Elsevier

Experimental Cell Research

Volume 316, Issue 17, 15 October 2010, Pages 2747-2759
Experimental Cell Research

Research Article
Defective DSB repair correlates with abnormal nuclear morphology and is improved with FTI treatment in Hutchinson-Gilford progeria syndrome fibroblasts

https://doi.org/10.1016/j.yexcr.2010.05.015Get rights and content

Abstract

Impaired DSB repair has been implicated as a molecular mechanism contributing to the accelerating aging phenotype in Hutchinson-Gilford progeria syndrome (HGPS), but neither the extent nor the cause of the repair deficiency has been fully elucidated. Here we perform a quantitative analysis of the steady-state number of DSBs and the repair kinetics of ionizing radiation (IR)-induced DSBs in HGPS cells. We report an elevated steady-state number of DSBs and impaired repair of IR-induced DSBs, both of which correlated strongly with abnormal nuclear morphology. We recreated the HGPS cellular phenotype in human coronary artery endothelial cells for the first time by lentiviral transduction of GFP-progerin, which also resulted in impaired repair of IR-induced DSBs, and which correlated with abnormal nuclear morphology. Farnesyl transferase inhibitor (FTI) treatment improved the repair of IR-induced DSBs, but only in HGPS cells whose nuclear morphology was also normalized. Interestingly, FTI treatment did not result in a statistically significant reduction in the higher steady-state number of DSBs. We also report a delay in localization of phospho-NBS1 and MRE11, MRN complex repair factors necessary for homologous recombination (HR) repair, to DSBs in HGPS cells. Our results demonstrate a correlation between nuclear structural abnormalities and the DSB repair defect, suggesting a mechanistic link that may involve delayed repair factor localization to DNA damage. Further, our results show that similar to other HGPS phenotypes, FTI treatment has a beneficial effect on DSB repair.

Introduction

Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder that causes rapid premature aging shortly after birth, recapitulating multiple pathologies associated with aging and resulting in a median lifespan of 13 years (reviewed in Pollex and Hegele [1]). The disease is caused by de novo mutations within exon 11 of the LMNA gene which partially activates a cryptic splice donor site, resulting in deletion of 50 amino acids from exon 11, with subsequent production of a “truncated” form of lamin A termed, progerin or lamin AΔ50 [2], [3]. LMNA encodes the A-type nuclear lamins, primarily lamins A and C. Along with the primary B-type lamins, lamin B1 and lamin B2, the A-type lamins form the nuclear lamina [4], [5], a scaffold-like structure that lines the inner nuclear membrane and also contributes to the nuclear matrix [6]. Multiple LMNA mutations produce nuclear structural irregularities, demonstrating that A-type lamins are intricately involved in nuclear structural organization (reviewed in Dechat et al. [7]). Furthermore, there are multiple lines of evidence indicating a central role for A-type lamins in chromatin organization (reviewed in Dechat et al. [7]).

Accordingly, HGPS cells exhibit altered nuclear structural characteristics and chromatin organization. Nuclear structural irregularities include changes in the spatial distribution of nuclear pore complexes [8] as well as modifications of the nuclear lamina [8], [9] that are likely responsible for the abnormal nuclear morphology observed in primary HGPS cells [2], [3], [8]. These structural irregularities are accompanied by functional changes, including reduced deformability of the nuclear lamina [9], increased nuclear stiffness and sensitivity to mechanical stress [10], and mitotic defects, including abnormal chromosome segregation, delays in cytokinesis, nuclear reassembly, and binucleation [11], [12]. Multiple chromatin organization changes have been described as well. Particularly, HGPS cells exhibit loss of peripheral heterochromatin [8], [13] that may be due to epigenetic changes, including up-regulation of H3K9me3 and H4K20me3, which define constitutive heterochromatin, and downregulation of H3K27me3, which defines facultative heterochromatin [13], [14], [15]. In addition to epigenetic changes, HP1α, which is usually associated with H3K9me3, is downregulated and partially dissociated [14], [15]. It is likely that these abnormalities influence nuclear biological processes that are dependent on proper nuclear structure and chromatin organization.

It has been demonstrated that the nuclear morphological changes and altered chromatin characteristics are not caused by lamin A haploinsufficiency but by the presence of progerin in a dominant gain-of-function fashion, possibly through its accumulation at the inner nuclear membrane (INM) [8], [14], where it appears to alter nuclear lamina structure [9]. Accordingly, reduction of the farnesylated form of progerin using farnesyl transferase inhibitors (FTIs) [16], [17], [18], [19], [20], [21], [22] and reduction of progerin using antisense morpholinos [14] improve nuclear abnormalities in HGPS cells as well as various disease phenotypes in HGPS mouse models. The success of FTIs in these studies and the current lack of any other therapeutic approach for HGPS have lead to a current phase II clinical trial examining the beneficial effect of FTIs in HGPS patients (ClinicalTrials.gov identifier: NCT00425607).

Interestingly, recent studies suggest that DSB accumulation due to impaired DSB repair is one of the mechanisms leading to the accelerated aging phenotype [19], [23], [24], [25]. DSB accumulation appears to be due at least in part to impaired localization of DSB repair factors including Rad51 and Rad50 [24]. It has also been shown that xeroderma pigmentosum group A (XPA), a nucleotide excision repair protein (NER), aberrantly localizes to a subset of DSBs in HGPS cells through interaction with chromatin and inhibits the localization of Rad51 and Rad50, perhaps through steric hindrance [24]. There is evidence to suggest that XPA does not localize to camptothecin (CPT)-induced DSBs, indicating that XPA-localized DSBs may be functionally different in origin or repair [24]. However, XPA-mediated interference with DSB repair does not fully explain the DSB repair problem in HGPS cells since repair of CPT-induced DSBs in HGPS cells is slower compared to wild type cells [24].

In this study, we performed a quantitative analysis of the steady-state number of DSBs and the repair kinetics of IR-induced DSBs in HGPS fibroblasts. We also examined whether the observed deviations from wild type for these characteristics correlated with abnormal nuclear morphology and whether they were caused by progerin. Further, we examined whether FTI treatment can decrease the steady-state DSB level and improve the repair kinetics of IR-induced DSBs in HGPS cells. Lastly, to expand on previous studies that showed that localization of repair factors to DSBs is impaired, we performed a quantitative analysis of the localization kinetics of the MRN repair complex factors, phospho-NBS1 and MRE1, to γ-irradiation-induced DSBs.

Section snippets

Cell culture

Primary dermal fibroblasts from HGPS donors (AG11513 and AG11498) and from apparently healthy donors (AG08470 and AG16409) (Coriell Cell Repository, Camden, NJ) were grown in Dulbecco's modified Eagle's medium (DMEM) supplemented with 15% fetal bovine serum, 1 mM l-glutamine, 1% Pen/Strep, 1% MEM nonessential amino acids. Cells were passaged every 4–6 days, and medium was changed every 2 days. Primary human coronary artery endothelial cells (Cambrex, East Rutherford, NJ) were cultured in

HGPS and wild-type fibroblasts with an abnormal nuclear morphology have elevated steady-state levels of DSBs

To investigate DNA damage repair in HGPS cells, we examined primary dermal fibroblasts derived from two HGPS patients (AG11513 and AG11498) and two apparently healthy individuals (AG08470 and AG16409) of similar age (Supplemental Fig. 3A). Since there is evidence that HGPS cellular phenotypes increase in severity with increasing passage number [8], we used HGPS and wild-type cells of similar population doubling (Supplemental Fig. 3A). The HGPS fibroblasts used in these experiments display

Discussion

Recent reports have demonstrated an accumulation of DSBs in HGPS cells which may be due to defective DSB repair [19], [23], [24], [25]. These observations are interesting since DNA damage accumulation may contribute to the increased levels of apoptosis and senescence observed in HGPS cells and in turn to the accelerated aging phenotype. Here, we set forth to examine the severity of the DSB repair defect and investigate the mechanism/s responsible. Quantitative analysis showed that HGPS

Acknowledgments

We would like to thank Dr. Ahmi Ben-Yehudah, Sandra Tavares-Varum, and Olga Momcilovic for helpful discussion. This research was supported by a grant from the National Institute of Child Health and Human Development, 1PO1HD047675.

References (38)

  • T. Dechat et al.

    Nuclear lamins: major factors in the structural organization and function of the nucleus and chromatin

    Genes Dev.

    (2008)
  • R.D. Goldman et al.

    Accumulation of mutant lamin A causes progressive changes in nuclear architecture in Hutchinson-Gilford progeria syndrome

    Proc. Natl Acad. Sci. USA

    (2004)
  • K.N. Dahl et al.

    Distinct structural and mechanical properties of the nuclear lamina in Hutchinson-Gilford progeria syndrome

    Proc. Natl Acad. Sci. USA

    (2006)
  • V.L. Verstraeten et al.

    Increased mechanosensitivity and nuclear stiffness in Hutchinson-Gilford progeria cells: effects of farnesyltransferase inhibitors

    Aging Cell

    (2008)
  • K. Cao et al.

    A lamin A protein isoform overexpressed in Hutchinson-Gilford progeria syndrome interferes with mitosis in progeria and normal cells

    Proc. Natl Acad. Sci. USA

    (2007)
  • T. Dechat et al.

    Alterations in mitosis and cell cycle progression caused by a mutant lamin A known to accelerate human aging

    Proc. Natl Acad. Sci. USA

    (2007)
  • M. Columbaro et al.

    Rescue of heterochromatin organization in Hutchinson-Gilford progeria by drug treatment

    Cell. Mol. Life Sci.

    (2005)
  • P. Scaffidi et al.

    Reversal of the cellular phenotype in the premature aging disease Hutchinson-Gilford progeria syndrome

    Nat. Med.

    (2005)
  • D.K. Shumaker et al.

    Mutant nuclear lamin A leads to progressive alterations of epigenetic control in premature aging

    Proc. Natl Acad. Sci. USA

    (2006)
  • Cited by (47)

    • Critical DNA damaging pathways in tumorigenesis

      2022, Seminars in Cancer Biology
    • Alteration of genetic recombination and double-strand break repair in human cells by progerin expression

      2020, DNA Repair
      Citation Excerpt :

      In the discussion that follows, we will assume that the majority of HDDs occur via crossovers. It has been shown [36,48] that recruitment of HR proteins to the site of DSBs is hampered in HGPS cells. We posit that one consequence of progerin-associated disruption of HR is a reduction in the recombination machinery’s ability to establish Holliday junction recombination intermediates at the site of HR, whether that site be a classic DSB, a stalled or collapsed replication fork, or perhaps a site of no lesion at all.

    • Abnormal nuclear morphology is independent of longevity in a zmpste24-deficient fish model of Hutchinson-Gilford progeria syndrome (HGPS)

      2018, Comparative Biochemistry and Physiology Part - C: Toxicology and Pharmacology
      Citation Excerpt :

      There was some variability in the amount of subcutaneous fat tissue between individuals, but specific loss of adipose tissue in the mutants was not evident (Fig. 3E to H). HGPS patient-derived cells and Zmpste24-deficient mice show an increased basal level of DNA damage and radiation sensitivity (Liu et al., 2005; Liu, Rusinol, Sinensky, Wang, and Zou, 2006; Constantinescu, Csoka, Navara, and Schatten, 2010; Noda et al., 2015). To investigate the radiosensitivity of zmpste24-deficient medaka, we irradiated 10-month-old mutant and WT fish with increasing X-ray doses (Table S3).

    View all citing articles on Scopus
    View full text