Share this post on:

Ys are defective in LB1 silenced cells. UV irradiation induces cyclobutane pyrimidine dimers (CPDs) that are removed by the nucleotide excision repair (NER) pathway [33]. In order to determine if LB1 silenced cells were deficient in NER, we used a quantitative ELISA to measure the CPD content of genomic DNA isolated from control and LB1 silenced cells following irradiation with 20 J/m2 UV [24,25]. There was a significant delay of ,7 hr before the initiation of CPD clearance in silenced cells as compared to control cells (Fig. 3B). Clearance of CPDs was essentially complete 10457188 in control cells by 48 hrs post irradiation, but LB1 silenced cells required an additional 24195657 72 hr for complete CPD clearance. This delay in DNA repair is therefore the most likely cause of the significant increase in apoptosis in LB1 silenced cells at 48 hr following UV irradiation (Fig. 3A).These results suggest that LB1 silencing alone affected the initiation steps of both NER sub-pathways. The accumulation of phosphorylated pRPA32, which binds to the single stranded region opposite the nucleotide lesion Overall intracellular ADPR content,including both the free and the protein-bound during repair [27,30,33] was induced by UV. However silenced cells exhibited both a delay in and lower expression level of pRPA32 compared to control cells (Fig. 4). Interestingly, as expected cH2AX was transiently induced between 0 and 8 hours and was not detectable by 24 hours after UV irradiation in control cells. In contrast, cH2AX was induced between 0 and 8 hours in LB1 silenced cells and persisted until at least 48 hours after UV irradiation (Fig. 4 and 5). Taken together these data show that the Title Loaded From File levels of DNA damage repair factors involved in NER are significantly decreased in LB1 silenced cells. The lack of sufficient repair factors in LB1 silenced cells could explain the delayed response to the DNA damage caused by UV irradiation. Because of the delayed NER response in LB1 silenced cells, we analyzed the expression of these and other key factors involved in NER [36] by qRT-PCR of RNA isolated from cells 3 days after LB1 silencing (see Table 1). The activation of p53 suggested by the increase in p53 levels in silenced cells (Fig. 2) was confirmed by the significant increase in mRNA levels for TP53 (p53) and its effector gene CDKN1A (p21) (Table 1). The mRNA levels of two NER factors, DDB1 and ERCC6 (CSB), were significantly decreased by more than two-fold compared to control cells. The mRNA levels of other factors involved in NER such as DDB2, ERCC8 (CSA), XPA, RPA, and ERCC5 (XPG) were not significantly altered when comparing LB1 silenced and control cells Table I). In contrast, the expression of PCNA and POLH (Pol eta), the gene products of which are required for trans-lesion synthesis (TLS) [37?9] were significantly down regulated in LB1 silenced cells. The decrease in DDB1 and PCNA mRNA levels in silenced cells is consistent with the decreased protein levels in these cells (Fig. 2 and 4).LB1 silencing causes a delayed initiation of DNA damage repair foci in response to UV irradiationThe mRNA and protein analyses of factors involved in the DNA damage response suggested that some aspects of the NER pathway might be delayed or absent in LB1 silenced cells. Therefore we monitored the timing of the formation of 53BP1, pRPA32 and cH2AX foci, common components of both GGNER and TC-NER, in the nuclei of control and LB1 silenced cells following exposure to UV. Immunofluorescence analyses confirmed that LB1 silenced cells are deficient in DDB1 before and after UV.Ys are defective in LB1 silenced cells. UV irradiation induces cyclobutane pyrimidine dimers (CPDs) that are removed by the nucleotide excision repair (NER) pathway [33]. In order to determine if LB1 silenced cells were deficient in NER, we used a quantitative ELISA to measure the CPD content of genomic DNA isolated from control and LB1 silenced cells following irradiation with 20 J/m2 UV [24,25]. There was a significant delay of ,7 hr before the initiation of CPD clearance in silenced cells as compared to control cells (Fig. 3B). Clearance of CPDs was essentially complete 10457188 in control cells by 48 hrs post irradiation, but LB1 silenced cells required an additional 24195657 72 hr for complete CPD clearance. This delay in DNA repair is therefore the most likely cause of the significant increase in apoptosis in LB1 silenced cells at 48 hr following UV irradiation (Fig. 3A).These results suggest that LB1 silencing alone affected the initiation steps of both NER sub-pathways. The accumulation of phosphorylated pRPA32, which binds to the single stranded region opposite the nucleotide lesion during repair [27,30,33] was induced by UV. However silenced cells exhibited both a delay in and lower expression level of pRPA32 compared to control cells (Fig. 4). Interestingly, as expected cH2AX was transiently induced between 0 and 8 hours and was not detectable by 24 hours after UV irradiation in control cells. In contrast, cH2AX was induced between 0 and 8 hours in LB1 silenced cells and persisted until at least 48 hours after UV irradiation (Fig. 4 and 5). Taken together these data show that the levels of DNA damage repair factors involved in NER are significantly decreased in LB1 silenced cells. The lack of sufficient repair factors in LB1 silenced cells could explain the delayed response to the DNA damage caused by UV irradiation. Because of the delayed NER response in LB1 silenced cells, we analyzed the expression of these and other key factors involved in NER [36] by qRT-PCR of RNA isolated from cells 3 days after LB1 silencing (see Table 1). The activation of p53 suggested by the increase in p53 levels in silenced cells (Fig. 2) was confirmed by the significant increase in mRNA levels for TP53 (p53) and its effector gene CDKN1A (p21) (Table 1). The mRNA levels of two NER factors, DDB1 and ERCC6 (CSB), were significantly decreased by more than two-fold compared to control cells. The mRNA levels of other factors involved in NER such as DDB2, ERCC8 (CSA), XPA, RPA, and ERCC5 (XPG) were not significantly altered when comparing LB1 silenced and control cells Table I). In contrast, the expression of PCNA and POLH (Pol eta), the gene products of which are required for trans-lesion synthesis (TLS) [37?9] were significantly down regulated in LB1 silenced cells. The decrease in DDB1 and PCNA mRNA levels in silenced cells is consistent with the decreased protein levels in these cells (Fig. 2 and 4).LB1 silencing causes a delayed initiation of DNA damage repair foci in response to UV irradiationThe mRNA and protein analyses of factors involved in the DNA damage response suggested that some aspects of the NER pathway might be delayed or absent in LB1 silenced cells. Therefore we monitored the timing of the formation of 53BP1, pRPA32 and cH2AX foci, common components of both GGNER and TC-NER, in the nuclei of control and LB1 silenced cells following exposure to UV. Immunofluorescence analyses confirmed that LB1 silenced cells are deficient in DDB1 before and after UV.

Share this post on: