Elsevier

Experimental Cell Research

Volume 316, Issue 7, 15 April 2010, Pages 1190-1201
Experimental Cell Research

Research Article
Involvement of ZFPIP/Zfp462 in chromatin integrity and survival of P19 pluripotent cells

https://doi.org/10.1016/j.yexcr.2010.02.024Get rights and content

Abstract

Toti- or pluripotent cells proliferation and/or differentiation have been shown to be strongly related to nuclear chromatin organization and structure over the last past years.

We have recently identified ZFPIP/Zfp462 as a zinc finger nuclear factor necessary for correct cell division during early embryonic developmental steps of vertebrates. We thus questioned whether this factor was playing a general role during cell division or if it was somehow involved in embryonic cell fate or differentiation.

To achieve this goal, we performed a knock-down experiment in the pluripotent P19 and differentiated 3T3 cell lines, both expressing endogenous ZFPIP/Zfp462. Using specific shRNA directed against ZFPIP/Zfp462 transcripts, we demonstrated that depletion of this protein induced cell death in P19 but had no effect in 3T3 cells. In addition, in the absence of the protein, the P19 cells exhibited a complete destructuration of pericentromeric domains associated with a redistribution of the HP1α proteins and an increase in DNA satellites transcribed RNAs level.

These data suggested an instrumental role of ZFPIP/Zfp462 in maintaining the chromatin structure of pluripotent cells.

Introduction

We have recently identified ZFPIP/Zfp462 as a vertebrate-specific gene encoding a nuclear zinc finger protein that strongly interacts with Pbx1 [1]. The 250 kDa ZFPIP/Zfp462 protein contains a strikingly important number (34) of C2H2 zinc fingers distributed throughout the sequence and a putative Nuclear Localization Signal (NLS). Previous data have shown that ZFPIP/Zfp462 mRNA is expressed during development in the mouse with a maximum yield between E10.5 to E12.5. This expression is particularly abundant in brain, somites and limbs buds [1], [2]. ZFPIP/Zfp462 appeared to be an essential developmental gene in Xenopus laevis. Indeed, knocked-down expression of ZFPIP/Zfp462 in X. laevis embryos leads to a rapid cell division arrest coupled with important nuclear chromatin disorders: mitotic cells undergo abnormal mitosis, with aberrant metaphase, anaphase, incomplete chromosome segregation or conjoined nuclei [3]. Furthermore, decrease of ZFPIP/Zfp462 levels seems to impair cell cycle, inducing cell death with a mechanism described as “mitotic catastrophe” [4].

Analysis of the chromatin/cell division disturbance in whole animal using morpholinos remains difficult because of the important cellular variability in developing embryos [5]. Therefore, analysis of such mechanism in a cell culture model is an alternative approach. To this aim, we sought for a model of ZFPIP/Zfp462 expressing cell line and selected out the murine embryonal carcinoma P19 cell line which has already been widely used to study early events of embryonic differentiation [6], [7]. Indeed, these cells resemble the inner cell mass of the early embryo and can differentiate into different cell types according to the culture conditions, such as fibroblastic, muscular or neural cells [8], [9].

With this work, we demonstrated that ZFPIP/Zfp462 expression was cell-specific. Indeed, the protein was detected in P19, ES or 3T3 whereas no expression of the gene was detected in several vertebrate cell lines. Using a shRNA approach, we demonstrated that depletion of the protein in the P19 embryonic cells induced abnormal nucleus phenotype that was not observed in differentiated depleted 3T3 cells. Moreover, ZFPIP/Zfp462-depleted P19 cells exhibited disruption of nuclear heterochromatin, correlated to a delocalization of HP1α and an increase of DNA satellites transcription. Following these abnormal nuclear cell processes, the P19 cells died within 24 h.

These results demonstrated that (i) the absence of ZFPIP/Zfp462 led to a disorganization of pericentromeric domains in P19 cells which were subsequently not able to divide correctly and that (ii) the gene appeared vital in P19 pluripotent cells but not in 3T3 cells. The overall data clearly showed that ZFPIP/Zfp462 was an essential gene involved in chromatin structure modelling mechanisms occurring during cell division of pluripotent cells.

Section snippets

Plasmid constructs

The pCMV-N-ZFPIP and pCMV-C-ZFPIP vectors correspond to the N-terminal and the C-terminal regions (respectively N-ZFPIP and C-ZFPIP) of the human ZFPIP/Zfp462 encoding cDNA cloned into BamHI sites of the pCMVflag6c vector (Sigma) as described in [1]. The full-length human ZFPIP/Zfp462 cDNA was purified from the pT7TS-ZFPIP construct [3] and cloned into EcoRV and Acc65I sites of the pCMV-flag6b vector (pCMV-FL-ZFPIP).

The eukaryotic expression vector pEGFP-C3 (pGFP) from Clontech was used for

ZFPIP/Zfp462 is endogenously expressed in P19, 3T3 and mES cells

In order to identify a relevant cell line to our studies, we first performed quantitative RT-PCR experiments, using ZFPIP/Zfp462 mouse-specific primers [1] and RNA extracted from various cell lines. As shown in Fig. 1A, ZFPIP/Zfp462 mRNAs were detected in P19, 3T3 or ES cells in equivalent amounts to those observed in mouse E9.5 embryos. In contrast, none or a low amount of ZFPIP/Zfp462 mRNAs was detected in other mouse cell lines such as B16-F1 melanoma or K1737.

To complement these data,

Discussion

In the present work, we demonstrated that ZFPIP/Zfp462 knock-down was lethal for pluripotent P19 cells that died following important chromatin alterations through a non-apoptotic mechanism.

Acknowledgments

The authors thank Dr. Christian Jaulin for critical review of the manuscript and insightful discussion. Also, thank you to Drs. Michel Cohen-Tanoudji and Teddy Léguillier for providing mES cells. The authors thank Dr. Claire Piquet-Pellorce for its help in FACS experiments and Dr. Stéphanie Dutertre from the microcopy plateform of IFR 140.

JM and AL were supported by a grant from the “Conseil Régional de Bretagne.” This work was supported by the CNRS and by grants from “Rennes Métropole” and the

References (39)

  • M.V. Blagosklonny

    Mitotic arrest and cell fate: why and how mitotic inhibition of transcription drives mutually exclusive events

    Cell Cycle

    (2007)
  • D.R. Corey et al.

    Morpholino antisense oligonucleotides: tools for investigating vertebrate development

    Genome Biol.

    (2001)
  • M.W. McBurney

    P19 embryonal carcinoma cells

    Int. J. Dev. Biol.

    (1993)
  • B.F. Andrews

    Use of stem cells

    J. Ky. Med. Assoc.

    (2002)
  • E.M. Jones-Villeneuve et al.

    Retinoic acid induces embryonal carcinoma cells to differentiate into neurons and glial cells

    J. Cell Biol.

    (1982)
  • J. Pachernik et al.

    Neural differentiation of pluripotent mouse embryonal carcinoma cells by retinoic acid: inhibitory effect of serum

    Physiol. Res.

    (2005)
  • C. Kress et al.

    Nonpermissiveness for mouse embryonic stem (ES) cell derivation circumvented by a single backcross to 129/Sv strain: establishment of ES cell lines bearing the Omd conditional lethal mutation

    Mamm. Genome

    (1998)
  • A. Laurent et al.

    Interaction of ZFPIP with PBX1 is crucial for proper expression of neural genetic markers during Xenopus development

    Dev. Growth Differ.

    (2009)
  • J.F. Lima-Neto et al.

    Viability and cell cycle analysis of equine fibroblasts cultured in vitro

    Cell Tissue Bank

    (2009)
  • Cited by (7)

    • Acute lymphoblastic leukemia in a child with Weiss-Kruszka syndrome: Casual or causal association?

      2022, European Journal of Medical Genetics
      Citation Excerpt :

      Among these patients, the most common phenotypical findings were dysmorphic features, intellectual disabilities and cardiac defects. Zinc finger protein 462, a C2H2 type zinc finger transcription factor, is a relatively newly discovered vertebrate specific protein involved in the pluripotency and differentiation of embryonic stem cells by regulating SOX2, POU5F1/OCT4, and NANOG; by binding PBX1, ZNF462 prevents the heterodimerization of PBX1 and HOXA9 and their binding to DNA; ZNF462 regulates also neuronal development and neural cell differentiation (Nagase et al., 2001; Masse et al., 2010). For these motives, ZNF462 is believed to play an important role in embryonic development, especially in brain development, and it controls early patterning of the central nervous system (Cosemans et al., 2018; Laurent et al., 2009; Wang et al., 2017).

    • ZFPIP/Zfp462 is involved in P19 cell pluripotency and in their neuronal fate

      2011, Experimental Cell Research
      Citation Excerpt :

      While the structural role of ZFPIP/Zfp462 is difficult to analyze and is yet unknown, we have observed that it is associated with HP1 proteins. Indeed, our previous work showed that ZFPIP/Zfp462 is involved in HP1 distribution and chromocenter organization [17]. We can thus hypothesize that ZFPIP/Zfp462 might also be linked to the epigenetic machinery of the cell through the PcG repressive complexes signaling pathway.

    • Phenotype delineation of ZNF462 related syndrome

      2019, American Journal of Medical Genetics, Part A
    View all citing articles on Scopus
    View full text