The long-term goal of our research is to understand the mechanism of cellular senescence and differentiation in human epithelial cells. As the first step toward our goal, we have established an in vitro model of cellular senescence and differentiation with serially subcultured normal human oral keratinocytes (NHOK). NHOK underwent a maximum of 22+3 population doublings (PDs) through distinct phases of replication: exponential, senescing, senescent and terminal differentiation. Using our in vitro model, we have investigated the mode of senescence of NHOK in comparison to other human somatic cells, such as human diploid fibroblasts (HDF). In contrast to HDF, exponentially replicating NHOK showed telomerase activity and entered the senescent phase in the absence of telomere shortening. Also, in contrast to HOF, whose senescence program is triggered by p53 and p21WAF1/CIP1, senescent NHOK exhibited decreased levels of these proteins. These findings indicate that NHOK enter senescence in a telomere length-independent manner and through a distinct mechanism from that of HDF. We also found that subcultured NHOK exhibited the features of senescence and differentiation in a sequentially distinct manner, suggesting that separate mechanisms elicit these processes during in vitro passage. Accordingly, we raise a question: What genes constitute the molecular pathways of replicative senescence and differentiation in NHOK? This question will be addressed in the present proposal with the following specific aims: (1) To investigate the profile and the kinetics of gene expression in exponentially replicating, senescing, and senescent and terminally differentiating NHOK during serial subculture, (2) to identify the gene(s) associated with senescence and/or differentiation of NHOK in vitro, and (3) to determine whether the ectopic expression of the identified genes can trigger the senescence and differentiation programs in exponentially replicating NHOK and in immortalized human oral keratinocytes. With these specific aims, we expect to address the following questions: Which genes are involved in senescence and/or differentiation in NHOK? Are same type of gene(s) involved in both senescence and differentiation pathways or not? Can we induce senescence and/or differentiation of cells with specific genes identified by our specific aims? It is expected that the outcome of this proposal will develop into novel hypotheses with which we can further study the molecular mechanisms of cellular replication, senescence, and differentiation of human epithelial cells.

Agency
National Institute of Health (NIH)
Institute
National Institute of Dental & Craniofacial Research (NIDCR)
Type
Small Research Grants (R03)
Project #
5R03DE014635-02
Application #
6626070
Study Section
NIDCR Special Grants Review Committee (DSR)
Program Officer
Shirazi, Yasaman
Project Start
2002-07-01
Project End
2004-06-30
Budget Start
2003-07-01
Budget End
2004-06-30
Support Year
2
Fiscal Year
2003
Total Cost
$76,250
Indirect Cost
Name
University of California Los Angeles
Department
Dentistry
Type
Schools of Dentistry
DUNS #
092530369
City
Los Angeles
State
CA
Country
United States
Zip Code
90095
Kang, Mo K; Shin, Ki-Hyuk; Yip, Felix K et al. (2005) Normal human oral keratinocytes demonstrate abnormal DNA end joining activity during replicative senescence. Mech Ageing Dev 126:475-9
Shin, Ki-Hyuk; Kang, Mo K; Dicterow, Erica et al. (2004) Introduction of human telomerase reverse transcriptase to normal human fibroblasts enhances DNA repair capacity. Clin Cancer Res 10:2551-60
Kang, Mo K; Kameta, Ayako; Shin, Ki-Hyuk et al. (2004) Senescence occurs with hTERT repression and limited telomere shortening in human oral keratinocytes cultured with feeder cells. J Cell Physiol 199:364-70
Kameta, Ayako; Kang, Mo K; Shin, Ki-Hyuk et al. (2003) Absence of hamTERT gene expression is associated with promoter hypermethylation in immortal hamster buccal pouch epithelial cells. Int J Oncol 22:1351-6
Kang, Mo K; Kameta, Ayako; Baluda, Marcel A et al. (2003) Telomere shortening does not occur during postmaturational aging in situ in normal human oral fibroblasts. Mech Ageing Dev 124:873-6
Shin, K-H; Kang, M K; Dicterow, E et al. (2003) Hypermethylation of the hTERT promoter inhibits the expression of telomerase activity in normal oral fibroblasts and senescent normal oral keratinocytes. Br J Cancer 89:1473-8
Kang, Mo K; Kameta, Ayako; Shin, Ki-Hyuk et al. (2003) Senescence-associated genes in normal human oral keratinocytes. Exp Cell Res 287:272-81