Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 35(3): e21397, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33565176

RESUMEN

Sperm develop from puberty in the seminiferous tubules, inside the blood-testis barrier to prevent their recognition as "non-self" by the immune system, and it is widely assumed that human sperm-specific proteins cannot access the circulatory or immune systems. Sperm-specific proteins aberrantly expressed in cancer, known as cancer-testis antigens (CTAs), are often pursued as cancer biomarkers and therapeutic targets based on the assumption they are neoantigens absent from the circulation in healthy men. Here, we identify a wide range of germ cell-derived and sperm-specific proteins, including multiple CTAs, that are selectively deposited by the Sertoli cells of the adult mouse and human seminiferous tubules into testicular interstitial fluid (TIF) that is "outside" the blood-testis barrier. From TIF, the proteins can access the circulatory- and immune systems. Disruption of spermatogenesis decreases the abundance of these proteins in mouse TIF, and a sperm-specific CTA is significantly decreased in TIF from infertile men, suggesting that exposure of certain CTAs to the immune system could depend on fertility status. The results provide a rationale for the development of blood-based tests useful in the management of male infertility and indicate CTA candidates for cancer immunotherapy and biomarker development that could show sex-specific and male-fertility-related responses.


Asunto(s)
Antígenos de Neoplasias/análisis , Proteínas/análisis , Túbulos Seminíferos/metabolismo , Espermatozoides/química , Animales , Barrera Hematotesticular , Líquido Extracelular/química , Humanos , Inmunoterapia , Infertilidad Masculina/metabolismo , Masculino , Ratones , Neoplasias/terapia , Proteoma , Células de Sertoli/fisiología , Espermatogénesis , Testículo/metabolismo
2.
PLoS Biol ; 17(2): e3000002, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30763313

RESUMEN

Masculinization of the external genitalia in humans is dependent on formation of 5α-dihydrotestosterone (DHT) through both the canonical androgenic pathway and an alternative (backdoor) pathway. The fetal testes are essential for canonical androgen production, but little is known about the synthesis of backdoor androgens, despite their known critical role in masculinization. In this study, we have measured plasma and tissue levels of endogenous steroids in second trimester human fetuses using multidimensional and high-resolution mass spectrometry. Results show that androsterone is the principal backdoor androgen in the male fetal circulation and that DHT is undetectable (<1 ng/mL), while in female fetuses, there are significantly lower levels of androsterone and testosterone. In the male, intermediates in the backdoor pathway are found primarily in the placenta and fetal liver, with significant androsterone levels also in the fetal adrenal. Backdoor intermediates, including androsterone, are only present at very low levels in the fetal testes. This is consistent with transcript levels of enzymes involved in the alternate pathway (steroid 5α-reductase type 1 [SRD5A1], aldo-keto reductase type 1C2 [AKR1C2], aldo-keto reductase type 1C4 [AKR1C4], cytochrome P450 17A1 [CYP17A1]), as measured by quantitative PCR (qPCR). These data identify androsterone as the predominant backdoor androgen in the human fetus and show that circulating levels are sex dependent, but also that there is little de novo synthesis in the testis. Instead, the data indicate that placental progesterone acts as substrate for synthesis of backdoor androgens, which occurs across several tissues. Masculinization of the human fetus depends, therefore, on testosterone and androsterone synthesis by both the fetal testes and nongonadal tissues, leading to DHT formation at the genital tubercle. Our findings also provide a solid basis to explain why placental insufficiency is associated with disorders of sex development in humans.


Asunto(s)
Andrógenos/biosíntesis , Feto/fisiología , Masculinidad , Dihidrotestosterona/sangre , Dihidrotestosterona/metabolismo , Femenino , Humanos , Masculino , Redes y Vías Metabólicas , Ovario/metabolismo , Embarazo , Segundo Trimestre del Embarazo/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , Testículo/metabolismo
3.
FASEB J ; 34(8): 10373-10386, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32557858

RESUMEN

Male development, fertility, and lifelong health are all androgen-dependent. Approximately 95% of circulating testosterone is synthesized by the testis and the final step in this canonical pathway is controlled by the activity of the hydroxysteroid-dehydrogenase-17-beta-3 (HSD17B3). To determine the role of HSD17B3 in testosterone production and androgenization during male development and function we have characterized a mouse model lacking HSD17B3. The data reveal that developmental masculinization and fertility are normal in mutant males. Ablation of HSD17B3 inhibits hyperstimulation of testosterone production by hCG, although basal testosterone levels are maintained despite the absence of HSD17B3. Reintroduction of HSD17B3 via gene-delivery to Sertoli cells in adulthood partially rescues the adult phenotype, showing that, as in development, different cell-types in the testis are able to work together to produce testosterone. Together, these data show that HS17B3 acts as a rate-limiting-step for the maximum level of testosterone production by the testis but does not control basal testosterone production. Measurement of other enzymes able to convert androstenedione to testosterone identifies HSD17B12 as a candidate enzyme capable of driving basal testosterone production in the testis. Together, these findings expand our understanding of testosterone production in males.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/metabolismo , Células de Sertoli/metabolismo , Testículo/metabolismo , Testosterona/metabolismo , Andrógenos/metabolismo , Animales , Femenino , Fertilidad/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
BMC Dev Biol ; 19(1): 8, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30995907

RESUMEN

BACKGROUND: The interstitium of the mouse testis contains Leydig cells and a small number of steroidogenic cells with adrenal characteristics which may be derived from the fetal adrenal during development or may be a normal subset of the developing fetal Leydig cells. Currently it is not known what regulates development and/or proliferation of this sub-population of steroidogenic cells in the mouse testis. Androgen receptors (AR) are essential for normal testicular function and in this study we have examined the role of the AR in regulating interstitial cell development. RESULTS: Using a mouse model which lacks gonadotropins and AR (hpg.ARKO), stimulation of luteinising hormone receptors in vivo with human chorionic gonadotropin (hCG) caused a marked increase in adrenal cell transcripts/protein in a group of testicular interstitial cells. hCG also induced testicular transcripts associated with basic steroidogenic function in these mice but had no effect on adult Leydig cell-specific transcript levels. In hpg mice with functional AR, treatment with hCG induced Leydig cell-specific function and had no effect on adrenal transcript levels. Examination of mice with cell-specific AR deletion and knockdown of AR in a mouse Leydig cell line suggests that AR in the Leydig cells are likely to regulate these effects. CONCLUSIONS: This study shows that in the mouse the androgen receptor is required both to prevent development of testicular cells with adrenal characteristics and to ensure development of an adult Leydig cell phenotype.


Asunto(s)
Gonadotropina Coriónica/metabolismo , Desarrollo Embrionario/fisiología , Células Intersticiales del Testículo/citología , Hormona Luteinizante/metabolismo , Receptores Androgénicos/biosíntesis , Animales , Recuento de Células , Línea Celular Tumoral , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Modelos Animales , Fenotipo , Receptores Androgénicos/genética
5.
BMC Med ; 16(1): 23, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29429410

RESUMEN

BACKGROUND: Human fetal adrenal glands are highly active and, with the placenta, regulate circulating progesterone, estrogen and corticosteroids in the fetus. At birth the adrenals are essential for neonate salt retention through secretion of aldosterone, while adequate glucocorticoids are required to prevent adrenal insufficiency. The objective of this study was to carry out the first comprehensive analysis of adrenal steroid levels and steroidogenic enzyme expression in normal second trimester human fetuses. METHODS: This was an observational study of steroids, messenger RNA transcripts and proteins in adrenals from up to 109 second trimester fetuses (11 weeks to 21 weeks) at the Universities of Aberdeen and Glasgow. The study design was balanced to show effects of maternal smoking. RESULTS: Concentrations of 19 intra-adrenal steroids were quantified using liquid chromatography and mass spectrometry. Pregnenolone was the most abundant steroid while levels of 17α-hydroxyprogesterone, dehydroepiandrosterone sulphate (DHEAS) and progesterone were also high. Cortisol was present in all adrenals, but aldosterone was undetected and Δ4 androgens were low/undetected. CYP17A1, CYP21A2 and CYP11A1 were all highly expressed and the proteins localized to the adrenal fetal zone. There was low-level expression of HSD3B and CYP11B2, with HSD3B located mainly in the definitive zone. Maternal smoking altered fetal plasma adrenocorticotropic hormone (ACTH) (P = 0.052) and intra-adrenal progesterone, 17α-hydroxyprogesterone and 16α-hydroxyprogesterone, but not plasma or intra-adrenal cortisol, or intra-adrenal DHEAS. Fetal adrenal GATA6 and NR5A1 were increased by maternal smoking. CONCLUSIONS: The human fetal adrenal gland produces cortisol but very low levels of Δ4 androgens and no detectable aldosterone throughout the second trimester. The presence of cortisol in fetal adrenals suggests that adrenal regulation of circulating fetal ACTH remains a factor in development of congenital adrenal hyperplasia during the second trimester, while a relative lack of aldosterone explains the salt-wasting disorders frequently seen in extreme pre-term neonates. Finally, maternal smoking may alter fetal adrenal sensitivity to ACTH, which could have knock-on effects on post-natal health.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Aldosterona/metabolismo , Feto/efectos de los fármacos , Adulto , Aldosterona/análisis , Femenino , Humanos , Embarazo , Segundo Trimestre del Embarazo , Adulto Joven
6.
Development ; 141(10): 2139-49, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24803659

RESUMEN

Sertoli cells (SCs) regulate testicular fate in the differentiating gonad and are the main regulators of spermatogenesis in the adult testis; however, their role during the intervening period of testis development, in particular during adult Leydig cell (ALC) differentiation and function, remains largely unknown. To examine SC function during fetal and prepubertal development we generated two transgenic mouse models that permit controlled, cell-specific ablation of SCs in pre- and postnatal life. Results show that SCs are required: (1) to maintain the differentiated phenotype of peritubular myoid cells (PTMCs) in prepubertal life; (2) to maintain the ALC progenitor population in the postnatal testis; and (3) for development of normal ALC numbers. Furthermore, our data show that fetal LCs function independently from SC, germ cell or PTMC support in the prepubertal testis. Together, these findings reveal that SCs remain essential regulators of testis development long after the period of sex determination. These findings have significant implications for our understanding of male reproductive disorders and wider androgen-related conditions affecting male health.


Asunto(s)
Diferenciación Celular , Células Intersticiales del Testículo/fisiología , Epitelio Seminífero/citología , Células de Sertoli/fisiología , Maduración Sexual/fisiología , Testículo/citología , Testículo/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Masculino , Ratones , Ratones Desnudos , Ratones Transgénicos , Túbulos Seminíferos/citología
7.
Biol Reprod ; 96(4): 733-742, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28339967

RESUMEN

The placenta is a critical organ during pregnancy, essential for the provision of an optimal intrauterine environment, with fetal survival, growth, and development relying on correct placental function. It must allow nutritional compounds and relevant hormones to pass into the fetal bloodstream and metabolic waste products to be cleared. It also acts as a semipermeable barrier to potentially harmful chemicals, both endogenous and exogenous. Transporter proteins allow for bidirectional transport and are found in the syncytiotrophoblast of the placenta and endothelium of fetal capillaries. The major transporter families in the human placenta are ATP-binding cassette (ABC) and solute carrier (SLC), and insufficiency of these transporters may lead to deleterious effects on the fetus. Transporter expression levels are gestation-dependent and this is of considerable clinical interest as levels of drug resistance may be altered from one trimester to the next. This highlights the importance of these transporters in mediating correct and timely transplacental passage of essential compounds but also for efflux of potentially toxic drugs and xenobiotics. We review the current literature on placental molecular transporters with respect to their localization and ontogeny, the influence of fetal sex, and the relevance of animal models. We conclude that a paucity of information exists, and further studies are required to unlock the enigma of this dynamic organ.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas de Transporte de Membrana/metabolismo , Placenta/metabolismo , Femenino , Edad Gestacional , Humanos , Masculino , Proteínas de Transporte de Membrana/genética , Embarazo
8.
Arch Toxicol ; 91(11): 3645, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28980015

RESUMEN

During manuscript proofing, the following sentence was not deleted in the section "Results" at the end of the paragraph: "Both male and female hepatocytes responded in a similar fashion to cotinine, whereas male hepatocyte function was more sensitive to chrysene, fluorene and naphthalene than female hepatocytes".

9.
Arch Toxicol ; 91(11): 3633-3643, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28510779

RESUMEN

The liver is a dynamic organ which is both multifunctional and highly regenerative. A major role of the liver is to process both endo and xenobiotics. Cigarettes are an example of a legal and widely used drug which can cause major health problems for adults and constitute a particular risk to the foetus, if the mother smokes during pregnancy. Cigarette smoke contains a complex mixture of thousands of different xenobiotics, including nicotine and polycyclic aromatic hydrocarbons. These affect foetal development in a sex-specific manner, inducing sex-dependant molecular responses in different organs. To date, the effect of maternal smoking on the foetal liver has been studied in vitro using cell lines, primary tissue and animal models. While these models have proven to be useful, poor cell phenotype, tissue scarcity, batch-to-batch variation and species differences have led to difficulties in data extrapolation toward human development. Therefore, in this study we have employed hepatoblasts, derived from pluripotent stem cells, to model the effects of xenobiotics from cigarette smoke on human hepatocyte development. Highly pure hepatocyte populations (>90%) were produced in vitro and exposed to factors present in cigarette smoke. Analysis of ATP levels revealed that, independent of the sex, the majority of smoking derivatives tested individually did not deplete ATP levels below 50%. However, following exposure to a cocktail of smoking derivatives, ATP production fell below 50% in a sex-dependent manner. This was paralleled by a loss metabolic activity and secretory ability in both female and male hepatocytes. Interestingly, cell depletion was less pronounced in female hepatocytes, whereas caspase activation was ~twofold greater, indicating sex differences in cell death upon exposure to the smoking derivatives tested.


Asunto(s)
Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Fumar/efectos adversos , Adenosina Trifosfato/metabolismo , Diferenciación Celular , Células Cultivadas , Cotinina/toxicidad , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Femenino , Humanos , Masculino , Células Madre Pluripotentes/citología , Hidrocarburos Policíclicos Aromáticos/toxicidad , Factores Sexuales , alfa-Fetoproteínas/metabolismo
10.
Semin Cell Dev Biol ; 29: 55-65, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24598767

RESUMEN

Spermatogenesis is completely dependent on the pituitary hormone follicle-stimulating hormone (FSH) and androgens locally produced in response to luteinising hormone (LH). This dual control has been known since the 1930s and 1940s but more recent work, particularly using transgenic mice, has allowed us to determine which parts of the spermatogenic pathway are regulated by each hormone. During the first spermatogenic cycle after puberty both FSH and androgen act to limit the massive wave of germ cell apoptosis which occurs at this time. The established role of FSH in all cycles is to increase spermatogonial and subsequent spermatocyte numbers with a likely effect also on spermiation. Mice lacking FSH or its receptor are fertile, albeit with reduced germ cell numbers, and so this hormone is not an essential regulator of spermatogenesis but acts to optimise germ cell production Androgens also appear to regulate spermatogonial proliferation but, crucially, they are also required to allow spermatocytes to complete meiosis and form spermatids. Animals lacking androgen receptors fail to generate post-meiotic germ cells, therefore, and are infertile. There is also strong evidence that androgens act to ensure appropriate spermiation of mature spermatids. Androgen regulation of spermatogenesis is dependent upon action on the Sertoli cell but recent studies have shown that androgenic stimulation of the peritubular myoid cells is also essential for normal germ cells development. While FSH or androgen alone will both stimulate germ cell development, together they act synergistically to maximise germ cell number. The other hormones/local factors which can regulate spermatogenesis include activins and estrogens although their role in normal physiological regulation of this process needs to be more clearly established. Regulation of spermatogenesis in primates appears to be similar to that in rodents although the role of FSH may be greater. While our knowledge of hormone function during spermatogenesis is now well developed we still lack understanding of the mechanisms by which these hormones act to regulate this process.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Hormona Luteinizante/metabolismo , Receptores Androgénicos/genética , Espermatogénesis/fisiología , Espermatozoides/citología , Activinas/metabolismo , Animales , Apoptosis , Diferenciación Celular , Estrógenos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Células de Sertoli/fisiología
11.
Hum Reprod ; 31(2): 463-72, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26732622

RESUMEN

STUDY QUESTION: Do sex and maternal smoking effects on human fetal anogenital distance (AGD) persist in a larger study and how do these data integrate with the wider literature on perinatal human AGD, especially with respect to sex differences? SUMMARY ANSWER: Second trimester sex differences in AGD are broadly consistent with neonatal and infant measures of AGD and maternal cigarette smoking is associated with a temporary increase in male AGD in the absence of changes in circulating testosterone. WHAT IS KNOWN ALREADY: AGD is a biomarker of fetal androgen exposure, a reduced AGD in males being associated with cryptorchidism, hypospadias and reduced penile length. Normative fetal AGD data remain partial and windows of sensitivity of human fetal AGD to disruption are not known. STUDY DESIGN, SIZE, DURATION: The effects of fetal sex and maternal cigarette smoking on the second trimester (11-21 weeks of gestation) human fetal AGD were studied, along with measurement of testosterone and testicular transcripts associated with apoptosis and proliferation. PARTICIPANTS/MATERIALS, SETTING METHODS: AGD, measured from the centre of the anus to the posterior/caudal root of penis/clitoris (AGD(app)) was determined in 56 female and 70 male morphologically normal fetuses. These data were integrated with current literature on perinatal AGD in humans. MAIN RESULTS AND THE ROLE OF CHANCE: At 11-13 weeks of gestation male fetal AGD(app) was 61% (P< 0.001) longer than in females, increasing to 70% at 17-21 weeks. This sexual dimorphism was independent of growth characteristics (fetal weight, length, gonad weight). We confirmed that at 14-16 weeks of gestation male fetal AGD(app) was increased 28% (P < 0.05) by in utero cigarette smoke exposure. Testosterone levels were not affected by smoking. To develop normative data, our findings have been integrated with available data from in vivo ultrasound scans and neonatal studies. Inter-study variations in male/female AGD differences lead to the conclusion that normalization and standardization approaches should be developed to enable confidence in comparing data from different perinatal AGD studies. LIMITATIONS, REASONS FOR CAUTION: Sex differences, and a smoking-dependent increase in male fetal AGD at 14-16 weeks, identified in a preliminary study, were confirmed with a larger number of fetuses. However, human fetal AGD should, be re-assessed once much larger numbers of fetuses have been studied and this should be integrated with more detailed analysis of maternal lifestyle. Direct study of human fetal genital tissues is required for further mechanistic insights. WIDER IMPLICATIONS OF THE FINDINGS: Fetal exposure to cigarette smoke chemicals is known to lead to reduced fertility in men and women. Integration of our data into the perinatal human AGD literature shows that more work needs to be done to enable reliable inter-study comparisons. STUDY FUNDING/COMPETING INTERESTS: The study was supported by grants from the Chief Scientist Office (Scottish Executive, CZG/1/109 & CZG/4/742), NHS Grampian Endowments (08/02), the European Community's Seventh Framework Programme (FP7/2007-2013) under grant agreement no 212885 and the Medical Research Council, UK (MR/L010011/1). The authors declare they have no competing interests, be it financial, personal or professional.


Asunto(s)
Desarrollo Fetal/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Caracteres Sexuales , Humo/efectos adversos , Fumar/efectos adversos , Adulto , Canal Anal/anatomía & histología , Pesos y Medidas Corporales , Clítoris/anatomía & histología , Cotinina/sangre , Femenino , Humanos , Lactante , Masculino , Edad Materna , Pene/anatomía & histología , Embarazo , Segundo Trimestre del Embarazo , Testosterona
12.
BMC Med ; 13: 18, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25630355

RESUMEN

BACKGROUND: Maternal smoking is one of the most important modifiable risk factors for low birthweight, which is strongly associated with increased cardiometabolic disease risk in adulthood. Maternal smoking reduces the levels of the methyl donor vitamin B12 and is associated with altered DNA methylation at birth. Altered DNA methylation may be an important mechanism underlying increased disease susceptibility; however, the extent to which this can be induced in the developing fetus is unknown. METHODS: In this retrospective study, we measured concentrations of cobalt, vitamin B12, and mRNA transcripts encoding key enzymes in the 1-carbon cycle in 55 fetal human livers obtained from 11 to 21 weeks of gestation elective terminations and matched for gestation and maternal smoking. DNA methylation was measured at critical regions known to be susceptible to the in utero environment. Homocysteine concentrations were analyzed in plasma from 60 fetuses. RESULTS: In addition to identifying baseline sex differences, we found that maternal smoking was associated with sex-specific alterations of fetal liver vitamin B12, plasma homocysteine and expression of enzymes in the 1-carbon cycle in fetal liver. In the majority of the measured parameters which showed a sex difference, maternal smoking reduced the magnitude of that difference. Maternal smoking also altered DNA methylation at the imprinted gene IGF2 and the glucocorticoid receptor (GR/NR3C1). CONCLUSIONS: Our unique data strengthen studies linking in utero exposures to altered DNA methylation by showing, for the first time, that such changes are present in fetal life and in a key metabolic target tissue, human fetal liver. Furthermore, these data propose a novel mechanism by which such changes are induced, namely through alterations in methyl donor availability and changes in 1-carbon metabolism.


Asunto(s)
Carbono/metabolismo , Metilación de ADN/efectos de los fármacos , Feto/metabolismo , Hígado/metabolismo , Transferasas del Grupo 1-Carbono/metabolismo , Fumar/efectos adversos , Adulto , Peso Corporal , Cobalto/análisis , Femenino , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Hígado/química , Masculino , Transferasas del Grupo 1-Carbono/genética , Embarazo , ARN Mensajero/análisis , Receptores de Glucocorticoides/metabolismo , Estudios Retrospectivos , Factores Sexuales , Vitamina B 12/análisis
13.
Hum Reprod ; 29(7): 1471-89, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24847019

RESUMEN

STUDY QUESTION: How does maternal cigarette smoking disturb development of the human fetal ovary? SUMMARY ANSWER: Maternal smoking increases fetal estrogen titres and dysregulates several developmental processes in the fetal ovary. WHAT IS KNOWN ALREADY: Exposure to maternal cigarette smoking during gestation reduces human fetal ovarian cell numbers, germ cell proliferation and subsequent adult fecundity. STUDY DESIGN, SIZE, DURATION: The effects of maternal cigarette smoking on the second trimester human fetal ovary, fetal endocrine signalling and fetal chemical burden were studied. A total of 105 fetuses were studied, 56 from mothers who smoked during pregnancy and 49 from those who did not. PARTICIPANTS/MATERIALS, SETTING METHODS: Ovary, liver and plasma samples were collected from electively terminated, normally progressing, second trimester human fetuses. Circulating fetal hormones, levels of 73 fetal ovarian transcripts, protein localization, density of oocytes/primordial follicles and levels of 16 polycyclic aromatic hydrocarbons (PAHs) in the fetal liver were determined. MAIN RESULTS AND THE ROLE OF CHANCE: Circulating fetal estrogen levels were very high and were increased by maternal smoking (ANOVA, P = 0.055-0.004 versus control). Smoke exposure also dysregulated (two-way ANOVA, smoking versus gestation weeks interaction, P = 0.046-0.023) four fetal ovarian genes (cytochrome P450 scc [CYP11A1], NOBOX oogenesis homeobox [NOBOX], activator of apoptosis harakiri [HRK], nuclear receptor subfamily 2, group E, member 1 [NR2E1]), shifted the ovarian Inhibin ßA/inhibin α ratio (NHBA/INHA) transcript ratio in favour of activin (ANOVA, P = 0.049 versus control) and reduced the proportion of dominant-negative estrogen receptor 2 (ERß: ESR2) isoforms in half the exposed fetuses. PAHs, ligands for the aryl hydrocarbon receptor (AHR), were increased nearly 6-fold by maternal smoking (ANOVA, P = 0.011 versus control). A fifth transcript, COUP transcription factor 1 (nuclear receptor subfamily 2, group F, member 1: NR2F1, which contains multiple AHR-binding sites), was both significantly increased (ANOVA, P = 0.026 versus control) and dysregulated by (two-way ANOVA, smoking versus gestation weeks interaction, P = 0.021) maternal smoking. NR2F1 is associated with repression of FSHR expression and smoke-exposed ovaries failed to show the normal increase in FSHR expression during the second trimester. There was a significantly higher number of DEAD (Asp-Glu-Ala-Asp) box polypeptide 4 (DDX4) VASA-positive (ANOVA, P = 0.016 versus control), but not POU domain, class 1, transcription factor 1 (POU5F1) OCT3/4-positive, oocytes in smoke-exposed fetuses and this matched with a significantly higher number of primordial follicles (ANOVA, P = 0.024 versus control). LIMITATIONS, REASONS FOR CAUTION: The effects of maternal smoking on establishment of the maximum fetal primordial follicle pool cannot be reliably studied in our population since the process is not completed until 28 weeks of gestation and normal fetuses older than 21 weeks of gestation are not available for study. Our data suggest that some fetal ovaries are affected by smoke exposure while others are not, indicating that additional studies, with larger numbers, may show more significant effects. WIDER IMPLICATIONS OF THE FINDINGS: Fetal exposure to chemicals in cigarette smoke is known to lead to reduced fecundity in women. Our study suggests, for the first time, that this occurs via mechanisms involving activation of AHR, disruption of inhibin/activin and estrogen signalling, increased exposure to estrogen and dysregulation of multiple molecular pathways in the exposed human fetal ovary. Our data also suggest that alterations in the ESR2 positive and dominant negative isoforms may be associated with reduced sensitivity of some fetuses to increased estrogens and maternal smoking. STUDY FUNDING/COMPETING INTEREST(S): The study was supported by grants from the Chief Scientist Office (Scottish Executive, CZG/1/109, and CZG/4/742), NHS Grampian Endowments (08/02), the European Community's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 212885, a Society for Reproduction & Fertility summer studentship, Medical Research Scotland (research grant 354 FRG) and the Medical Research Council (WBS: U.1276.00.002.00001 and G1100357). The authors declare they have no competing interests, be it financial, personal or professional.


Asunto(s)
Exposición Materna/efectos adversos , Ovario/efectos de los fármacos , Fumar/efectos adversos , Adulto , Índice de Masa Corporal , Proliferación Celular , Cotinina/metabolismo , Estrógenos/metabolismo , Femenino , Desarrollo Fetal/efectos de los fármacos , Células Germinativas/citología , Humanos , Inmunohistoquímica , Recién Nacido , Ligandos , Hígado/metabolismo , Oocitos/citología , Folículo Ovárico/embriología , Ovario/embriología , Ovario/patología , Fenotipo , Hidrocarburos Policíclicos Aromáticos , Embarazo , Segundo Trimestre del Embarazo , Transducción de Señal , Productos de Tabaco
14.
Lancet Planet Health ; 8(1): e5-e17, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38199723

RESUMEN

BACKGROUND: Perfluoroalkyl and polyfluoroalkyl substances are classed as endocrine disrupting compounds but continue to be used in many products such as firefighting foams, flame retardants, utensil coatings, and waterproofing of food packaging. Perfluoroalkyl exposure aberrantly modulates lipid, metabolite, and bile acid levels, increasing susceptibility to onset and severity of metabolic diseases, such as diabetes and metabolic dysfunction-associated steatotic liver disease. To date, most studies in humans have focused on perfluoroalkyl-exposure effects in adults. In this study we aimed to show if perfluoroalkyls are present in the human fetal liver and if they have metabolic consequences for the human fetus. METHODS: In this cross-sectional study, human fetal livers from elective termination of pregnancies at the Aberdeen Pregnancy Counselling Service, Aberdeen, UK, were analysed by both targeted (bile acids and perfluoroalkyl substances) and combined targeted and untargeted (lipids and polar metabolites) mass spectrometry based metabolomic analyses, as well as with RNA-Seq. Only fetuses from normally progressing pregnancies (determined at ultrasound scan before termination), terminated for non-medical reasons, from women older than 16 years, fluent in English, and between 11 and 21 weeks of gestation were collected. Women exhibiting considerable emotional distress or whose fetuses had anomalies identified at ultrasound scan were excluded. Stringent bioinformatic and statistical methods such as partial correlation network analysis, linear regression, and pathway analysis were applied to this data to investigate the association of perfluoroalkyl exposure with hepatic metabolic pathways. FINDINGS: Fetuses included in this study were collected between Dec 2, 2004, and Oct 27, 2014. 78 fetuses were included in the study: all 78 fetuses were included in the metabolomics analysis (40 female and 38 male) and 57 fetuses were included in the RNA-Seq analysis (28 female and 29 male). Metabolites associated with perfluoroalkyl were identified in the fetal liver and these varied with gestational age. Conjugated bile acids were markedly positively associated with fetal age. 23 amino acids, fatty acids, and sugar derivatives in fetal livers were inversely associated with perfluoroalkyl exposure, and the bile acid glycolithocholic acid was markedly positively associated with all quantified perfluoroalkyl. Furthermore, 7α-hydroxy-4-cholesten-3-one, a marker of bile acid synthesis rate, was strongly positively associated with perfluoroalkyl levels and was detectable as early as gestational week 12. INTERPRETATION: Our study shows direct evidence for the in utero effects of perfluoroalkyl exposure on specific key hepatic products. Our results provide evidence that perfluoroalkyl exposure, with potential future consequences, manifests in the human fetus as early as the first trimester of gestation. Furthermore, the profiles of metabolic changes resemble those observed in perinatal perfluoroalkyl exposures. Such exposures are already linked with susceptibility, initiation, progression, and exacerbation of a wide range of metabolic diseases. FUNDING: UK Medical Research Council, Horizon Europe Program of the European Union, Seventh Framework Programme of the European Union, NHS Grampian Endowments grants, European Partnership for the Assessment of Risks from Chemicals, Swedish Research Council, Formas, Novo Nordisk Foundation, and the Academy of Finland.


Asunto(s)
Fluorocarburos , Enfermedades Metabólicas , Adulto , Embarazo , Humanos , Femenino , Masculino , Estudios Transversales , Metaboloma , Escocia , Ácidos y Sales Biliares , Fluorocarburos/efectos adversos
15.
PLoS One ; 18(9): e0290846, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37656709

RESUMEN

Sertoli cells support the development of sperm and the function of various somatic cells in the interstitium between the tubules. Sertoli cells regulate the function of the testicular vasculature and the development and function of the Leydig cells that produce testosterone for fertility and virility. However, the Sertoli cell-derived factors that regulate these cells are largely unknown. To define potential mechanisms by which Sertoli cells could support testicular somatic cell function, we aimed to identify Sertoli cell-enriched proteins in the testicular interstitial fluid (TIF) between the tubules. We previously resolved the proteome of TIF in mice and humans and have shown it to be a rich source of seminiferous tubule-derived proteins. In the current study, we designed bioinformatic strategies to interrogate relevant proteomic and genomic datasets to identify Sertoli cell-enriched proteins in mouse and human TIF. We analysed proteins in mouse TIF that were significantly reduced after one week of acute Sertoli cell ablation in vivo and validated which of these are likely to arise primarily from Sertoli cells based on relevant mouse testis RNASeq datasets. We used a different, but complementary, approach to identify Sertoli cell-enriched proteins in human TIF, taking advantage of high-quality human testis genomic, proteomic and immunohistochemical datasets. We identified a total of 47 and 40 Sertoli cell-enriched proteins in mouse and human TIF, respectively, including 15 proteins that are conserved in both species. Proteins with potential roles in angiogenesis, the regulation of Leydig cells or steroidogenesis, and immune cell regulation were identified. The data suggests that some of these proteins are secreted, but that Sertoli cells also deposit specific proteins into TIF via the release of extracellular vesicles. In conclusion, we have identified novel Sertoli cell-enriched proteins in TIF that are candidates for regulating somatic cell-cell communication and testis function.


Asunto(s)
Células de Sertoli , Testículo , Humanos , Masculino , Animales , Ratones , Líquido Extracelular , Proteómica , Semen
17.
Mol Hum Reprod ; 17(10): 620-5, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21527436

RESUMEN

Measurement of tissue mRNA transcript levels is critically dependent upon the normalization strategy used. For real-time polymerase chain reaction (PCR) (RT-PCR) this commonly depends upon identification of stable (non-variable) endogenous reference genes, with housekeeping genes (HKGs) the most commonly used. In this report we describe the use of an external mRNA standard to identify stable HKGs in the human fetal gonad. Total RNA was extracted from second trimester human fetal gonads and a standard amount of luciferase mRNA was added at the start of the extraction process. Levels of luciferase were then measured relative to each of seven HKGs (SDHA, TBP, B2M, PMM1, SFRS4, HMBS and UBC) by RT-PCR. When normalized to tissue weight, HKG expression was constant across fetal ages. Measurement of overall variation in transcript expression showed that PMM1 was the most stable HKG in the ovary while B2M was most stable in the testis. Re-analysis of the data using GeNorm and NormFinder algorithms showed that two of the top three most stable HKGs were the same using all three methods. This study describes a method for identification of endogenous, stable reference genes for RT-PCR studies of transcript expression levels which is objective and not dependent on prior assumptions of HKG expression. This technique is likely to be applicable to most tissues and, in this case, identifies suitable HKGs for studies into human gonadal development.


Asunto(s)
Genes Esenciales , Gónadas/embriología , ARN Mensajero/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Femenino , Perfilación de la Expresión Génica/normas , Regulación del Desarrollo de la Expresión Génica , Humanos , Luciferasas/genética , Masculino
18.
Reproduction ; 141(1): 37-46, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20956578

RESUMEN

The testes are essential endocrine regulators of fetal masculinization and male development and are, themselves, subject to hormonal regulation during gestation. This review focuses, primarily, on this latter control of testicular function. Data available suggest that, in most mammalian species, the testis goes through a period of independent function before the fetal hypothalamic-pituitary-gonadal axis develops at around 50% of gestation. This pituitary-independent phase coincides with the most critical period of fetal masculinization. Thereafter, the fetal testes appear to become pituitary hormone-dependent, concurrent with declining Leydig cell function, but increasing Sertoli cell numbers. The two orders of mammals most commonly used for these types of studies (rodents and primates) appear to represent special cases within this general hypothesis. In terms of testicular function, rodents are born 'early' before the pituitary-dependent phase of fetal development, while the primate testis is dependent upon placental gonadotropin released during the pituitary-independent phase of development.


Asunto(s)
Feto/metabolismo , Hormonas/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Transducción de Señal , Testículo/metabolismo , Animales , Animales Recién Nacidos , Humanos , Sistema Hipotálamo-Hipofisario/embriología , Recién Nacido , Células Intersticiales del Testículo/metabolismo , Masculino , Hormonas Hipofisarias/metabolismo , Hormonas Placentarias/metabolismo , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Hormonas Testiculares/metabolismo , Testículo/embriología
19.
Reprod Biol Endocrinol ; 7: 88, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19712470

RESUMEN

BACKGROUND: Testicular microliths are calcifications found within the seminiferous tubules. In humans, testicular microlithiasis (TM) has an unknown etiology but may be significantly associated with testicular germ cell tumors. Factors inducing microlith development may also, therefore, act as susceptibility factors for malignant testicular conditions. Studies to identify the mechanisms of microlith development have been hampered by the lack of suitable animal models for TM. METHODS: This was an observational study of the testicular phenotype of different mouse models. The mouse models were: cryptorchid mice, mice lacking androgen receptors (ARs) on the Sertoli cells (SCARKO), mice with a ubiquitous loss of androgen ARs (ARKO), hypogonadal (hpg) mice which lack circulating gonadotrophins, and hpg mice crossed with SCARKO (hpg.SCARKO) and ARKO (hpg.ARKO) mice. RESULTS: Microscopic TM was seen in 94% of hpg.ARKO mice (n=16) and the mean number of microliths per testis was 81+/-54. Occasional small microliths were seen in 36% (n=11) of hpg testes (mean 2+/-0.5 per testis) and 30% (n=10) of hpg.SCARKO testes (mean 8+/-6 per testis). No microliths were seen in cryptorchid, ARKO or SCARKO mice. There was no significant effect of FSH or androgen on TM in hpg.ARKO mice. CONCLUSION: We have identified a mouse model of TM and show that lack of endocrine stimulation is a cause of TM. Importantly, this model will provide a means with which to identify the mechanisms of TM development and the underlying changes in protein and gene expression.


Asunto(s)
Andrógenos/farmacología , Hipogonadismo/fisiopatología , Litiasis/patología , Enfermedades Testiculares/patología , Andrógenos/administración & dosificación , Animales , Criptorquidismo/genética , Criptorquidismo/fisiopatología , Dihidrotestosterona/administración & dosificación , Dihidrotestosterona/farmacología , Modelos Animales de Enfermedad , Femenino , Gonadotropinas/genética , Gonadotropinas/fisiología , Humanos , Hipogonadismo/genética , Litiasis/genética , Masculino , Ratones , Ratones Noqueados , Receptores Androgénicos/genética , Receptores Androgénicos/fisiología , Células de Sertoli/metabolismo , Enfermedades Testiculares/genética , Testículo/efectos de los fármacos , Testículo/metabolismo , Testículo/patología , Testosterona/administración & dosificación , Testosterona/farmacología
20.
Placenta ; 78: 10-17, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30955705

RESUMEN

INTRODUCTION: The placenta controls nutrient transfer between mother and fetus via membrane transporters. Appropriate transplacental passage of nutrients is essential for fetal growth and development. We investigated whether transporter transcript levels in human placenta-liver pairs from first and early second trimester pregnancies exhibit gestational age- or fetal sex-specific profiles and whether these are dysregulated by maternal smoking. METHODS: In a step-change for the field, paired placenta and fetal livers from 54 electively terminated, normally-progressing pregnancies (7-20 weeks of gestation, Scottish Advanced Fetal Research Study, REC 15/NS/0123) were sexed and cigarette smoking-exposure confirmed. Thirty-six nutrient transporter transcripts were quantified using RT-qPCR. RESULTS: While fetal, liver and placenta weights were not altered by maternal smoking, levels of transporter transcripts changed with fetal age and sex in the placenta and fetal liver and their trajectories were altered if the mother smoked. Placental levels of glucose uptake transporters SLC2A1 and SLC2A3 increased in smoking-exposed fetuses while smoking was associated with altered levels of amino acid and fatty acid transporter genes in both tissues. SLC7A8, which exchanges non-essential amino acids in the fetus for essential amino acids from the placenta, was reduced in smoking-exposed placentas while transcript levels of four hepatic fatty acid uptake transporters were also reduced by smoking. DISCUSSION: This data shows that fetal sex and age and maternal smoking are associated with altered transporter transcript levels. This could influence nutrient transport across the placenta and subsequent uptake by the fetal liver, altering trophic delivery to the growing fetus.


Asunto(s)
Feto/metabolismo , Hígado/metabolismo , Proteínas de Transporte de Membrana/genética , Nutrientes/metabolismo , Placenta/metabolismo , Complicaciones del Embarazo , Fumar , Adulto , Estudios de Casos y Controles , Femenino , Expresión Génica , Edad Gestacional , Humanos , Proteínas de Transporte de Membrana/metabolismo , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/metabolismo , Fumar/efectos adversos , Fumar/genética , Fumar/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA