Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 15: 1327800, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38654926

RESUMO

Introduction: Azoospermia, characterized by an absence of sperm in the ejaculate, represents the most severe form of male infertility. While surgical sperm retrieval in obstructive azoospermia (OA) is successful in the majority of cases, patients with non-obstructive azoospermia (NOA) show retrieval rates of only about 50% and thus frequently have unnecessary surgery. Surgical intervention could be avoided if patients without preserved spermatogenesis are identified preoperatively. This prospective study aimed to discover biomarkers in seminal plasma that could be employed for a non-invasive differential diagnosis of OA/NOA in order to rationalize surgery recommendations and improve success rates. Methods: All patients signed written informed consent, underwent comprehensive andrological evaluation, received human genetics to exclude relevant pathologies, and patients with azoospermia underwent surgical sperm retrieval. Using label-free LC-MS/MS, we compared the proteomes of seminal plasma samples from fertile men (healthy controls (HC), n=8) and infertile men diagnosed with 1) OA (n=7), 2) NOA with successful sperm retrieval (mixed testicular atrophy (MTA), n=8), and 3) NOA without sperm retrieval (Sertoli cell-only phenotype (SCO), n=7). Relative abundance changes of two candidate markers of sperm retrieval, HSPA2 and LDHC, were confirmed by Western Blot. Results: We found the protein expression levels of 42 proteins to be significantly down-regulated (p ≤ 0.05) in seminal plasma from SCO NOA patients relative to HC whereas only one protein was down-regulated in seminal plasma from MTA patients. Analysis of tissue and cell expression suggested that the testis-specific proteins LDHC, PGK2, DPEP3, and germ-cell enriched heat-shock proteins HSPA2 and HSPA4L are promising biomarkers of spermatogenic function. Western blotting revealed a significantly lower abundance of LDHC and HSPA2 in the seminal plasma of men with NOA (SCO and MTA) compared to controls. Discussion: The results indicate that certain testis-specific proteins when measured in seminal plasma, could serve as indicators of the presence of sperm in the testis and predict the success of sperm retrieval. Used in conjunction with conventional clinical assessments, these proteomic biomarkers may assist in the non-invasive diagnosis of idiopathic male infertility.


Assuntos
Azoospermia , Biomarcadores , Proteômica , Sêmen , Humanos , Masculino , Azoospermia/metabolismo , Azoospermia/diagnóstico , Sêmen/metabolismo , Sêmen/química , Biomarcadores/metabolismo , Biomarcadores/análise , Biomarcadores/sangue , Adulto , Proteômica/métodos , Estudos Prospectivos , Recuperação Espermática , Estudos de Casos e Controles , Espermatogênese/fisiologia
2.
Front Endocrinol (Lausanne) ; 13: 898876, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35685219

RESUMO

The long-standing knowledge that Sertoli cells determine fetal testosterone production levels is not widespread, despite being first reported over a decade ago in studies of mice. Hence any ongoing use of testosterone as a marker of Leydig cell function in fetal testes is inappropriate. By interrogating new scRNAseq data from human fetal testes, we demonstrate this situation is also likely to be true in humans. This has implications for understanding how disruptions to either or both Leydig and Sertoli cells during the in utero masculinization programming window may contribute to the increasing incidence of hypospadias, cryptorchidism, testicular germ cell tumours and adult infertility. We recently discovered that activin A levels directly govern androgen production in mouse Sertoli cells, because the enzymes that drive the conversion of the precursor androgen androstenedione to generate testosterone are produced exclusively in Sertoli cells in response to activin A. This minireview addresses the implications of this growing understanding of how in utero exposures affect fetal masculinization for future research on reproductive health, including during programming windows that may ultimately be relevant for organ development in males and females.


Assuntos
Ativinas , Células de Sertoli , Testículo , Androgênios , Animais , Humanos , Masculino , Camundongos , Testículo/patologia , Testosterona
3.
Curr Opin Cell Biol ; 77: 102104, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35671587

RESUMO

Spermatozoa are comprised of many unique proteins not expressed elsewhere. Sperm-specific proteins are first expressed at puberty, after the development of immune tolerance to self-antigens, and have been assumed to remain confined inside the seminiferous tubules, protected from immune cell recognition by various mechanisms of testicular immune privilege. However, new data has shown that sperm-specific proteins are released by the tubules into the surrounding interstitial fluid; from here they can contact immune cells, potentially promote immune tolerance, and enter the circulation. These new findings have clinical implications for diagnostics and therapeutics targeted at a specific class of proteins known as cancer-testis antigens (CTA), the opportunity to identify new communication pathways in the testis, and to discover new ways to monitor testis function.


Assuntos
Neoplasias , Sêmen , Humanos , Imunoterapia , Masculino , Neoplasias/diagnóstico , Neoplasias/terapia , Túbulos Seminíferos , Espermatozoides
4.
Mol Cell Endocrinol ; 544: 111556, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35031431

RESUMO

Testicular Leydig cells (LCs) are the principal source of circulating testosterone in males. LC steroidogenesis maintains sexual function, fertility and general health, and is influenced by various paracrine factors. The leukemia inhibitory factor receptor (LIFR) is expressed in the testis and activated by different ligands, including leukemia inhibitory factor (LIF), produced by peritubular myoid cells. LIF can modulate LC testosterone production in vitro under certain circumstances, but the role of consolidated signalling through LIFR in adult LC function in vivo has not been established. We used a conditional Lifr allele in combination with adenoviral vectors expressing Cre-recombinase to generate an acute model of LC Lifr-KO in the adult mouse testis, and showed that LC Lifr is not required for short term LC survival or basal steroidogenesis. However, LIFR-signalling negatively regulates steroidogenic enzyme expression and maximal gonadotrophin-stimulated testosterone biosynthesis, expanding our understanding of the intricate regulation of LC steroidogenic function.


Assuntos
Células Intersticiais do Testículo , Testosterona , Animais , Fator Inibidor de Leucemia/metabolismo , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Receptores de OSM-LIF/metabolismo , Testículo/metabolismo , Testosterona/metabolismo
5.
FASEB J ; 35(3): e21397, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33565176

RESUMO

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.


Assuntos
Antígenos de Neoplasias/análise , Proteínas/análise , Túbulos Seminíferos/metabolismo , Espermatozoides/química , Animais , Barreira Hematotesticular , Líquido Extracelular/química , Humanos , Imunoterapia , Infertilidade Masculina/metabolismo , Masculino , Camundongos , Neoplasias/terapia , Proteoma , Células de Sertoli/fisiologia , Espermatogênese , Testículo/metabolismo
6.
PLoS Genet ; 13(11): e1007078, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29136647

RESUMO

The katanin microtubule-severing proteins are essential regulators of microtubule dynamics in a diverse range of species. Here we have defined critical roles for the poorly characterised katanin protein KATNAL2 in multiple aspects of spermatogenesis: the initiation of sperm tail growth from the basal body, sperm head shaping via the manchette, acrosome attachment, and ultimately sperm release. We present data suggesting that depending on context, KATNAL2 can partner with the regulatory protein KATNB1 or act autonomously. Moreover, our data indicate KATNAL2 may regulate δ- and ε-tubulin rather than classical α-ß-tubulin microtubule polymers, suggesting the katanin family has a greater diversity of function than previously realised. Together with our previous research, showing the essential requirement of katanin proteins KATNAL1 and KATNB1 during spermatogenesis, our data supports the concept that in higher order species the presence of multiple katanins has allowed for subspecialisation of function within complex cellular settings such as the seminiferous epithelium.


Assuntos
Katanina/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos/genética , Animais , Células Germinativas/metabolismo , Haploidia , Infertilidade Masculina/metabolismo , Katanina/genética , Masculino , Camundongos , Microtúbulos/metabolismo , Isoformas de Proteínas , Epitélio Seminífero/metabolismo , Espermatogênese/genética , Espermatozoides/metabolismo , Testículo/metabolismo , Tubulina (Proteína)/metabolismo
7.
PLoS Genet ; 9(7): e1003628, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935508

RESUMO

Alternative splicing of precursor messenger RNA (pre-mRNA) is common in mammalian cells and enables the production of multiple gene products from a single gene, thus increasing transcriptome and proteome diversity. Disturbance of splicing regulation is associated with many human diseases; however, key splicing factors that control tissue-specific alternative splicing remain largely undefined. In an unbiased genetic screen for essential male fertility genes in the mouse, we identified the RNA binding protein RBM5 (RNA binding motif 5) as an essential regulator of haploid male germ cell pre-mRNA splicing and fertility. Mice carrying a missense mutation (R263P) in the second RNA recognition motif (RRM) of RBM5 exhibited spermatid differentiation arrest, germ cell sloughing and apoptosis, which ultimately led to azoospermia (no sperm in the ejaculate) and male sterility. Molecular modelling suggested that the R263P mutation resulted in compromised mRNA binding. Within the adult mouse testis, RBM5 localises to somatic and germ cells including spermatogonia, spermatocytes and round spermatids. Through the use of RNA pull down coupled with microarrays, we identified 11 round spermatid-expressed mRNAs as putative RBM5 targets. Importantly, the R263P mutation affected pre-mRNA splicing and resulted in a shift in the isoform ratios, or the production of novel spliced transcripts, of most targets. Microarray analysis of isolated round spermatids suggests that altered splicing of RBM5 target pre-mRNAs affected expression of genes in several pathways, including those implicated in germ cell adhesion, spermatid head shaping, and acrosome and tail formation. In summary, our findings reveal a critical role for RBM5 as a pre-mRNA splicing regulator in round spermatids and male fertility. Our findings also suggest that the second RRM of RBM5 is pivotal for appropriate pre-mRNA splicing.


Assuntos
Processamento Alternativo/genética , Diferenciação Celular/genética , Infertilidade Masculina/genética , Motivos de Nucleotídeos/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Células Germinativas/patologia , Humanos , Infertilidade Masculina/patologia , Masculino , Camundongos , Modelos Moleculares , Mutação , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/química , Espermátides/metabolismo , Espermátides/patologia , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética
8.
PLoS One ; 7(7): e41718, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22860010

RESUMO

The production of mature sperm is reliant on androgen action within the testis, and it is well established that androgens act on receptors within the somatic Sertoli cells to stimulate male germ cell development. Mice lacking Sertoli cell androgen receptors (AR) show late meiotic germ cell arrest, suggesting Sertoli cells transduce the androgenic stimulus co-ordinating this essential step in spermatogenesis. This study aimed to identify germ cell proteins responsive to changes in testicular androgen levels and thereby elucidate mechanisms by which androgens regulate meiosis. Testicular androgen levels were suppressed for 9 weeks using testosterone and estradiol-filled silastic implants, followed by a short period of either further androgen suppression (via an AR antagonist) or the restoration of intratesticular testosterone levels. Comparative proteomics were performed on protein extracts from enriched meiotic cell preparations from adult rats undergoing androgen deprivation and replacement in vivo. Loss of androgenic stimulus caused changes in proteins with known roles in meiosis (including Nasp and Hsp70-2), apoptosis (including Diablo), cell signalling (including 14-3-3 isoforms), oxidative stress, DNA repair, and RNA processing. Immunostaining for oxidised DNA adducts confirmed spermatocytes undergo oxidative stress-induced DNA damage during androgen suppression. An increase in PCNA and an associated ubiquitin-conjugating enzyme (Ubc13) suggested a role for PCNA-mediated regulation of DNA repair pathways in spermatocytes. Changes in cytoplasmic SUMO1 localisation in spermatocytes were paralleled by changes in the levels of free SUMO1 and of a subunit of its activating complex, suggesting sumoylation in spermatocytes is modified by androgen action on Sertoli cells. We conclude that Sertoli cells, in response to androgens, modulate protein translation and post-translational events in spermatocytes that impact on their metabolism, survival, and completion of meiosis.


Assuntos
Androgênios/fisiologia , Estradiol/fisiologia , Meiose , Proteoma/metabolismo , Espermatogênese , Testosterona/fisiologia , Animais , Apoptose , Células Cultivadas , RNA Helicases DEAD-box/metabolismo , Adutos de DNA/metabolismo , Reparo do DNA , Masculino , Estresse Oxidativo , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Processamento de Proteína Pós-Traducional , Proteômica , Splicing de RNA , Ratos , Ratos Sprague-Dawley , Proteína SUMO-1/metabolismo , Espermatócitos/metabolismo , Espermatócitos/fisiologia
9.
PLoS Genet ; 8(5): e1002697, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22654668

RESUMO

Spermatogenesis is a complex process reliant upon interactions between germ cells (GC) and supporting somatic cells. Testicular Sertoli cells (SC) support GCs during maturation through physical attachment, the provision of nutrients, and protection from immunological attack. This role is facilitated by an active cytoskeleton of parallel microtubule arrays that permit transport of nutrients to GCs, as well as translocation of spermatids through the seminiferous epithelium during maturation. It is well established that chemical perturbation of SC microtubule remodelling leads to premature GC exfoliation demonstrating that microtubule remodelling is an essential component of male fertility, yet the genes responsible for this process remain unknown. Using a random ENU mutagenesis approach, we have identified a novel mouse line displaying male-specific infertility, due to a point mutation in the highly conserved ATPase domain of the novel KATANIN p60-related microtubule severing protein Katanin p60 subunit A-like1 (KATNAL1). We demonstrate that Katnal1 is expressed in testicular Sertoli cells (SC) from 15.5 days post-coitum (dpc) and that, consistent with chemical disruption models, loss of function of KATNAL1 leads to male-specific infertility through disruption of SC microtubule dynamics and premature exfoliation of spermatids from the seminiferous epithelium. The identification of KATNAL1 as an essential regulator of male fertility provides a significant novel entry point into advancing our understanding of how SC microtubule dynamics promotes male fertility. Such information will have resonance both for future treatment of male fertility and the development of non-hormonal male contraceptives.


Assuntos
Adenosina Trifosfatases/genética , Infertilidade Masculina/genética , Células de Sertoli , Espermatogênese/genética , Adenosina Trifosfatases/metabolismo , Animais , Linhagem Celular Tumoral , Mapeamento Cromossômico , Expressão Gênica , Células Germinativas/citologia , Células Germinativas/metabolismo , Humanos , Katanina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microtúbulos/genética , Microtúbulos/metabolismo , Mutagênese , Fenótipo , Polimorfismo de Nucleotídeo Único , Epitélio Seminífero/metabolismo , Epitélio Seminífero/patologia , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Espermátides/metabolismo , Espermátides/patologia
10.
PLoS Genet ; 8(5): e1002698, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22654669

RESUMO

Katanin is an evolutionarily conserved microtubule-severing complex implicated in multiple aspects of microtubule dynamics. Katanin consists of a p60 severing enzyme and a p80 regulatory subunit. The p80 subunit is thought to regulate complex targeting and severing activity, but its precise role remains elusive. In lower-order species, the katanin complex has been shown to modulate mitotic and female meiotic spindle dynamics and flagella development. The in vivo function of katanin p80 in mammals is unknown. Here we show that katanin p80 is essential for male fertility. Specifically, through an analysis of a mouse loss-of-function allele (the Taily line), we demonstrate that katanin p80, most likely in association with p60, has an essential role in male meiotic spindle assembly and dissolution and the removal of midbody microtubules and, thus, cytokinesis. Katanin p80 also controls the formation, function, and dissolution of a microtubule structure intimately involved in defining sperm head shaping and sperm tail formation, the manchette, and plays a role in the formation of axoneme microtubules. Perturbed katanin p80 function, as evidenced in the Taily mouse, results in male sterility characterized by decreased sperm production, sperm with abnormal head shape, and a virtual absence of progressive motility. Collectively these data demonstrate that katanin p80 serves an essential and evolutionarily conserved role in several aspects of male germ cell development.


Assuntos
Adenosina Trifosfatases , Células Germinativas , Infertilidade Masculina/genética , Microtúbulos , Espermatogênese/genética , Espermatozoides , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Animais , Astenozoospermia/genética , Expressão Gênica , Células Germinativas/citologia , Células Germinativas/metabolismo , Katanina , Masculino , Meiose/genética , Camundongos , Microtúbulos/genética , Microtúbulos/metabolismo , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Oligospermia/genética , Subunidades Proteicas/genética , Motilidade dos Espermatozoides/genética , Espermatozoides/citologia , Espermatozoides/metabolismo , Espermatozoides/patologia , Fuso Acromático/genética , Testículo/metabolismo
11.
Endocrinology ; 152(4): 1670-83, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21325043

RESUMO

Spermatogenesis is absolutely dependent on FSH and androgens; suppression of these hormones inhibits germ cell development and thus sperm production. The final release of spermatids by the Sertoli cell, a process known as spermiation, is particularly sensitive to hormone suppression. To define the molecular mechanisms that mediate FSH and androgen effects in the Sertoli cell, we investigated the expression and regulation of micro-RNAs (miRNAs), small noncoding RNAs that regulate protein translation and modify cellular responses. By array analysis, we identified 23 miRNAs up-regulated more than 2-fold after hormone suppression in vivo and in vitro in primary Sertoli cell cultures. The regulation of four of these miRNAs (miR-23b, -30c, -30d, and -690) was confirmed by quantitative RT-PCR. Bioinformatic analysis of potential targets of hormonally regulated miRNAs identified genes important for focal adhesion and regulation of the actin cytoskeleton, processes known to be intimately associated with adhesion of spermatids to Sertoli cells. Two of the identified genes, Pten, an intracellular phosphatase, and Eps15, a mediator of endocytosis, were down-regulated by the withdrawal of hormones in vivo and possess miR-23b target sites in their 3'-untranslated regions. Overexpression of miR-23b in vitro resulted in decreased translation of PTEN and EPS15 protein as assessed by Western blot and luciferase analysis. We conclude that FSH and androgens act on Sertoli cells in stage VIII to control the expression of miRNAs that operate in a coordinated manner to regulate cell adhesion pathways and male fertility and that miRNA transcription is a new paradigm in the hormone dependence of spermiation.


Assuntos
Androgênios/farmacologia , Hormônio Foliculoestimulante/farmacologia , MicroRNAs/genética , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Animais , Western Blotting , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Biologia Computacional , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Túbulos Seminíferos/citologia , Testículo/citologia , Testículo/metabolismo
12.
J Endocrinol ; 190(3): 759-70, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17003277

RESUMO

Spermiation is the final step of spermatogenesis and culminates in the disengagement (release) of elongated spermatids from Sertoli cells into the seminiferous tubule lumen. Spermiation failure, wherein spermatids are retained by Sertoli cells instead of releasing, occurs after hormone suppression. The mechanisms involved in spermatid disengagement and retention are not well understood. We previously showed that beta(1)-integrin is associated with spermatids until the point of disengagement, but the ectoplasmic specialisation junction (ES) is not. The aims of this paper are to further characterise the complex that is present immediately prior to spermatid disengagement by identifying the alpha-integrin form dimerised with beta(1)-integrin, localising focal adhesion kinase (FAK) and determining if microtubules are involved. Adult Sprague-Dawley rats received testosterone and oestradiol implants and an FSH antibody for 7 days to suppress testicular testosterone and FSH and induce spermiation failure. Control rats were treated with saline. Immunohistochemical analysis showed that alpha(6)-integrin and a phosphorylated form of FAK (FAK-Tyr(397)) are present between late spermatids and Sertoli cells after ES removal, until the point of disengagement, and both proteins remain associated with retained spermatids after spermiation failure induced by hormone suppression. Using dual-label immunofluorescence, tubulins (and thus microtubules) were observed to co-localise with ES, but were neither associated with elongated spermatids just prior to release nor with retained spermatids following hormone suppression. These results suggest that microtubules are not involved in the final release of spermatids from Sertoli cells. We conclude that spermatid release during spermiation is mediated by a 'disengagement complex' containing alpha(6)beta(1)-integrin and phospho-FAK, the function of which can be affected by gonadotrophin suppression.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/análise , Integrina alfa6beta1/análise , Epitélio Seminífero/química , Células de Sertoli/química , Espermátides/química , Espermatogênese/fisiologia , Animais , Biomarcadores/análise , Western Blotting/métodos , Estradiol/farmacologia , Imunofluorescência , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Hormônio Foliculoestimulante/farmacologia , Integrina alfa6beta1/metabolismo , Masculino , Microtúbulos/química , Fosforilação , Ratos , Ratos Sprague-Dawley , Espermátides/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Testosterona/antagonistas & inibidores , Testosterona/farmacologia , Tubulina (Proteína)/análise
13.
J Clin Endocrinol Metab ; 91(10): 3962-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16895950

RESUMO

CONTEXT: Male hormonal contraception via gonadotropin and intratesticular androgen withdrawal disrupts spermatogenesis at two principal sites: 1) spermatogonial maturation, and 2) spermiation. OBJECTIVE: The objective of this study was to explore the relative dependence of each stage of germ cell development on FSH and LH/intratesticular androgen action. DESIGN, SETTING, AND PARTICIPANTS: Eighteen men enrolled in this prospective, randomized 14-wk study at Prince Henry's Institute. INTERVENTIONS: Subjects (n = 6/group) were assigned to 6 wk of 1) testosterone (T) implant (4 x 200 mg sc once)+depot medroxy progesterone acetate (DMPA; 150 mg im once); 2) T implant+DMPA+FSH (300 IU sc twice weekly); and 3) T implant+DMPA+human chorionic gonadotropin (hCG; 1000 IU sc twice weekly as an LH substitute). Men then underwent a vasectomy and testicular biopsy with previously reported control data used for comparison. MAIN OUTCOME MEASURES: Germ cell number (assessed by the optical disector stereological approach) and intratesticular androgen levels were determined. RESULTS: T+DMPA alone significantly suppressed type B spermatogonia, preleptotene through to pachytene spermatocytes, and round spermatids from control (P < 0.05). All germ cell subtypes were maintained at control levels by either FSH or LH activity, except pachytene spermatocytes, which were found to be lower in the hCG vs. FSH (P < 0.01) and control groups (P < 0.05). CONCLUSIONS: FSH and LH maintained spermatogenesis independently in this gonadotropin-suppressed model. Compared with LH, FSH showed better maintenance of pachytene spermatocyte number, whereas improved conversion to round spermatids was suggested with hCG treatment. Future contraceptive treatment strategies must consider independent regulation of spermatogenesis by both FSH and LH/intratesticular androgens for maximum efficacy.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Hormônio Luteinizante/fisiologia , Espermatogênese , Espermatogônias/fisiologia , Adulto , Gonadotropina Coriônica/farmacologia , Estradiol/sangue , Hormônio Foliculoestimulante/sangue , Humanos , Hormônio Luteinizante/sangue , Masculino , Acetato de Medroxiprogesterona/administração & dosagem , Pessoa de Meia-Idade , Estudos Prospectivos , Contagem de Espermatozoides , Testículo/química , Testosterona/administração & dosagem , Testosterona/sangue
14.
J Clin Endocrinol Metab ; 90(10): 5647-55, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16030154

RESUMO

CONTEXT: Combination of a GnRH antagonist (acyline), types I and II, 5alpha-reductase inhibitor (dutasteride) or levonorgestrel (LNG) with testosterone (T) treatment may augment the suppression of spermatogenesis and intratesticular (iT) steroids. OBJECTIVE: The objective of this study was to assess the effects of combined hormonal contraceptive regimens on germ cell populations and iT steroids. DESIGN, SETTING, AND PARTICIPANTS: Twenty-nine normal health men enrolled in this prospective, randomized, 14-wk study at the University of Washington. INTERVENTION(S): Twenty-two men (n = 5-6/group) received 8 wk of T enanthate (TE; 100 mg, i.m., weekly) combined with 1) 125 microg LNG daily, orally; 2) 125 microg LNG plus 0.5 mg dutasteride daily, orally; 3) 300 microg/kg acyline twice weekly, s.c.; or 4) 125 microg LNG daily, orally, plus 300 microg/kg acyline twice weekly, s.c. Subjects then underwent a vasectomy and testicular biopsy. Control men (n = 7) proceeded directly to surgery. MAIN OUTCOME MEASURE(S): The main outcome measures were germ cells and iT steroids [T, dihydrotestosterone, 3alpha- and beta-androstanediol (Adiol), and estradiol (E2)]. RESULTS: High iT levels of all androgens (6- to 123-fold serum levels) and E2 (407-fold serum levels) were found in control men. iTT (1.9-2.6% control; P < 0.001) and iT3betaAdiol (16-34% control; P < 0.05) levels decreased with all treatments. iT dihydrotestosterone (13-29% control; P < 0.05) and iT3alphaAdiol (44-47% control; P < 0.05) levels decreased with all but the TE plus LNG treatment. iTE2 levels decreased only in the TE plus acyline group (28% control; P = 0.01). Germ cells from type B spermatogonia onward were suppressed, with no differences between groups found. Variable sites of impairment of germ cell progression were evident between men (spermagonial maturation, meiosis 1 entry, and spermiation). Other than a negative correlation between iT3alphaAdiol and haploid germ cell number (P < 0.006), no correlations between germ cell number and gonadotropins, sperm concentration, or iT steroids were found. CONCLUSIONS: A similar high testicular:serum gradient exists for E2 and T in normal men, and 8 wk of gonadotropin suppression markedly reduces iTT, with 5alpha-reduced androgens and E2 levels decreasing to a much lesser degree. The heterogeneity of the germ cell response, regardless of treatment, gonadotropins or iT steroids, points to the individual sensitivity of sites in germ cell development, which is worthy of additional exploration.


Assuntos
Inibidores de 5-alfa Redutase , Inibidores Enzimáticos/farmacologia , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Levanogestrel/farmacologia , Congêneres da Progesterona/farmacologia , Espermatogênese/efeitos dos fármacos , Esteroides/metabolismo , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testosterona/farmacologia , Adulto , Androstanos/sangue , Azasteroides/farmacologia , Di-Hidrotestosterona/sangue , Dutasterida , Estradiol/sangue , Células Germinativas/efeitos dos fármacos , Humanos , Masculino , Estudos Prospectivos , Células de Sertoli/efeitos dos fármacos , Testosterona/sangue , Vasectomia
15.
Fertil Steril ; 81(6): 1595-603, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15193483

RESUMO

OBJECTIVE: To quantify germ cell loss and the extent of testicular fibrosis in vasectomized patients of varying obstructive intervals. DESIGN: Retrospective study. SETTING: Specialized male reproductive surgery and endocrinology service. PATIENT(S): Thirty-four vasectomized patients 1-20 years after surgery and 10 normal subjects. INTERVENTION(S): Thirty-four testicular biopsies taken at the time of vasectomy reversal (vasovasostomy). Control biopsies taken from 10 normal men at the time of vasectomy. MAIN OUTCOME MEASURE(S): Stereological assessment of testicular germ cell populations and testicular fibrosis. RESULT(S): Vasectomy caused a significant decrease in germ cells in the later stages of spermatogenesis, with significant reductions in pachytene spermatocytes (by 18%), round spermatids (by 40%), elongating spermatids (by 23%), and elongated spermatids (by 39%). The loss of spermatids showed a significant relationship with obstructive interval. A significant 2.7-fold increase in total (peritubular plus interstitial) fibrosis was observed, which showed a positive relationship with obstructive interval. Decreased germ cell populations and fibrosis did not seem to be related to sperm antibody levels nor to sperm counts obtained up to 2 years after vasovasostomy. CONCLUSION(S): Vasal obstruction results in significant reductions in germ cells in the later stages of spermatogenesis and increases in testicular fibrosis, both worsening with an increasing obstructive interval. Testicular damage after vasectomy might impact upon the prospects for reversal.


Assuntos
Espermatogênese , Testículo/patologia , Vasectomia , Adulto , Anticorpos/análise , Fibrose , Hormônios/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Período Pós-Operatório , Estudos Retrospectivos , Contagem de Espermatozoides , Espermátides/patologia , Espermatozoides/imunologia , Fatores de Tempo , Vasovasostomia
16.
FASEB J ; 16(11): 1389-97, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12205030

RESUMO

Steroids play a critical role in gonadal differentiation in birds, reptiles, and amphibia whereas gonadal differentiation in mammals is thought to be determined by genetic mechanisms. The gonads of female mice incapable of synthesizing estrogens due to disruption of the aromatase gene (ArKO) provide a unique model to test the role of estrogen in regulating the gonadal phenotype. We have shown that in the absence of estrogen, genetically female mice develop testicular tissue within their ovaries. The ovaries develop cells that possess structural and functional characteristics of testicular interstitial cells and of seminiferous tubule-like structures lined with Sertoli cells. Moreover, the ovaries express mRNA for the testis-specific Sertoli cell transcription factor Sox 9 and espin protein, which is specific for inter-Sertoli cell junctions. The development of the testicular tissue in this model can be reverted/postponed by replacing estrogen. When ArKO female mice were fed a diet containing phytoestrogens, the appearance of Leydig and Sertoli cells was postponed and reduced. Furthermore, administration of estradiol-17beta decreased the number of Sertoli and Leydig cells in the ovaries. These findings constitute definitive evidence that estrogen plays a critical role in maintaining female somatic interstitial and granulosa cells in the eutherian ovary.


Assuntos
Estrogênios/farmacologia , Isoflavonas , Ovário/citologia , Animais , Aromatase/genética , Diferenciação Celular/efeitos dos fármacos , Estradiol/farmacologia , Estrogênios/fisiologia , Estrogênios não Esteroides/farmacologia , Feminino , Gonadotropinas/sangue , Células da Granulosa/ultraestrutura , Proteínas de Grupo de Alta Mobilidade/biossíntese , Proteínas de Grupo de Alta Mobilidade/genética , Imuno-Histoquímica , Células Intersticiais do Testículo/ultraestrutura , Masculino , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/análise , Proteínas dos Microfilamentos/imunologia , Ovário/efeitos dos fármacos , Ovário/metabolismo , Ovário/ultraestrutura , Fenótipo , Fitoestrógenos , Preparações de Plantas , RNA Mensageiro/biossíntese , Fatores de Transcrição SOX9 , Células de Sertoli/ultraestrutura , Testículo/química , Testículo/citologia , Testículo/ultraestrutura , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
17.
Biol Reprod ; 67(3): 820-8, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12193390

RESUMO

Spermatogenesis in the rat consists of 14 unique morphologic cellular associations between Sertoli cells and developing germ cells within the seminiferous epithelium. The complexity of the cellular associations leads to difficulty in the isolation of individual cells at a defined stage of development for the study of their unique patterns of gene or protein expression. Thus, laser-capture microdissection is an ideal technique to permit such analysis. This study used laser-capture microdissection and real-time reverse transcription-polymerase chain reaction (RT-PCR) to quantitate the stage-specific expression of a series of genes of functional significance in hormonal regulation and cell-cell interactions in spermatogenesis, including cathepsin-L, CREM-tau, transition protein-1, androgen receptor, beta1-integrin, N-cadherin, and hypoxanthine phosphoribosyltransferase (HPRT). Frozen sections (10 micro m) were obtained from normal adult rat testes. Laser-capture microdissection (LCM) was used to capture all cells in cross-sections of seminiferous tubules that were grouped into stages I-V, VII-VIII, and IX-XIII. Transition protein-1 expression was lowest during stages I-V and increased 5.9-fold during stages VII-VIII and IX-XIII (P < 0.01). Cathepsin-L expression was highest during stages I-V and VII-VIII, falling 4.9-fold during stages IX-XIII (P < 0.05). Similarly, CREM-tau expression was highest during stages I-V and VII-VIII, falling 1.6-fold during stages IX-XIII (P < 0.05). A novel CREM-tau isoform lacking the phosphorylation domain was also characterized but was not stage-specific. beta1-Integrin, N-cadherin, and androgen receptor expression did not change between the spermatogenic stages examined. HPRT housekeeper expression was lowest during stages I-V but increased 1.5-fold during stages VII-VIII and IX-XIII (P < 0.05). This study is the first to apply LCM and real-time RT-PCR analysis to quantitate stage-specific changes in the expression of multiple genes in the seminiferous epithelium.


Assuntos
Expressão Gênica , Proteínas Repressoras , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogênese/genética , Animais , Caderinas/genética , Catepsina L , Catepsinas/genética , Proteínas Cromossômicas não Histona/genética , Modulador de Elemento de Resposta do AMP Cíclico , Cisteína Endopeptidases , Proteínas de Ligação a DNA/genética , Hipoxantina Fosforribosiltransferase/genética , Integrina beta1/genética , Lasers , Masculino , Microscopia de Fluorescência , Ratos , Ratos Sprague-Dawley , Receptores Androgênicos/genética
18.
Endocrinology ; 143(8): 2913-21, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12130556

RESUMO

Estrogen is synthesized in the testis, both in Leydig cells and seminiferous epithelium, and its importance in spermatogenesis is highlighted by the phenotype of the aromatase knockout (ArKO) mouse. These mice are unable to synthesize endogenous estrogens. The males develop postmeiotic defects by 18 wk of age. We hypothesized that maintenance of spermatogenesis in younger animals may be mediated by exogenous estrogenic substances. Dietary soy meal, contained in almost all commercial rodent diets, provides a source of estrogenic isoflavones. We thus investigated spermatogenesis in wild-type and ArKO mice raised on a diet containing soy, compared with a soy-free diet, to elucidate the biological action of phytoestrogens on the testis. In ArKO mice, dietary phytoestrogens could partially prevent disruptions to spermatogenesis, in that they prevented the decline in germ cell numbers. They also seemed to maintain Sertoli cell function, and they blocked elevations in FSH. The impairment of spermatogenesis seen in soy-free ArKOs occurred in the absence of a decreased gonadotropic stimulus, suggesting that the effects of dietary phytoestrogens are independent of changes to the pituitary-gonadal axis. Our study highlights the importance of estrogen in spermatogenesis and shows that relatively low levels of dietary phytoestrogens have a biological effect in the testis.


Assuntos
Aromatase/fisiologia , Estrogênios não Esteroides/farmacologia , Isoflavonas , Testículo/efeitos dos fármacos , Animais , Aromatase/genética , Estrogênios/fisiologia , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Fitoestrógenos , Preparações de Plantas , Células de Sertoli/fisiologia , Contagem de Espermatozoides , Espermatogênese/efeitos dos fármacos , Testículo/patologia , Testículo/fisiologia
19.
Mol Cell Endocrinol ; 191(1): 27-33, 2002 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-12044916

RESUMO

The steroidogenic pathway within the ovary gives rise to progestins, androgens and oestrogens, all of which act via specific nuclear receptors to regulate reproductive function and maintain fertility. The precise role of oestrogen in the ovary remains to be elucidated, hence the data presented here which arises from studies designed to resolve this issue. Oestrogens signal via two receptor subtypes ERalpha and ERbeta, both of which are present in the ovary. ERbeta, the most abundant mRNA, is primarily expressed by GC where it transduces signals from ovarian-derived and exogenous oestrogens. Specific roles for each of the ERs in the ovary have yet to be established, despite ER knockout studies indicating both are required for normal function. The ArKO mouse is a model of oestrogen insufficiency. These mice are infertile as a result of arrested folliculogenesis (at the antral stage) and a failure to ovulate. Trans/re-differentiation of somatic cells in the ovary gives rise to Sertoli cell-like and Leydig cell-like cells within abnormal follicular structures. Disruption to the balance of sex steroids in the ovary is likely to facilitate this phenotype. Future studies will focus on the regulation of somatic cell differentiation, assigning roles to individual ERs and establishing definitive targets of oestrogen action in the ovary.


Assuntos
Estrogênios/metabolismo , Ovário/fisiologia , Receptores de Estrogênio/metabolismo , Receptores de Esteroides/metabolismo , Animais , Feminino , Camundongos , Camundongos Knockout , Modelos Biológicos , Fenótipo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/genética , Receptores de Esteroides/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA