7,099 research outputs found

    Down-regulation of the Lamin A/C in neuroblastoma triggers the expansion of tumor initiating cells

    Get PDF
    Tumor-initiating cells constitute a population within a tumor mass that shares properties with normal stem cells and is considered responsible for therapy failure in many cancers. We have previously demonstrated that knockdown of the nuclear envelope component Lamin A/C in human neuroblastoma cells inhibits retinoic acid-mediated differentiation and results in a more aggressive phenotype. In addition, Lamin A/C is often lost in advanced tumors and changes in the nuclear envelope composition occur during tumor progression. Based on our previous data and considering that Lamin A/C is expressed in differentiated tissues, we hypothesize that the lack of Lamin A/C could predispose cells toward a stem-like phenotype, thus influencing the development of tumor-initiating cells in neuroblastoma. This paper demonstrates that knockdown of Lamin A/C triggers the development of a tumor-initiating cell population with self-renewing features in human neuroblastoma cells. We also demonstrates that the development of TICs is due to an increased expression of MYCN gene and that in neuroblastoma exists an inverse relationship between LMNA and MYCN expression

    Lamin A/C sustains PcG protein architecture, maintaining transcriptional repression at target genes

    Get PDF
    Beyond its role in providing structure to the nuclear envelope, lamin A/C is involved in transcriptional regulation. However, its cross talk with epigenetic factors--and how this cross talk influences physiological processes--is still unexplored. Key epigenetic regulators of development and differentiation are the Polycomb group (PcG) of proteins, organized in the nucleus as microscopically visible foci. Here, we show that lamin A/C is evolutionarily required for correct PcG protein nuclear compartmentalization. Confocal microscopy supported by new algorithms for image analysis reveals that lamin A/C knock-down leads to PcG protein foci disassembly and PcG protein dispersion. This causes detachment from chromatin and defects in PcG protein-mediated higher-order structures, thereby leading to impaired PcG protein repressive functions. Using myogenic differentiation as a model, we found that reduced levels of lamin A/C at the onset of differentiation led to an anticipation of the myogenic program because of an alteration of PcG protein-mediated transcriptional repression. Collectively, our results indicate that lamin A/C can modulate transcription through the regulation of PcG protein epigenetic factors

    Altered modulation of lamin A/C-HDAC2 interaction and p21 expression during oxidative stress response in HGPS

    Get PDF
    Defects in stress response are main determinants of cellular senescence and organism aging. In fibroblasts from patients affected by Hutchinson-Gilford progeria, a severe LMNA-linked syndrome associated with bone resorption, cardiovascular disorders, and premature aging, we found altered modulation of CDKN1A, encoding p21, upon oxidative stress induction, and accumulation of senescence markers during stress recovery. In this context, we unraveled a dynamic interaction of lamin A/C with HDAC2, an histone deacetylase that regulates CDKN1A expression. In control skin fibroblasts, lamin A/C is part of a protein complex including HDAC2 and its histone substrates; protein interaction is reduced at the onset of DNA damage response and recovered after completion of DNA repair. This interplay parallels modulation of p21 expression and global histone acetylation, and it is disrupted by LMNAmutations leading to progeroid phenotypes. In fact, HGPS cells show impaired lamin A/C-HDAC2 interplay and accumulation of p21 upon stress recovery. Collectively, these results link altered physical interaction between lamin A/C and HDAC2 to cellular and organism aging. The lamin A/C-HDAC2 complex may be a novel therapeutic target to slow down progression of progeria symptoms

    Reduced expression of lamin A/C correlates with poor histological differentiation and prognosis in primary gastric carcinoma

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Lamin A/C is very important in DNA replication, RNA dependent transcription and nuclear stabilization. Reduced or absent lamin A/C expression has been found to be a common feature of a variety of different cancers. To investigate the role of lamin A/C in gastric carcinoma (GC) pathogenesis, we analyzed the correlations between the lamin A/C expression level and clinicopathological factors and studied its prognostic role in primary GC.</p> <p>Methods</p> <p>The expression of lamin A/C at mRNA level was detected by the reverse transcription-polymerase chain reaction (RT-PCR) and real time RT-PCR, and western blot was used to examine the protein expression. Lamin A/C expression and its prognostic significance were investigated by performing immunohistochemical analysis on a total of 126 GC clinical tissue samples.</p> <p>Results</p> <p>Both lamin A/C mRNA and protein expression were downregulated in the majority of tumours compared with corresponding normal gastric tissues (<it>p </it>= 0.011 and <it>p </it>= 0.036, respectively). Real time RT-PCR further validated that downregulation of lamin A/C is associated with poor histological differentiation (r = 0.438, <it>p </it>= 0.025). The immunohistochemical staining showed an evident decrease of lamin A/C expression in 55.6% (70/126) GC cases. Importantly, the negative lamin A/C expression correlated strongly with histological classification (r = 0.361, <it>p </it>= 0.034). Survival analysis revealed that patients with lamin A/C downregulation have a poorer prognosis (<it>p </it>= 0.034). In addition, lamin A/C expression was found to be an independent prognostic factor by multivariate analysis.</p> <p>Conclusion</p> <p>Data of this study suggest that lamin A/C is involved in the pathogenesis of GC, and it may serve as a valuable biomarker for assessing the prognosis for primary GC.</p

    SPATIAL ORGANIZATION OF LAMIN A/C IN HODGKIN’S LYMPHOMA AND MULTIPLE MYELOMA

    Get PDF
    Genomic Instability is a complex phenomenon observed both at the DNA and the chromosome level. It is responsible for the generation of mutations and chromosomal rearrangements, which can potentially lead to tumor development. A common genomic instability feature found in some malignant tumors is the shortening of the telomeres, the end of the chromosomes. Shortening of telomeres leads to the formation of anaphase bridges, subsequent breakage and initiation new breakage-bridge-fusion (BFB) cycles. Classical Hodgkin’s lymphoma (cHL) and Multiple Myeloma (MM) are two blood tumor both characterized by genomic instability. cHL is a B-Cell lymphoma comprised of mononuclear Hodgkin cells (H) and bi- to multi-nucleated Reed-Sternberg (RS) cells, both characterized by numerical (nCIN) and structural chromosomal instability (sCIN). Transition from H to RS cells is correlated with aberrant number of mitotic spindles, aberrant 3D telomere organization, increase in the number of telomere aggregates and telomere loss. Advanced shortening of telomeres leads disruption of 3D interaction of the telomere with TRF2, a member of the shelterin proteins responsible for the protection of the chromosome ends from the DNA damage repair system. MM, an incurable plasma cell disorder, is also characterized by CIN, microsatellite instability (MSI), and telomere attrition. Lamin A/C, a nuclear envelope protein and component of the nuclear matrix, is important for the assembly of the mitotic spindles and for the maintenance of the 3D telomeres architecture by binding the shelterin protein TRF2. Due to lamin A/C involvement in the maintenance of genomic stability, our goal was to investigate lamin A/C 3D spatial organization in cHL and MM, and understand whether the localized uncapping of TRF2 from telomeres leads to their detachment from the nuclear matrix component lamin A/C causing genomic instability. Western blot analysis and qRT-PCR analysis revealed that H and RS are characterized by an overall overexpression of lamin A/C when compared to the B-lymphocytes control. Three dimensional (3D) fluorescent microscopy in cHL and MM cells revealed that the regular homogeneous lamin A/C pattern identified in normal activated B-lymphocytes, was replaced by an aberrant lamin A/C 3D spatial distribution characterized by the presence of internal lamin A/C structures. Different lamin A/C patterns where observed where the internal lamin A/C sub-divided the nucleus in to two or multiple compartments, suggesting a possible involvement of lamin A/C in the multi-nucleation process. To investigate lamin A/C involvement in the multinucleation process and transition from H to RS, the downregulation of lamin A/C with siRNA was performed. Downregulation of lamin A/C affected the transition from H to RS cells, as the number of RS cells found after lamin silencing was decreased. Telomere 3D structure and nuclear organization were also affected by the deregulation of lamin A/C. H and RS cells were also characterized by unbinding of telomere-TRF2-lamin A/C when compared to the healthy control. Lack of co-localization of telomere-TRF2-lamin A/C confirms lamin A/C involvement in genomic instability, and suggests that localized absence of TRF2 could be causal to the genome remodeling in cHL

    Identification of cyclin D3 as a new interaction partner of lamin A/C

    Get PDF
    Lamin A/C is a major component of the nuclear lamina. An intact nuclear lamina has been proposed to be necessary for muscle differentiation. Cyclin D3 is known to be upregulated in differentiated muscle cells and to form insoluble complexes with cell-cycle regulatory factors in these cells. We have examined the possibility of direct binding interactions between lamin A/C and cyclin D3 by in vitro binding assays and co-immunoprecipitation studies with muscle cells. Our results indicate that cyclin D3 binds specifically to amino acid residues 383–474 of lamin A/C and associates with lamin A/C in muscle cells. The identification of cyclin D3 as a novel binding partner of lamin A/C has important implications for a role for lamin A/C in muscle differentiation

    Lamin A/C is Dispensable to Mechanical Repression of Adipogenesis

    Get PDF
    Mesenchymal stem cells (MSCs) maintain the musculoskeletal system by differentiating into multiple lineages, including osteoblasts and adipocytes. Mechanical signals, including strain and low-intensity vibration (LIV), are important regulators of MSC differentiation via control exerted through the cell structure. Lamin A/C is a protein vital to the nuclear architecture that supports chromatin organization and differentiation and contributes to the mechanical integrity of the nucleus. We investigated whether lamin A/C and mechanoresponsiveness are functionally coupled during adipogenesis in MSCs. siRNA depletion of lamin A/C increased the nuclear area, height, and volume and decreased the circularity and stiffness. Lamin A/C depletion significantly decreased markers of adipogenesis (adiponectin, cellular lipid content) as did LIV treatment despite depletion of lamin A/C. Phosphorylation of focal adhesions in response to mechanical challenge was also preserved during loss of lamin A/C. RNA-seq showed no major adipogenic transcriptome changes resulting from LIV treatment, suggesting that LIV regulation of adipogenesis may not occur at the transcriptional level. We observed that during both lamin A/C depletion and LIV, interferon signaling was downregulated, suggesting potentially shared regulatory mechanism elements that could regulate protein translation. We conclude that the mechanoregulation of adipogenesis and the mechanical activation of focal adhesions function independently from those of lamin A/C

    Expression of Lamin A/C in early-stage breast cancer and its prognostic value

    Get PDF
    Purpose: Lamins A/C, a major component of the nuclear lamina, plays key roles in maintaining nuclear integrity, regulation of gene expression, cell proliferation and apoptosis. Reduced lamin A/C expression in cancer has been reported to be a sign of poor prognosis. However, its clinical significance in breast cancer remains to be defined. This study aimed to evaluate expression and prognostic significance of lamin A/C in early-stage breast cancer.Methods: Using immunohistochemical staining of tissue microarrays, expression of lamin A/C was evaluated in a large well-characterised series of early-stage operable breast cancer (n=938) obtained from Nottingham Primary Breast Carcinoma Series. Association of lamin A/C expression with clinicopathological parameters and outcome was evaluated.Results: Positive expression rate of lamin A/C in breast cancer was 42.2% (n=398). Reduced/loss of expression of lamin A/C was significantly associated with high histological grade (p [less than] 0.001), larger tumour size (p=0.004), poor Nottingham Prognostic Index (NPI) score (p [less than] 0.001), lymphovascular invasion (p=0.014) and development of distant metastasis (p=0.027). Survival analysis showed that reduced/loss of expression of lamin A/C was significantly associated with shorter breast cancer specific survival (p=0.008).Conclusion: This study suggests lamin A/C plays a role in breast cancer and loss of its expression is associated with variables of poor prognosis and shorter outcome

    Failure of lamin A/C to functionally assemble in R482L mutated familial partial lipodystrophy fibroblasts: altered intermolecular interaction with emerin and implications for gene transcription

    Get PDF
    Familial partial lipodystrophy is an autosomal dominant disease caused by mutations of the LMNA gene encoding alternatively spliced lamins A and C. Abnormal distribution of body fat and insulin resistance characterize the clinical phenotype. In this study, we analyzed primary fibroblast cultures from a patient carrying an R482L lamin A/C mutation by a morphological and biochemical approach. Abnormalities were observed consisting of nuclear lamin A/C aggregates mostly localized close to the nuclear lamina. These aggregates were not bound to either DNA-containing structures or RNA splicing intranuclear compartments. In addition, emerin did not colocalize with nuclear lamin A/C aggregates. Interestingly, emerin failed to interact with lamin A in R482L mutated fibroblasts in vivo, while the interaction with lamin C was preserved in vitro, as determined by coimmunoprecipitation experiments. The presence of lamin A/C nuclear aggregates was restricted to actively transcribing cells, and it was increased in insulin-treated fibroblasts. In fibroblasts carrying lamin A/C nuclear aggregates, a reduced incorporation of bromouridine was observed, demonstrating that mutated lamin A/C in FPLD cells interferes with RNA transcription
    • …
    corecore