Cargando…

Dysfunctional telomeres in primary cells from Fanconi anemia FANCD2 patients

BACKGROUND: Fanconi anemia (FA) is characterized by sensitivity to DNA cross-linking agents, mild cellular, and marked clinical radio sensitivity. In this study we investigated telomeric abnormalities of non-immortalized primary cells (lymphocytes and fibroblasts) derived from FA patients of the FA-...

Descripción completa

Detalles Bibliográficos
Autores principales: Joksic, Ivana, Vujic, Dragana, Guc-Scekic, Marija, Leskovac, Andreja, Petrovic, Sandra, Ojani, Maryam, Trujillo, Juan P, Surralles, Jordi, Zivkovic, Maja, Stankovic, Aleksandra, Slijepcevic, Predrag, Joksic, Gordana
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3511208/
https://www.ncbi.nlm.nih.gov/pubmed/22980747
http://dx.doi.org/10.1186/2041-9414-3-6
_version_ 1782251556297506816
author Joksic, Ivana
Vujic, Dragana
Guc-Scekic, Marija
Leskovac, Andreja
Petrovic, Sandra
Ojani, Maryam
Trujillo, Juan P
Surralles, Jordi
Zivkovic, Maja
Stankovic, Aleksandra
Slijepcevic, Predrag
Joksic, Gordana
author_facet Joksic, Ivana
Vujic, Dragana
Guc-Scekic, Marija
Leskovac, Andreja
Petrovic, Sandra
Ojani, Maryam
Trujillo, Juan P
Surralles, Jordi
Zivkovic, Maja
Stankovic, Aleksandra
Slijepcevic, Predrag
Joksic, Gordana
author_sort Joksic, Ivana
collection PubMed
description BACKGROUND: Fanconi anemia (FA) is characterized by sensitivity to DNA cross-linking agents, mild cellular, and marked clinical radio sensitivity. In this study we investigated telomeric abnormalities of non-immortalized primary cells (lymphocytes and fibroblasts) derived from FA patients of the FA-D2 complementation group, which provides a more accurate physiological assessment than is possible with transformed cells or animal models. RESULTS: We analyzed telomere length, telomere dysfunction-induced foci (TIFs), sister chromatid exchanges (SCE), telomere sister chromatid exchanges (T-SCE), apoptosis and expression of shelterin components TRF1 and TRF2. FANCD2 lymphocytes exhibited multiple types of telomeric abnormalities, including premature telomere shortening, increase in telomeric recombination and aberrant telomeric structures ranging from fragile to long-string extended telomeres. The baseline incidence of SCE in FANCD2 lymphocytes was reduced when compared to control, but in response to diepoxybutane (DEB) the 2-fold higher rate of SCE was observed. In contrast, control lymphocytes showed decreased SCE incidence in response to DEB treatment. FANCD2 fibroblasts revealed a high percentage of TIFs, decreased expression of TRF1 and invariable expression of TRF2. The percentage of TIFs inversely correlated with telomere length, emphasizing that telomere shortening is the major reason for the loss of telomere capping function. Upon irradiation, a significant decrease of TIFs was observed at all recovery times. Surprisingly, a considerable percentage of TIF positive cells disappeared at the same time when incidence of γ-H2AX foci was maximal. Both FANCD2 leucocytes and fibroblasts appeared to die spontaneously at higher rate than control. This trend was more evident upon irradiation; the percentage of leucocytes underwent apoptosis was 2.59- fold higher than that in control, while fibroblasts exhibited a 2- h delay before entering apoptosis. CONCLUSION: The results of our study showed that primary cells originating from FA-D2 patients display shorten telomeres, elevated incidence of T-SCEs and high frequency of TIFs. Disappearance of TIFs in early response to irradiation represent distinctive feature of FANCD2 cells that should be examined further.
format Online
Article
Text
id pubmed-3511208
institution National Center for Biotechnology Information
language English
publishDate 2012
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-35112082012-12-01 Dysfunctional telomeres in primary cells from Fanconi anemia FANCD2 patients Joksic, Ivana Vujic, Dragana Guc-Scekic, Marija Leskovac, Andreja Petrovic, Sandra Ojani, Maryam Trujillo, Juan P Surralles, Jordi Zivkovic, Maja Stankovic, Aleksandra Slijepcevic, Predrag Joksic, Gordana Genome Integr Research BACKGROUND: Fanconi anemia (FA) is characterized by sensitivity to DNA cross-linking agents, mild cellular, and marked clinical radio sensitivity. In this study we investigated telomeric abnormalities of non-immortalized primary cells (lymphocytes and fibroblasts) derived from FA patients of the FA-D2 complementation group, which provides a more accurate physiological assessment than is possible with transformed cells or animal models. RESULTS: We analyzed telomere length, telomere dysfunction-induced foci (TIFs), sister chromatid exchanges (SCE), telomere sister chromatid exchanges (T-SCE), apoptosis and expression of shelterin components TRF1 and TRF2. FANCD2 lymphocytes exhibited multiple types of telomeric abnormalities, including premature telomere shortening, increase in telomeric recombination and aberrant telomeric structures ranging from fragile to long-string extended telomeres. The baseline incidence of SCE in FANCD2 lymphocytes was reduced when compared to control, but in response to diepoxybutane (DEB) the 2-fold higher rate of SCE was observed. In contrast, control lymphocytes showed decreased SCE incidence in response to DEB treatment. FANCD2 fibroblasts revealed a high percentage of TIFs, decreased expression of TRF1 and invariable expression of TRF2. The percentage of TIFs inversely correlated with telomere length, emphasizing that telomere shortening is the major reason for the loss of telomere capping function. Upon irradiation, a significant decrease of TIFs was observed at all recovery times. Surprisingly, a considerable percentage of TIF positive cells disappeared at the same time when incidence of γ-H2AX foci was maximal. Both FANCD2 leucocytes and fibroblasts appeared to die spontaneously at higher rate than control. This trend was more evident upon irradiation; the percentage of leucocytes underwent apoptosis was 2.59- fold higher than that in control, while fibroblasts exhibited a 2- h delay before entering apoptosis. CONCLUSION: The results of our study showed that primary cells originating from FA-D2 patients display shorten telomeres, elevated incidence of T-SCEs and high frequency of TIFs. Disappearance of TIFs in early response to irradiation represent distinctive feature of FANCD2 cells that should be examined further. BioMed Central 2012-09-14 /pmc/articles/PMC3511208/ /pubmed/22980747 http://dx.doi.org/10.1186/2041-9414-3-6 Text en Copyright ©2012 joksic et al.; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research
Joksic, Ivana
Vujic, Dragana
Guc-Scekic, Marija
Leskovac, Andreja
Petrovic, Sandra
Ojani, Maryam
Trujillo, Juan P
Surralles, Jordi
Zivkovic, Maja
Stankovic, Aleksandra
Slijepcevic, Predrag
Joksic, Gordana
Dysfunctional telomeres in primary cells from Fanconi anemia FANCD2 patients
title Dysfunctional telomeres in primary cells from Fanconi anemia FANCD2 patients
title_full Dysfunctional telomeres in primary cells from Fanconi anemia FANCD2 patients
title_fullStr Dysfunctional telomeres in primary cells from Fanconi anemia FANCD2 patients
title_full_unstemmed Dysfunctional telomeres in primary cells from Fanconi anemia FANCD2 patients
title_short Dysfunctional telomeres in primary cells from Fanconi anemia FANCD2 patients
title_sort dysfunctional telomeres in primary cells from fanconi anemia fancd2 patients
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3511208/
https://www.ncbi.nlm.nih.gov/pubmed/22980747
http://dx.doi.org/10.1186/2041-9414-3-6
work_keys_str_mv AT joksicivana dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT vujicdragana dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT gucscekicmarija dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT leskovacandreja dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT petrovicsandra dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT ojanimaryam dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT trujillojuanp dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT surrallesjordi dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT zivkovicmaja dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT stankovicaleksandra dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT slijepcevicpredrag dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients
AT joksicgordana dysfunctionaltelomeresinprimarycellsfromfanconianemiafancd2patients