Relationships of Species of the Paramecium aurelia Complex (Protozoa, Ph. Ciliophora, Cl. Oligohymenophorea) Based on Sequences of the Histone H4 Gene Fragment

2006 ◽  
Vol 54 (1) ◽  
pp. 37-42 ◽  
Author(s):  
Ewa Przyboś ◽  
Agnieszka Maciejewska ◽  
Bogumiła Skotarczak
2003 ◽  
Vol 90 (5) ◽  
pp. 387-389 ◽  
Author(s):  
Jude M. Przyborski ◽  
Kathrin Bartels ◽  
Michael Lanzer ◽  
Katherine T. Andrews

1996 ◽  
Vol 10 (3) ◽  
pp. 425-435 ◽  
Author(s):  
Nicole Chaubet ◽  
Martine Flenet ◽  
Bernadette Clement ◽  
Pierre Brignon ◽  
Claude Gigot

1987 ◽  
Vol 7 (3) ◽  
pp. 1048-1054
Author(s):  
A Seiler-Tuyns ◽  
B M Paterson

The mouse histone H4 gene, when stably transformed into L cells on the PSV2gpt shuttle vector, is cell cycle regulated in parallel with the endogenous H4 genes. This was determined in exponentially growing pools of transformants fractionated into cell cycle-specific stages by centrifugal elutriation, a method for purifying cells at each stage of the cell cycle without the use of treatments that arrest growth. Linker additions in the 5' noncoding region of the H4 RNA or in the coding region of the gene did not affect the cell cycle-regulated expression of the modified H4 gene even though the overall level of expression was altered. However, replacing the H4 promoter with the human alpha-2 globin promoter, so that the histone transcript produced by the chimeric gene remains essentially unchanged, resulted in the constitutive expression of H4 mRNA during all phases of the cell cycle with no net increase in H4 mRNA levels during the G1-to-S transition. From these results we conclude that all the information necessary for the cell cycle-regulated expression of the H4 gene is contained in the 5.2-kilobase subclone used in these studies with 228 nucleotides of 5'-flanking DNA and that the increase in H4 mRNA during the G1-to-S transition in the cell cycle is mediated by the H4 promoter and not by the increased stability of the H4 RNA.


1985 ◽  
Vol 5 (2) ◽  
pp. 380-389
Author(s):  
S M Hanly ◽  
G C Bleecker ◽  
N Heintz

We have examined the nucleotide sequences necessary for transcription of a human histone H4 gene in vitro. Maximal transcription of the H4 promoter requires, in addition to the TATA box and cap site, promoter elements between 70 and 110 nucleotides upstream from the transcription initiation site. These distal promoter elements are recognized preferentially in extracts from synchronized S-phase HeLa cells. The inability of non-S-phase nuclear extracts to recognize the H4 upstream sequences reflects a specific lack of a transcription factor which interacts with those sequences. These results indicate that the cell cycle regulation of human histone gene expression involves both a specific transcription factor and distal transcription signals in the H4 promoter.


2005 ◽  
Vol 74 (2) ◽  
pp. 121-126 ◽  
Author(s):  
O New Lee ◽  
Keisuke Nemoto ◽  
Nobuo Sugiyama

Biochemistry ◽  
1997 ◽  
Vol 36 (47) ◽  
pp. 14447-14455 ◽  
Author(s):  
Bo Guo ◽  
Janet L. Stein ◽  
André J. van Wijnen ◽  
Gary S. Stein

DNA ◽  
1984 ◽  
Vol 3 (3) ◽  
pp. 215-222
Author(s):  
ANNE SEILER-TUYNS ◽  
PANAGIOTIS PANTAZIS ◽  
WILLIAM BONNER ◽  
DEAN HAMER ◽  
AJIT KUMAR

2003 ◽  
Vol 23 (22) ◽  
pp. 8110-8123 ◽  
Author(s):  
Partha Mitra ◽  
Rong-Lin Xie ◽  
Ricardo Medina ◽  
Hayk Hovhannisyan ◽  
S. Kaleem Zaidi ◽  
...  

ABSTRACT At the G1/S phase cell cycle transition, multiple histone genes are expressed to ensure that newly synthesized DNA is immediately packaged as chromatin. Here we have purified and functionally characterized the critical transcription factor HiNF-P, which is required for E2F-independent activation of the histone H4 multigene family. Using chromatin immunoprecipitation analysis and ligation-mediated PCR-assisted genomic sequencing, we show that HiNF-P interacts with conserved H4 cell cycle regulatory sequences in vivo. Antisense inhibition of HiNF-P reduces endogenous histone H4 gene expression. Furthermore, we find that HiNF-P utilizes NPAT/p220, a substrate of the cyclin E/cyclin-dependent kinase 2 (CDK2) kinase complex, as a key coactivator to enhance histone H4 gene transcription. The biological role of HiNF-P is reflected by impeded cell cycle progression into S phase upon antisense-mediated reduction of HiNF-P levels. Our results establish that HiNF-P is the ultimate link in a linear signaling pathway that is initiated with the growth factor-dependent induction of cyclin E/CDK2 kinase activity at the restriction point and culminates in the activation of histone H4 genes through HiNF-P at the G1/S phase transition.


Sign in / Sign up

Export Citation Format

Share Document