacity attributed to these compounds.


Subject(s)
Adiponectin/metabolism , Endocrine Glands/drug effects , Endocrine Glands/metabolism , Glycine max/chemistry , Isoflavones/pharmacology , Testis/drug effects , Testis/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Androgens/biosynthesis , Animals , Blood Glucose/metabolism , Blotting, Western , Body Weight/drug effects , Cell Cycle Proteins/biosynthesis , Cell Separation , Estradiol/blood , In Vitro Techniques , Insulin/blood , Leptin/metabolism , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Rats , Rats, Long-Evans , Receptors, Estrogen/biosynthesis , Testosterone/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...