Research Article
Toward a generalized computational workflow for exploiting transient pockets as new targets for small molecule stabilizers: Application to the homogentisate 1,2-dioxygenase mutants at the base of rare disease Alkaptonuria

https://doi.org/10.1016/j.compbiolchem.2017.08.008Get rights and content

Highlights

  • A new approach to the treatment of Alkaptonuria is proposed by use of Pharmacological Chaperones (PCs).

  • Transient pockets at the surface of mutated enzyme are exploited as targets for PC.

  • A workflow employing ready-to-use tools is proposed.

Abstract

Alkaptonuria (AKU) is an inborn error of metabolism where mutation of homogentisate 1,2-dioxygenase (HGD) gene leads to a deleterious or misfolded product with subsequent loss of enzymatic degradation of homogentisic acid (HGA) whose accumulation in tissues causes ochronosis and degeneration. There is no licensed therapy for AKU. Many missense mutations have been individuated as responsible for quaternary structure disruption of the native hexameric HGD. A new approach to the treatment of AKU is here proposed aiming to totally or partially rescue enzyme activity by targeting of HGD with pharmacological chaperones, i.e. small molecules helping structural stability. Co-factor pockets from oligomeric proteins have already been successfully exploited as targets for such a strategy, but no similar sites are present at HGD surface; hence, transient pockets are here proposed as a target for pharmacological chaperones. Transient pockets are detected along the molecular dynamics trajectory of the protein and filtered down to a set of suitable sites for structural stabilization by mean of biochemical and pharmacological criteria. The result is a computational workflow relevant to other inborn errors of metabolism requiring rescue of oligomeric, misfolded enzymes.

Introduction

Pharmacological chaperones (PCs) are small molecules designed to facilitate the correct folding of a protein and to re-establish its functionality (Aymami et al., 2013). Their role is similar to physiological chaperones: instead of macromolecules they are small molecules with the task of stabilizing an already folded protein through binding (Petsko et al., 2009) and subsequent increase in non-covalent interactions among protein structural segments. PCs, obeying to the Lipinski rule of five (Lipinski, 2009), have a low molecular weight giving the advantage of being administered orally with a non-invasive treatment (Leeson and Springthorpe, 2007).

PCs should not be confused with chemical chaperones. The latter are non-specific molecules, that exert their stabilization action on protein structures without binding to a specific site; trehalose (Crowe, 2007) and glycerol (Ohnishi et al., 1999) are well known examples. The non-specific mechanism of action of chemical chaperones requires a high concentration for them be effective, preventing their use as therapeutics.

Most of the PCs currently in clinical trial or already registered as therapeutics are designed to target active (Germain and Fan, 2009) or co-factor binding sites (Santos-Sierra et al., 2012). They are analogues of physiological substrates or co-factors, therefore binding with high affinity and selectivity to the target binding site.

It has been shown that the binding of an inhibitor to an enzyme may stabilize the protein against denaturation, leading to a greater stability of the structure (Ringe and Petsko, 2009), as proven also by the improvement in the crystallization process of inhibitor-bound enzymes (Hassel et al., 2007). Thus, enzyme inhibitors are good candidates as PCs provided that they are reversible competitive, hence allowing for the presence of a small amount of free enzyme to be available for substrate binding. Again, especially in the case of severely destabilized structures, a very high concentration of such inhibitor acting as PC would be necessary, shifting the equilibrium toward the bound, inactive form of the enzyme, thus neutralizing the stabilization effect.

Currently, use of co-factor sites as targets for PC designed as analogues of natural binders has proven to be the most suitable strategy in small molecule-assisted stabilization of protein structures, as for the case of Tafamidis and Transthyretin (TTR) (Bulawa et al., 2012). Transthyretin-related hereditary amyloidosis (ATTR) is an autosomal neurodegenerative disease caused by mutations in the TTR gene leading to loss of three-dimensional structure at both tertiary and quaternary level, which in turn leads to protein aggregation causing cell death. The commercially available PC Tafamidis exploits the co-factor binding site for thyroxine (T4) located at the protomer–protomer interface, increasing the number of inter-chain interactions, which in turn stabilize the tetramer structure and prevent aggregation. Such example well illustrates why modulation of protein–protein interface by PCs is becoming a new important branch in drug discovery, especially for diseases characterized by quaternary structure instability (Bier et al., 2015, Makley and Gestwicki, 2013)

Unfortunately, co-factor sites are not always available in protein structures while PCs may, ideally, bind anywhere on the surface of a protein to give stabilization; hence, the search for alternative, non-biologically active binding sites is needed (Metz et al., 2012). A test case is possibly the hexameric homogentisate 1,2-dioxygenase (HGD), whose activity deficiency due to gene mutations leads to Alkaptonuria (AKU), an inborn error of metabolism resulting in the accumulation of homogentisic acid (HGA) and ochronosis (Mitri et al., 2017, Braconi et al., 2015, Braconi et al., 2017). AKU is a multisystemic disease (Bernardini et al., 2015) in which also secondary amyloidosis (Millucci et al., 2012, Millucci et al., 2014a, Millucci et al., 2014b) and angiogenesis occur (Millucci et al., 2016), with an easy diagnosis (Jacomelli et al., 2017, Millucci et al., 2014c) but no current licensed therapy (Ranganath et al., 2016). Several missense mutations have been individuated as responsible for HGD quaternary structure disruption (see Fig. 1) (Ranganath et al., 2016). Contrarily to TTR, no co-factor sites are present at HGD enzyme surface to be exploited for PC targeting, therefore alternative approaches must be sought.

A new approach to the treatment of AKU is here proposed by targeting the mutated HGD by PCs aiming to hexamer stabilization, seeking for alternative pockets showing along the molecular dynamics simulation of the protein in a transient fashion, i.e. transient pockets (Bernini et al., 2014).

Section snippets

Molecular dynamics simulations

Molecular dynamics simulations were carried out in the AMBER99 sb force field employing the Gromacs software suite (Hess et al., 2008). For human HGD the X-ray crystallography structure with PDB ID: 1EY2 was used after reconstruction of missing segments by using P. putida X-ray crystallography structure (PDB ID: 4AQ2) as template. For method validation, human C220Y p53 crystal structure was used (PDB ID: 2J1X). Proteins metal centers (Fe(II) and Zn(II) for HGD and p53, respectively) were

Method set up and validation

The pocket detection method here proposed was first set-up and validated on p53 protein. P53 is a transcription factor that regulates the cell cycle acting as a tumour suppressor (Culmsee et al., 2001). The tumour appearance is always related to missense mutations on p53 gene and, therefore, to perturbations in its regulatory pathways. In particular, the cancer-related mutation Y220C of p53 has been studied in depth (Joerger and Fersht, 2007); here, the replacement of a surface tyrosine with

Conclusions

AKU is an ultra-rare multisystemic disease due to HGD mutations and characterized by a high degree of complexity. Such features lead to a difficulty in finding a cure and the development of novel drugs (Santucci et al., 2017, Laschi et al., 2016). As a strategy to overcome such obstacles, computational techniques can be implemented at low costs in the drug discovery pipeline, making the development of therapies for ultra-rare disease more affordable.

In the present study, several different

Supporting information

The following files are available free of charge. Supplementary figures and tables. PDB file of HGD/small molecules structures reported in Table 4 and Fig. 6.

Competing interests

The authors declare that they have no competing interest.

Ethics statement

The authors declare that no human subjects or human data are used in this study.

Founding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

References (63)

  • G. Bernardini et al.

    Homogentisate 1,2 dioxygenase is expressed in brain: implications in alkaptonuria

    J. Inherit. Metab. Dis.

    (2015)
  • D. Bier et al.

    Small-Molecule stabilization of the 14-3-3/Gab2 protein-protein interaction (PPI) interface

    Chem. Med. Chem.

    (2015)
  • A. Borrel et al.

    PockDrug: a model for predicting pocket druggability that overcomes pocket estimation uncertainties

    J. Chem. Inf. Model.

    (2015)
  • G.P. Brady et al.

    Fast prediction and visualization of protein binding pockets with PASS

    J. Comput. Aided Mol. Des. P. F.W.

    (2000)
  • D. Bromley et al.

    An in silico algorithm for identifying stabilizing pockets in proteins: test case, the Y220C mutant of the p53 tumor suppressor protein

    Protein Eng. Des. Sel.

    (2016)
  • C.E. Bulawa et al.

    Tafamidis, a potent and selective transthyretin kinetic stabilizer that inhibits the amyloid cascade

    Proc. Natl. Acad. Sci.

    (2012)
  • J.H. Crowe

    Trehalose as a chemical chaperone

    Mol. Aspects Stress Response: Chaperones, Membr. Networks

    (2007)
  • C. Culmsee et al.

    A synthetic inhibitor of p53 protects neurons against death induced by ischemic and excitotoxic insults, and amyloid β-peptid

    J. Neurochem.

    (2001)
  • W.L. DeLano

    PyMOL(TM) Molecular Graphics System Version 1.7.2.1

    (2014)
  • D.A. Erlanson et al.

    Ligand efficiency metrics and their use in fragment optimizations

    Fragment-Based Drug Discov.

    (2016)
  • S. Eyrisch et al.

    Transient pockets on protein surfaces involved in protein–protein interaction

    J. Med. Chem.

    (2007)
  • S. Genheden et al.

    The MM/PBSA and MM/GBSA methods to estimate ligand-binding affinities

    Expert Opin. Drug Discov.

    (2015)
  • D.P. Germain et al.

    Pharmacological chaperone therapy by active-site-specific chaperones in Fabry disease: in vitro and preclinical studies

    Int. J. Clin. Pharmacol. Ther.

    (2009)
  • D. Ghersi et al.

    Improving accuracy and efficiency of blind protein-ligand docking by focusing on predicted binding sites

    Proteins

    (2009)
  • A.K. Ghose et al.

    A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases

    J. Comb. Chem.

    (1999)
  • A.M. Hassel et al.

    Crystallization of protein–ligand complexe

    Acta Crystallogr. D Biol. Crystallogr.

    (2007)
  • B. Hess et al.

    Gromacs 4: Algorithms for highly efficient, load-balanced, and scalable molecular simulation

    J. Chem. Theory Comput.

    (2008)
  • H.A. Hussein et al.

    PockDrug-Server: a new web server for predicting pocket druggability on holo and apo proteins

    Nucleic Acids Res.

    (2015)
  • G. Jacomelli et al.

    Quick diagnosis of alkaptonuria by homogentisic acid determination in urine paper spots

    JIMD Rep.

    (2017)
  • J.H. Jeoung et al.

    Visualizing the substrate-, superoxo-, alkylperoxo-, andproduct-bound states at the nonheme Fe(II) site of homogentisate dioxygenase

    Pnas

    (2013)
  • A.C. Joerger et al.

    Structure–function–rescue: the diverse nature of common p53 cancer mutant

    Oncogene

    (2007)
  • Cited by (15)

    • Transient pockets as mediators of gas molecules routes inside proteins: The case study of dioxygen pathway in homogentisate 1,2-dioxygenase and its implication in Alkaptonuria development

      2020, Computational Biology and Chemistry
      Citation Excerpt :

      In this protein, the O2 is supposed to reach the active site trough the central opening placed along the threefold axis of the hexamer allowing for the transport of the gas from the surface to the core of the quaternary structure followed by diffusion toward the active site through a hydrophobic pore connecting the central opening with the deep end of the active site, although no description of the mechanism at atomic level has been provided yet (Jeoung et al., 2013). Our previous work on HGD showed how the protein dynamics is conducive of the opening of transient pockets with remarkable druggability (Bernini et al., 2017) (Fig. 1). Furthermore, a transient pocket was found to be located between the central opening and the deep end of the active site.

    • AKUImg: A database of cartilage images of Alkaptonuria patients

      2020, Computers in Biology and Medicine
      Citation Excerpt :

      In this context, behavioral/clinical data and image repositories can constitute useful guides to generate an exhaustive and dynamic picture of the individual, to identify new potential biomarkers, to collect and share information derived from different research groups. AKU is an ultra-rare autosomal recessive metabolic disease [3] with a very low prevalence (1:1000000-250000) [4], caused by mutation in the structure of homogentisate 1,2-dioxygenase (HGD) [4,5], an enzyme involved in the metabolism of the two amino acids tyrosine and phenylalanine, precisely in the step of oxidation of homogentisic acid (HGA) to maleyl acetoacetate. The deficient activity of HGD enzyme leads to the accumulation of HGA, which undergoes oxidation and polymerization, forming a dark-brown pigmentation in different connective tissues with a phenomenon called “ochronosis”.

    • Cell and tissue models of alkaptonuria

      2020, Drug Discovery Today: Disease Models
      Citation Excerpt :

      The activity of antioxidants [20,26,31] and methotrexate [24] was assayed in vitro to evaluate their role in counteracting HGA-related oxidative stress and amyloidosis, respectively. Furthermore, the activity of nitisinone analogues was tested in vitro [49], and an silico approach highlighted potential binding sites for drugs acting as pharmacological chaperones for the enzyme HGD [50] (Fig. 1). Similarities between AKU and OA at the molecular level were found [38,46,47], which reinforced the significance of findings in AKU and the potential application of similar workflows for more common diseases.

    • Alkaptonuria

      2024, Nature Reviews Disease Primers
    View all citing articles on Scopus
    1

    First authors.

    View full text