scholarly journals Different Combinations of Regulatory Elements May Account For Expression of the Glycoprotein Hormone α-Subunit Gene in Primate and Horse Placenta

1990 ◽  
Vol 4 (10) ◽  
pp. 1480-1487 ◽  
Author(s):  
Robert A. Fenstermaker ◽  
Todd A. Farmerie ◽  
Colin M. Clay ◽  
Debora L. Hamernik ◽  
John H. Nilson
1997 ◽  
Vol 11 (11) ◽  
pp. 1669-1680
Author(s):  
Paul R. Budworth ◽  
Patrick G. Quinn ◽  
John H. Nilson

Abstract Trophoblast-specific expression of the humanα -subunit glycoprotein hormone gene requires a tightly linked array of five different regulatory elements [trophoblast-specific element (TSE), α-activating element (αACT), a tandem cAMP response element (CRE), junctional regulatory element (JRE), and a CCAAT box]. We examined their contextual contributions to trophoblast-specific expression by using transfection assays to evaluate activity of systematic block replacement mutations made within the 1500-bp 5′-flanking region of the human α-subunit gene. While all five elements were required for full activity, only the TSE and JRE displayed trophoblast specificity. Interestingly, the TSE-binding protein has limited tissue distribution whereas a JRE-binding protein appears trophoblast specific. Likewise, replacement studies with an AP-1 element that binds heterodimers of jun and fos indicated that this element was incapable of compensating for either the tandem CRE or JRE. This preference for both CRE- and JRE-binding proteins provides another avenue for configuring an α-subunit promoter with trophoblast specificity. Additional analysis with a cAMP response element binding protein (CREB)-Gal4 fusion protein further underscored the importance of CREB as well as suggested that transcriptional contributions come from both the DNA-binding domain and transactivation domain of this protein. We also examined the interactive nature of the pentameric array by placing a 15-bp random sequence between each element. Remarkably, only the insertion 3′ of the CCAAT box diminished promoter activity. This suggested the absence of direct interactions between the transcriptional factors that bind each element in the array. It also suggested that the CCAAT box is position-dependent relative to the TATA box. This position dependence appeared cell-specific, as it was not manifest in a gonadotrope cell line (αT3–1 cells). Thus, the CCAAT box also has tissue-specific characteristics that assist in targeting expression of the α-subunit gene to trophoblasts. Together, these data suggest that multiple characteristics of a complex pentameric array of regulatory elements endow the α-subunit promoter with trophoblast specificity and maximal activity.


1991 ◽  
Vol 44 (2) ◽  
pp. 231-237 ◽  
Author(s):  
John H. Nilson ◽  
Joseph A. Bokar ◽  
Colin M. Clay ◽  
Todd A. Farmerie ◽  
Robert A. Fenstermaker ◽  
...  

1998 ◽  
Vol 142 (1-2) ◽  
pp. 141-152 ◽  
Author(s):  
William M Wood ◽  
Janet M Dowding ◽  
Virginia D Sarapura ◽  
Michael T McDermott ◽  
David F Gordon ◽  
...  

1991 ◽  
Vol 7 (1) ◽  
pp. 27-34 ◽  
Author(s):  
Y. Kato ◽  
T. Ezashi ◽  
T. Hirai ◽  
T. Kato

ABSTRACT The gene for the common α subunit of the porcine anterior pituitary glycoprotein hormones was cloned from a genomic library constructed in EMBL3. The nucleotide sequence of the entire coding sequence of the porcine common α-subunit gene was determined in addition to one intron and 1059 and 160 bp of the 5′-and 3′-flanking regions respectively. Southern blot analysis of the porcine genomic DNA indicated that the common α-subunit gene is present as a single copy. The transcriptional unit of the porcine common α subunit spanned about 14kb and contained four exons interrupted by three introns of about 11.5, 1.2 and 0.4kb. The short untranslated sequence in the first exon and the location of the exon/intron junctions at amino acid residues +9/+10 and +71/+72 were highly conserved among the rat, human and bovine common α-subunit genes. In the proximal portion of the 5′-flanking region, one TATA box and one CCAAT box were present. A steroid-responsive element was not found up to 1059 bases upstream from the transcription start site. The potential AP-1 and AP-2 factor-responsive elements were present at three and one positions respectively in the 5′-flanking region. This feature suggests that hypothalamic gonadotrophin-releasing hormone stimulates the expression of the common α-subunit gene predominantly by a signal-transduction system, with the protein kinase C cascade and factors AP-1 and AP-2 as mediators. The cyclic AMP-responsive element was also present at two positions, but a single base substitution was found in each sequence compared with the consensus sequence. The porcine common α-subunit gene has a structure distinct from its counterparts, the porcine FSH-β and LH-β genes, reflecting differential control of their synthesis during gametogenesis.


1990 ◽  
Vol 4 (4) ◽  
pp. 573-582 ◽  
Author(s):  
Bogi Andersen ◽  
Giulia Catignani Kennedy ◽  
Debora L. Hamernik ◽  
Joseph A. Bokar ◽  
Robert Bohinski ◽  
...  

2000 ◽  
Vol 20 (10) ◽  
pp. 3331-3344 ◽  
Author(s):  
Mark S. Roberson ◽  
Makiko Ban ◽  
Tong Zhang ◽  
Jennifer M. Mulvaney

ABSTRACT The aim of these studies was to elucidate a role for epidermal growth factor (EGF) signaling in the transcriptional regulation of the glycoprotein hormone α subunit gene, a subunit of chorionic gonadotropin. Studies examined the effects of EGF and the adenylate cyclase activator forskolin on the expression of a transfected α subunit reporter gene in a human choriocarcinoma cell line (JEG3). At maximal doses, administration of EGF resulted in a 50% increase in a subunit reporter activity; forskolin administration induced a fivefold activation; the combined actions of EGF and forskolin resulted in synergistic activation (greater than eightfold) of the α subunit reporter. Mutagenesis studies revealed that the cyclic AMP response elements (CRE) were required and sufficient to mediate EGF-forskolin-induced synergistic activation. The combined actions of EGF and forskolin resulted in potentiated activation of extracellular signal-regulated kinase (ERK) enzyme activity compared with EGF alone. Specific blockade of ERK activation was sufficient to block EGF-forskolin-induced synergistic activation of the α subunit reporter. Pretreatment of JEG3 cells with a p38 mitogen-activated protein kinase inhibitor did not influence activation of the α reporter. However, overexpression of c-Jun N-terminal kinase (JNK)-interacting protein 1 as a dominant interfering molecule abolished the synergistic effects of EGF and forskolin on the α subunit reporter. CRE binding studies suggested that the CRE complex consisted of CRE binding protein and EGF-ERK-dependent recruitment of c-Jun–c-Fos (AP-1) to the CRE. A dominant negative form of c-Fos (A-Fos) that specifically disrupts c-Jun–c-Fos DNA binding inhibited synergistic activation of the α subunit. Thus, synergistic activation of the α subunit gene induced by EGF-forskolin requires the ERK and JNK cascades and the recruitment of AP-1 to the CRE binding complex.


Sign in / Sign up

Export Citation Format

Share Document