Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
MycoKeys ; 58: 1-26, 2019.
Article in English | MEDLINE | ID: mdl-31534413

ABSTRACT

Shiraiaceae is an important family in Pleosporales (Dothideomycetes), which includes medical fungi and plant pathogens. Two hypocrellin-producing taxa, Shiraia bambusicola and a novel genus Rubroshiraia gen. nov., typified by Rubroshiraia bambusae are treated in this article. Maximum likelihood analysis, generated via RAxML (GTR+G model), using a combined SSU, LSU, TEF1 and RPB2 sequence dataset, shows that Rubroshiraia is close to Shiraia and belongs to the family Shiraiaceae. Descriptions, illustrations and a taxonomic key are provided for the genera in Shiraiaceae. Rubroshiraia morphologically differs from Shiraia in having small and dark ascostromata and filiform ascospores. Production of the ascostromatal metabolites, hypocrellin A and B, were examined by HPLC and spectrophotometer. The content of hypocrellin A and B of specimen HKAS 102255 (R. bambusae) is twice that produced by HKAS 102253 (S. bambusicola). To clarify the relationship between R. bambusae and Hypocrella bambusae, type material of the latter was examined and provided the illustration.

2.
Oncotarget ; 10(15): 1491-1506, 2019 Feb 19.
Article in English | MEDLINE | ID: mdl-30863497

ABSTRACT

TSPX is a tumor suppressor gene located at Xp11.22, a prostate cancer susceptibility locus. It is ubiquitously expressed in most tissues but frequently downregulated in various cancers, including lung, brain, liver and prostate cancers. The C-terminal acidic domain (CAD) of TSPX is crucial for the tumor suppressor functions, such as inhibition of cyclin B/CDK1 phosphorylation and androgen receptor transactivation. Currently, the exact role of the TSPX CAD in transcriptional regulation of downstream genes is still uncertain. Using different variants of TSPX, we showed that overexpression of either TSPX, that harbors a CAD, or a CAD-truncated variant (TSPX[∆C]) drastically retarded cell proliferation in a prostate cancer cell line LNCaP, but cell death was induced only by overexpression of TSPX. Transcriptome analyses showed that TSPX or TSPX[∆C] overexpression downregulated multiple cancer-drivers/oncogenes, including MYC and MYB, in a CAD-dependent manner and upregulated various tumor suppressors in a CAD-independent manner. Datamining of transcriptomes of prostate cancer specimens in the Cancer Genome Atlas (TCGA) dataset confirmed the negative correlation between the expression level of TSPX and those of MYC and MYB in clinical prostate cancer, thereby supporting the hypothesis that the CAD of TSPX plays an important role in suppression of cancer-drivers/oncogenes in prostatic oncogenesis.

3.
Asian J Androl ; 21(3): 260-269, 2019.
Article in English | MEDLINE | ID: mdl-29974883

ABSTRACT

The Y-located testis-specific protein Y-encoded (TSPY) and its X-homologue TSPX originated from the same ancestral gene, but act as a proto-oncogene and a tumor suppressor gene, respectively. TSPY has specialized in male-specific functions, while TSPX has assumed the functions of the ancestral gene. Both TSPY and TSPX harbor a conserved SET/NAP domain, but are divergent at flanking structures. Specifically, TSPX contains a C-terminal acidic domain, absent in TSPY. They possess contrasting properties, in which TSPY and TSPX, respectively, accelerate and arrest cell proliferation, stimulate and inhibit cyclin B-CDK1 phosphorylation activities, have no effect and promote proteosomal degradation of the viral HBx oncoprotein, and exacerbate and repress androgen receptor (AR) and constitutively active AR variant, such as AR-V7, gene transactivation. The inhibitory domain has been mapped to the carboxyl acidic domain in TSPX, truncation of which results in an abbreviated TSPX exerting positive actions as TSPY. Transposition of the acidic domain to the C-terminus of TSPY results in an inhibitory protein as intact TSPX. Hence, genomic mutations/aberrant splicing events could generate TSPX proteins with truncated acidic domain and oncogenic properties as those for TSPY. Further, TSPY is upregulated by AR and AR-V7 in ligand-dependent and ligand-independent manners, respectively, suggesting the existence of a positive feedback loop between a Y-located proto-oncogene and male sex hormone/receptors, thereby amplifying the respective male oncogenic actions in human cancers and diseases. TSPX counteracts such positive feedback loop. Hence, TSPY and TSPX are homologues on the sex chromosomes that function at the two extremes of the human oncogenic spectrum.


Subject(s)
Carcinogenesis/genetics , Cell Cycle Proteins/genetics , Chromosomes, Human, Y/genetics , DNA-Binding Proteins/genetics , Testis/metabolism , Humans , Male , Proto-Oncogene Mas
4.
Hum Mol Genet ; 26(5): 901-912, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28169398

ABSTRACT

The gonadoblastoma gene, testis-specific protein Y-encoded (TSPY), on the Y chromosome and its X-homologue, TSPX, are cell cycle regulators and function as a proto-oncogene and a tumor suppressor respectively in human oncogenesis. TSPY and TSPX competitively bind to the androgen receptor (AR) and AR variants, such as AR-V7, at their conserved SET/NAP domain, and exacerbate and repress the transactivation of the AR/AR-V7 target genes in ligand dependent and independent manners respectively. The inhibitory domain has been mapped to the carboxyl acidic domain of TSPX, truncation of which renders TSPX to be stimulatory while its transposition to the C-terminus of TSPY results in an inhibitory hybrid protein. TSPY and TSPX co-localize with the endogenous AR, in the presence of ligand, on the promoters and differentially regulate the expression of the endogenous AR target genes in the androgen-responsive LNCaP prostate cancer cells. Transcriptome analysis shows that TSPY and TSPX expressions differentially affect significant numbers of canonical pathways, upstream regulators and cellular functions. Significantly, among the common ones, TSPY activates and TSPX inhibits numerous growth-related and oncogenic canonical pathways and cellular functions in the respective cell populations. Hence, TSPY and TSPX exert opposing effects on the transactivation functions of AR and AR-Vs important for various physiological and disease processes sensitive to male sex hormone actions, thereby not only affecting the pathogenesis of male-specific prostate cancer but also likely contributing to sex differences in the health and diseases of man.


Subject(s)
Cell Cycle Proteins/genetics , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Transcriptome/genetics , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Chromosomes, Human, X/genetics , Chromosomes, Human, Y/genetics , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Gonadoblastoma/genetics , Humans , Male , Nuclear Proteins/biosynthesis , Prostatic Neoplasms/pathology , Protein Domains/genetics , Proto-Oncogene Mas , Receptors, Androgen/biosynthesis , Testis/growth & development , Testis/pathology , Transcriptional Activation/genetics
5.
Hum Mol Genet ; 24(3): 685-97, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25267720

ABSTRACT

The Hirschsprung disease (HSCR) is a complex congenital disorder, arising from abnormalities in enteric nervous system (ENS) development. There is a gender disparity among the patients, with the male to female ratio as high as 5 : 1. Loss-of-function mutations of HSCR genes and haploinsufficiency of their gene products are the primary pathogenic mechanisms for disease development. Recent studies identified over half of the HSCR disease susceptibility genes as targets for the sex-determining factor SRY, suggesting that this Y-encoded transcription factor could be involved in sexual dimorphism in HSCR. Among the SRY targets, the tyrosine kinase receptor RET represents the most important disease gene, whose mutations account for half of the familial and up to one-third of the sporadic forms of HSCR. RET is regulated by a distal and a proximal enhancer at its promoter, in which PAX3 and NKX2-1 are the resident transcription factors respectively. We show that the SRY-box 10 (SOX10) co-activator interacts and forms transcriptional complexes with PAX3 and NKX2-1 in a sequence-independent manner and exacerbates their respective transactivation activities on the RET promoter. SRY competitively displaces SOX10 in such transcription complexes and represses their regulatory functions on RET. Hence SRY could be a Y-located negative modifier of RET expression; and if it is ectopically expressed during ENS development, such SRY repression could result in RET protein haploinsufficiency and promotion of HSCR development, thereby contributing to sexual dimorphism in HSCR.


Subject(s)
Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Proto-Oncogene Proteins c-ret/genetics , SOXE Transcription Factors/metabolism , Sex-Determining Region Y Protein/metabolism , Chromosomes, Human, Y/metabolism , Female , Humans , Male , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Proto-Oncogene Proteins c-ret/metabolism , SOXE Transcription Factors/genetics , Sex Characteristics , Sex-Determining Region Y Protein/genetics , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Cell Rep ; 8(3): 723-33, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25088423

ABSTRACT

Male sex determination is mediated sequentially by sex-determining region Y (SRY) and related SRY-box 9 (SOX9) transcription factors. To understand the gene regulatory hierarchy for SRY and SOX9, a series of chromatin immunoprecipitation and whole-genome promoter tiling microarray (ChIP-Chip) experiments were conducted with mouse gonadal cells at the time of sex determination. SRY and SOX9 bind to the promoters of many common targets involved in testis differentiation and regulate their expression in Sertoli cells. SRY binds to various ovarian differentiation genes and represses their activation through WNT/ß-catenin signaling. Sertoli cell-Sertoli cell junction signaling, important for testis cord formation, is the top canonical pathway among the SRY and SOX9 targets. Hence, SRY determines Sertoli cell fate by repressing ovarian and activating testicular differentiation genes, promotes early Sertoli cells to form testis cord, and then passes on its functions to SOX9, which regulates common targets and activates its own gene regulatory program, beyond SRY actions, in sex determination.


Subject(s)
SOX9 Transcription Factor/metabolism , Sertoli Cells/cytology , Sex Determination Processes , Sex-Determining Region Y Protein/metabolism , Animals , Cell Differentiation , Female , Gene Expression Regulation, Developmental , Male , Mice , SOX9 Transcription Factor/genetics , Sertoli Cells/metabolism , Sex-Determining Region Y Protein/genetics , Wnt Signaling Pathway
7.
Hum Pathol ; 45(9): 1847-58, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25017435

ABSTRACT

There is a significant sex disparity favoring males among hepatocellular carcinoma (HCC) patients. Although various risk factors have been identified, the exact etiology of such sexual dimorphism(s) in HCC is uncertain. Previous studies showed that overexpression of the Y-located protooncogene, testis-specific protein Y encoded (TSPY), promotes cell proliferation and oncogenesis whereas its X-located homologue, TSPYhomologue X (TSPX), retards cell cycle and oncogenic progression. Furthermore, TSPX promotes proteasomal degradation of hepatitis B virus-encoded X oncoprotein and hence could serve as a tumor suppressor in virus-associated HCC. Using immunohistochemistry and reverse-transcription polymerase chain reaction analysis, we had examined the expression of TSPY and TSPX with reference to other established biomarkers in HCC and related liver cancers. Our results demonstrated that 55 (19.2%) of 287 male cases were TSPY positive in immunohistochemistry of tissue arrays, and 15 (46.9%) of 32 male cases were TSPY positive in reverse-transcription polymerase chain reaction analysis of clinical samples. TSPY expression was closely associated with the expression of HCC biomarkers, such as glypican 3. In contrast, TSPX expression was down-regulated in 54.5% of total tumor/nontumorous paired samples (18/33) and negatively associated with those of TSPY, glypican 3, and forkhead box M1 (FOXM1) and was positively associated with that of a tumor suppressor, insulin-like growth factor binding protein 3. The present findings support the hypothesis that the oncogenic events leading to an ectopic activation of the Y-located protooncogene TSPY and/or inactivating mutation/epigenetic silencing of the X-located tumor suppressor gene TSPX could collectively contribute to the sexual dimorphism(s) in HCC and related liver cancers in male-biased manners.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/genetics , Cholangiocarcinoma/genetics , Liver Neoplasms/genetics , Nuclear Proteins/genetics , Adult , Aged , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/metabolism , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , DNA-Binding Proteins , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Glypicans/genetics , Glypicans/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Nuclear Proteins/metabolism , Protein Isoforms , Sequence Analysis, DNA , Sex Factors , Tissue Array Analysis
8.
Methods Mol Biol ; 1067: 185-202, 2013.
Article in English | MEDLINE | ID: mdl-23975793

ABSTRACT

Chromatin immunoprecipitation and hybridization of high-density promoter microarray (ChIP-chip) is a powerful strategy to identify target genes for specific transcription factors and other DNA-binding nuclear proteins in a genome-wide manner. Services of core facilities have greatly enhanced the accessibility of these technologies to new investigators to the field. The Mpeak modeling is a simple and efficient computer program, capable of identifying chromatin-binding peaks in ChIP-chip datasets. It utilizes advanced statistical computation, but yet offers a simple procedure with user inputs on parameters in its operation. The Mpeak-fitted signals are tabulation in convenient formats and can be visualized in various genome-display graphic programs, including SignalMap and Genome Browser, and analyzed together with other datasets, such as microarray expression patterns. Several research groups have used the Mpeak program in their respective ChIP-chip studies. The various features of Mpeak will be illustrated with ChIP-chip datasets from a study designed to identify the target genes for the sex-determining factor, SRY, in mouse embryonic gonads at the time of sex determination.


Subject(s)
Chromatin Immunoprecipitation/methods , Genes , Genomics/methods , Oligonucleotide Array Sequence Analysis/methods , Sex-Determining Region Y Protein/metabolism , Software , Animals , Binding Sites , Chromatin/metabolism , Humans , Mice , Promoter Regions, Genetic
9.
Methods Mol Biol ; 825: 3-16, 2012.
Article in English | MEDLINE | ID: mdl-22144232

ABSTRACT

Gonadal sex differentiation is an important developmental process, in which a bipotential primordial gonad undergoes two distinct pathways, i.e., testicular and ovarian differentiation, dependent on its genetic sex. Techniques of isolating fetal gonads at various developmental stages are valuable for studies on the molecular events involved in cell-fate determination, sex-specific somatic and germ-cell differentiation and structural organization. Here we describe various procedures for isolation of embryonic gonads at different developmental stages from embryos of timed-pregnant mice. The isolated fetal gonads can be used for a variety of studies, such as organ culture, gene and protein expression. As examples of applications, we describe the immunofluorescence detection of SOX9 expression in gonadal tissue sections and microRNAs profiling/expression in fetal gonads at a critical stage for sex determination.


Subject(s)
Gonads/embryology , Gonads/metabolism , Animals , Cytological Techniques , Female , Fetus/anatomy & histology , Fetus/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Gonads/anatomy & histology , Male , Mice , MicroRNAs/genetics , Pregnancy , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sex Determination Processes/genetics , Sex Differentiation/genetics , Time Factors
10.
Syst Biol Reprod Med ; 57(1-2): 27-34, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21204751

ABSTRACT

The gonadoblastoma locus on the human Y chromosome (GBY) is postulated to serve normal functions in spermatogenesis, but could exert oncogenic properties in predisposing susceptible germ cells to tumorigenesis in incompatible niches such as streaked gonads in XY sex reversed patients or dysfunctional testis in males. The testis-specific protein Y-linked (TSPY) repeat gene has recently been demonstrated to be the putative gene for GBY, based on its location on the GBY critical region, expression patterns in early and late stages of gonadoblastoma and ability to induce gonadoblastoma-like structures in the ovaries of transgenic female mice. Over-expression of TSPY accelerates G(2)/M progression in the cell cycle by enhancing the mitotic cyclin B-CDK1 kinase activities. Currently the normal functions of TSPY in spermatogenesis are uncertain. Expression studies of TSPY, and its X-homologue, TSPX, in normal human testis suggest that TSPY is co-expressed with cyclin B1 in spermatogonia and various stages of spermatocytes while TSPX is principally expressed in Sertoli cells in the human testis. The co-expression pattern of TSPY and cyclin B1 in spermatogonia and spermatocytes suggest respectively that 1) TSPY is important for male spermatogonial cell replication and renewal in the testis; and 2) TSPY could be a catalyst/meiotic factor essential for augmenting the activities of cyclin B-cyclin dependent kinases, important for the differentiation of the spermatocytes in prophase I and in preparation for consecutive rounds of meiotic divisions without an intermediate interphase during spermatogenesis.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomes, Human, Y/genetics , Gonadoblastoma/genetics , Spermatogenesis/genetics , Animals , CDC2 Protein Kinase/metabolism , COS Cells , Cell Cycle Proteins/physiology , Chlorocebus aethiops , Cyclin B1/genetics , Cyclin B1/metabolism , DNA-Binding Proteins , Female , Humans , Male , Nuclear Proteins
11.
Bioinformatics ; 26(4): 585-6, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20028690

ABSTRACT

SUMMARY: Serial analysis of gene expression (SAGE) provides an alternative, with additional advantages, to microarray gene expression studies. GonadSAGE is the first publicly available web-based SAGE database on male gonad development that covers six male mouse embryonic gonad stages, including E10.5, E11.5, E12.5, E13.5, E15.5 and E17.5. The sequence coverage of each SAGE library is beyond 150K, 'which is the most extensive sequence-based male gonadal transcriptome to date'. An interactive web interface with customizable parameters is provided for analyzing male gonad transcriptome information. Furthermore, the data can be visualized and analyzed with the other genomic features in the UCSC genome browser. It represents an integrated platform that leads to a better understanding of male gonad development, and allows discovery of related novel targets and regulatory pathways.


Subject(s)
Databases, Genetic , Gene Expression , Genomics/methods , Gonads/growth & development , Software , Animals , Embryonic Development/genetics , Male , Mice
12.
FASEB J ; 23(11): 4029-38, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19661285

ABSTRACT

Monoamine oxidase A (MAO A), encoded by the X chromosome, catalyzes the oxidative deamination of monoamine neurotransmitters, such as serotonin, and plays a critically important role in brain development and functions. Abnormal MAO A activity has been implicated in several neuropsychiatric disorders, such as depression, autism, and attention deficit hyperactivity disorder, which show sexual dimorphism. However, the molecular basis for these disease processes is unclear. Recently, we found that MAO A was a putative target gene directly regulated by a transcription factor encoded by the sex-determining region Y (SRY) gene located on the Y chromosome. We demonstrated that SRY activates both MAO A-promoter and catalytic activities in a human male neuroblastoma BE(2)C cell line. A functional SRY-binding site in the MAO A core promoter was identified and validated by electrophoretic mobility shift and chromatin immunoprecipitation (ChIP) analyses. Coimmunoprecipitation and ChIP assays showed that SRY and Sp1 form a transcriptional complex and synergistically activate MAO A transcription. This is the first study demonstrating that the Y-encoded transcription factor SRY is capable of regulating an X-located gene, suggesting a novel molecular mechanism for sexual dimorphism in neural development, brain functions, and initiation/progression of neural disorders associated with MAO A dysfunction.


Subject(s)
Genes, sry/physiology , Monoamine Oxidase/metabolism , Cell Line, Tumor , Humans , Male , Monoamine Oxidase/genetics , Promoter Regions, Genetic , SOX Transcription Factors/genetics , Sp1 Transcription Factor/physiology
13.
J Genet Genomics ; 36(4): 193-202, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19376480

ABSTRACT

The sex-determining region Y (SRY) is the gene on the Y chromosome responsible for switching on male sex determination during mammalian embryogenesis. In its absence, ovaries develop in the embryo. Hence, ovarian determination and differentiation is considered to be a default, or passive, developmental pathway. Recently this classical paradigm of sex determination has been challenged with the discovery of the R-spondin 1 (RSPO1) as an active ovarian determinant. Mutations of RSPO1 cause a female-to-male sex reversal. RSPO1 synergizes with WNT4 in activating an ovarian development in the bipotential gonad via the canonical Wnt signaling. Early studies showed that SRY represses such Wnt signaling, but also generated discrepancies on whether only mouse Sry is capable of inhibiting such Wnt signaling and whether both human and mouse SRY proteins are able to interact with beta-catenin, the intracellular messenger responsible for executing the Wnt signals. Our studies show that both human SRY and mouse Sry are capable of repressing the Rspo1/Wnt/beta-catenin signaling. However, the repression activities vary among different SRY/Sry proteins and paradoxically related to the presence and/or size of an acidic/glutamine-rich domain. The HMG box of human SRY could bind directly to beta-catenin while the mouse Sry binds to beta-catenin via its HMG box and glutamine-rich domain. The results clarify some of the initial discrepancies, and raise the possibility that SRY interacts with beta-catenin in the nucleus and represses the transcriptional activation of the Rspo1/Wnt target genes involved in ovarian determination, thereby switching on testis determination.


Subject(s)
Down-Regulation , Sex Determination Processes , Sex-Determining Region Y Protein/metabolism , Signal Transduction , Thrombospondins/metabolism , beta Catenin/metabolism , Animals , Female , Gonads/metabolism , Humans , Male , Mice , Protein Binding , Protein Structure, Tertiary , Sex-Determining Region Y Protein/genetics , Thrombospondins/genetics , beta Catenin/genetics
14.
J Genet Genomics ; 36(4): 215-27, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19376482

ABSTRACT

Despite the identification of key genes such as Sry integral to embryonic gonadal development, the genomic classification and identification of chromosomal activation of this process is still poorly understood. To better understand the genetic regulation of gonadal development, we performed Serial Analysis of Gene Expression (SAGE) to profile the genes and novel transcripts, and an average of 152,000 tags from male embryonic gonads at E10.5 (embryonic day 10.5), E11.5, E12.5, E13.5, E15.5 and E17.5 were analyzed. A total of 275,583 non-singleton tags that do not map to any annotated sequence were identified in the six gonad libraries, and 47,255 tags were mapped to 24,975 annotated sequences, among which 987 sequences were uncharacterized. Utilizing an unsupervised pattern identification technique, we established molecular staging of male gonadal development. Rather than providing a static descriptive analysis, we developed algorithms to cluster the SAGE data and assign SAGE tags to a corresponding chromosomal position; these data are displayed in chromosome graphic format. A prominent increase in global genomic activity from E10.5 to E17.5 was observed. Important chromosomal regions related to the developmental processes were identified and validated based on established mouse models with developmental disorders. These regions may represent markers for early diagnosis for disorders of male gonad development as well as potential treatment targets.


Subject(s)
Gene Expression Profiling , Mice/embryology , Mice/genetics , Testis/embryology , Animals , Chromosome Mapping , Female , Gene Expression Regulation, Developmental , Male , Mice/growth & development , Molecular Sequence Data , Testis/growth & development , Transcription, Genetic
15.
Birth Defects Res C Embryo Today ; 87(1): 114-22, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19306348

ABSTRACT

The gonadoblastoma (GBY) locus is the only oncogenic locus on the human Y chromosome. It is postulated to serve a normal function in the testis, but could exert oncogenic effects in dysgenetic gonads of individuals with intersex and/or dysfunctional testicular phenotypes. Recent studies establish the testis-specific protein Y-encoded (TSPY) gene to be the putative gene for GBY. TSPY serves normal functions in male stem germ cell proliferation and differentiation, but is ectopically expressed in early and late stages of gonadoblastomas, testicular carcinoma in situ (the premalignant precursor for all testicular germ cell tumors), seminomas, and selected nonseminomas. Aberrant TSPY expression stimulates protein synthetic activities, accelerates cell proliferation, and promotes tumorigenicity in athymic mice. TSPY binds to type B cyclins, enhances an activated cyclin B-CDK1 kinase activity, and propels a rapid G(2)/M transition in the cell cycle. TSPY also counteracts the normal functions of its X-homologue, TSPX, which also binds to cyclin B and modulates the cyclin B-CDK1 activity to insure a proper G(2)/M transition in the cell cycle. Hence, ectopic expression and actions of the Y-located TSPY gene in incompatible germ cells, such as those in dysgenetic or ovarian environments and dysfunctional testis, disrupt the normal cell cycle regulation and predispose the host cells to tumorigenesis. The contrasting properties of TSPY and TSPX suggest that somatic cancers, such as intracranial germ cell tumors, melanoma, and hepatocellular carcinoma, with detectable TSPY expression could exhibit sexual dimorphisms in the initiation and/or progression of the respective oncogenesis.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomes, Human, Y/genetics , Gonadoblastoma/genetics , Ovarian Neoplasms/genetics , Testicular Neoplasms/genetics , Animals , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cell Proliferation , Cyclin B/metabolism , Female , Gonadoblastoma/metabolism , Gonadoblastoma/pathology , Humans , Male , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Seminoma/genetics , Seminoma/metabolism , Seminoma/pathology , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology
16.
Int J Exp Pathol ; 89(5): 342-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18808526

ABSTRACT

Testicular germ cell tumours (TGCTs) are prevalent cancers among young men. Currently, there is no reliable animal model for TGCTs. To establish such animal models, we have explored the possibility of intratubular testicular transplantation as means to deliver tumour cells into the seminiferous tubules of host animals. Our results demonstrated that transplanted cells could effectively populate the testis of a recipient mouse and develop into TGCTs. In addition, the donor cells could be transfected with a specific transgene before transplantation, thereby providing an approach to evaluate the specific effects of gene functions in the oncogenic processes. Hence, depending on selection of specific donor cells or mixtures of donor cells, transplantation models of TGCTs could be significant for studies on the pathogenesis, diagnosis and therapies of such a prevalent and important cancer in men.


Subject(s)
Models, Animal , Neoplasm Transplantation/methods , Seminiferous Tubules , Seminoma , Testicular Neoplasms , Adult , Animals , Biomarkers, Tumor/analysis , Cell Line, Tumor , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , Mice , Mice, Nude
17.
Hum Pathol ; 38(10): 1470-81, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17521702

ABSTRACT

The testis-specific protein Y-encoded (TSPY) gene is the putative gene for the gonadoblastoma locus on the Y chromosome (GBY) that predisposes dysgenetic gonads of intersex patients to gonadoblastoma development. TSPY is expressed at high levels in gonadoblastoma tissues, supporting its possible oncogenic function in this type of germ cell tumors. To explore the possibility that this Y chromosome gene is also involved in pathogenesis of the more common testicular germ cell tumors (TGCTs), we have conducted various expression studies using immunohistochemistry, Western blotting, and reverse transcription-polymerase chain reaction analysis on 171 cases of TGCTs and selected normal testis controls. Our results demonstrated that TSPY protein is abundantly expressed in the precursor, carcinoma in situ or intratubular germ cell neoplasia unclassified, and seminoma, but only minimally or not expressed in various types of nonseminomas. TSPY coexpresses with established germ cell tumor markers (such as placental-like alkaline phosphatase, c-KIT, OCT4) and proliferative markers (such as Ki-67 and cyclin B1) in the same tumor cells at both RNA and protein levels. Ectopic TSPY expression in cultured cells up-regulates progrowth genes, including those at chromosome 12p13, frequently gained/amplified in TGCTs. Our results suggest that TSPY, in combination with other markers, could be an important marker for diagnosis and subclassification of TGCTs and support its role in the pathogenesis of both gonadoblastoma and TGCTs.


Subject(s)
Biomarkers, Tumor/analysis , Cell Cycle Proteins/biosynthesis , Neoplasms, Germ Cell and Embryonal/metabolism , Testicular Neoplasms/metabolism , Blotting, Western , Cell Cycle Proteins/genetics , Gene Expression , Humans , Immunohistochemistry , Male , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology
18.
Urol Oncol ; 25(2): 141-6, 2007.
Article in English | MEDLINE | ID: mdl-17349529

ABSTRACT

The testis-specific protein Y-encoded gene (TSPY) is a tandem repeat gene located at the critical region for the gonadoblastoma locus on Y chromosome that predisposes the dysgenetic gonads of intersex individuals to oncogenesis. The expression and molecular properties of TSPY suggest that it is the putative gene for the gonadoblastoma locus on Y chromosome. In this study, we examined the expression of TSPY and other germ cell tumor markers in 4 cases of gonadoblastoma using immunostaining techniques. Our results showed that TSPY expression was closely associated with initiation and various stages of gonadoblastoma development. TSPY protein localized with established germ cell tumor markers, such as the placental alkaline phosphatase, c-KIT, and OCT3/4, in the same tumor cells of both gonadoblastoma and adjacent carcinoma in situ, the precursor for germ cell tumors. These findings support the candidacy of TSPY as the gene for the gonadoblastoma locus on Y chromosome and suggest that TSPY could be a significant marker for these types of germ cell tumors.


Subject(s)
Carcinoma in Situ/pathology , Cell Cycle Proteins/metabolism , Germ Cells/pathology , Gonadoblastoma/pathology , Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/pathology , Adolescent , Carcinoma in Situ/metabolism , Cell Cycle Proteins/genetics , Child , Female , Gonadoblastoma/metabolism , Humans , Male , Neoplasms, Germ Cell and Embryonal/metabolism , Testicular Neoplasms/metabolism
19.
Wei Sheng Wu Xue Bao ; 46(5): 827-30, 2006 Oct.
Article in Chinese | MEDLINE | ID: mdl-17172038

ABSTRACT

Alpha-amylase are of considerable commercial value. It can be produced by a wide variety of microorganisma. The alpha-amylase gene (amyE) from Bacillus licheniformis, which is widely used for the industrial hydrolysis of starch, was mutated (amyEM), then amplified by PCR and inserted into pBV220 and pPIC9k to obtain the recombinant vector pBV220-amyEM and pPIC9k-amyEM. These recombinant vectors were transformed into corresponding competent cell E. coli DH5alpha and P. pastoris GS115 respectively. The resulting recombinant strains, DH5alpha/pBV220-amyEM and GS115/ pPIC9k-amyEM, were then screened by measuring the enzymatic activity and SDS-PAGE. DH5alpha/pBV220-amyEM was induced by temperature and GS115/pPIC9k-amyEM by methanol. In contrast to the parent cells, the a-amylases were expressed in both the recombinant strains. In E. coli the molecular weight was approximately 55kDa; optimal temperature and pH of the recombinant a-amylase were 80 degrees C - 90 degrees C and 6.0 respectively. The recombinant amylase had high activity in pH 5.0 - 5.5 compared to wild type. In Pichia pastoris, the recombinant amylase was secreted to the medium; molecular weight was 60kDa for the putative post-translational modifications; optimal pH shifted to 5.5. The specific activities of alpha-amylase produced by E. coli and P. pastoris were 8.1U/mg and 102U/mg respectively. This result indicated that the alpha-amylase were secreted into the culture medium with high efficiency in the recombinant P. pastoris High activity in high temperature and lower pH properties impart the recombinant amylase potential applications in industry.


Subject(s)
Escherichia coli/genetics , Mutation , Pichia/genetics , Recombinant Proteins/biosynthesis , alpha-Amylases/genetics , Hydrogen-Ion Concentration , Temperature , alpha-Amylases/metabolism
20.
Mol Cell Endocrinol ; 257-258: 35-46, 2006 Sep 26.
Article in English | MEDLINE | ID: mdl-16904257

ABSTRACT

Sry encodes a putative transcription factor that switches on testis differentiation during embryogenesis. Currently, the mechanism(s) by which Sry mediates such developmental process is still uncertain. To understand its gene regulation mechanism, we have utilized an in vitro affinity chromatography and proteomic strategy to identify and characterize Sry binding proteins from the mouse testis potentially involved in the formation of an Sry transcriptional complex(es). Our study has consistently identified the poly(ADP-ribose) polymerase 1 (PARP-1) as an Sry interactive protein. PARP-1 is expressed in mouse fetal gonads at the time of sex determination and co-localized with Sry in the nuclei of pre-Sertoli cells. PARP-1 could be co-immunoprecipitated with Sry in cultured cells. The interactive domains have been mapped to the HMG box of Sry and the zinc fingers of the PARP-1 protein, respectively. The Sry-PARP-1 interaction is evolutionarily conserved and it interferes with the ability of Sry in binding to its consensus sequence. In the presence of its substrate, PARP-1 poly(ADP-ribosyl)ates Sry and minimizes severely its DNA-binding activities. PARP-1 represses Sry-mediated transactivation of a reporter gene in cultured cells. Hence, PARP-1 could modulate the regulatory function(s) of Sry on its target genes in this developmental pathway.


Subject(s)
Poly(ADP-ribose) Polymerases/metabolism , Sex-Determining Region Y Protein/metabolism , Sex-Determining Region Y Protein/physiology , Animals , Animals, Newborn , Cells, Cultured , Cloning, Molecular , DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fetal Development , Gene Expression Regulation, Developmental , Gonads/metabolism , HMG-Box Domains , Humans , Male , Mice , Poly (ADP-Ribose) Polymerase-1 , Protein Binding , Rats , Regulatory Sequences, Nucleic Acid , Testis/metabolism , Transcriptional Activation , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...