Review
Modulating transcription factor activity: Interfering with protein-protein interaction networks

https://doi.org/10.1016/j.semcdb.2018.07.019Get rights and content

Abstract

Biophysical parameters that govern transcription factors activity are binding locations across the genome, dwelling time at these regulatory elements and specific protein-protein interactions. Most molecular strategies used to develop small compounds that block transcription factors activity have been based on biochemistry and cell biology methods that that do not take into consideration these key biophysical features. Here, we review the advance in the field of transcription factor biology and describe how their interactome and transcriptional regulation on a genome wide scale have been deciphered. We suggest that this new knowledge has the potential to be used to implement innovative research drug discovery program.

Introduction

Transcription factors (TFs) are proteins that coordinate gene expression in a spatial and temporal manner in specific cell types. These proteins act as the control panel of the genome, finely tuning transcriptional regulation and directly shaping organism phenotypes by inducing or maintaining cell fate [1]. Over the past four decades, TFs have been grouped mostly according to their homology domains and phylogenetic conservation [2,3]. However, recently a novel classification has emerged based on TF’s ability to modify the chromatin landscape. This alternative ontology is based on three functional features: local chromatin access (pioneer TFs), self-sustained remodelling (settler TFs) and sporadic occupancy/selective gene activation (migrant TFs) [4,5]. Such a classification relies on two key parameters: sequential protein-protein and protein-DNA interactions which are both essential to modulating gene activation or repression. In addition to this functional classification, TFs display other molecular characteristics that deeply influence their activity:

  • 1)

    TFs are part of distinct regulatory networks that also elicit multiple simultaneous protein-protein interactions (PPIs).

  • 2)

    TFs interactions are interchangeable, facilitated mostly by their intrinsically disordered structural characteristics.

  • 3)

    The nature of TFs protein-protein interactions and their affinity for DNA binding motifs defines gene target selectivity and chromatin binding dynamics.

In this review, we explain why knowledge around these individual components are essential for successful pharmacological targeting of TFs. We propose here a molecular strategy designed to disrupt TF activity via simultaneous interference of multiple protein-protein interactions.

Section snippets

Defining TFs interactomes

Transcription factors are embedded into PPI networks which enable them to display a tightly coordinated activity at enhancers or promoters. These interactions involve other TFs, chromatin remodellers and co-factors to modulate a specific transcriptional response at a precise time window depending on specific cell types. Within these PPIs networks, some TFs play a central role by acting as communication hubs in combination with direct and indirect recruitment of protein partners that establish

Redundancy mechanisms

One of the major characteristics of TFs is their ability to share functional redundancy. In yeast model systems, it has been shown that TF genetic disruption only affects 3% of their putative target [27]. This phenomenon is due to the fact that closely related members of the same phylogenetic group can act interchangeably in a loss of function scenario, to partially rescue the missing TF. This has been defined as paralogue connectivity in yeast [28]. There are some examples of TF redundancy in

Current status of transcription factors pharmacomodulation

The dogma that pharmacological modulation of TFs is near impossible, is progressively being challenged as illustrated by the rising number of reports on TFs chemical inhibition [[36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48]]. Until now, a major concern of developing a drug targeting a specific TF was the risk of interfering with the expression levels of potentially hundreds of target genes. The recent explosion of knowledge in genomics has presented opportunities

The landscape of TF pharmacomodulation in industry

Pharmacomodulation of TF activity can be achieved by modulating their expression (pre-or post-transcriptionally), changing nuclear translocation dynamics, impeding DNA binding activity or interfering with protein partner recruitment. These different avenues of intervention are attempted mostly on nuclear receptors such as the estrogen or androgen receptors, signal transducers such as STAT3, more recently NOTCH effector RBPJ. Other targets have been tumour suppressors (p53), oncogenes (cMyc,

Conclusion

Many TFs are implicated in the aetiology of human rare diseases, which collectively represent large cohorts of patients. This is a strong indicator that these molecular hubs are viable molecular targets relevant to human health with a huge therapeutic potential that has remained untapped so far. It has been nearly 5 decades since the discovery of the first transcription factor and the idea of drugging this class of protein has been around ever since. What has dramatically changed is the

Acknowledgment

This work was supported by grants from the NHMRC (APP1107643 to MF). MF is supported by a NHMRC Career Developmental Fellowship (APP1111169).

References (86)

  • F. Spitz et al.

    Transcription factors: from enhancer binding to developmental control

    Nat. Rev. Genet.

    (2012)
  • E. Wingender

    Classification of eukaryotic transcription factors

    Mol. Biol. (Mosk.)

    (1997)
  • E. Wingender et al.

    TFClass: a classification of human transcription factors and their rodent orthologs

    Nucleic Acids Res.

    (2015)
  • S. Rieck et al.

    PIQ-ing into chromatin architecture

    Nat. Biotechnol.

    (2014)
  • R.I. Sherwood

    Discovery of directional and nondirectional pioneer transcription factors by modeling DNase profile magnitude and shape

    Nat. Biotechnol.

    (2014)
  • A.J. Walhout

    Unraveling transcription regulatory networks by protein-DNA and protein-protein interaction mapping

    Genome Res.

    (2006)
  • H. Yu

    High-quality binary protein interaction map of the yeast interactome network

    Science

    (2008)
  • D. Park

    Comparative interactomics analysis of protein family interaction networks using PSIMAP (protein structural interactome map)

    Bioinformatics

    (2005)
  • M.F. Berger

    Variation in homeodomain DNA binding revealed by high-resolution analysis of sequence preferences

    Cell

    (2008)
  • A. Bartlett

    Mapping genome-wide transcription-factor binding sites using DAP-seq

    Nat. Protoc.

    (2017)
  • D.S. Johnson et al.

    Genome-wide mapping of in vivo protein-DNA interactions

    Science

    (2007)
  • A. Jolma

    DNA-dependent formation of transcription factor pairs alters their binding specificity

    Nature

    (2015)
  • T. Ravasi

    An atlas of combinatorial transcriptional regulation in mouse and man

    Cell

    (2010)
  • J.P. Lambert et al.

    A novel proteomics approach for the discovery of chromatin-associated protein networks

    Mol. Cell Proteomics

    (2009)
  • H. Mohammed

    Endogenous purification reveals GREB1 as a key estrogen receptor regulatory factor

    Cell Rep.

    (2013)
  • J. Overman

    Pharmacological targeting of the transcription factor SOX18 delays breast cancer in mice

    Elife

    (2017)
  • P.E. Wright et al.

    Intrinsically disordered proteins in cellular signalling and regulation

    Nat. Rev. Mol. Cell Biol.

    (2015)
  • A.C. Ferreon et al.

    Modulation of allostery by protein intrinsic disorder

    Nature

    (2013)
  • J.M. Vaquerizas et al.

    A census of human transcription factors: function, expression and evolution

    Nat. Rev. Genet.

    (2009)
  • K. Xia et al.

    Impacts of protein-protein interaction domains on organism and network complexity

    Genome Res.

    (2008)
  • J. Chen

    Single-molecule dynamics of enhanceosome assembly in embryonic stem cells

    Cell

    (2014)
  • E.E. Swinstead

    Steroid receptors reprogram FoxA1 occupancy through dynamic chromatin transitions

    Cell

    (2016)
  • S.C. Biddie

    Transcription factor AP1 potentiates chromatin accessibility and glucocorticoid receptor binding

    Mol. Cell

    (2011)
  • T.C. Voss

    Dynamic exchange at regulatory elements during chromatin remodeling underlies assisted loading mechanism

    Cell

    (2011)
  • H.J. Dyson et al.

    Coupling of folding and binding for unstructured proteins

    Curr. Opin. Struct. Biol.

    (2002)
  • R.S. Spolar et al.

    Coupling of local folding to site-specific binding of proteins to DNA

    Science

    (1994)
  • W.S. Wu et al.

    Functional redundancy of transcription factors explains why most binding targets of a transcription factor are not affected when the transcription factor is knocked out

    BMC Syst. Biol.

    (2015)
  • R. Kafri et al.

    Preferential protection of protein interaction network hubs in yeast: evolved functionality of genetic redundancy

    Proc. Natl. Acad. Sci. U. S. A.

    (2008)
  • B. Hosking

    Sox7 and Sox17 are strain-specific modifiers of the lymphangiogenic defects caused by Sox18 dysfunction in mice

    Development

    (2009)
  • M. Maconochie et al.

    Paralogous Hox genes: function and regulation

    Annu. Rev. Genet.

    (1996)
  • C. Zhang

    A clustering property of highly-degenerate transcription factor binding sites in the mammalian genome

    Nucleic Acids Res.

    (2006)
  • I. Maeso et al.

    Deep conservation of cis-regulatory elements in metazoans

    Philos. Trans. R. Soc. Lond. B: Biol. Sci.

    (2013)
  • S. Domene

    Enhancer turnover and conserved regulatory function in vertebrate evolution

    Philos. Trans. R. Soc. Lond. B: Biol. Sci.

    (2013)
  • E. Cannavo

    Shadow enhancers are pervasive features of developmental regulatory networks

    Curr. Biol.

    (2016)
  • D.M. Wellik et al.

    Hox10 and Hox11 genes are required to globally pattern the mammalian skeleton

    Science

    (2003)
  • H.D. Arndt

    Small molecule modulators of transcription

    Angew. Chemie

    (2006)
  • T. Berg

    Inhibition of transcription factors with small organic molecules

    Curr. Opin. Chem. Biol.

    (2008)
  • F. Bernal

    A stapled p53 helix overcomes HDMX-mediated suppression of p53

    Cancer Cell

    (2010)
  • A.L. Kung

    Small molecule blockade of transcriptional coactivation of the hypoxia-inducible factor pathway

    Cancer Cell

    (2004)
  • H. Li

    Discovery of small-molecule inhibitors selectively targeting the DNA-binding domain of the human androgen receptor

    J. Med. Chem.

    (2014)
  • K. Miyoshi

    Stat3 as a therapeutic target for the treatment of psoriasis: a clinical feasibility study with STA-21, a Stat3 inhibitor

    J. Invest. Dermatol.

    (2011)
  • K.A. Siddiquee

    An oxazole-based small-molecule Stat3 inhibitor modulates Stat3 stability and processing and induces antitumor cell effects

    ACS Chem. Biol.

    (2007)
  • L.T. Vassilev

    In vivo activation of the p53 pathway by small-molecule antagonists of MDM2

    Science

    (2004)
  • Cited by (39)

    • Genome-wide analysis of MdABF Subfamily and functional identification of MdABF1 in drought tolerance in apple

      2022, Environmental and Experimental Botany
      Citation Excerpt :

      The target gene of this study, MdABF1 had the highest homology with AtABF3 (Fig. 1A) and localized to the nucleus of Arabidopsis roots (Fig. 4) which was coincident with localization in Populus euphratica (Y.Y. Yang, Li et al., 2020; Y. Yang, Li et al., 2020). This result indicated that MdABF1 may function as a positive regulator of downstream gene expression (Francois et al., 2020). It is reported that ABF transcription factors can respond to drought stress through ABA signaling transduction (Fujita et al., 2005).

    View all citing articles on Scopus
    View full text