HMG proteins: dynamic players in gene regulation and differentiation

https://doi.org/10.1016/j.gde.2005.08.007Get rights and content

Core histones package the genome into nucleosomes and control its accessibility to transcription factors. High mobility group proteins (HMGs) are, after histones, the second most abundant chromatin proteins and exert global genomic functions in establishing active or inactive chromatin domains. It is becoming increasingly clear that they also specifically control the expression of a limited number of genes. Moreover, they contribute to the fine tuning of transcription in response to rapid environmental changes. They do so by interacting with nucleosomes, transcription factors, nucleosome-remodelling machines, and with histone H1.

Introduction

Chromatin functions as an integrated platform to process a variety of endogenous and exogenous signals, which are transformed into operational instructions for the accessibility, retrieval and execution of the biological programs that are stored as four-letter information in the genome of eukaryotes. DNA and the core histones (H2A, H2B, H3 and H4) make up the basic chromatin unit, the nucleosome. Nucleosomes are modified, ‘remodelled’ and organized into high-order structures by huge and diverse protein complexes. They are also bound non-stoichiometrically and transiently by histone H1 and the high mobility group (HMG) proteins, ‘architectural’ factors that organize chromatin by appropriately bending and plasticizing DNA.

HMG proteins belong to three families (HMGA, HMGB and HMGN), members of each family having different structures but broadly similar functions. All HMGs are small nuclear proteins less than 30 kDa and undergo extensive post-translational modification. They bind to chromatin in a dynamic and reversible way, right inside nucleosomes (in the case of HMGN), to multiprotein complexes of transcription factors and cofactors (HMGA) or to both nucleosomes and transcription factors (HMGB). They have both global genomic functions in establishing active or inactive chromatin domains and specific control functions on a limited number of genes. HMGs compete with histone H1 for chromatin binding sites and affect its dynamics. The HMG–H1–nucleosome web of interactions might provide flexibility to chromatin structure and, most importantly, a fine tuning of gene transcription in response to rapid environmental changes.

The structure and dynamics of all nuclear macromolecular assemblages — from the individual nucleosomes to the nucleus as a whole — contain ‘epigenetic’ information: information that ultimately derives from a genetic (DNA sequence) source — although is not genetic in itself — and to which the environment and the history of the organism might contribute in significant ways. Epigenetic information is the spice that turns a uniform endowment of genetic information for all cells of a multicellular organism into a rich diversity of cell identities and functions.

A huge amount of work in the past 15 years has focused on epigenetic modification of DNA and core histones, and basic discovery continues at an unabated pace. The next simplest hierarchical level, the association to chromatin of architectural proteins, has been explored to a lesser degree. As far as it concerns the authors of this review, this is a good thing: a concise essay on HMGs can still be written. Likewise, the role of histone H1 was reviewed recently [1••].

Section snippets

HMG families

‘High mobility group’ proteins were discovered more than 30 years ago as acid-extractable components of chromatin that had high electrophoretic mobility [2]. There are three families of HMG proteins (Figure 1), which have been renamed with systematic reference to the domains they contain:

  • 1.

    HMGA proteins contain AT-hooks, nine amino acid segments that are unstructured in solution but bind AT-rich DNA stretches in the minor groove

  • 2.

    HMGB proteins contain HMG Boxes, 80 amino acid domains that bind into

HMGA

HMGAs are abundant early in embryonic life and are expressed at low levels in most adult cells [3].

HMGAs are directly involved in the transcriptional control of specific genes. They are key actors within enhanceosomes, which are complex assemblages of transcription factors and cofactors on nucleosome-free control regions of genes. The best-studied enhanceosome is situated on the interferon β (IFN-β) promoter: it contains HMGA1a together with the transcription factors NF-κB, IRF (interferon

Mammalian HMGBs

Mammalian HMGBs are characterized by two tandem HMG box domains followed by a long acidic tail (Figure 1). The two boxes are structurally similar, and their motions are completely independent; the acidic tail appears to be unstructured [24].

HMG box domains are present in many proteins, and contain three α-helices that fold up into a typical wedge shape, the concave surface of which can delve into the minor groove of DNA [25]. All HMG boxes, whether present alone, in tandem or embedded into

Non-mammalian HMGBs

Recently, our understanding of the transcriptional control exerted by mammalian HMGBs has been vastly increased by the study of HMG box proteins of Drosophila and yeast. Whereas HMGN proteins appear to be late inventions in evolution, and are typical of vertebrates only, HMGB proteins are as old as multicellular animals — even sponges have a clearly recognizable HMGB with two tandem HMG boxes [45]. Plants, fungi and unicellular eukaryotes rarely have proteins containing tandem HMGB boxes.

HMGN

HMGNs unfold higher-order chromatin structure and enhance several DNA-dependent activities: transcription, and DNA repair and replication [59]. HMGNs are the only non-histone proteins known to specifically bind inside the nucleosomes — between the histone core and the DNA; the H3 histone tail and H2B are involved in the interaction. Stretches of contiguous nucleosomes contain a uniform population of HMGN homodimers, either HMGN1 or HMGN2, but not mixed HMGN1/2 populations; HMGN1 and HMGN2 are

Conclusion: a web of architectural proteins

It has progressively become clear that HMGs (10 proteins, including the splice variants) and histone H1 (eight similar but subtly different proteins) play similar yet opposing roles within chromatin. Chromatin is an active and dynamic structure, which in living cells is constantly kept far from equilibrium by ATP-consuming machineries. The accessibility of DNA within chromatin depends at the most basic level on the localization and mobility of nucleosomes and on the post-translational

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

The authors thank Luca Sessa for critical reading of the manuscript. Research in the authors’ laboratory is supported by the Italian Association for Cancer Research (AIRC), Fondazione del Monte dei Paschi di Siena, Fondazione Cariplo, and the Ministries for Health and for Education, University and Research.

Glossary

Consensus half-site/full-site
Steroid hormone receptors (class I) bind preferentially as preformed homodimers to a hexameric core DNA sequence (HRE, hormone response element) arranged as an inverted repeat. In contrast, class II and orphan receptors can recognize the hexameric core HRE arranged as an inverted repeat, a direct repeat or a single HRE half-site. Class I receptors can also bind to half-sites, but with much reduced efficiency. Once bound to hormone and DNA, nuclear receptors enhance

References (70)

  • M. Kruppa et al.

    Nhp6, an HMG1 protein, functions in SNR6 transcription by RNA polymerase III in S. cerevisiae

    Mol Cell

    (2001)
  • G.S. Fragiadakis et al.

    Nhp6 facilitates Aft1 binding and Ssn6 recruitment, both essential for FRE2 transcriptional activation

    EMBO J

    (2004)
  • R. Belotserkovskaya et al.

    Facts about FACT and transcript elongation through chromatin

    Curr Opin Genet Dev

    (2004)
  • M. Bustin

    Chromatin unfolding and activation by HMGN* chromosomal proteins

    Trends Biochem Sci

    (2001)
  • S. Lehtonen et al.

    HMG-17 is an early marker of inductive interactions in the developing mouse kidney

    Differentiation

    (2001)
  • G.H. Goodwin et al.

    A new group of chromatin-associated proteins with a high content of acidic and basic amino acids

    Eur J Biochem

    (1973)
  • R. Sgarra et al.

    Nuclear phosphoproteins HMGA and their relationship with chromatin structure and cancer

    FEBS Lett

    (2004)
  • J. Martinez Hoyos et al.

    Identification of the genes up- and down-regulated by the high mobility group A1 (HMGA1) proteins: tissue specificity of the HMGA1-dependent gene regulation

    Cancer Res

    (2004)
  • M.Z. Whitley et al.

    A striking similarity in the organization of the E-selectin and beta interferon gene promoters

    Mol Cell Biol

    (1994)
  • H. Lewis et al.

    Cooperativity between two NF-κB complexes, mediated by high-mobility group protein I(Y), is essential for cytokine-induced expression of the E-selectin promoter

    Mol Cell Biol

    (1994)
  • N.R. Treff et al.

    Human KIT ligand promoter is positively regulated by HMGA1 in breast and ovarian cancer cells

    Oncogene

    (2004)
  • J.L. Attema et al.

    The human IL-2 gene promoter can assemble a positioned nucleosome that becomes remodeled upon T cell activation

    J Immunol

    (2002)
  • M.I. Darville et al.

    An octamer motif is required for activation of the inducible nitric oxide synthase promoter in pancreatic β-cells

    Endocrinology

    (2004)
  • D. Foti et al.

    Lack of the architectural factor HMGA1 causes insulin resistance and diabetes in humans and mice

    Nat Med

    (2005)
  • G. Baldassarre et al.

    Negative regulation of BRCA1 gene expression by HMGA1 proteins accounts for the reduced BRCA1 protein levels in sporadic breast carcinoma

    Mol Cell Biol

    (2003)
  • S. Battista et al.

    High mobility group A1 proteins downregulate RAG2 gene expression

    Biochem J

    (2005)
  • K. Rothermund et al.

    c-Myc-independent restoration of multiple phenotypes by two c-Myc target genes with overlapping functions

    Cancer Res

    (2005)
  • R. Reeves et al.

    Architectural transcription factor HMGI(Y) promotes tumor progression and mesenchymal transition of human epithelial cells

    Mol Cell Biol

    (2001)
  • D.D. Edberg et al.

    Dynamic and differential in vivo modifications of the isoform HMGA1a and HMGA1b chromatin proteins

    J Biol Chem

    (2005)
  • R. Sgarra et al.

    Increase of HMGA1a protein methylation is a distinctive characteristic of leukaemic cells induced to undergo apoptosis

    Cell Death Differ

    (2003)
  • M. Harrer et al.

    Dynamic interaction of HMGA1a proteins with chromatin

    J Cell Sci

    (2004)
  • R. Sgarra et al.

    Discovering high mobility group A molecular partners in tumour cells

    Proteomics

    (2005)
  • J. Liu et al.

    Hmga1 is required for normal sperm development

    Mol Reprod Dev

    (2003)
  • M.A. Tessari et al.

    Transcriptional activation of the cyclin A gene by the architectural transcription factor HMGA2

    Mol Cell Biol

    (2003)
  • S. Knapp et al.

    The long acidic tail of high mobility group box 1 (HMGB1) protein forms an extended and flexible structure that interacts with specific residues within and between the HMG boxes

    Biochemistry

    (2004)
  • Cited by (0)

    View full text