Drug Discovery: From worms to fish to mice

An multi-species approach can be used to identify small molecules with properties that might prove useful for the treatment of some neuromuscular diseases.
  1. Guy M Benian  Is a corresponding author
  2. Hyojung J Choo
  1. Department of Pathology, Emory University, United States
  2. Department of Cell Biology, Emory University, United States

Muscle contraction is a complicated process that starts with an electrical signal called an action potential entering a muscle cell, and ends with thin filaments sliding past thick filaments to make the cell shorter. A central part of this process involves the action potential activating a protein called a dihydropyridine (DHP) receptor, which then activates a calcium-ion channel called a ryanodine receptor. The activation of this channel in an organelle called the sarcoplasmic reticulum results in the release of calcium ions (Ca2+) into the cytoplasm of the cell. These ions go on to activate muscle contraction through a mechanism that involves promoting the interaction between thick and thin filaments (see Figure 1A).

Muscle contraction and a 'multi-species discovery pipeline' for drug screening.

(A) Ryanodine receptor 1 (RyR1; blue) is a channel protein that releases calcium ions (yellow) from the sarcoplasmic reticulum (SR) when activated by a dihydropyridine receptor (DHP; green). Muscle tissue contains thick filaments (blue) and thin filaments (orange) arranged in units called sarcomeres. The calcium ions cause structures on the thick filaments called myosin heads (not shown) to bind to the thin filaments and pull them so that the muscle contracts. The DHP receptor is activated by action potentials (red spikes) travelling along a structure called a T-tubule (blue). (B) Drug screening was performed sequentially using null mutants in C. elegans (top), zebrafish (middle), and mouse myotubes (bottom). A screen of 3700 compounds in C. elegans identified 74 compounds that enabled the mutants to grow. Testing many of these 74 compounds on zebrafish revealed two compounds that improved the swimming performance of the mutant animals. Both compounds were known to be inhibitors of a protein kinase called p38, and both were found to induce the release of calcium ions in mutant mouse muscle cells.

Congenital muscle diseases in humans can result from mutations in at least 20 genes, but mutations in the gene for ryanodine receptor 1 (RyR1) are the most common cause of such myopathies (Jungbluth et al., 2018; Robinson et al., 2006). Mutations in this gene cause malignant hyperthermia (a rare severe reaction that can occur during anesthesia), central core disease and a range of other myopathies that can result in severe disabilities and early mortality. There are currently no effective therapies for any of these conditions. Now, in eLife, James Dowling (Hospital for Sick Children and the University of Toronto) and colleagues – including Jonathan Volpatti as first author – report how they have used a 'multi-species discovery pipeline' to identify two compounds that might be effective in treating these patients (Volpatti et al., 2020).

The pipeline involved three species: the worm C. elegans, the zebrafish and mouse cells (see Figure 1B). C. elegans has just one type of ryanodine receptor (humans have three) and mutants that lack unc-68, the gene for this receptor, move much less than wild-type worms (Maryon et al., 1996). To make the phenotype more robust, Volpatti et al. exposed the mutant animals to a DHP inhibitor called nemadipine-A that induces larval growth arrest in mutant animals but not in wild-type animals. The researchers screened 3700 compounds to identify those that permitted mutant worms that had been exposed to nemadipine-A to reach adulthood. Initially, the screen revealed 278 compounds, but this number dropped to 74 after additional testing. Compounds that inhibited a protein kinase called p38 were over-represented in this sample. To explore if off-target effects might be responsible for their results, Volpatti et al. used RNAi to knock-down orthologs of p38 in worms. Again the mutant worms reached adulthood, confirming that the results were likely due to p38 being inhibited and not due to off-target effects.

Many of the 74 compounds identified from the worm screen were then tested for their ability to improve the movement of zebrafish that lacked a gene called ryr1b that encodes one of the ryanodine receptors: these mutants are normally poor swimmers (Hirata et al., 2007). Two of the p38 inhibitors were successful.

Volpatti et al. then moved to a mouse cell line called C2C12. These cells are myoblasts that can be differentiated into mature functional muscle cells, and the researchers used CRISPR/Cas9 to create mutants in which the gene Ryr1 had been knocked out. When caffeine is administered to wild-type C2C12 cells they release calcium ions, but this does not happen with the mutant cells. However, when the mutant cells were treated with either of the p38 inhibitors, they released calcium ions. The researchers verified that the two compounds were likely specifically inhibiting p38 by observing the release of calcium ions from the knockout cells when any one of the three isoforms of p38 were knocked down by siRNA.

The role of p38 during the formation of muscle tissue has been studied intensively. p38 activates various factors that regulate muscle formation (Keren et al., 2006); it also activates adult muscle stem cells and promotes their self-renewal (Jones et al., 2005). Thus, p38 has been proposed as a potential therapeutic target for muscular dystrophies. Activation of p38 has been detected in mouse models of two forms of muscular dystrophy (Duchenne Muscular Dystrophy and Limb Girdle Muscular Dystrophy R6). Moreover, deletion of the gene for the alpha isoform of p38 results in reduced myopathy in both muscular dystrophy models via the inhibition of apoptosis (Wissing et al., 2014). Although the role of p38 in the pathology of the various myopathies related to RyR1 is not explored by Volpatti et al., the inhibition of apoptosis or the regulation of muscle formation may not be relevant, since their results were obtained on fully differentiated muscle cells. A more likely explanation is that p38 inhibition results in increased expression or activity of other Ca2+ channels.

A weakness of this study is that the screens were conducted on models that completely lack expression of RyR1, whereas none of the patients with malignant hyperthermia or RyR1-related myopathies are nulls. Recognizing this problem, Volpatti et al. plan to re-do their screens on mice or cells carrying missense mutations analogous to those found in malignant hyperthermia (Lopez et al., 2018), or the more complex mutations found in patients with RyR1-related myopathies (see, for example, Brennan et al., 2019). An intriguing question is how can the inhibition of p38 result in the release of more Ca2+ in animals or cells that have no RyR1 expression? As mentioned in the previous paragraph, it is possible that the inhibition of p38 increases the release of Ca2+ from the sarcoplasmic reticulum through other Ca2+channels such as the IP3R and STIM1/ORAI1 channels (which are found in both C. elegans and C2C12 cells), and RyR3 channel, which is also found in C2C12 cells.

Volpatti et al. conducted their drug screens rapidly and in mutant animals displaying phenotypes relevant to those found in human patients. For example, the screen of 3700 compounds in C. elegans was completed in just two weeks.

It is likely that their strategy of screening in C. elegans first, followed by zebrafish and cultured cells, could be used to screen drugs for many human genetic diseases that are due to mutations in proteins with orthologs in C. elegans. (About 40% of C. elegans proteins have human orthologs). This is particularly true for human diseases that display developmental or neuromuscular defects, as development is fast (fertilized egg to adult in three days) and occurs in distinct stages (embryo, four larval stages and adult), and there are powerful methods for studying the locomotion of these animals (see, for example, Restif et al., 2014). This presages another case in which worms will lead the way!

References

Article and author information

Author details

  1. Guy M Benian

    Guy M Benian is in the Department of Pathology and the Department of Cell Biology, Emory University, Atlanta, United States

    For correspondence
    pathgb@emory.edu
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-8236-3176
  2. Hyojung J Choo

    Hyojung J Choo is in the Department of Cell Biology, Emory University, Atlanta, United States

    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-6280-0332

Publication history

  1. Version of Record published: May 6, 2020 (version 1)

Copyright

© 2020, Benian and Choo

This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 1,051
    views
  • 113
    downloads
  • 2
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Guy M Benian
  2. Hyojung J Choo
(2020)
Drug Discovery: From worms to fish to mice
eLife 9:e57481.
https://doi.org/10.7554/eLife.57481

Further reading

    1. Immunology and Inflammation
    2. Medicine
    Joanna C Porter, Jamie Inshaw ... Venizelos Papayannopoulos
    Research Article

    Background:

    Prinflammatory extracellular chromatin from neutrophil extracellular traps (NETs) and other cellular sources is found in COVID-19 patients and may promote pathology. We determined whether pulmonary administration of the endonuclease dornase alfa reduced systemic inflammation by clearing extracellular chromatin.

    Methods:

    Eligible patients were randomized (3:1) to the best available care including dexamethasone (R-BAC) or to BAC with twice-daily nebulized dornase alfa (R-BAC + DA) for seven days or until discharge. A 2:1 ratio of matched contemporary controls (CC-BAC) provided additional comparators. The primary endpoint was the improvement in C-reactive protein (CRP) over time, analyzed using a repeated-measures mixed model, adjusted for baseline factors.

    Results:

    We recruited 39 evaluable participants: 30 randomized to dornase alfa (R-BAC +DA), 9 randomized to BAC (R-BAC), and included 60 CC-BAC participants. Dornase alfa was well tolerated and reduced CRP by 33% compared to the combined BAC groups (T-BAC). Least squares (LS) mean post-dexamethasone CRP fell from 101.9 mg/L to 23.23 mg/L in R-BAC +DA participants versus a 99.5 mg/L to 34.82 mg/L reduction in the T-BAC group at 7 days; p=0.01. The anti-inflammatory effect of dornase alfa was further confirmed with subgroup and sensitivity analyses on randomised participants only, mitigating potential biases associated with the use of CC-BAC participants. Dornase alfa increased live discharge rates by 63% (HR 1.63, 95% CI 1.01–2.61, p=0.03), increased lymphocyte counts (LS mean: 1.08 vs 0.87, p=0.02) and reduced circulating cf-DNA and the coagulopathy marker D-dimer (LS mean: 570.78 vs 1656.96 μg/mL, p=0.004).

    Conclusions:

    Dornase alfa reduces pathogenic inflammation in COVID-19 pneumonia, demonstrating the benefit of cost-effective therapies that target extracellular chromatin.

    Funding:

    LifeArc, Breathing Matters, The Francis Crick Institute (CRUK, Medical Research Council, Wellcome Trust).

    Clinical trial number:

    NCT04359654.

    1. Medicine
    Ruijie Zeng, Yuying Ma ... Hao Chen
    Research Article

    Background:

    Adverse effects of proton pump inhibitors (PPIs) have raised wide concerns. The association of PPIs with influenza is unexplored, while that with pneumonia or COVID-19 remains controversial. Our study aims to evaluate whether PPI use increases the risks of these respiratory infections.

    Methods:

    The current study included 160,923 eligible participants at baseline who completed questionnaires on medication use, which included PPI or histamine-2 receptor antagonist (H2RA), from the UK Biobank. Cox proportional hazards regression and propensity score-matching analyses were used to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs).

    Results:

    Comparisons with H2RA users were tested. PPI use was associated with increased risks of developing influenza (HR 1.32, 95% CI 1.12–1.56) and pneumonia (hazard ratio [HR] 1.42, 95% confidence interval [CI] 1.26–1.59). In contrast, the risk of COVID-19 infection was not significant with regular PPI use (HR 1.08, 95% CI 0.99–1.17), while the risks of severe COVID-19 (HR 1.19. 95% CI 1.11–1.27) and mortality (HR 1.37. 95% CI 1.29–1.46) were increased. However, when compared with H2RA users, PPI users were associated with a higher risk of influenza (HR 1.74, 95% CI 1.19–2.54), but the risks with pneumonia or COVID-19-related outcomes were not evident.

    Conclusions:

    PPI users are associated with increased risks of influenza, pneumonia, as well as COVID-19 severity and mortality compared to non-users, while the effects on pneumonia or COVID-19-related outcomes under PPI use were attenuated when compared to the use of H2RAs. Appropriate use of PPIs based on comprehensive evaluation is required.

    Funding:

    This work is supported by the National Natural Science Foundation of China (82171698, 82170561, 81300279, 81741067, 82100238), the Program for High-level Foreign Expert Introduction of China (G2022030047L), the Natural Science Foundation for Distinguished Young Scholars of Guangdong Province (2021B1515020003), the Guangdong Basic and Applied Basic Research Foundation (2022A1515012081), the Foreign Distinguished Teacher Program of Guangdong Science and Technology Department (KD0120220129), the Climbing Program of Introduced Talents and High-level Hospital Construction Project of Guangdong Provincial People’s Hospital (DFJH201923, DFJH201803, KJ012019099, KJ012021143, KY012021183), and in part by VA Clinical Merit and ASGE clinical research funds (FWL).