Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Nov 22.
Published in final edited form as: Mol Genet Genomics. 2008 Jul 10;280(3):10.1007/s00438-008-0361-2. doi: 10.1007/s00438-008-0361-2

Computational identification of transcription frameworks of early committed spermatogenic cells

Claudia Lalancette 1,2, Adrian E Platts 3,4, Yi Lu 5, Shiyong Lu 6, Stephen A Krawetz 7,8,9
PMCID: PMC3837529  NIHMSID: NIHMS529249  PMID: 18615256

Abstract

It is known that transcription factors (TFs) work in cooperation with each other to govern gene expression and thus single TF studies may not always reflect the underlying biology. Using microarray data obtained from two independent studies of the first wave of spermatogenesis, we tested the hypothesis that co-expressed spermatogenic genes in cells committed to differentiation are regulated by a set of distinct combinations of TF modules. A computational approach was designed to identify over-represented module combinations in the promoter regions of genes associated with transcripts that either increase or decrease in abundance between the first two major spermatogenic cell types: spermatogonia and spermatocytes. We identified five TFs constituting four module combinations that were correlated with expression and repression of similarly regulated genes. These modules were biologically assessed in the context that they represent the key transcriptional mediators in the developmental transition from the spermatogonia to spermatocyte.

Keywords: Bioinformatics, Spermatogenesis, Transcriptional regulation, Frameworks

Introduction

Within the seminiferous tubule of the testis, spermatogenesis is marked by the succession of cellular events that lead to the production of viable and fertile spermatozoa (Holstein et al. 2003). In mouse the first wave of spermatogenesis from birth is complete in approximately 33 days and in man the first wave encompasses approximately 64 days. Spermatogenesis is initiated in the basal compartment of the germinal epithelium, with asymmetric division of spermatogonial stem cells Asingle (As). This gives rise to daughter cells Apaired (Ap) that embark on the differentiative pathway (see de Rooij 2001). This first stage of spermatogenesis is characterized by the multiplication of spermatogonia through mitotic divisions. Each of the cells generated by a spermatogenic stem cell remains linked together by cytoplasmic bridges, until the later stages of spermiogenesis. Mitotic divisions appear to randomly occur between the different clusters of spermatogonia as Ap spermatogonia divide yielding Aaligned (Aal) spermatogonia. With each division, the spermatogonia migrate further into the seminiferous tubule, toward the lumen, but they are unable to cross the tight junction between adjacent Sertoli cells that separate the basal and the luminal compartment. At any given moment Aal spermatogonia will differentiate into A1 spermatogonia which will then undergo a series of synchronized mitoses that give rise to type B spermatogonia. After the last mitotic division, the cells enter the meiotic phase of spermatogenesis that after two reduction divisions gives rise to a population of haploid round spermatids. Whereas the first meiotic division occurs over a long period and cells can be isolated in relatively pure form, the second reduction division giving rise to the round spermatid is compressed. The spermatids are then morphologically restructured shedding their cytoplasm as their chromatin condenses, the acrosome and flagellum form until they are released into the lumen of the seminiferous tubule as the terminally differentiated spermatozoa.

This complex differentiative process requires the induction of many genes regulated by mechanisms capable of restricting expression to specific stages of the spermatogenic pathway (Krawetz et al. 1999). For example, using a gain-of-function screen (Schulz et al. 2004) forced expression of numerous genes in Drosophila germ cells as well as in somatic progenitor cells, caused defects in early spermatogenesis.

Transcription factors (TFs) mediate gene expression by binding to their cognate sites within the promoter region. Many TFs belonging to major families such as CREB, heat-shock, Sox, zinc finger, homeo domain and basic helix-loop-helix, have been associated with the expression of spermatogenic genes (Maclean and Wilkinson 2005). One of the intensively studied TFs driving expression of spermatogenic genes is the testis-specific form of CREM, i.e., CREMT. The importance of this TF was shown by the arrest of spermatogenic cells at the round spermatid stage in male mice when CREM was inactivated (Blendy et al. 1996; Nantel et al. 1996). CREMT is required for the transcription of postmeiotic genes including the protamines (Prm1, Prm2), the transition proteins (Tnp1, Tnp2), proacrosin and calspermin (Sassone-Corsi 1998). The importance of these and other TFs like Plzf (Buaas et al. 2004; Costoya et al. 2004), Hsf1 and Hsf2 (Zhang et al. 2002) is established.

TFs rarely operate in isolation. Complex patterns of regulation are cooperatively achieved through the action of n-element transcription modules or frameworks (Werner et al. 2003). These generate a binding structure sensitive to the states of potentially numerous regulatory pathways within the promoter region of a gene (Arnone and Davidson 1997). For example, it is likely that TFs work in concert as a transcription network to contextualize the expression of acrosin. Mutating the acrosin promoter SREBP2gc binding site was shown to decrease acrosin expression in spermatogenic cells (Wang et al. 2004). In vitro experiments have also suggested several other co-regulatory TFs of acrosin, including Tet-1 and YY1 (Nayernia et al. 1994; Schulten et al. 1999, 2001).

Two primary strategies for mapping regulatory networks have been developed. They are (1) analyzing the promoter regions of coordinately expressed genes for common TF binding sites and (2) identifying highly conserved sites in the promoter regions of orthologous genes. Both strategies rely on the postulate that transcription of similarly expressed genes when considered over sufficient genes can be statistically associated with sets of similar TFs.

The KSPMM database of spermatogenic promoter modules and motifs is a searchable web-based resource for the comparative analysis of promoter regions and their constituent transfactor elements in developing male germ cells (Lu et al. 2006b). The system is populated with promoter sequences from the database of transcription start sites (DBTSS) (Suzuki et al. 2004) and Transfac (Wingender et al. 1996) binding site matrix models to identify TF modules present in proximal promoter regions of genes coordinately expressed during spermatogenesis. This approach was adopted to assess whether other trans-acting factors may be involved in spermatogenic gene expression. A novel algorithm was used to determine over-representation of TF modules of co-expressed genes (Lu et al. 2006a; Naismith et al. 2008) from two independent microarray datasets for the first wave of murine spermatogenesis. Transcription frameworks governing the expression of spermatogenic genes from spermatogonia to spermatocytes were identified. The TFs were then confirmed using a proteomic strategy. The results of this study revealed a discrete set of TFs that are likely coordinated to govern gene expression during the first morphological progression of spermatogenesis.

Materials and methods

In-silico identification of transcription factor modules driving spermatogenic genes

Gene expression from two independent microarray time-course studies encompassing the first round of murine spermatogenesis were selected for analysis (Schultz et al. 2003a; Shima et al. 2004). The data from the three duplicate MG-U74 A, B and C microarrays for 11 time points between day 0 and day 56 (GEO Series GSE926) were employed as one dataset. A similar dataset covering ten time-points from day 1 to adult (GEO Series GSE640) was used as the validating dataset. Data from both sources were assigned to reflect the first two stages of development, spermatogonia (days 0–8), and spermatocytes (days 10–21) at which the germ cells are first observed. Comparisons between the median expression of genes across replicates and samples at these two different stages were undertaken. Those genes exhibiting stable expression within each stage and at least a twofold change in expression between stages (P < 0.01) were selected as exhibiting consistent stage linked modulated transcription.

Promoters for the genes of interest were obtained by querying the DBTSS on murine genome build 5 (May 2004). Analysis encompassed 1 kb 5′ of the transcription start site (TSS) and 200 bp 3′ of TSS. Where multiple TSSs were evidenced for a gene, the promoter sequences for all start sites were used as independent promoters. Candidate TF sites were identified using a threshold of a ≥0.96 match to the position weight matrices (Lu et al. 2006a). A single exception, the GATA family members bind essentially identical sequences, such that at ≥0.96 they are considered identical. Accordingly they were considered a single class the GATA-C (Class). To reduce complexity, transcription modules composed of binary elements were initially considered. This criterion enabled the identification of potentially functional hetero or homodimeric modules from a nonspecific separation model that permitted a distance range of no more than 200 bp and no less than 5 bp between two TFs (Frech et al. 1997; Klingenhoff et al. 1999). All possible module combinations were catalogued. The correlation between the expression changes, either positive, or negative, common to genes having a conserved subset of modules was then determined using a series of contingency tables (Lu et al. 2006a). The co-incidence matrices were highly biased. For example, the absence of a module-combination with no significant change in expression was more likely to be observed. To reduce type I error a Liddell measure (Liddell 1976) was used to determine a P value, and threshold. This was set at P < 0.005. This test has been widely used for the analysis of clinical trial data and is based on the maximum likelihood estimate of a single parameter, in this case co-incidence, and provides greater power when compared to an exact test without randomization (Liddell 1976). The simplest module combinations capable of predicting expression were then considered further. Association of module combinations and genes changing in expression were visualized using the Osprey Network Visualization System, version 1.2.0 (Breitkreutz et al. 2003). SymAtlas (http://www.symatlas.gnf.org/SymAtlas) of the Genomic Institute of the Novartis Research Foundation was used to determine the tissue specificity of the transcripts retained in the analysis.

Isolation of spermatogenic stage-specific cells and nuclear protein extraction

Pachytene spermatocytes and round spermatids were isolated from adult CL/BL6 mice by unit density gravity sedimentation as described (Wykes and Krawetz 2003). The purity of the fractions was assessed through optical microscopy to ascertain the absence of contamination by testicular somatic cells (Sertoli cells, Leidyg cells). Fractions were typically of >90% pure. Spermatogonia were a gift from Dr. John McCarrey (University of Texas at San Antonio, USA). Nuclear protein extraction, used the Panomics Nuclear extraction kit and essentially as described by the manufacturer (Panomics Inc., Redwood City, CA). In brief, the cells were first washed in PBS then resuspended in Buffer A supplemented with 1 mM DTT, protease inhibitors and 0.4% IGEPAL to lyse the cells without affecting the nucleus. After centrifugation, the supernatant was removed and the nuclear pellet incubated in a high salt buffer supplemented with 1 mM DTT and protease inhibitors to extract the nuclear proteins comprising the TFs. These were then collected by centrifugation. The supernatant obtained after cell lysis was reserved for future analysis. Supernatant and nuclear protein extracts were stored at –20°C for subsequent use. Proteins were quantified using the Bradford assay.

Biological identification of spermatogenic transcription factors

Validation of spermatogenic TFs constituting the modules identified in-silico was carried out using the Panomic's TranSignal Protein/DNA Combo Arrays essentially as described by the manufacturer (Panomics Inc., Redwood City, CA). Briefly, nuclear proteins were incubated in the presence of a biotinylated DNA probe mix containing 345 TF binding site oligonucleotides. The protein/DNA complexes were purified away from unbound DNA probes, then the proteins released from the complex. The specifically bound DNA probes were then isolated, denatured then hybridized to an array containing 345 complementary TF binding sites. Subsequent to hybridization, the specifically hybridized sequences were detected by streptavidin–HRP chemiluminescence.

Western blot confirmation

Nuclear proteins (Panomics Inc., Redwood City, CA) were resolved on a 10% SDS-PAGE gel then transferred to Amersham Hybond ECL membranes (GE Healthcare Life Sciences, NJ). Detection of Stat1 and Stat3 employed the Stat Antibody Sampler according to the manufacturer's recommend protocol (Cell Signaling Technology, MA). Protein complexes were detected using the ECL Advance Western Blotting Detection (GE Healthcare Life Sciences, NJ).

Results

Microarray technology has enabled the identification of numerous genes associated with each stage of spermatogenesis. However, the transcriptional regulation of these genes remains poorly characterized. Using the initial microarray data describing the spermatogenic transcriptome (Schultz et al. 2003a; Shima et al. 2004), we selected the concordant group of transcripts that increase or decrease in abundance at least twofold after commitment to spermatogenesis, i.e., from spermatogonia to spermatocytes. Surprisingly only 160 transcripts were concordant between datasets and thus retained for analysis. TF modules, i.e., frameworks, within the promoter regions of these genes were then identified.

A summary of the number of promoters and module combinations that were identified in association with the change in expression between the two spermatogenic cell types is summarized in Table 1. Only one correlating framework was identified in the promoters of the genes encoding transcripts that increased from spermatogonia to spermatocytes. A greater number of correlating module combinations in the promoters of genes that decreased in expression from spermatogonia to spermatocytes was observed.

Table 1.

Data summary of module combinations that correlate with changes in expression from spermatogonia to spermatocytes

Stage and direction of >twofold expression Promoters in category Module combinations
Spermatogonia–spermatocytes: increase 77 1
Spermatogonia–spermatocytes: decrease 83 3

The modules represent the combination of five TFs, PAX2, GATA-C, STAT3, STAT1, and AREB6 defining the following modules PAX2-GATA-C + STAT3-GATAC; STAT1-GATA-C module along with either a GATA-CPAX2, STAT3-STAT3, or AREB6-GATA-C. While STAT1 and AREB6 were solely associated with a decrease in transcript levels, PAX2, GATA-C and STAT3 were associated with both the increase and decrease in expression. This may reflect that these factors work together in both positive and negative combinations to limit expression within a specified range.

Increasing levels of transcripts from spermatogonia to spermatocytes

As shown in Fig. 1, as meiosis begins, a single transcription framework, PAX2-GATA-C + STAT3-GATA-C, was common among the promoters of the 70 genes that exhibited an increase in transcript abundance from spermatogonia to spermatocytes. A subset of genes, i.e., Bad, Nphp1, Lrrc28, Tsga8, Sumo, Pde1c, Capbpip1 were DBTSS classified as containing two promoters and both were considered. A list of the genes containing this framework, their SymAtlas level of expression in testis and ontology is summarized in Table 2. These genes were representative of a broad range of ontologies including transport, cell cycle, metabolism, protein biosynthesis, protein phosphorylation, RNA processing, signal transduction, transcription, cell organization, biogenesis, and protein degradation.

Fig. 1.

Fig. 1

Transcription factor (TF) module combinations of encoded transcripts that increase in abundance. TF module combinations overrepresented in the promoters of genes that increase in expression from spermatogonia to spermatocytes were determined. They are represented as an interaction map

Table 2.

Summary of the properties of transcripts increasing in expression from spermatogonia to spermatocytes

AffyID Gene name Gene ID Testis expression (SymAtlas) Function
104346_at Acyl-coenzyme A binding domain containing 6 Acbd6 Testis-specific
97811_at ADP-ribosylation factor GTPase activating protein 3 Arfgap3 Median Vesicle-mediated transport
109772_at Aquaporin 11 Aqp11 Testis-specific Transport
104328_at Aquaporin 9 Aqp9 Overexpressed testis Water transport
99670_at Bcl-associated death promoter Bad Median Induction of apoptosis
133861_at BTB (POZ) domain containing 12 Btbd12 Overexpressed testis
98959_at C21orf19-like protein C21orf19-like protein
104029_at Calmegin Clgn Overexpressed testis Proteolysis and peptidolysis
109455_at CAP-binding protein complex interacting protein 1 Capbpip1
93499_at Capping protein (actin filament) muscle Z-line, alpha 1 Capza1 Overexpressed testis Actin cytoskeleton organization and biogenesis
113828_at Carnitine palmitoyltransferase 1b Cpt1b Median Fatty acid beta-oxidation
116319_at CDP-diacylglycerol synthase 1 Cds1 Median Phospholipid biosynthesis
97377_at Coilin Coil Overexpressed testis Regulation of transcription
95309_at Dynein, axonemal, heavy chain 8 Dnahc8 Testis-specific
95662_at EST X83328 EST X83328
96918_at Fructose bisphosphatase 1 Fbp1 Gluconeogenesis
104310_at Glucose 6 phosphatase G6pc3 Overexpressed testis
106663_at Glutathione S-transferase, theta 3 Gstt3 Overexpressed testis
103397_at HIV-1 Rev binding protein AU045498 Hrb Overexpressed testis mRNA-nucleus export
130609_at Inositol hexaphosphate kinase 1 Ihpk1 Median Myo-inositol metabolism
113957_at Insulin-like 6 Insl6 Testis-specific Regulation of transcription
113122_i_at Interleukin 33 Il33 Median
103656_at LanC (bacterial lantibiotic synthetase component C)-like 1 Lancl1 Median G-protein coupled receptor protein signaling pathway
116115_at Leucine rich repeat containing 28 Lrrc28 Median
110229_at Leupaxin Lpxn Overexpressed testis Signal transduction
93675_at Male germ cell-associated kinase Mak Testis-specific Protein amino acid phosphorylation
111893_at Mitochondrial ribosomal protein L1 Mrpl1 Median Protein biosynthesis
99153_at Mitochondrial ribosomal protein L53 Mrpl53 Median
116680_at Mitogen-activated protein kinase 8 interacting protein 2 Mapk8ip2 Overexpressed testis Vesicle-mediated transport
106572_at Myotubularin related protein 6 Mtmr6 Overexpressed testis Protein amino acid dephosphorylation
106256_at NDC80 kinetochore complex component Nuf2
98614_at Nephronophthisis 1 (juvenile) homolog Nphp1 Testis-specific Actin cytoskeleton organization and biogenesis
109727_at PHD finger protein 2 Phf2 Median Regulation of transcription
105240_at Phosphodiesterase 1C Pde1c Overexpressed testis Signal transduction
97965_at Phospholipase A2, group VI Pla2g6 Overexpressed testis Phospholipid metabolism
111239_at Poliovirus receptor-related 3 Pvrl3 Testis-specific
111288_at Poly(rC) binding protein 3 AlphaCP-3 Pcbp3 Overexpressed testis mRNA metabolism
113953_at Potassium channel, subfamily K, member 4 Kcnk4 Median Potassium ion transport
93207_at Preproacrosin Acr Testis-specific Proteolysis and peptidolysis
113964_at Protein phosphatase 1, regulatory (inhibitor) subunit 11 Ppp1r11
93658_at Protein tyrosine phosphatase, non-receptor type 20 Ptpn20 Testis-specific
103881_at Pyrophosphatase (inorganic) 2 Ppa2 Overexpressed testis
95539_at RAB3A interacting protein Rab3ip Median
109997_at Rab9 effector protein with kelch motifs Rabepk Median
113638_at Rag1/Nwc fusion Rag1 Median
140878_at RAN binding protein 17 Ranbp17 Testis-specific Protein-nucleus import
99591_i_at Retinol dehydrogenase 11 Rdh11 Overexpressed testis Retinol metabolism
104544_at Ribosomal protein L39-like protein Rpl39 Low Protein biosynthesis
107508_at RIKEN cDNA 0710001D07 RIKEN cDNA 0710001D07 Testis-specific
111023_at RIKEN cDNA 1700022C21 RIKEN cDNA 1700022C21 Testis-specific
115373_at RIKEN cDNA 1700027N10 RIKEN cDNA 1700027N10 Overexpressed testis
97210_at RIKEN cDNA 1700037H04 RIKEN cDNA 1700037H04 Overexpressed testis
108060_at RIKEN cDNA 1810030N24 gene RIKEN cDNA 1810030N24 Overexpressed testis
96640_at RIKEN cDNA 3110001A13 RIKEN cDNA 3110001A13 Median
129854_at RIKEN cDNA 6820408C15 RIKEN cDNA 6820408C15 Testis-specific
117204_at RWD domain containing 2 Rwdd2 Overexpressed testis
113152_at Serine/threonine kinase 39 Stk39 Overexpressed testis Protein amino acid phosphorylation
101412_at SH3-domain GRB2-like 3 Sh3gl3 Overexpressed testis Signal transduction
112181_at SMEK homolog 1 Smek1
105805_at Sperm associated antigen 16 Spag16 Overexpressed testis
133455_at Sperm associated antigen 17 Spag17 Median
101850_at Sperm autoantigenic protein 17 Sp17 Spa17 Testis-specific Signal transduction
136288_at Sphingosine-1-phosphate phosphotase 2 Sgpp2 Median
110159_at SUMO/sentrin specific peptidase 2 Sumo
99531_at Synaptogyrin 4 Syngr4 Testis-specific Transport
112945_at Testis specific gene a8 Tsga8 Testis-specific
113799_at Tetratricopeptide repeat domain 26 Ttc26 Overexpressed testis
109128_at THO complex 5 Thoc5
92821_at Ubiquitin specific peptidase 2 Usp2 Overexpressed testis Ubiquitin-dependent protein catabolism
113166_at Zinc finger, matrin type 5 Zmat5 Overexpressed testis

Affymetrix probe ID, gene name and gene ID are presented for each gene analyzed. Expression in testis compared to other mouse tissues from GNF SymAtlas database and the cellular function, when available from NIH David are summarized

Decreasing levels of transcripts from spermatogonia to spermatocytes

As summarized in Fig. 2 and Table 3, three transcription frameworks were common among the promoters of 83 genes that presented a decrease in transcript abundance from spermatogonia to spermatocytes. A subset of genes, i.e., Ddx3, Notch2, Ric8b, Otud5, Cugbp2, Tcf12 was attributed in DBTSS as containing two promoter regions. As above, both were considered. Interestingly all frameworks contained the STAT1-GATA-C module accompanied with either a GATA-C-PAX2, STAT3-STAT3, or AREB6-GATA-C module. Fourteen genes were associated with all module combinations. Ontology groups associated with each framework included signal transduction, transport, transcription, metabolism, RNA processing, protein biosynthesis, and protein degradation.

Fig. 2.

Fig. 2

TF module combinations of encoded transcripts that decrease in abundance. TF module combinations overrepresented in the promoters of genes that decrease in expression from spermatogonia to spermatocytes were determined. They are represented as an interaction map. While the majority of genes are represented by a single unique combination, multiple combinations of modules are observed for others as revealed by a series of interconnecting nodes

Table 3.

Summary of the properties of transcripts decreasing in expression from spermatogonia to spermatocytes

AffyID Gene name Gene ID Testis expression (SymAtlas) Function
116914_at A disintegrin-like and metallopeptidase (reprolysin type) with thrombospondin type 1 motif, 5 Adamts5 Median Proteolysis and peptidolysis
107969_at Activated leukocyte cell adhesion molecule Alcam Low Signal transduction
94304_at Annexin A6 Anxa6 Low Blood coagulation
111383_at BTB (POZ) domain containing 3 Btbd3 Median
102248_f_at Calcium/calmodulin-dependent serine protein kinase (MAGUK family) DXPri1 Cask Median Protein amino acid phosphorylation
101542_f_at DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 3 Ddx3 Low
93534_at Decorin Dcn Low Organogenesis
99191_at EP300 interacting inhibitor of differentiation 1 Eid1 Low
92852_at Fibronectin 1 Fn1 Low Signal transduction
100986_at Four and a half LIM domains 2 Fhl2 Median Transcription
102402_at Glioblastoma amplified sequence AV006093 Gbas Low Metabolism
99109_at Immediate early response 2 Ier2 Median
92614_at Inhibitor of DNA binding 3 Id3 Low Development
135201_at Integrin, beta-like 1 Itgbl1 Median Integrin-mediated signaling pathway
94345_at Interleukin 6 signal transducer Il6st Low Signal transduction
106222_at Ipoma HMGIC fusion partner-like 2 Lhfpl2 Low
97247_at Isochorismatase domain containing 1 Idsoc1 Low
98911_at Janus kinase 1 Jak1 Median Intprotein amino acid phosphorylation
104464_s_at KDEL (Lys-Asp-Glu-Leu) endoplasmic reticulum protein retention receptor Kdelr3 Median Intracellular protein transport
99577_at Kit ligand Kitl
99956_at Kit oncogene Kit Median Protein amino acid phosphorylation
92366_at Lamin alpha 2 Lama2 n.a Cell adhesion
101948_at Laminin B1 subunit 1 Lamb1-1 Median
95135_at Mid1 interacting protein 1 [gastrulation specific g12-like (zebrafish)] Mid1ip1 Low
93041_at Minichromosome maintenance deficient 4 homolog Mcm4 Low Regulation of transcription
97990_at Myosin, heavy polypeptide 11 Myh11 Median Cytoskeleton organization and biogenesis
101930_at Nuclear factor I/X Nfix Low Regulation of transcription
95109_at Nucleolar protein 5A Nol5a Low Regulation of transcription
108076_at Nucleoporin 210 Nup210 Low
106557_at OAF homolog Oaf Median
96813_f_at OTU domain containing 5 Otud5 Low
113047_at PDZ domain containing RING finger 3 Pdzrn3 Low
102395_at Peripheral myelin protein Pmp22 Low Negative regulation of cell proliferation
114557_at PHD finger protein 16 Phf16 n.a
95079_at Platelet derived growth factor receptor, alpha polypeptide Pdgfra Low Protein amino acid phosphorylation
102990_at Procollagen, type III, alpha 1 Col3a1 Median Histogenesis and organogenesis;organogenesis
105660_at Procollagen, type IV, alpha 3 Col4aebp
136277_at Procollagen, type IV, alpha 4 Col4a4 Median Regulation of transcription
112304_at Procollagen-lysine, 2-oxoglutarate 5-dioxygenase 1 Plod1 Low
97496_f_at Protein Kinase C delta-binding protein Prkcdbp Median
97844_at Regulator of g-protein signaling 2 Rgs2 Median Cell cycle
115426_at Resistance to inhibitors of cholinesterase 8 homolog B Ric8b Median
104716_at Retinol binding protein 1 Rbp1 Median Retinoid metabolism
133065_at Rho GTPase activating protein 10 Arhgap10 Median
116381_at Ribosomal protein S6 kinase polypeptide 6 Rps6ka6 Overexpressed testis Signal transduction
109975_at RIKEN cDNA 2310045A20 RIKEN cDNA 2310045A20 Median
116890_at RIKEN cDNA 2600003E23 RIKEN cDNA 2600003E23 Median
96207_at RNA binding motif, single stranded interacting protein 1 Rbms1 Low Regulation of translation
98923_at RNA terminal phosphate cyclase-like 1 Rcl1 Low
98600_at S100 calcium binding protein A11 S100a11 Low Negative regulation of cell
109669_at SEC24 related gene family, member D Sec24d Low Intracellular protein transport
93574_at Serine (or cysteine) peptidase inhibitor, clade F, member 1 Serpinf1 Median Cell proliferation
106281_f_at Serine incorporator 5 Serinc5 Low
96812_at Smoothened homolog Smo Low G-protein coupled receptor protein signaling pathway
111448_f_at Special AT-rich sequence binding protein 1 Satb1 Median Regulation of transcription
115354_at Sphingomyelin phosphodiesterase, acid-like 3B Smpdl3d Median
108488_at SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily d, member 3 Smarcd3 Low Regulation of transcription
103781_at Syntaxin 4A Stx4a Low Intracellular protein transport
115703_at TAF9B RNA polymerase II, TATA box binding protein (TBP)-associated factor Taf9b Overexpressed testis
98555_at Tetratricopeptide repeat domain 3 Ttc3 Low
104601_at Thrombomodulin Thbd Low Blood coagulation
112472_at TNFAIP3 interacting protein 2 Tnip2 Median Negative regulation of viral genome replication
98981_s_at Transcription factor 12 Tcf12 Median Regulation of transcription
103050_at Transcription factor 21 Tcf21 Median Regulation of transcription
113196_at Transmembrane protein 119 Tmem119 Low
100039_at Transmembrane protein 4 Tmem4 Low
113139_at Tribbles homolog 2 Trib2 Low
115520_at Tripartite motif protein 34 Trim34 n.a
93595_at Tripeptidyl peptidase I Cln2 Tpp1 Low
96766_s_at TYRO3 protein tyrosine kinase 3 Tyro3 Median Signal transduction
97544_at Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide Ywhaz Low RAS protein signal transduction
103955_at Crystallin, lambda 1 Cryl1 n.a Fatty acid metabolism
104030_at Patched homolog 1 (Drosophila) Ptch1 Median
104188_at Notch homolog 2 (Drosophila) Notch2 Median Regulation of transcription
113982_at Odd-skipped related 2 (Drosophila) Osr2 Overexpressed testis
93600_at Leptin receptor overlapping transcript Leprot Median
93806_at SH3-binding domain glutamic acid-rich protein like Sh3grbl Median Protein complex assembly
95117_at Insulin-like growth factor 2 receptor Igf2r Low Signal transduction
96285_at Myeloid-associated differentiation marker Myadm Low
96632_at Mortality factor 4 like 2 Morf4l2 n.a
97255_at CUG triplet repeat, RNA binding protein 2 Cugbp2 Median mRNA splice site selection
98038_at High mobility group box 3 Hmg3 Median Regulation of transcription
101509_at Von Hippel-Lindau binding protein 1 Vbp1 Low Protein folding

Affymetrix probe ID, gene name and gene ID are presented for each gene analyzed. Expression in testis compared to other mouse tissues from the GNF SymAtlas database and the cellular function, where available from NIH David are summarized

Identification of transcription factors present in spermatogenic cell nuclei

To ascertain whether the TFs were present in spermatogenic cell nuclei, spermatogenic cells were isolated from testis and interrogated with Panomic's TranSignal Protein/DNA Combo Arrays. The results are shown in Fig. 3. All of the TFs identified by the computational approach were represented on the 345-element array. Patterns of TFs present in type A spermatogonia and spermatocytes were very similar. TFs previously shown to be present in spermatogenic cells, such as SP1 (J3), YY1 (J2), E2F (J1) and Ahr/Arnt (J4) were detected (Persengiev et al. 1996; Schulten et al. 2001; Schultz et al. 2003b; El-Darwish et al. 2006). TFs including PAX2 (G21), GATA1 (G6), GATA2 (I6), and STAT1/STAT3 (M22) were detected in both cell types, while AREB6 (B10) was not. It is of note that both STAT1 and STAT3 were detected using a binding site common to both TFs, whereas STAT1 and STAT3 specific sequences appeared absent. STAT proteins are primarily cytosolic and translocate to the nucleus upon activation through phosphorylation and dimerization (Desrivieres et al. 2006). The presence of STATs was verified by Western analysis, using antibodies specific for STAT1 and STAT3. Both cytosolic and nuclear fractions for each cell type were resolved by SDS-PAGE, transferred to nitrocellulose membranes and then processed for immunodetection of STAT proteins. As shown in Fig. 4, both STAT1 and STAT3 are present in spermatogenic cells.

Fig. 3.

Fig. 3

Identification of TFs present in adult mouse spermatogenic cells. Nuclear protein fractions from (a) spermatogonia and (b) spermatocytes were interrogated using a protein/DNAbinding assay. The interaction of 345 elements spotted on the array was assessed by hybridization to a corresponding set of biotinylated probes

Fig. 4.

Fig. 4

Immunodetection of STAT1 and STAT3 in nuclear extracts from adult mouse spermatogenic cells. Spg spermatogonia, Spc spermatocytes, Rnd Spd round spermatids were isolated by unit gravity sedimentation. Nuclear extracts, Nucl, were then prepared from each cell type. Equivalent quantities of protein supernatant SN and Nucl extracts obtained were separated on SDS-PAGE, transferred to Nitrocellulose, then the presence of STAT1 and STAT3 assessed by immunodetection. The hybridizing portion is shown

Discussion

An in silico strategy was developed to mine microarray data for common transcriptional control elements. The utility of this strategy was previously validated shown using a yeast cell cycle dataset where known TFs were indentified (Lu et al. 2006a). Having validated this strategy, the question becomes, can transcriptional frameworks that demarcate differentiation be identified? To directly address this question, two studies of the first wave of murine spermatogenesis were identified and the changes in the transcriptional profiles from spermatogonia to spermatocyte compared. This resolved several combinations of TF modules embedded within promoters, i.e., frameworks that were strikingly correlated with their coordinate change in expression from the spermatogonial to spermatocyte stage of spermatogenesis. When taken individually, the TFs that comprise the frameworks identified, i.e., PAX2, GATA-C, STAT1, STAT3, and AREB6, have many binding sites in the promoters analyzed. However, the modules they constitute and identified using our computational approach are generally present once or twice in the promoters. Thus, considering modules instead of the individual binding sites eliminated the possibility that the transcription factors were identified because their binding sites were present many times in the promoter regions analyzed. A total of 160 transcripts changed expression in a concordant manner. This somewhat low level of concordance was unexpected since both studies used the same approach for RNA extraction through the first wave of spermatogenesis and the same microarray platform. This likely reflects the high stringency of the statistical bounds employed in this study to minimize type 1 error.

Analysis of microarray data from isolated adult spermatogenic cells (Lee et al. 2006) and the first wave of spermatogenesis (Schultz et al. 2003a; Shima et al. 2004) corroborates the presence of the majority of these TFs. Interestingly, GATA1 and GATA2 proteins were present in accordance with transcriptome data. Of the five TFs computationally predicted, four were validated as present in spermatogonia and spermatocyte nuclei using a protein/DNA array. Recently the presence of GATA binding sites in spermatocyte-specific genes was reported (Lee et al. 2006), and mRNAs corresponding to the GATA family members have been identified (Schultz et al. 2003a; Shima et al. 2004). Their over-representation in module combinations regulating spermatogenic cell expression is novel. Other TFs that were identified have been associated with spermatogenesis. For example PAX2 was detected in a testis-specific manner in the rainbow trout (Baron et al. 2005) and shown to be important in the formation and maintenance of the male reproductive tract in mammals (Kobayashi and Behringer 2003). STAT3 has been detected in adult mouse testis (Murphy et al. 2005) and is suggested to be involved in the self-renewal of spermatogenic cells in Drosophila (Tulina and Matunis 2001). STAT1 was detected in mature spermatozoa (D'Cruz et al. 2001) and as summarized by Western analysis in Fig. 4, both STAT1 and STAT3 are present in cells of the spermatogenic lineage.

The protein/DNA array enabled the validation of the majority of the modules identified as changing in gene expression from spermatogonia to spermatocytes, in addition to identifying other TFs present in murine spermatogenic cells. This method has been successfully applied in other studies to identify pathways by which IL-13 down-regulates the inducible nitric oxide synthase gene (Shao et al. 2007), the TFs downstream of the protease activated receptor in mouse urinary bladder during inflammation (Saban et al. 2007), and the cis-elements regulated by toxic nitric oxide (NO) concentrations in neuroblastoma cells (Dhakshinamoorthy et al. 2007). In general, the module combinations identified in-silico in the promoters of genes that change between spermatogonia and spermatocytes were validated. The sole exception was the AREB6-GATA-C + STAT1-GATA-C combination. AREB6 was not detected in either of the spermatogenic cell types using the protein/DNA array method. Perhaps another currently uncharacterized TF binds to this location. Irrespective, within this framework the STAT1-GATA-C module was validated. Furthermore, the majority of the genes associated with this module were associated with either one or both of the other two modules to form functional frameworks. It is possible that the STAT1-GATA-C module in itself is sufficient to down-regulate those genes. Whether all module combinations are required to down-regulate the 14 genes associated with all three frameworks remains to be determined.

Other spermatogenic-specific TFs have been identified, but their consensus binding site sequences largely remain unknown and the computational identification of transcription frameworks must be afforded this consideration. For example, Sohlh1, a basic helix-loop-helix TF detected in oocytes and spermatogonia, was recently suggested to be involved in differentiation of spermatogonia to spermatocytes (Ballow et al. 2006a). Similarly, Sohlh2, is only detected in spermatogonia in the male (Ballow et al. 2006b). At present, the binding sites for these TFs remains to be fully elucidated.

Extending this approach to determine transcription frameworks governing the expression of coregulated genes in adult spermatogenic cells is the clear next step. To date only one microarray dataset from isolated male germ cells has become publicly available (Namekawa et al. 2006) and caution must be exercised as recent studies suggest differences in the “behavior” of spermatogenic cells during pubertal and adult spermatogenesis (Jahnukainen et al. 2004; Yoshida et al. 2006; Ebata et al. 2007). This will require careful consideration.

Analysis of combinations of corelated modules in the promoters of coregulated genes enables the determination of potential frameworks involved in gene expression in a tissue/cell specific context. The multi-factor composition of all the significantly detected module combinations has highlighted the extent to which the conjunction of TFs permit tissue-specific contextualization of regulation to be achieved with even a relatively limited Transcription Factor vocabulary. The crosstalk between multiple transcription networks may permit relatively ubiquitous binding sequences such as those targeted by GATA TFs to exert a highly stage-specific influence to fine tune and specify gene expression.

Acknowledgments

This work was supported in part by NIH grant HD36512 to SAK. C.L is supported in part by the Deans Postdoctoral Recruiting Award from Wayne State School of Medicine.

Contributor Information

Claudia Lalancette, Center for Molecular Medicine and Genetics, Detroit, USA claudia@compbio.med.wayne.edu; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, 275 East Hancock, Detroit, MI 48201, USA.

Adrian E. Platts, Center for Molecular Medicine and Genetics, Detroit, USA Department of Obstetrics and Gynecology, Wayne State University School of Medicine, 275 East Hancock, Detroit, MI 48201, USA.

Yi Lu, Computer Science Department, Prairie View A and M University, Prairie View, USA.

Shiyong Lu, Department of Computer Science, College of Liberal Arts and Sciences, Wayne State University, Detroit, USA.

Stephen A. Krawetz, Center for Molecular Medicine and Genetics, Detroit, USA Department of Obstetrics and Gynecology, Wayne State University School of Medicine, 275 East Hancock, Detroit, MI 48201, USA; Institute for Scientific Computing, Wayne State University, Detroit, USA.

References

  1. Arnone MI, Davidson EH. The hardwiring of development: organization and function of genomic regulatory systems. Development. 1997;124:1851–1864. doi: 10.1242/dev.124.10.1851. [DOI] [PubMed] [Google Scholar]
  2. Ballow D, Meistrich ML, Matzuk M, Rajkovic A. Sohlh1 is essential for spermatogonial differentiation. Dev Biol. 2006a;294:161–167. doi: 10.1016/j.ydbio.2006.02.027. [DOI] [PubMed] [Google Scholar]
  3. Ballow DJ, Xin Y, Choi Y, Pangas SA, Rajkovic A. Sohlh2 is a germ cell-specific bHLH transcription factor. Gene Expr Patterns. 2006b;6:1014–1018. doi: 10.1016/j.modgep.2006.04.007. [DOI] [PubMed] [Google Scholar]
  4. Baron D, Houlgatte R, Fostier A, Guiguen Y. Large-scale temporal gene expression profiling during gonadal differentiation and early gametogenesis in rainbow trout. Biol Reprod. 2005;73:959–966. doi: 10.1095/biolreprod.105.041830. [DOI] [PubMed] [Google Scholar]
  5. Blendy JA, Kaestner KH, Weinbauer GF, Nieschlag E, Schutz G. Severe impairment of spermatogenesis in mice lacking the CREM gene. Nature. 1996;380:162–165. doi: 10.1038/380162a0. [DOI] [PubMed] [Google Scholar]
  6. Breitkreutz BJ, Stark C, Tyers M. Osprey: a network visualization system. Genome Biol. 2003;4:R22. doi: 10.1186/gb-2003-4-3-r22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Buaas FW, Kirsh AL, Sharma M, McLean DJ, Morris JL, Griswold MD, de Rooij DG, Braun RE. Plzf is required in adult male germ cells for stem cell self-renewal. Nat Genet. 2004;36:647–652. doi: 10.1038/ng1366. [DOI] [PubMed] [Google Scholar]
  8. Costoya JA, Hobbs RM, Barna M, Cattoretti G, Manova K, Sukhwani M, Orwig KE, Wolgemuth DJ, Pandolfi PP. Essential role of Plzf in maintenance of spermatogonial stem cells. Nat Genet. 2004;36:653–659. doi: 10.1038/ng1367. [DOI] [PubMed] [Google Scholar]
  9. D'Cruz OJ, Vassilev AO, Uckun FM. Members of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway are present and active in human sperm. Fertil Steril. 2001;76:258–266. doi: 10.1016/s0015-0282(01)01896-9. [DOI] [PubMed] [Google Scholar]
  10. de Rooij DG. Proliferation and differentiation of spermatogonial stem cells. Reproduction. 2001;121:347–354. doi: 10.1530/rep.0.1210347. [DOI] [PubMed] [Google Scholar]
  11. Desrivieres S, Kunz C, Barash I, Vafaizadeh V, Borghouts C, Groner B. The biological functions of the versatile transcription factors STAT3 and STAT5 and new strategies for their targeted inhibition. J Mammary Gland Biol Neoplasia. 2006;11:75–87. doi: 10.1007/s10911-006-9014-4. [DOI] [PubMed] [Google Scholar]
  12. Dhakshinamoorthy S, Sridharan SR, Li L, Ng PY, Boxer LM, Porter AG. Protein/DNA arrays identify nitric oxide-regulated cis-element and trans-factor activities some of which govern neuroblastoma cell viability. Nucleic Acids Res. 2007;35:5439–5451. doi: 10.1093/nar/gkm594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Ebata KT, Zhang X, Nagano MC. Male germ line stem cells have an altered potential to proliferate and differentiate during postnatal development in mice. Biol Reprod. 2007;76:841–847. doi: 10.1095/biolreprod.106.058305. [DOI] [PubMed] [Google Scholar]
  14. El-Darwish KS, Parvinen M, Toppari J. Differential expression of members of the E2F family of transcription factors in rodent testes. Reprod Biol Endocrinol. 2006;4:63. doi: 10.1186/1477-7827-4-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Frech K, Danescu-Mayer J, Werner T. A novel method to develop highly speciffic models for regulatory units detects a new LTR in GenBank which contains a functional promoter. J Mol Biol. 1997;270:674–687. doi: 10.1006/jmbi.1997.1140. [DOI] [PubMed] [Google Scholar]
  16. Holstein AF, Schulze W, Davidoff M. Understanding spermato-genesis is a prerequisite for treatment. Reprod Biol Endocrinol. 2003;1:107. doi: 10.1186/1477-7827-1-107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Jahnukainen K, Chrysis D, Hou M, Parvinen M, Eksborg S, Soder O. Increased apoptosis occurring during the first wave of spermatogenesis is stage-specific and primarily affects midpachytene spermatocytes in the rat testis. Biol Reprod. 2004;70:290–296. doi: 10.1095/biolreprod.103.018390. [DOI] [PubMed] [Google Scholar]
  18. Klingenhoff A, Frech K, Quandt K, Werner T. Functional promoter modules can be detected by formal models independent of overall nucleotide sequence similarity. Bioinformatics. 1999;15:180–186. doi: 10.1093/bioinformatics/15.3.180. [DOI] [PubMed] [Google Scholar]
  19. Kobayashi A, Behringer RR. Developmental genetics of the female reproductive tract in mammals. Nat Rev Genet. 2003;4:969–980. doi: 10.1038/nrg1225. [DOI] [PubMed] [Google Scholar]
  20. Krawetz SA, Kramer JA, McCarrey JR. Reprogramming the male gamete genome: a window to successful gene therapy. Gene. 1999;234:1–9. doi: 10.1016/s0378-1119(99)00147-x. [DOI] [PubMed] [Google Scholar]
  21. Lee TL, Alba D, Baxendale V, Rennert OM, Chan WY. Application of transcriptional and biological network analyses in mouse germ-cell transcriptomes. Genomics. 2006;88:18–33. doi: 10.1016/j.ygeno.2006.03.008. [DOI] [PubMed] [Google Scholar]
  22. Liddell D. Practical tests of 2 × 2 contingency tables. Statistician. 1976;25:295–304. [Google Scholar]
  23. Lu Y, Lu S, Platts AE, Krawetz SA. Proceedings of the sixth international conference on data mining. IEEE Computer Society Press; Hong Kong: 2006a. Mining correlation between motifs and gene expression. [Google Scholar]
  24. Lu Y, Platts AE, Ostermeier GC, Krawetz SA. K-SPMM: a database of murine spermatogenic promoters modules & motifs. BMC Bioinformatics. 2006b;7:238. doi: 10.1186/1471-2105-7-238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Maclean JA, 2nd, Wilkinson MF. Gene regulation in spermato-genesis. Curr Top Dev Biol. 2005;71:131–197. doi: 10.1016/S0070-2153(05)71005-X. [DOI] [PubMed] [Google Scholar]
  26. Murphy K, Carvajal L, Medico L, Pepling M. Expression of Stat3 in germ cells of developing and adult mouse ovaries and testes. Gene Expr Patterns. 2005;5:475–482. doi: 10.1016/j.modgep.2004.12.007. [DOI] [PubMed] [Google Scholar]
  27. Naismith L, Lalancette C, Platts AE, Krawetz SA. The KLAB toolbox: a suite of in-house software applications for epigenetic analysis. Syst Biol Reprod Med. 2008;54:97–108. doi: 10.1080/19396360801935644. [DOI] [PubMed] [Google Scholar]
  28. Namekawa SH, Park PJ, Zhang LF, Shima JE, McCarrey JR, Griswold MD, Lee JT. Postmeiotic sex chromatin in the male germ-line of mice. Curr Biol. 2006;16:660–667. doi: 10.1016/j.cub.2006.01.066. [DOI] [PubMed] [Google Scholar]
  29. Nantel F, Monaco L, Foulkes NS, Masquilier D, LeMeur M, Henriksen K, Dierich A, Parvinen M, Sassone-Corsi P. Spermiogenesis deficiency and germ-cell apoptosis in CREM-mutant mice. Nature. 1996;380:159–162. doi: 10.1038/380159a0. [DOI] [PubMed] [Google Scholar]
  30. Nayernia K, Nieter S, Kremling H, Oberwinkler H, Engel W. Functional and molecular characterization of the transcriptional regulatory region of the proacrosin gene. J Biol Chem. 1994;269:32181–32186. [PubMed] [Google Scholar]
  31. Persengiev SP, Raval PJ, Rabinovitch S, Millette CF, Kilpatrick DL. Transcription factor Sp1 is expressed by three different developmentally regulated messenger ribonucleic acids in mouse spermatogenic cells. Endocrinology. 1996;137:638–646. doi: 10.1210/endo.137.2.8593813. [DOI] [PubMed] [Google Scholar]
  32. Saban R, Simpson C, Davis CA, Dozmorov I, Maier J, Fowler B, Ihnat MA, Hurst RE, Wershil BK, Saban MR. Transcription factor network downstream of protease activated receptors (PARs) modulating mouse bladder inXammation. BMC Immunol. 2007;8:17. doi: 10.1186/1471-2172-8-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Sassone-Corsi P. CREM: a master-switch governing male germ cells differentiation and apoptosis. Semin Cell Dev Biol. 1998;9:475–482. doi: 10.1006/scdb.1998.0200. [DOI] [PubMed] [Google Scholar]
  34. Schulten HJ, Engel W, Nayernia K, Burfeind P. Yeast one-hybrid assay identifies YY1 as a binding factor for a proacrosin promoter element. Biochem Biophys Res Commun. 1999;257:871–873. doi: 10.1006/bbrc.1999.0556. [DOI] [PubMed] [Google Scholar]
  35. Schulten HJ, Nayernia K, Reim K, Engel W, Burfeind P. Assessment of promoter elements of the germ cell-specific proacrosin gene. J Cell Biochem. 2001;83:155–162. doi: 10.1002/jcb.1226. [DOI] [PubMed] [Google Scholar]
  36. Schultz N, Hamra FK, Garbers DL. A multitude of genes expressed solely in meiotic or postmeiotic spermatogenic cells offers a myriad of contraceptive targets. Proc Natl Acad Sci USA. 2003a;100:12201–12206. doi: 10.1073/pnas.1635054100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Schultz R, Suominen J, Varre T, Hakovirta H, Parvinen M, Toppari J, Pelto-Huikko M. Expression of aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator messenger ribonucleic acids and proteins in rat and human testis. Endocrinology. 2003b;144:767–776. doi: 10.1210/en.2002-220642. [DOI] [PubMed] [Google Scholar]
  38. Schulz C, Kiger AA, Tazuke SI, Yamashita YM, Pantalena-Filho LC, Jones DL, Wood CG, Fuller MT. A misexpression screen reveals effects of bag-of-marbles and TGF beta class signaling on the Drosophila male germ-line stem cell lineage. Genetics. 2004;167:707–723. doi: 10.1534/genetics.103.023184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Shao L, Guo Z, Geller DA. Transcriptional suppression of cytokine-induced iNOS gene expression by IL-13 through IRF-1/ISRE signaling. Biochem Biophys Res Commun. 2007;362:582–586. doi: 10.1016/j.bbrc.2007.07.203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Shima JE, McLean DJ, McCarrey JR, Griswold MD. The murine testicular transcriptome: characterizing gene expression in the testis during the progression of spermatogenesis. Biol Reprod. 2004;71:319–330. doi: 10.1095/biolreprod.103.026880. [DOI] [PubMed] [Google Scholar]
  41. Suzuki Y, Yamashita R, Sugano S, Nakai K. DBTSS, database of transcriptional start sites: progress report 2004. Nucleic Acids Res. 2004;32:D78–D81. doi: 10.1093/nar/gkh076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Tulina N, Matunis E. Control of stem cell self-renewal in Drosophila spermatogenesis by JAK-STAT signaling. Science. 2001;294:2546–2549. doi: 10.1126/science.1066700. [DOI] [PubMed] [Google Scholar]
  43. Wang H, San Agustin JT, Witman GB, Kilpatrick DL. Novel role for a sterol response element binding protein in directing spermatogenic cell-specific gene expression. Mol Cell Biol. 2004;24:10681–10688. doi: 10.1128/MCB.24.24.10681-10688.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Werner T, Fessele S, Maier H, Nelson PJ. Computer modeling of promoter organization as a tool to study transcriptional coregulation. FASEB J. 2003;17:1228–1237. doi: 10.1096/fj.02-0955rev. [DOI] [PubMed] [Google Scholar]
  45. Wingender E, Dietze P, Karas H, Knuppel R. TRANSFAC: a database on transcription factors and their DNA binding sites. Nucleic Acids Res. 1996;24:238–241. doi: 10.1093/nar/24.1.238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Wykes SM, Krawetz SA. Separation of spermatogenic cells from adult transgenic mouse testes using unit-gravity sedimentation. Mol Biotechnol. 2003;25:131–138. doi: 10.1385/MB:25:2:131. [DOI] [PubMed] [Google Scholar]
  47. Yoshida S, Sukeno M, Nakagawa T, Ohbo K, Nagamatsu G, Suda T, Nabeshima Y. The first round of mouse spermatogenesis is a distinctive program that lacks the self-renewing spermatogonia stage. Development. 2006;133:1495–1505. doi: 10.1242/dev.02316. [DOI] [PubMed] [Google Scholar]
  48. Zhang Y, Huang L, Zhang J, Moskophidis D, Mivechi NF. Targeted disruption of hsf1 leads to lack of thermotolerance and defines tissue-specific regulation for stress-inducible Hsp molecular chaperones. J Cell Biochem. 2002;86:376–393. doi: 10.1002/jcb.10232. [DOI] [PubMed] [Google Scholar]

RESOURCES