Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Andrology ; 7(4): 463-468, 2019 07.
Article in English | MEDLINE | ID: mdl-30786164

ABSTRACT

BACKGROUND: The microRNA-371a-3p (miR-371a-3p) has been reported to be an informative liquid biopsy (serum and plasma) molecular biomarker for both diagnosis and follow-up of patients with a malignant (testicular) germ cell tumor ((T)GCT). It is expressed in all histological cancer elements, with the exception of mature teratoma. However, normal testis, semen, and serum of males with a disrupted testicular integrity without a TGCT may contain miR-371a-3p levels above threshold, of which the cellular origin is unknown. OBJECTIVES: Therefore, a series of relevant tissues (frozen and formalin-fixed paraffin-embedded (FFPE), when available) from the complete male urogenital tract (i.e., kidney to urethra and testis to urethra) and semen was investigated for miR-371a-3p levels using targeted quantitative RT-PCR (qRT-PCR). MATERIALS AND METHODS: In total, semen of males with normospermia (n = 11) and oligospermia (n = 3) was investigated, as well as 88 samples derived from 32 different patients. The samples represented one set of tissues related to the entire male urogenital tract (11 anatomical locations), three sets for 10 locations, and four sets for six locations. RESULTS: All testis parenchyma (n = 17) cases showed low miR-371a-3p levels. Eight out of 14 (57%) semen samples showed detectable miR-371a-3p levels, irrespective of the amount of motile spermatozoa, but related to sperm concentration and matched Johnsen score (Spearman's rho correlation coefficient 0.849 and 0.871, p = 0.000, respectively). In all other tissues investigated, miR-371a-3p could not be detected. DISCUSSION: This study demonstrates that the miR-371a-3p in healthy adult males is solely derived from the germ cell compartment. CONCLUSIONS: The observation is important in the context of applying miR-371a-3p as molecular liquid biopsy biomarker for diagnosis and follow-up of patients with malignant (T)GCT. Moreover, miR-371a-3p might be an informative seminal biomarker for testicular germ cell composition.


Subject(s)
Genitalia, Male/metabolism , MicroRNAs/metabolism , Semen/metabolism , Urinary Tract/metabolism , Humans , Male , Oligospermia/metabolism , Reference Values
2.
Hum Reprod ; 32(12): 2561-2573, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29121256

ABSTRACT

STUDY QUESTION: What is the prevalence of malignant testicular germ cell tumors (TGCT) and its precursors, (pre-) germ cell neoplasia in situ (GCNIS), in late teenagers and adults who have androgen insensitivity syndrome (AIS) and the impact of an individual's genetic susceptibility to development of TGCT? SUMMARY ANSWER: No GCNIS or TGCT was diagnosed, but pre-GCNIS was identified in 14 and 10% of complete and partial AIS patients, respectively, and was associated with a higher genetic susceptibility score (GSS), with special attention for KITLG (rs995030) and ATFZIP (rs2900333). WHAT IS KNOWN ALREADY: Many adult women with AIS decline prophylactic gonadectomy, while data regarding the incidence, pathophysiology and outcomes of TGCT in postpubertal individuals with AIS are lacking. The relevance of genetic factors, such as single nucleotide polymorphisms (SNPs), in predisposing AIS individuals to TGCT is unknown. STUDY DESIGN, SIZE, DURATION: This multicenter collaborative study on prophylactically removed gonadal tissue was conducted in a pathology lab specialized in germ cell tumor biology. PARTICIPANTS/MATERIALS, SETTING, METHODS: Material from 52 postpubertal individuals with molecularly confirmed AIS (97 gonadal samples) was included; the median age at surgery was 17.5 (14-54) years. Immunohistochemical studies and high-throughput profiling of 14 TGCT-associated SNPs were performed. The main outcome measures were the prevalence of pre-GCNIS, GCNIS and TGCT, and its correlation with a GSS, developed based on the results of recent genome-wide association studies. MAIN RESULTS AND ROLE OF CHANCE: The earliest recognizable change preceding GCNIS, referred to as pre-GCNIS, was present in 14% of individuals with complete and 10% of those with partial AIS at a median age of 16 years. No GCNIS or invasive TGCT were found. The median GSS was significantly greater for those with, compared to those without, pre-GCNIS (P = 0.01), with an overlap between groups. Our data suggest important roles for risk alleles G at KITLG (rs995030) and C at ATFZIP (rs2900333), among the 14 studied TGCT-associated SNPs. LARGE SCALE DATA: N/A. LIMITATIONS REASONS FOR CAUTION: A limited number of cases were included. WIDER IMPLICATIONS OF THE FINDINGS: Our data suggest that the prevalence of pre-GCNIS in individuals with AIS beyond puberty is around 15%. Genetic susceptibility likely contributes to pre-GCNIS development in AIS but factors related to malignant progression remain unclear. Although data in older patients remain scarce, malignant progression appears to be a rare event, although the natural history of the premalignant lesion remains unknown. Therefore, the practice of routine prophylactic gonadectomy in adults with AIS appears questionable and the patient's preference, after having been fully informed, should be decisive in this matter. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by research grants from the Research Foundation Flanders (FWO) (to M.C.), the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq G0D6713N) (to B.B.M. and M.C.) and the European Society for Pediatric Endocrinology (ESPE), granted by Novo Nordisk AB (to J.K.). There are no competing interests.


Subject(s)
Androgen-Insensitivity Syndrome/diagnosis , Androgen-Insensitivity Syndrome/genetics , Neoplasms, Germ Cell and Embryonal/diagnosis , Neoplasms, Germ Cell and Embryonal/genetics , Polymorphism, Single Nucleotide , Testicular Neoplasms/diagnosis , Testicular Neoplasms/genetics , Adolescent , Adult , Alleles , Androgen-Insensitivity Syndrome/complications , Androgen-Insensitivity Syndrome/epidemiology , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Middle Aged , Neoplasms, Germ Cell and Embryonal/complications , Neoplasms, Germ Cell and Embryonal/epidemiology , Phenotype , Prevalence , Sexual Maturation , Stem Cell Factor/genetics , Testicular Neoplasms/complications , Testicular Neoplasms/epidemiology , Young Adult
3.
Andrology ; 3(1): 85-91, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25382245

ABSTRACT

microRNAs (miRs) are short non-coding RNA molecules (≈21 nucleotides) involved in regulation of translation. As such they are crucial for normal cell development and differentiation as well as cellular maintenance. Dysregulation of miRs has been reported in various diseases, including cancer. Interestingly, miRs can be informative as tumor classifiers and disease biomarkers. Recent studies demonstrated the presence of miRs in body fluids like serum, thus providing a putative non-invasive tool to study and monitor disease state. Earlier targeted studies by several independent groups identified specific embryonic miRs as characteristic for germ cell tumors (GCT) (miR-371-2-3 & miR-302/367 clusters). This study reports a high-throughput miR profiling (≈750 miRs) approach on serum from testicular germ cell tumor patients (14 seminoma and 10 non-seminoma) and controls (n = 11), aiming at independent identification of miRs as candidate biomarkers for testicular GCT. A magnetic bead capture system was used to isolate miRs from serum. Subsequently, the TaqMan Array Card 3.0 platform was used for profiling. The previously identified miRs 371 and 372 were confirmed to be specifically elevated in serum from germ cell tumor patients. In addition, several novels miRs were identified that were discriminative between germ cell cancer and controls: miR-511, -26b, -769, -23a, -106b, -365, -598, -340, and let-7a. In conclusion, this study validates the power of the embryonic miRs 371 and 372 in detecting malignant GCT (SE and NS) based on serum miR levels and identifies several potential novel miR targets.


Subject(s)
Biomarkers, Tumor/blood , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , MicroRNAs/blood , Neoplasms, Germ Cell and Embryonal/blood , Seminoma/blood , Testicular Neoplasms/blood , Base Sequence , Biomarkers, Tumor/genetics , Case-Control Studies , Discriminant Analysis , Humans , Male , MicroRNAs/genetics , Molecular Sequence Data , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Predictive Value of Tests , Seminoma/genetics , Seminoma/pathology , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology
4.
Br J Cancer ; 105(6): 854-63, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21847120

ABSTRACT

BACKGROUND: OCT3/4 (POU5F1) is an established diagnostic immunohistochemical marker for specific histological variants of human malignant germ cell tumours (GCTs), including the seminomatous types and the stem cell component of non-seminomas, known as embryonal carcinoma. OCT3/4 is crucial for the regulation of pluripotency and the self-renewal of normal embryonic stem- and germ cells. Detection of expression of this transcription factor is complicated by the existence of multiple pseudogenes and isoforms. Various claims have been made about OCT3/4 expression in non-GCTs, possibly related to using nonspecific detection methods. False-positive findings undermine the applicability of OCT3/4 as a specific diagnostic tool in a clinical setting. In addition, false-positive findings could result in misinterpretation of pluripotency regulation in solid somatic cancers and their stem cells. Of the three identified isoforms--OCT4A, OCT4B and OCT4B1--only OCT4A proved to regulate pluripotency. Up until now, no convincing nuclear OCT4A protein expression has been shown in somatic cancers or tissues. METHODS: This study investigates expression of the various OCT3/4 isoforms in GCTs (both differentiated and undifferentiated) and somatic (non-germ cell) cancers, including representative cell lines and xenografts. RESULTS: Using specific methods, OCT4A and OCT4B1 are shown to be preferentially expressed in undifferentiated GCTs. The OCT4B variant shows no difference in expression between GCTs (either differentiated or undifferentiated) and somatic cancers. In spite of the presence of OCT4A mRNA in somatic cancer-derived cell lines, no OCT3/4 protein is detected. Significant positive correlations between all isoforms of OCT3/4 were identified in both tumours with and without a known stem cell component, possibly indicating synergistic roles of these isoforms. CONCLUSION: This study confirms that OCT4A protein only appears in seminomatous GCTs, embryonal carcinoma and representative cell lines. Furthermore, it emphasises that in order to correctly assess the presence of functional OCT3/4, both isoform specific mRNA and protein detection are required.


Subject(s)
Biomarkers, Tumor/metabolism , Octamer Transcription Factor-3/metabolism , Carcinoma, Embryonal/metabolism , Cell Line, Tumor , Humans , Male , Neoplasms, Germ Cell and Embryonal , Octamer Transcription Factor-3/genetics , Protein Isoforms/metabolism , Pseudogenes , RNA, Messenger/metabolism , Seminoma/metabolism , Sensitivity and Specificity
5.
Int J Androl ; 34(4 Pt 2): e160-74, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21631526

ABSTRACT

OCT3/4, NANOG, SOX2 and, most recently, LIN28 have been identified as key regulators of pluripotency in mammalian embryonic and induced stem cells, and are proven to be crucial for generation of the mouse germ-cell lineage. These factors are a hallmark of certain histological types of germ-cell tumours (GCTs). Here, we report novel information on the temporal and spatial expression pattern of LIN28 during normal human male germ-cell development as well as various types of GCTs. To investigate LIN28 expression, immunohistochemical analyses and quantitative proximity ligation assay-based TaqMan protein assays were applied on snap-frozen and formalin-fixed, paraffin-embedded samples as well as representative cell lines. LIN28 was found in primordial germ cells, gonocytes and pre-spermatogonia, in contrast to OCT3/4 and NANOG, which were found only in the first two stages. LIN28 was also found in all precursor lesions (carcinoma in situ and gonadoblastoma) of type II GCTs, as well as the invasive components seminoma and the non-seminomatous elements embryonal carcinoma and yolk sac tumour. Choriocarcinoma showed a heterogeneous pattern, while teratomas and spermatocytic seminomas (type III GCTs) were negative. This expression pattern suggests that LIN28 is associated with malignant behaviour of type II GCTs. Cell line experiments involving siRNA knockdown of LIN28, OCT3/4 and SOX2 showed that LIN28 plays a role in the maintenance of the undifferentiated state of both seminoma and embryonal carcinoma, closely linked to, and likely upstream of OCT3/4 and NANOG. In conclusion, LIN28 regulates the differentiation status of seminoma and embryonal carcinoma and is likely to play a related role in normal human germ-cell development.


Subject(s)
DNA-Binding Proteins/metabolism , Germ Cells/metabolism , Neoplasms, Germ Cell and Embryonal/metabolism , Testicular Neoplasms/pathology , Biomarkers, Tumor/analysis , Carcinoma in Situ/pathology , Carcinoma, Embryonal/pathology , Cell Differentiation , Cells, Cultured , Choriocarcinoma/pathology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endodermal Sinus Tumor/pathology , Germ Cells/chemistry , Gonadoblastoma , Homeodomain Proteins/biosynthesis , Humans , Male , Nanog Homeobox Protein , Neoplasms, Germ Cell and Embryonal/pathology , Octamer Transcription Factor-3/biosynthesis , Organic Cation Transport Proteins/biosynthesis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/biosynthesis , Seminoma/pathology , Spermatogonia , Testis/chemistry , Testis/metabolism , Testis/pathology
6.
Int J Androl ; 34(4 Pt 2): e189-203, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21649665

ABSTRACT

Malignant germ-cell tumours arise from a neoplastic precursor, the carcinoma in situ, and develop into seminomas and/or non-seminomas (embryonal carcinomas, teratomas, yolk-sac tumours and choriocarcinomas). Based on histological and clinical findings, it has been postulated that seminomas can eventually transform into non-seminomas. Here, we used the cell line TCam-2 as model for seminomas and interrogated their differentiation potential. We demonstrate that TCam-2 cells are able to differentiate into mixed non-seminomatous lineages after supplementing the media with TGF-ß1, EGF and FGF4. On a molecular level, the differentiation is initiated by repression of BMP/SMAD signalling. As a consequence, BLIMP1, a molecule known to inhibit the differentiation of murine primordial germ cells, is down-regulated and differentiation-inhibiting histone modifications are lost. The appearance of multinucleated giant cells and the expression of marker genes indicate that cells differentiate predominantly into extra-embryonic choriocarcinoma-like cells. This is most likely due to the presence of components of the Hippo pathway, TEAD4 and YAP1. These molecules have been described to trigger extra-embryonic fate determination in the murine system. This study supports the model that seminomas indeed have an intrinsic ability to transform into a non-seminoma. In addition, the data suggest that the transformation does not require an additional mutation, but can be triggered by changes in the tumour microenvironment.


Subject(s)
Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 4/pharmacology , Neoplasms, Germ Cell and Embryonal/pathology , Seminoma/pathology , Transforming Growth Factor beta1/pharmacology , Adaptor Proteins, Signal Transducing/biosynthesis , Biomarkers/metabolism , Bone Morphogenetic Protein Receptors/metabolism , Cell Differentiation , Cell Line, Tumor , Choriocarcinoma/embryology , DNA-Binding Proteins/biosynthesis , Epidermal Growth Factor/metabolism , Fibroblast Growth Factor 4/metabolism , Giant Cells , Histones/metabolism , Humans , Male , Muscle Proteins/biosynthesis , Polymerase Chain Reaction , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Signal Transduction , Smad Proteins/metabolism , TEA Domain Transcription Factors , Testicular Neoplasms , Transcription Factors/biosynthesis , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment
7.
Int J Androl ; 34(4 Pt 2): e234-51, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21564133

ABSTRACT

Human type II germ cell tumours (GCTs) originate from an embryonic germ cell, either as a primordial germ cell or gonocyte. This start determines the biological as well as clinical characteristics of this type of cancer, amongst others their totipotency as well as their overall (exceptional) sensitivity to DNA damaging agents. The histology of the precursor lesion, either carcinoma in situ or gonadoblastoma, depends on the level of testicularization (i.e. testis formation) of the gonad. The impact of either intrinsic (genetic) - and environmental factors involved in the pathogenesis is demonstrated by disorders of sex development as well as testicular dysgenesis syndrome as risk factors, including cryptorchidism, hypospadias and disturbed fertility as parameters. This knowledge allows identification of individuals at risk for development of this type of cancer, being a population of interest for screening. Factors known to regulate pluripotency during embryogenesis are proven to be of diagnostic value for type II GCTs, including OCT3/4, even applicable for non-invasive screening. In addition, presence of stem cell factor, also known as KITLG, allows distinction between delayed matured germ cells and the earliest stages of malignant transformation. This is of special interest because of the identified association between development of type II GCTs of the testis and a limited number of single nucleotide polymorphisms, including some likely related to KITL. Transition from the precursor lesion to an invasive cancer is associated with gain of the short arm of chromosome 12, in which multiple genes might be involved, including KRAS2 and possibly NANOG (pseudogenes). While most precursor lesions will progress to an invasive cancer, only a limited number of cancers will develop treatment resistance. Putative explanatory mechanisms are identified, including presence of microsatellite instability, BRAF mutations, apoptosis suppression and p21 sub-cellular localization. It remains to be investigated how these different pathways integrate to each other and how informative they are at the patient-individual level. Further understanding will allow development of more targeted treatment, which will benefit quality of life of these young cancer patients.


Subject(s)
Neoplasms, Germ Cell and Embryonal/metabolism , Signal Transduction , Testicular Neoplasms/metabolism , Biomarkers, Tumor/genetics , Carcinoma in Situ/genetics , Carcinoma in Situ/therapy , Humans , Male , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/therapy , Risk Factors , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology , Testicular Neoplasms/therapy
8.
J Pathol ; 216(1): 43-54, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18566970

ABSTRACT

Carcinoma in situ (CIS) of the testis is the pre-invasive stage of type II testicular germ cell tumours (TGCTs) of adolescents and adults. These tumours are the most frequently diagnosed cancer in Caucasian adolescents and young adults. In dysgenetic gonads, the precursor of type II GCTs can be either CIS or a lesion known as gonadoblastoma (GB). CIS/GB originates from a primordial germ cell (PGC)/gonocyte, ie an embryonic cell. CIS can be cured by local low-dose irradiation, with limited side effects on hormonal function. Therefore, strategies for early diagnosis of CIS are essential. Various markers are informative to diagnose CIS in adult testis by immunohistochemistry, including c-KIT, PLAP, AP-2gamma, NANOG, and POU5F1 (OCT3/4). OCT3/4 is the most informative and consistent in presence and expression level, resulting in intense nuclear staining. In the case of maturational delay of germ cells, frequently present in gonads of individuals at risk for type II (T)GCTs, use of these markers can result in overdiagnosis of malignant germ cells. This demonstrates the need for a more specific diagnostic marker to distinguish malignant germ cells from germ cells showing maturation delay. Here we report the novel finding that immunohistochemical detection of stem cell factor (SCF), the c-KIT ligand, is informative in this context. This was demonstrated in over 400 cases of normal (fetal, neonatal, infantile, and adult) and pathological gonads, as well as TGCT-derived cell lines, specifically in cases of CIS and GB. Both membrane-bound and soluble SCF were expressed, suggestive of an autocrine loop. SCF immunohistochemistry can be a valuable diagnostic tool, in addition to OCT3/4, to screen for precursor lesions of TGCTs, especially in patients with germ cell maturation delay.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma in Situ/diagnosis , Gonadoblastoma/diagnosis , Stem Cell Factor/analysis , Testicular Neoplasms/diagnosis , Adolescent , Adult , Biomarkers, Tumor/genetics , Gonadoblastoma/genetics , Gonadoblastoma/metabolism , Humans , Immunohistochemistry , Infant , Male , Reverse Transcriptase Polymerase Chain Reaction , Testicular Neoplasms/metabolism
9.
J Pathol ; 215(1): 21-30, 2008 May.
Article in English | MEDLINE | ID: mdl-18348160

ABSTRACT

Combined action of SOX and POU families of transcription factors plays major roles in embryonic development. In embryonic stem cells, the combination of SOX2 and POU5F1 (OCT3/4) is essential for maintaining the undifferentiated state by activating pluripotency-linked genes, and inhibition of genes involved in differentiation. Besides embryonic stem cells, POU5F1 is also present in early germ cells, primordial germ cells, and gonocytes, where it has a role in suppression of apoptosis. Here we demonstrate that SOX2 is absent in germ cells of human fetal gonads, and as expected carcinoma in situ (CIS), ie the precursor lesion of testicular germ cell tumours of adolescents and adults (TGCTs), and seminoma. Based on genome-wide expression profiling, SOX17 was found to be present, instead of SOX2, in early germ cells and their malignant counterparts, CIS and seminoma. Immunohistochemistry, western blot analysis, and quantitative RT-PCR showed that SOX17 is a suitable marker to distinguish seminoma from embryonal carcinoma, confirmed in representative cell lines. Aberrant SOX2 expression can be present in Sertoli cells when associated with CIS, which can be misdiagnosed as embryonal carcinoma. In conclusion, this study demonstrates the absence of SOX2 in human embryonic and malignant germ cells, which express SOX17 in conjunction with POU5F1. This finding has both diagnostic and developmental biological implications. It allows the identification of seminoma-like cells from embryonal carcinoma based on a positive marker and might be the explanation for the different function of POU5F1 in normal and malignant germ cells versus embryonic stem cells.


Subject(s)
DNA-Binding Proteins/genetics , Germ Cells/metabolism , HMGB Proteins/genetics , High Mobility Group Proteins/genetics , Stem Cells/metabolism , Transcription Factors/genetics , Adult , Biomarkers, Tumor/analysis , Blotting, Western/methods , Carcinoma in Situ/metabolism , Cell Line , Cell Line, Tumor , Female , Gene Expression , Gene Expression Profiling , Germinoma/metabolism , Humans , Immunohistochemistry , Male , Octamer Transcription Factor-3/genetics , Oligonucleotide Array Sequence Analysis , Ovary/embryology , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors , SOXF Transcription Factors , Seminoma/metabolism , Testicular Neoplasms/metabolism , Testis/embryology
10.
J Pathol ; 213(3): 319-28, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17893849

ABSTRACT

Testicular germ cell tumours (GCTs) of adolescents and adults can be subdivided into seminomas (referred to as dysgerminomas of the ovary) and non-seminomas, all referred to as type II GCTs. They originate from carcinoma in situ (CIS), being the malignant counterparts of primordial germ cells (PGCs)/gonocytes. The invasive components mimic embryogenesis, including the stem cell component embryonal carcinoma (EC), the somatic lineage teratoma (TE), and the extra-embryonic tissues yolk sac tumour (YST) and choriocarcinoma (CH). The other type is the so-called spermatocytic seminomas (SS, type III GCT), composed of neoplastic primary spermatocytes. We reported previously that the miRNAs hsa-miR 371-373 cluster is involved in overruling cellular senescence induced by oncogenic stress, allowing cells to become malignant. Here we report the first high-throughput screen of 156 microRNAs in a series of type II and III GCTs (n = 69, in duplicate) using a quantitative PCR-based approach. After normalization to allow inter-sample analysis, the technical replicates clustered together, and the previous hsa-miRNA 371-373 cluster finding was confirmed. Unsupervised cluster analysis demonstrated that the cell lines are different from the in vivo samples. The in vivo samples, both normal and malignant, clustered predominantly based on their maturation status. This parallels normal embryogenesis, rather than chromosomal anomalies in the tumours. miRNAs within a single cluster showed a similar expression pattern, implying common regulatory mechanisms. Normal testicular tissue expressed most discriminating miRNAs at a higher level than SE and SS. Moreover, differentiated non-seminomas showed overexpression of discriminating miRNAs. These results support the model that miRNAs are involved in regulating differentiation of stem cells, retained in GCTs.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , MicroRNAs/analysis , Neoplasms, Germ Cell and Embryonal/genetics , Oligonucleotide Array Sequence Analysis , Testicular Neoplasms/genetics , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cell Line, Tumor , Cluster Analysis , Endodermal Sinus Tumor/genetics , Endodermal Sinus Tumor/pathology , Female , Germinoma/genetics , Germinoma/pathology , Humans , Male , Neoplasms, Germ Cell and Embryonal/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Seminoma/genetics , Seminoma/pathology , Teratoma/genetics , Teratoma/pathology , Testicular Neoplasms/pathology
11.
Int J Androl ; 30(4): 304-14; discussion 314-5, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17573854

ABSTRACT

The dogma of genome functionality has recently been challenged by identification of non-protein-encoding RNAs, including mi(cro)RNAs. These relatively small sequences interact with mRNA and in the mammalian system, are involved in fine-tuning the process of translation. miRNAs have been found to be of crucial importance for normal development, including stem cell formation. Recent interesting fundamental observations will be discussed in this paper, as well as their impact on the genesis of human germ cell tumours (GCTs), in particular those of the adult testis, seminomas and non-seminomas (type II), and spermatocytic seminomas (type III). miRNA cluster 371-373 is specifically involved in inhibition of cellular senescence induced by oncogenic stress in the type II GCTs. This explains the unusual presence of wild type P53, characteristic of this type of solid cancer. Specific sets of differentiating miRNA were found to characterize the various differentiation lineages within the GCTs, which simulate normal embryonic development.


Subject(s)
MicroRNAs/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms/genetics , Testicular Neoplasms/genetics , DNA, Neoplasm/genetics , Genome, Human , Humans , Male , Mutation , RNA, Neoplasm/genetics
12.
Oncogene ; 22(12): 1880-91, 2003 Mar 27.
Article in English | MEDLINE | ID: mdl-12660824

ABSTRACT

Gain of 12p material is invariably associated with testicular germ cell tumors (TGCTs) of adolescents and adults, most usually as an isochromosome 12p. We analyzed TGCTs with i(12p) using a global approach to expression profiling targeting chromosomes (comparative expressed sequence hybridization, CESH). This indicated overexpression of genes from 12p11.2-p12.1 relative to testis tissue and fibroblasts. The nonseminoma subtype showed higher levels of expression than seminomas. Notably, 12p11.2-p12.1 is amplified in about 10% of TGCTs and CESH analysis of such amplicon cases showed high levels of overexpression from this region. Microarray analysis, including cDNA clones representing most UniGene clusters from 12p11.2-p12.1, was applied to DNA and RNA from 5 TGCTs with amplification of 12p11.2-p12.1 and seven TGCTs with gain of the entire short arm of chromosome 12. Expression profiles were consistent with the CESH data and overexpression of EST595078, MRPS35 and LDHB at 12p11.2-p12.1 was detected in most TGCTs. High-level overexpression of BCAT1 was specific to nonseminomas and overexpression of genes such as CMAS, EKI1, KRAS2, SURB7 and various ESTs correlated with their amplification. Genes such as CCND2, GLU3, LRP6 and HPH1 at 12p13 were also overexpressed. The overexpressed sequences identified, particularly those in the region amplified, represent candidate genes for involvement in TGCT development.


Subject(s)
Chromosomes, Human, Pair 12 , Gene Amplification , Gene Expression Profiling , Germinoma/genetics , Testicular Neoplasms/genetics , Adolescent , Adult , Humans , Male , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
13.
J Clin Oncol ; 19(12): 3029-36, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11408498

ABSTRACT

PURPOSE: High-dose chemotherapy (HD-Ctx) followed by autologous peripheral-blood stem-cell (PBSC) transplantation is currently investigated in patients with poor prognosis or relapsed metastatic germ cell tumor (GCTs). This study analyzed the presence and the clinical importance of contaminating tumor cells in PBSC preparations used to support HD-Ctx in GCT patients. PATIENTS AND METHODS: Seven targets for reverse transcription polymerase chain reaction (RT-PCR)-based detection of GCT cells were able to detect seminomatous and different histologic variants of nonseminomatous tumor cells. PBSC preparations from 57 patients were investigated for the presence of contaminating tumor cells using this set of targets, including beta human chorionic gonadotropin (beta-hCG), fibronectin (EDB variant), epidermal growth factor receptor (EGFR), CD44 (v8 to 10 variant), germ cell and placental alkaline phosphatase (AP), human endogenous retrovirus type K (ENV and GAG), and XIST. Samples of PBSC preparations from four healthy donors for allogenic transplantations as well as blood specimens from 10 healthy volunteers served as negative controls. RESULTS: Fifty patients (43 first-line and seven second-line Ctx) were assessable. Combining all RT-PCR results, 29 PBSC preparations (58%) were positive for tumor-specific amplification products (HERV-K 0, fibronectin 4, XIST 14, beta-hCG 19, AP 19, CD44 24, EGFR 26). Ten (35%) of 29 patients who underwent transplantation with positive PBSC preparations and seven (33%) of 21 patients with negative PBSC preparations have suffered relapse or progression (not significant [ns]). With a median follow-up of 22 months (2 to 66) post-HD-Ctx projected 3-year survival rates are 68% (RT-PCR+) and 58% (RT-PCR-) (ns). None of the 10 control peripheral-blood samples showed positivity for any of the targets studied. CONCLUSION: GCT cells can be detected in more than 50% of PBSC preparations using a RT-PCR approach with multiple targets. Despite the presence of tumor cells, retransplantation of the PBSC products did not effect long-term outcome. Factors such as responsiveness to chemotherapy and tumor mass seem to overcome the importance of potentially re-infused tumor cells.


Subject(s)
Germinoma/therapy , Hematopoietic Stem Cell Transplantation , Leukapheresis , Neoplastic Cells, Circulating , Reverse Transcriptase Polymerase Chain Reaction , Adolescent , Adult , Biomarkers, Tumor , Case-Control Studies , DNA Primers , Disease-Free Survival , Follow-Up Studies , Germinoma/mortality , Humans , Male , Middle Aged , Sensitivity and Specificity , Survival Rate
14.
Am J Pathol ; 157(4): 1155-66, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11021820

ABSTRACT

Human testicular germ-cell tumors of young adults (TGCTs), both seminomas and nonseminomas, are characterized by 12p overrepresentation, mostly as isochromosomes, of which the biological and clinical significance is still unclear. A limited number of TGCTs has been identified with an additional high-level amplification of a restricted region of 12p including the K-RAS proto-oncogene. Here we show that the incidence of these restricted 12p amplifications is approximately 8% in primary TGCTs. Within a single cell formation of i(12p) and restricted 12p amplification is mutually exclusive. The borders of the amplicons cluster in short regions, and the amplicon was never found in the adjacent carcinoma in situ cells. Seminomas with the restricted 12p amplification virtually lacked apoptosis and the tumor cells showed prolonged in vitro survival like seminoma cells with a mutated RAS gene. However, no differences in proliferation index between these different groups of seminomas were found. Although patients with a seminoma containing a homogeneous restricted 12p amplification presented at a significantly younger age than those lacking it, the presence of a restricted 12p amplification/RAS mutation did not predict the stage of the disease at clinical presentation and the treatment response of primary seminomas. In 55 primary and metastatic tumors from 44 different patients who failed cisplatinum-based chemotherapy, the restricted 12p amplification and RAS mutations had the same incidence as in the consecutive series of responding patients. These data support the model that gain of 12p in TGCTs is related to invasive growth. It allows tumor cells, in particular those showing characteristics of early germ cells (ie, the seminoma cells), to survive outside their specific microenvironment. Overexpression of certain genes on 12p probably inhibits apoptosis in these tumor cells. However, the copy numbers of the restricted amplification of 12p and K-RAS mutations do not predict response to therapy and survival of the patients.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Gene Amplification , Genes, ras/genetics , Germinoma/genetics , Mutation , Testicular Neoplasms/genetics , Adult , Apoptosis/genetics , Cell Division/genetics , Cell Division/physiology , Cell Survival/genetics , Genetic Variation , Germinoma/pathology , Humans , Male , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Staging , Proto-Oncogene Mas , Testicular Neoplasms/pathology
15.
Am J Hum Genet ; 64(5): 1445-52, 1999 May.
Article in English | MEDLINE | ID: mdl-10205278

ABSTRACT

In female mammalian cells, one of the two X chromosomes is inactivated to compensate for gene-dose effects, which would be otherwise doubled compared with that in male cells. In somatic lineages in mice, the inactive X chromosome can be of either paternal or maternal origin, whereas the paternal X chromosome is specifically inactivated in placental tissue. In human somatic cells, X inactivation is mainly random, but both random and preferential paternal X inactivation have been reported in placental tissue. To shed more light on this issue, we used PCR to study the methylation status of the polymorphic androgen-receptor gene in full-term human female placentas. The sites investigated are specifically methylated on the inactive X chromosome. No methylation was found in microdissected stromal tissue, whether from placenta or umbilical cord. Of nine placentas for which two closely apposed samples were studied, X inactivation was preferentially maternal in three, was preferentially paternal in one, and was heterogeneous in the remaining five. Detailed investigation of two additional placentas demonstrated regions with balanced (1:1 ratio) preferentially maternal and preferentially paternal X inactivation. No differences in ratio were observed in samples microdissected to separate trophoblast and stromal tissues. We conclude that methylation of the androgen receptor in human full-term placenta is specific for trophoblastic cells and that the X chromosome can be of either paternal or maternal origin.


Subject(s)
Dosage Compensation, Genetic , Receptors, Androgen/genetics , Trophoblasts , DNA Methylation , Female , Humans , Male , Placenta/cytology , Placenta/metabolism , Pregnancy , Receptors, Androgen/metabolism
16.
FEBS Lett ; 437(1-2): 107-11, 1998 Oct 16.
Article in English | MEDLINE | ID: mdl-9804181

ABSTRACT

Insulin-like growth factors are involved in the paracrine growth regulation of human breast tumor cells. IGF2 is imprinted in most tissues, and shows expression of the paternal allele only. To investigate whether disruption of this monoallelic IGF2 expression is involved in breast cancer development, a series of primary tumors and adjacent, histologically normal, breast tissue samples, as well as matched primary in vitro fibroblast cultures were studied. Biallelic expression (partial) of IGF2 was found in the majority of in vivo samples, and corresponding fibroblast cultures, while monoallelic expression was found in a normal breast sample. In contrast, H19, a closely apposed, but reciprocally imprinted gene, assumed to be regulated by a common control element, showed retention of monoallelic H19 expression in all in vivo and in the majority of in vitro samples. These data indicate that IGF2, but not H19, is prone to loss of imprinting in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast/metabolism , Genomic Imprinting , Insulin-Like Growth Factor II/metabolism , Muscle Proteins/metabolism , RNA, Untranslated , Cells, Cultured , Fibroblasts/metabolism , Humans , RNA, Long Noncoding , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
17.
APMIS ; 106(1): 187-95; discussion 196-7, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9524578

ABSTRACT

Genomic imprinting refers to the parental origin-specific functional difference between the paternally and maternally-derived mammalian haploid genome. Normal embryogenesis depends on the presence of both a paternal and a maternal copy of particular chromosomal regions, containing the so-called imprinted genes. Genomic imprinting is established somewhere in the maturation from a primordial germ cell to a mature gamete, either spermatid or oocyte. We discuss the value of testicular cancers, especially those derived from the germ cell lineage, as a model to study erasement of the biparental pattern of genomic imprinting as present in the zygote and establishment of the paternal pattern during spermatogenesis. In addition, we will present data on the presence of X-inactivation in these cancers.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genomic Imprinting , Germinoma/genetics , Testicular Neoplasms/genetics , Animals , Cell Differentiation/genetics , Germinoma/pathology , Humans , Male , Mice , Testicular Neoplasms/pathology
18.
APMIS ; 106(1): 207-13; discussion 213-5, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9524580

ABSTRACT

An accurate and relatively simple method to detect testicular germ cell tumours of adolescents and adults (TGCTs) could be useful for early detection and may help to avoid unnecessary orchidectomies. We report a highly sensitive reverse-transcription polymerase chain reaction assay for the 1.5 kb transcript of the platelet-derived growth factor-alpha receptor for molecular diagnosis of TGCTs. As a simulation of the clinical application of the assay the approach was applied on through-cut-biopsies from orchidectomy specimens. In a series of 31 specimens, the 1.5 kb transcript was detected in all samples containing either carcinoma in situ, or an invasive TGCT, with mature teratoma as only exception. No expression was detected in normal parenchyma. On the basis of the through-cut-biopsies the assay shows a sensitivity of 0.87 and a specificity of 1.00. The positive and negative predictive values of the test are 1.00 and 1.00. For carcinoma in situ alone these values are 0.86, 1.00, 1.00, and 0.80, respectively. The figures at least equal the results obtained with the most sensitive morphological/enzyme-histochemical study of duplicate biopsies. We conclude that the 1.5 kb transcript of the platelet-derived growth factor-alpha receptor is a useful molecular marker for TGCTs, and therefore of interest in a clinical setting.


Subject(s)
Biomarkers, Tumor , Germinoma/genetics , Receptors, Platelet-Derived Growth Factor/analysis , Receptors, Platelet-Derived Growth Factor/genetics , Testicular Neoplasms/genetics , Adolescent , Adult , Germinoma/diagnosis , Humans , Male , Polymerase Chain Reaction , Receptor, Platelet-Derived Growth Factor alpha , Testicular Neoplasms/diagnosis
19.
Am J Pathol ; 151(2): 581-90, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9250171

ABSTRACT

In female mammalian cells, inactivation of one of the X chromosomes compensates the increased dosage of X-linked genes as compared with their male counterparts. This process is initiated by the X-inactive specific transcripts of the xist/XIST gene in cis, resulting in methylation of specific sites of genes to be silenced. However, in male germ cells, X inactivation is established by xist/XIST expression only. We investigated the X inactivation pattern in human testicular tumors of different histogenesis by analysis of XIST expression and methylation of the androgen receptor gene. XIST was expressed only in tumors derived from the germ cell lineage with supernumerical X chromosomes: seminomas, nonseminomas, and spermatocytic seminomas. Although low expression was present in testicular parenchyma with spermatogenesis, XIST was expressed at a higher level in parenchyma with carcinoma in situ, the precursor lesion of seminomas and nonseminomas. Despite the consistent expression of XIST in germ-cell-derived tumors with gain of X chromosomes, methylation of the androgen receptor gene was present in all differentiated but only in a proportion of the undifferentiated nonseminomas. This differential pattern of methylation was also found in a number of representative cell lines. Our data indicate that the counting mechanism resulting in X inactivation is functional in testicular cancers of different histogenesis. Moreover, the differentiation-dependent pattern of X inactivation as reported during normal development in the case of multiple X chromosomes by methylation is retained in these tumors. We conclude therefore that X inactivation allows the excessive gain of X chromosomes found in germ-cell-derived tumors of the adult testis. In addition, this offers an interesting model to study the fundamental mechanisms of these processes.


Subject(s)
Carcinoma in Situ/genetics , RNA, Untranslated , Receptors, Androgen/metabolism , Testicular Neoplasms/genetics , Transcription Factors/genetics , X Chromosome , Adult , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Female , Humans , Male , Methylation , RNA, Long Noncoding , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Transcription Factors/biosynthesis
20.
Can J Nurs Res ; 29(1): 29-46, 1997.
Article in English | MEDLINE | ID: mdl-9287518

ABSTRACT

This paper describes the development and psychometric testing of an instrument designed to measure healthy lifestyle in adolescents. The Adolescent Lifestyle Questionnaire (ALQ) was tested on 292 adolescents residing in eastern Canada using item analysis, factor analysis, and reliability measures. Factor analysis isolated seven dimensions to a healthy lifestyle in adolescents, which accounted for 56.0% of the variance in the 43-item measure. The seven factors were: identity awareness, nutrition, physical participation, safety, health awareness, social support, and stress management. The alpha reliability coefficient for the total scale is .91; alpha coefficients for the subscales range from .60 to .88. The instrument warrants further testing and development with different adolescent populations. The instrument will enable researchers to investigate lifestyle patterns in adolescents and to assess the impact of interventions of lifestyle change in this population.


Subject(s)
Attitude to Health , Life Style , Psychology, Adolescent , Adolescent , Adolescent Nutritional Physiological Phenomena , Adult , Child , Factor Analysis, Statistical , Female , Humans , Male , Nursing Evaluation Research , Reproducibility of Results , Safety , Sampling Studies , Social Identification , Social Support , Stress, Psychological/prevention & control , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL
...