Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Hum Reprod ; 33(11): 1963-1974, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30247578

ABSTRACT

STUDY QUESTION: Are infertile men with reduced semen quality at risk of a further decrease in testicular function? SUMMARY ANSWER: Infertile men with severely reduced semen quality risk further deterioration of semen quality 15 years after treatment for infertility, and a lower baseline sperm concentration was associated with a more pronounced increase in LH and decrease in testosterone/LH ratio at follow-up. WHAT IS KNOWN ALREADY: Male factors account for up to 50% of human infertility. The most common finding is spermatogenic failure (SgF) yet the life course of semen quality and testosterone production in such men has not been described. STUDY DESIGN, SIZE, DURATION: A follow-up study of men with SgF was performed 15 years after the initial infertility assessment between January 1995 and December 2000. PARTICIPANTS/MATERIALS, SETTING, METHODS: Hospital records were used to identify potential participants in the study. A total of 137 men with primary male infertility due to SgF and 70 controls with good semen quality from couples with female factor infertility who attended a tertiary referral centre were included: the participation rate was 31% and 26%, respectively. The men provided semen samples and underwent a physical examination. Blood samples were taken to measure levels of reproductive hormones (FSH, LH, testosterone, sex hormone-binding globulin, estradiol and inhibin B). Current results were compared with results from the initial assessments. MAIN RESULTS AND THE ROLE OF CHANCE: At the time of follow up the SgF men had significantly lower Leydig cell capacity than the control group as well as much lower semen quality. For the SgF men, between baseline sampling and follow up, the median sperm concentration decreased from 1.9 to 0.6 mill/ml and total sperm count from 7.7 to 2.0 million (P = 0.019 and 0.012, respectively), and 10% developed azoospermia. Calculated free testosterone (cFT), but not total testosterone (tT) decreased in the SgF group by ~0.6% (95% CI 0.1-1.2%) per year. In the SgF group, LH increased by 1.6% (CI 0.9-2.3%) annually, and consequently tT/LH and cFT/LH ratios had decreased by 1.3% (CI 0.5-2.1) and 2.1% (CI 1.2-3.0%), respectively. The increase in LH and the decreases in tT/LH and cFT/LH ratios were more pronounced in men with lower baseline sperm concentrations. LIMITATIONS, REASONS FOR CAUTION: We consider the case group as representative of infertile men not in need of testosterone treatment at baseline investigation, but do not have information on those that chose not to participate in the follow-up study. There were alterations in some hormone analysis methods during the follow-up period that may introduce uncertainty in interpretation of long-term changes in hormone levels despite rigorous quality control. The validity of the control group suffers from a lack of hormone values at baseline. Also, at follow-up, for practical reasons only one semen sample could be obtained, which makes the effect estimate more uncertain and there is a risk of non-differential misclassification. WIDER IMPLICATIONS OF THE FINDINGS: Without being able to predict individual outcomes, it is prudent to consider sperm cryopreservation or advise not to postpone fertility treatment when men present with infertility due to impaired semen quality. Whether partly compensated Leydig cell insufficiency in men with SgF will eventually develop into overt testosterone deficiency cannot be determined from our study. STUDY FUNDING/COMPETING INTEREST(s): Aase and Einar Danielsen (Grant no. 10-001053), Nordic Research Committee (Grant no. 5109), The Kirsten and Freddie Johansen Fund, and Rigshospitalet's Research Fund (grant no. R24-A812). There are no competing interests.


Subject(s)
Infertility, Male/blood , Leydig Cells/physiology , Sperm Count/statistics & numerical data , Sperm Motility/physiology , Adult , Case-Control Studies , Female , Follicle Stimulating Hormone/blood , Humans , Infertility, Male/physiopathology , Longitudinal Studies , Luteinizing Hormone/blood , Male , Testosterone/blood
2.
Andrology ; 6(1): 176-183, 2018 01.
Article in English | MEDLINE | ID: mdl-29179257

ABSTRACT

Testicular germ cell cancer (TGCC) is derived from germ cell neoplasia in situ (GCNIS), which arises due to niche disturbances affecting the Sertoli cells. It is believed that exogenous endocrine factors have a crucial role in governing neoplastic transformation but on a strong hereditary background. Follicle-stimulating hormone (FSH) is the major regulatory hormone of the Sertoli cells. FSH signalling-related single-nucleotide polymorphisms (SNPs) have previously been shown to affect FSH action in men at different levels. We aimed to investigate whether three FSH-related SNPs (FSHR 2039A>G, FSHR -29G>A and FSHB -211G>T) are associated with development of TGCC. A total of 752 Danish and German patients with TGCC from two tertiary andrological referral centres were included. Three control groups comprising 2020 men from the general population, 679 fertile men and 417 infertile men, were also included. Chi-squared test was performed to compare genotype- and allele frequencies. Kruskal-Wallis test was performed to compare age at diagnosis. Patients with TGCC had a higher frequency of the A-allele of FSHR 2039A>G compared to the group of fertile men with an AA-genotype frequency of 30.2% vs. 22.0%, respectively, p = 0.002. This variant is associated with higher FSH receptor activity. The distribution of the FSHR 2039A>G did not differ significantly between the patients with TGCC and the infertile or the general population. The frequency of the two other SNPs did not differ between patient with TGCC and any of the control groups. No differences were detected between genotypes and age distribution or histological subtype of the tumours. In conclusion, we observed that a genetic variant associated with FSHR activity may modulate the susceptibility to TGCC.


Subject(s)
Genetic Predisposition to Disease/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, FSH/genetics , Testicular Neoplasms/genetics , Adolescent , Adult , Genotype , Humans , Male , Middle Aged , Retrospective Studies , Young Adult
3.
Endocr Connect ; 7(1): 16-25, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28874401

ABSTRACT

Human sperm cell function must be precisely regulated to achieve natural fertilization. Progesterone released by the cumulus cells surrounding the egg induces a Ca2+ influx into human sperm cells via the CatSper Ca2+-channel and thereby controls sperm function. Multiple chemical UV filters have been shown to induce a Ca2+ influx through CatSper, thus mimicking the effect of progesterone on Ca2+ signaling. We hypothesized that these UV filters could also mimic the effect of progesterone on sperm function. We examined 29 UV filters allowed in sunscreens in the US and/or EU for their ability to affect acrosome reaction, penetration, hyperactivation and viability in human sperm cells. We found that, similar to progesterone, the UV filters 4-MBC, 3-BC, Meradimate, Octisalate, BCSA, HMS and OD-PABA induced acrosome reaction and 3-BC increased sperm penetration into a viscous medium. The capacity of the UV filters to induce acrosome reaction and increase sperm penetration was positively associated with the ability of the UV filters to induce a Ca2+ influx. None of the UV filters induced significant changes in the proportion of hyperactivated cells. In conclusion, chemical UV filters that mimic the effect of progesterone on Ca2+ signaling in human sperm cells can similarly mimic the effect of progesterone on acrosome reaction and sperm penetration. Human exposure to these chemical UV filters may impair fertility by interfering with sperm function, e.g. through induction of premature acrosome reaction. Further studies are needed to confirm the results in vivo.

4.
Andrology ; 5(6): 1105-1114, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28992366

ABSTRACT

Perceived stress has been associated with decreased semen quality but the mechanisms have not been elucidated. It is not known whether cortisol, the major stress hormone in humans, can act directly via receptors in the testis, and whether variants in the gene encoding the glucocorticoid receptor (NR3C1) can possibly modulate the effect. To address these questions, we investigated the expression of the glucocorticoid receptor in human testicular tissue, including adult and fetal samples (n = 20) by immunohistochemical staining, and in silico analysis of publicly available datasets. In the adult testis NR3C1 protein was detected in peritubular cells, a subset of Leydig cells, Sertoli cells (weak), and spermatogonia, but not in spermatids. The NR3C1 expression pattern in fetal testis samples differed by a notably stronger reaction in Sertoli cells, lack of staining in gonocytes but the presence in a subset of pro-spermatogonia, and the almost absent reaction in nascent peritubular cells. In parallel, we explored the association between adult testicular function and three single nucleotide NR3C1 polymorphisms (BcII [rs41423247], 9ß [rs6198], and Tth111I [rs10052957]) affecting glucocorticoid sensitivity. Testicular function was determined by semen analysis and reproductive hormone profiling in 893 men from the general population. The NR3C1 SNP BclI was associated with semen quality in an over-dominant manner with heterozygotes having better semen parameters compared to both homozygote constellations, and with sperm motility showing the strongest association. This association was supported by a higher inhibin B and inhibin B/FSH ratio, as well as a lower FSH in BclI heterozygotes. The SNPs 9ß and Tth111I were not associated with semen parameters. Although the clinical impact of the findings is limited, the results substantiate a suggested link between stress and testicular function. Hence this investigation should be regarded as a discovery study generating hypotheses for future studies.


Subject(s)
Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Semen Analysis , Testis/metabolism , Adolescent , Adult , Fetus , Genotype , Humans , Male , Polymorphism, Single Nucleotide , Young Adult
6.
Andrology ; 3(1): 19-26, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25538016

ABSTRACT

Basic research results can provide new ideas and hypotheses to be examined in epidemiological studies. We conducted a survey among testicular cancer researchers on hypotheses concerning the etiology of this malignancy. All researchers on the mailing list of Copenhagen Testis Cancer Workshops and corresponding authors of PubMed-indexed articles identified by the search term 'testicular cancer' and published within 10 years (in total 2750 recipients) were invited to respond to an e-mail-based survey. Participants of the 8th Copenhagen Testis Cancer Workshop in May 2014 were subsequently asked to rate the plausibility of the suggested etiologic hypotheses on a scale of 1 (very implausible) to 10 (very plausible). This report describes the methodology of the survey, the score distributions by individual hypotheses, hypothesis group, and the participants' major research fields, and discuss the hypotheses that scored as most plausible. We also present plans for improving the survey that may be repeated at a next international meeting of experts in testicular cancer. Overall 52 of 99 (53%) registered participants of the 8th Copenhagen Testis Cancer Workshop submitted the plausibility rating form. Fourteen of 27 hypotheses were related to exposures during pregnancy. Hypotheses with the highest mean plausibility ratings were either related to pre-natal exposures or exposures that might have an effect during pregnancy and in post-natal life. The results of the survey may be helpful for triggering more specific etiologic hypotheses that include factors related to endocrine disruption, DNA damage, inflammation, and nutrition during pregnancy. The survey results may stimulate a multidisciplinary discussion about new etiologic hypotheses of testicular cancer.


Subject(s)
Research Personnel/psychology , Testicular Neoplasms/etiology , Consensus , Female , Humans , Male , Pregnancy , Prenatal Exposure Delayed Effects , Risk Assessment , Risk Factors , Surveys and Questionnaires
7.
Ann Oncol ; 26(4): 737-742, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25542924

ABSTRACT

BACKGROUND: Screening programmes for contralateral carcinoma in situ (CIS) testis in patients with unilateral germ-cell cancer (GCC) have never been evaluated. We investigated the effect of screening for contralateral CIS in a large nation-wide, population-based study. PATIENTS AND METHODS: A contralateral single-site biopsy was offered to 4130 patients in whom GCC had been diagnosed in 1984-2007 (screened cohort); 462 patients in whom GCC was diagnosed in 1984-1988 comprised the unscreened cohort. Cases with CIS were offered radiotherapy. Initially CIS-negative biopsies in patients with metachronous GCC were revised according to today's standards. Risk for metachronous GCC was estimated using cumulative incidence and the Cox proportional hazards model. RESULTS: In the screened cohort, contralateral CIS was found in 181 (4.4%) patients. The cumulative incidence of metachronous GCC after 20 years was 1.9% in the screened cohort and 3.1% in the unscreened cohort (P = 0.097), hazard ratio (HR) for the unscreened cohort: 1.59 (P = 0.144). Expert revision with contemporary methodology of CIS-negative biopsy samples from patients with metachronous cancer revealed CIS in 17 out of 45 (38%) cases. Decreased risks for metachronous GCC were related to older age at diagnosis (HR 0.52 per 10 years, P < 0.001) and chemotherapy (HR 0.35, P = 0.002). Limitations include the small number of patients in the unscreened cohort and the retrospective study design. CONCLUSIONS: Our evaluation of a national population-based screening programme for contralateral CIS in patients with testicular cancer showed no significant difference in the risk for metachronous GCC between a screened and an unscreened cohort. Single-site biopsy including modern immunohistochemistry does not identify all cases of CIS.


Subject(s)
Carcinoma in Situ/diagnosis , Carcinoma in Situ/epidemiology , Early Detection of Cancer , Neoplasms, Germ Cell and Embryonal/epidemiology , Neoplasms, Multiple Primary/diagnosis , Neoplasms, Multiple Primary/epidemiology , Testicular Neoplasms/epidemiology , Adult , Carcinoma in Situ/therapy , Cohort Studies , Combined Modality Therapy , Denmark/epidemiology , Female , Follow-Up Studies , Humans , Incidence , Male , Neoplasm Staging , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/therapy , Neoplasms, Multiple Primary/therapy , Prognosis , Risk Assessment , Testicular Neoplasms/pathology , Testicular Neoplasms/therapy
8.
J Assist Reprod Genet ; 31(5): 533-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24687878

ABSTRACT

MAIN PURPOSE AND RESEARCH QUESTION: To determine whether the true fusogen Syncytin-1 and its receptor (ASCT-2) is present in human gametes using qRT-PCR, immunoblotting and immunofluorescence. METHODS: Donated oocytes and spermatozoa, originating from a fertility center in tertiary referral university hospital, underwent qRT-PCR, immunoblotting and immunofluorescence analyzes. RESULTS: Quantitative RT-PCR of sperm samples from sperm donors showed that syncytin-1 is present in all samples, however, protein levels varied between donors. Syncytin-1 immunoreactivity predominates in the sperm head and around the equatorial segment. The receptor ASCT-2 is expressed in the acrosomal region and in the sperm tail. Moreover, ASCT-2, but not syncytin-1, is expressed in oocytes and the mRNA level increases with increasing maturity of the oocytes. CONCLUSIONS: Syncytin and its receptor are present in human gametes and localization and temporal appearance is consistent with a possible role in fusion between oocyte and sperm.


Subject(s)
Amino Acid Transport System ASC/genetics , Gene Products, env/genetics , Oocytes/physiology , Pregnancy Proteins/genetics , Spermatozoa/physiology , Adult , Amino Acid Transport System ASC/metabolism , Female , Fertilization/physiology , Gene Expression Regulation, Developmental , Gene Products, env/metabolism , Humans , Male , Pregnancy Proteins/metabolism , Sperm Head/physiology
9.
Br J Cancer ; 110(3): 668-78, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24292451

ABSTRACT

BACKGROUND: Developmental arrest of fetal germ cells may lead to neoplastic transformation and formation of germ cell tumours via carcinoma in situ (CIS) cells. Normal fetal germ cell development requires complete erasure and re-establishment of DNA methylation. In contrast to normal spermatogonia, the genome of CIS cells remains unmethylated in the adult testis. We here investigated the possible active and passive pathways that can sustain the CIS genome hypomethylated in the adult testis. METHODS: The levels of 5-methyl-cytosine (5mC) and 5-hydroxy-methyl-cytosine (5hmC) in DNA from micro-dissected CIS cells were assessed by quantitative measurements. The expression of TET1, TET2, APOBEC1, MBD4, APEX1, PARP1, DNMT1, DNMT3A, DNMT3B and DNMT3L in adult testis specimens with CIS and in human fetal testis was investigated by immunohistochemistry and immunofluorescence. RESULTS: DNA from micro-dissected CIS cells contained very low levels of 5hmC produced by ten eleven translocation (TET) enzymes. CIS cells and fetal germ cells expressed the suggested initiator of active demethylation, APOBEC1, and the base excision repair proteins MBD4, APEX1 and PARP1, whereas TETs - the alternative initiators were absent. Both maintenance and de novo methyltransferases were detected in CIS cells. CONCLUSION: The data are consistent with the presence of an active DNA de-methylation pathway in CIS cells. The hypomethylated genome of CIS cells may contribute to phenotypic plasticity and invasive capabilities of this testicular cancer precursor.


Subject(s)
Carcinoma in Situ/genetics , DNA Methylation/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Testicular Neoplasms/genetics , Carcinoma in Situ/pathology , Cell Differentiation , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Fetus/metabolism , Fetus/pathology , Genome, Human , Humans , Immunohistochemistry , Male , Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/pathology , Testis/metabolism , Testis/pathology
10.
Cytogenet Genome Res ; 142(2): 107-11, 2014.
Article in English | MEDLINE | ID: mdl-24335095

ABSTRACT

Tortoiseshell coat color is normally restricted to female cats due to X-linkage of the gene that encodes the orange coat color. Tortoiseshell male cats do, however, occur at a low frequency among tortoiseshell cats because of chromosome aberrations similar to the Klinefelter syndrome in man: the extra X chromosome of a 39,XXY karyotype introduces the possibility of an orange and a non-orange allele which produce the mixture of orange and non-orange coat spotting known as tortoiseshell. We analyzed the chromosome complement of a fibroblast culture and did histological examinations of testicular tissue from a tortoiseshell male cat referred to us. Chromosome analysis using RBA-banding consistently revealed a 39,XXY karyotype. Histological examinations of testis biopsies from this cat showed degeneration of the tubules, hyperplasia of the interstitial tissue, and complete loss of germ cells. Immunostaining using anti-vimentin and anti-VASA (DDX4) showed that only Sertoli cells and no germ cells were observed in the testicular tubules. As no sign of spermatogenesis was detected, we conclude that this is a classic case of a sterile, male tortoiseshell cat with a 39,XXY chromosome complement.


Subject(s)
Cats/genetics , Chromosome Aberrations/veterinary , Hair Color/genetics , Klinefelter Syndrome/veterinary , Testis/pathology , Animals , DEAD-box RNA Helicases/immunology , Germ Cells/cytology , Karyotype , Klinefelter Syndrome/genetics , Male , Sertoli Cells/pathology , Spermatogenesis/genetics , Vimentin/immunology
11.
Andrology ; 1(4): 615-23, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23606456

ABSTRACT

In the basic clinical work-up of infertile couples, a semen analysis is mandatory and the sperm concentration is one of the most essential variables to be determined. Sperm concentration is usually assessed by manual counting using a haemocytometer and is hence labour intensive and may be subjected to investigator bias. Here we show that image cytometry can be used to accurately measure the sperm concentration of human semen samples with great ease and reproducibility. The impact of several factors (pipetting, mixing, round cell content, sperm concentration), which can influence the read-out as well as inter-operator and -cytometer variation on two different image cytometers (NC-3000 and SP-100) were evaluated. Furthermore, 725 semen samples were assessed both by manual assessment (WHO recommended method) and by image cytometry and tight correlations between the measured concentrations were shown. Moreover, by evaluation of repeated measurements it appeared that image cytometry produced more consistent and accurate measurements than manual counting of human spermatozoa concentration. In conclusion, image cytometry provides an appealing substitute of manual counting by providing reliable, robust and easy measurement of human sperm concentration.


Subject(s)
Image Cytometry , Semen/cytology , Sperm Count/methods , Automation, Laboratory , Equipment Design , Humans , Image Cytometry/instrumentation , Linear Models , Male , Observer Variation , Predictive Value of Tests , Reproducibility of Results , Sperm Count/instrumentation
12.
Andrologia ; 44(2): 78-85, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21486421

ABSTRACT

Prompted by the recently reported expression of POU5F1 (OCT3/4) in epididymis, a panel of markers for carcinoma in situ (CIS) testis and testicular germ cell tumours (TGCT), including AP-2γ(TFAP2C), NANOG, OCT3/4, KIT, placental-like alkaline phosphatase (PLAP), M2A/PDPN and MAGE-A4 were examined by immunohistochemistry or in situ hybridisation in urogenital epithelia, which may interfere with detection of CIS cells in semen. In addition to OCT3/4, the expression of AP-2γ and NANOG or their variants was detected in urogenital epithelia, while other CIS markers, including PLAP/alkaline phosphatase were absent. A combination of immunocytological staining for AP-2γ or OCT3/4 and rapid cytochemical alkaline phosphatase reaction was subsequently developed. This approach was tested in 22 patients with TGCT. In 14 patients (63.6%), double stained cells were found and thus the method was proven suitable for the detection of CIS cells in semen. In conclusion, transcription factors related to pluripotency and undifferentiated state of cells, which most likely have several variants or modifications, are unexpectedly detected using currently available antibodies in urogenital epithelial cells which may be shed into semen. Combining the immunohistochemical nuclear markers with a rapid cytochemical alkaline phosphatase reaction for detection of CIS cells in ejaculates may provide a more reliable diagnostic method.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma in Situ/diagnosis , Homeodomain Proteins/analysis , Neoplasms, Germ Cell and Embryonal/diagnosis , Semen/chemistry , Staining and Labeling/methods , Testicular Neoplasms/diagnosis , Transcription Factor AP-2/analysis , Alkaline Phosphatase/analysis , Humans , Immunohistochemistry , Isoenzymes/analysis , Male , Nanog Homeobox Protein , Octamer Transcription Factor-3/analysis , Semen/cytology , Testis/enzymology
13.
Int J Androl ; 34(4 Pt 2): e21-30; discussion e30-1, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21696398

ABSTRACT

Testicular cancer (TC) is usually diagnosed after manifestation of an overt tumour. Tumour formation is preceded by a pre-invasive and asymptomatic stage, carcinoma in situ (CIS) testis, except for very rare subtypes. The CIS cells are located within seminiferous tubules but can be exfoliated and detected in ejaculates with specific CIS markers. We have built a high throughput framework involving automated immunocytochemical staining, scanning microscopy and in silico image analysis allowing automated detection and grading of CIS-like stained objects in semen samples. In this study, 1175 ejaculates from 765 subfertile men were tested using this framework. In 5/765 (0.65%) cases, CIS-like cells were identified in the ejaculate. Three of these had bilateral testicular biopsies performed and CIS was histologically confirmed in two. In total, 63 bilateral testicular biopsy were performed in conjunction with analysis of the ejaculates because of infertility work-up. Histological analysis of the biopsies for the presence of CIS yielded a test sensitivity of 0.67 and a specificity of 0.98. In addition, ejaculates from 45 patients with clinical signs of an overt TC were investigated and yielded a slightly lower sensitivity (0.51), possibly because of obstruction. We conclude that this novel non-invasive test combining automated immunocytochemistry and advanced image analysis allows identification of TC at the CIS stage with a high specificity, but a negative test does not completely exclude CIS. On the basis of the results, we propose that the assay could be offered to subfertile men and other patients who are at increased risk of TC.


Subject(s)
Carcinoma in Situ/diagnosis , Diagnostic Imaging/methods , Infertility, Male/pathology , Neoplasms, Germ Cell and Embryonal/diagnosis , Semen Analysis/methods , Testicular Neoplasms/diagnosis , Adult , Alkaline Phosphatase/analysis , Biopsy , Carcinoma in Situ/pathology , Cells, Cultured , High-Throughput Screening Assays/methods , Humans , Male , Microscopy , Neoplasms, Germ Cell and Embryonal/pathology , Semen/cytology , Staining and Labeling/methods , Testicular Neoplasms/pathology
14.
Br J Cancer ; 103(8): 1269-76, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20823885

ABSTRACT

BACKGROUND: The majority of testicular germ cell cancers develop through a pre-invasive carcinoma in situ (CIS) stage. The CIS cell is a neoplastic counterpart of foetal germ cells. During their development, foetal germ cells undergo extensive and essential epigenetic modifications, but little is known about epigenetic patterns in CIS cells. METHODS: Immunohistochemistry was used to investigate epigenetic patterns in CIS, germ cell tumours, normal adult and foetal testicular tissue. RESULTS: CIS cells show low levels of DNA methylation and repressive histone modifications H3K9me2 and H3K27me3, but high levels of H3K9 acetylation, H3K4 methylation and H2A.Z, which all are associated with an activated and accessible chromatin structure. Collectively this renders a permissive chromatin structure and in accordance high levels of RNA polymerase II activity and proliferation (Ki-67 and mitotic index) is observed in CIS cells. Epigenetic patterns similar to that of CIS cells were observed in human gonocytes present within sex cords in foetal testes but correspond to migrating primordial germ cell in mice. Development of overt tumours involves epigenetic repression of the chromatin. CONCLUSION: CIS cells have a permissive and foetal-like chromatin structure, which is associated with a high transcriptional and proliferative activity, likely empowering neoplastic transformation. Developmental epigenetic cues in foetal germ cells are substantially different between humans and mice.


Subject(s)
Carcinoma in Situ/metabolism , Chromatin/metabolism , Fetus/metabolism , Germ Cells/metabolism , Testicular Neoplasms/metabolism , Adult , Carcinoma in Situ/pathology , Cell Proliferation , Chromatin Assembly and Disassembly/physiology , DNA Methylation , Epigenesis, Genetic/physiology , Gestational Age , Histones/metabolism , Humans , Male , Neoplasms, Germ Cell and Embryonal/metabolism , Protein Processing, Post-Translational , Puberty/metabolism , Testicular Neoplasms/pathology , Transcriptional Activation
15.
Int J Androl ; 33(1): e207-15, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19780863

ABSTRACT

The aim of this study was to investigate endothelial lipase (EL, LIPG) and lipoprotein lipase (LPL) mRNA and protein expression in normal human testis and testicular germ cell tumours (GCT). Both EL and LPL were expressed in normal seminiferous tubules and in the interstitial compartment. EL mRNA and protein were found in all germ cells as well as in Sertoli and Leydig cells. EL mRNA was abundant in pre-invasive carcinoma in situ (CIS) cells and GCTs, and EL protein was present in the cytoplasm of these cells. LPL mRNA was also relatively abundant in germ cells, Sertoli cells, CIS cells and GCTs. The LPL protein, however, was restricted to the cell membranes of pachytene spermatocytes and spermatids in normal tubules, absent from CIS cells and scarcely represented in tumours. The distribution of LPL protein in non-seminomas resembled the distribution of OCT3/4, a marker of embryonal carcinoma. The results suggest that both EL and LPL participate in the supply of nutrients and steroidogenesis in the testes, and that especially EL may be important for the supply of cholesterol for testosterone production in the Leydig cells. The partial cellular separation of the expression of the two lipases in normal testis suggests the existence of distinct biological roles, perhaps developmentally regulated, as indicated by the LPL expression in GCTs with embryonic features. A high expression of EL and abundance of lipid in tubules with CIS may have a diagnostic value.


Subject(s)
Lipase/genetics , Lipoprotein Lipase/genetics , Testicular Neoplasms/metabolism , Testis/metabolism , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Endothelium/metabolism , Germ Cells/metabolism , Humans , Leydig Cells/cytology , Leydig Cells/metabolism , Leydig Cells/pathology , Lipase/metabolism , Lipoprotein Lipase/metabolism , Male , Neoplasms, Germ Cell and Embryonal/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Nitric Oxide Synthase Type III , RNA, Messenger/metabolism , Seminiferous Tubules/metabolism , Seminoma/metabolism , Seminoma/pathology , Sertoli Cells/cytology , Sertoli Cells/metabolism , Sertoli Cells/pathology , Spermatocytes/metabolism , Testicular Neoplasms/pathology , Testis/cytology
16.
Histopathology ; 47(1): 48-56, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15982323

ABSTRACT

AIMS: NANOG is a key regulator of embryonic stem cell (ESC) self-renewal and pluripotency. Our recent genome-wide gene expression profiling study of the precursor of testicular germ cell tumours, carcinoma in situ testis (CIS), showed close similarity between ESC and CIS, including high NANOG expression. In the present study we analysed the protein expression of NANOG during normal development of human testis and in a large series of neoplastic/dysgenetic specimens. METHODS AND RESULTS: We detected abundant expression of NANOG in CIS and in CIS-derived testicular tumours with marked differences; seminoma and embryonal carcinoma were strongly positive, differentiated somatic elements of teratoma were negative. We provide evidence for the fetal origin of testicular cancer as we detected strong expression of NANOG in fetal gonocytes up to gestational week 20, with subsequent down-regulation occurring earlier than for OCT-4. We detected no expression at the protein level in normal testis. CONCLUSIONS: NANOG is a new marker for testicular CIS and germ cell tumours and the high level of NANOG along with OCT-4 are determinants of the stem cell-like pluripotency of the preinvasive CIS cell. Timing of NANOG down-regulation in fetal gonocytes suggests that NANOG may act as a regulatory factor up-stream to OCT-4.


Subject(s)
Carcinoma in Situ/pathology , DNA-Binding Proteins/analysis , Germinoma/pathology , Homeodomain Proteins/analysis , Testicular Neoplasms/pathology , Adolescent , Adult , Alkaline Phosphatase , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Child , Child, Preschool , DNA-Binding Proteins/genetics , Fetus , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Germinoma/genetics , Germinoma/metabolism , Gestational Age , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Isoenzymes/analysis , Male , Nanog Homeobox Protein , Octamer Transcription Factor-3 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Transcription Factor AP-2 , Transcription Factors/analysis
17.
Br J Cancer ; 92(10): 1934-41, 2005 May 23.
Article in English | MEDLINE | ID: mdl-15856041

ABSTRACT

The carcinoma in situ (CIS) cell is the common precursor of nearly all testicular germ cell tumours (TGCT). In a previous study, we examined the gene expression profile of CIS cells and found many features common to embryonic stem cells indicating that initiation of neoplastic transformation into CIS occurs early during foetal life. Progression into an overt tumour, however, typically first happens after puberty, where CIS cells transform into either a seminoma (SEM) or a nonseminoma (N-SEM). Here, we have compared the genome-wide gene expression of CIS cells to that of testicular SEM and a sample containing a mixture of N-SEM components, and analyse the data together with the previously published data on CIS. Genes showing expression in the SEM or N-SEM were selected, in order to identify gene expression markers associated with the progression of CIS cells. The identified markers were verified by reverse transcriptase-polymerase chain reaction and in situ hybridisation in a range of different TGCT samples. Verification showed some interpatient variation, but combined analysis of a range of the identified markers may discriminate TGCT samples as SEMs or N-SEMs. Of particular interest, we found that both DNMT3B (DNA (cytosine-5-)-methyltransferase 3 beta) and DNMT3L (DNA (cytosine-5-)-methyltransferase 3 like) were overexpressed in the N-SEMs, indicating the epigenetic differences between N-SEMs and classical SEM.


Subject(s)
Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Germinoma/genetics , Germinoma/pathology , Seminoma/genetics , Seminoma/pathology , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/genetics , Genome , Humans , In Situ Hybridization , Male , Oligonucleotide Array Sequence Analysis , Puberty , Reverse Transcriptase Polymerase Chain Reaction
18.
Hum Reprod ; 20(3): 579-82, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15650041

ABSTRACT

The incidence of testicular cancer is rising. Despite a high cure rate, efforts should be made to obtain diagnosis at the pre-invasive intratubular carcinoma in situ (CIS) stage, as the disease is potentially lethal and treatment has severe side-effects, especially regarding reproductive function. CIS diagnosis is presently only possible by a surgical biopsy of the testis. Immunocytological staining for transcription factor activator protein (AP-2gamma), previously identified as a marker for neoplastic germ cells, was performed in centrifuged samples of ejaculates obtained from 104 andrological patients, including patients with testicular cancer and subfertility. Cells positive for AP-2gamma were found only in semen samples from patients diagnosed a priori with testicular neoplasms and, surprisingly, in a 23 year old control subject with oligozoospermia and no symptoms of a germ cell tumour. Testicular biopsies performed during the follow-up of this patient revealed widespread CIS in one testicle, thus proving a potential diagnostic value of the new marker. For the first time, a patient without clinical symptoms of testicular neoplasia was diagnosed at the pre-invasive CIS stage using a new, simple method based on immunocytological staining of a semen sample for AP-2gamma, a novel marker for CIS. The value of this method for diagnostic use in the clinic requires further careful validation in a large series of patients and controls, but the preliminary results are promising.


Subject(s)
Carcinoma in Situ/complications , Carcinoma in Situ/diagnosis , DNA-Binding Proteins/analysis , Infertility, Male/complications , Semen/chemistry , Testicular Neoplasms/complications , Testicular Neoplasms/diagnosis , Transcription Factors/analysis , Adult , Biomarkers, Tumor/analysis , Carcinoma in Situ/pathology , Humans , Immunohistochemistry/methods , Male , Neoplasm Staging , Oligospermia/complications , Staining and Labeling , Testicular Neoplasms/pathology , Transcription Factor AP-2
19.
Mol Hum Reprod ; 10(6): 423-31, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15123780

ABSTRACT

Virtually all testicular germ cell tumours originate from a common precursor, the carcinoma in situ (CIS) cell. The precise nature of the molecular mechanisms leading to CIS remains largely unknown. We performed the first systematic analysis of gene expression in testis with CIS compared to normal testis by the differential display (DDRT-PCR) method, with subsequent analysis by RT-PCR and in situ hybridization (ISH). In tissue containing CIS we identified overexpression of 28 mRNA, some previously reported in CIS and a number of genes not previously described in germ cell neoplasia, including the novel expressed sequence tag (EST) OIC1 (Overexpressed In CIS). The genes could be grouped functionally into genes involved in cell growth, proliferation, differentiation, immunological response, and genes with unknown biological function. Examples of overexpressed genes are SFRP1 that is involved in Wnt signalling and IGFBP6, which is of importance for fetal growth and inhibits cell growth through insulin-like growth factor-II. ISH analysis showed that both mRNA were localized to CIS cells. The results of our search for differentially expressed genes in CIS demonstrated a number of genes linked to testicular development (e.g. DCN, IGFBP6, SFRP1, SALL1), supporting our hypothesis that the origin of CIS is probably associated with disturbances of the fetal development of the testis.


Subject(s)
Carcinoma in Situ/genetics , Gene Expression Regulation, Neoplastic , Testicular Neoplasms/genetics , Testis/pathology , Testis/physiology , Adolescent , Adult , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Gene Expression Profiling , Humans , In Situ Hybridization , Male , Middle Aged , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology
20.
Eur J Histochem ; 47(3): 215-22, 2003.
Article in English | MEDLINE | ID: mdl-14514412

ABSTRACT

LANCL1 (LanC-like protein 1) is related to the bacterial LanC (lanthionine synthetase C) family, which is involved in the biosynthesis of antimicrobial peptides. Highest expression levels of LANCL1 are found in testes and brain, two organs that exist behind blood-tissue barriers. In the mouse, the establishment of an impermeable blood-testis barrier occurs between day 10-16 post natal (pn). Differential display analysis showed that the expression level of LANCL1 mRNA in mouse testes was very low until day 16 pn, but increased gradually from day 16 pn to reach a maximum on days 22-24 pn followed by a slight reduction from day 26 pn to adult animals. Thus, the expression of LANCL1 mRNA is initiated following the establishment of the blood-testis barrier. In situ hybridisation revealed that LANCL1 mRNA was induced in diplotene spermatocytes, which appear for the first time in mouse testes between days 18 and 20 pn, verifying the expression profile determined by differential display. LANCL1 mRNA level remained high in the meiotic division phase and in early round spermatids, but was down regulated in elongating spermatids and it was undetectable in step 9 elongating spermatids in stage IX (as defined by Russel et al., 1990). The steady decrease in expression level from round spermatids in stage I to elongating spermatids in stage IX suggested that LANCL1 mRNA was not transcribed in spermatids. LANCL1 expression in rat testes was initiated already in pachytene spermatocytes in stage IX, but otherwise similar to mouse.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Germ Cells/cytology , Germ Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Testis/cytology , Testis/metabolism , Animals , In Situ Hybridization , In Situ Hybridization, Fluorescence , Male , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Testis/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL