Volume 21, number 2
 Views: (Visited 8 times, 9 visits today)  

Alzahrani A. Identification of Therapeutic Agents Targeting Mitogen-Activated Protein Kinase 14 in the Treatment and Management of Asthma. Biotech Res Asia 2024;21(2).
Manuscript received on : 28-02-2024
Manuscript accepted on : 20-06-2024
Published online on:  02-07-2024

Plagiarism Check: Yes

Reviewed by: Dr. Binit Patel

Second Review by: Dr. Mohamed Nader

Final Approval by: Dr. Muhammad Hamayun

How to Cite    |   Publication History    |   PlumX Article Matrix

Identification of Therapeutic Agents Targeting Mitogen-Activated Protein Kinase 14 in the Treatment and Management of Asthma

Abdulrahman Alzahrani*

Department of Applied Medical Sciences, Applied College, Al-Baha University, Al-Baha City, Kingdom of Saudi Arabia.

corresponding Author E-mail:abdulrahmanmuidh@gmail.com

ABSTRACT: Mitogen-activated protein Kinase 14 (MAPK14) plays a pivotal role in the pathophysiology of asthma, influencing inflammation, airway remodeling, and bronchial hyperresponsiveness, highlighting its significance as a potential therapeutic target in asthma management. This study aimed to identify the inhibitory compounds from a small library of small molecule drugs curated from the DrugBank. We conducted a comprehensive exploration of the protein structure, cavity detection, molecular docking, ADMET predictions, and functional assays pertinent to lead molecules and MAPK14. Through protein structure homology modeling, the MAPK14 model exhibited exemplary quality, supported by high GMQE (0.90) and QMEANDisCo (0.82 ± 0.05) scores. Cavity detection highlighted prominent features, guiding subsequent molecular docking studies. Notably, Cobicistat emerged as a potential inhibitor, displaying strong binding affinity (-8.6 kcal/mol) across multiple binding pockets on MAPK14. ADMET predictions underscored its drug-like properties, while cytotoxicity assays on normal lung cells revealed its benign nature. Further investigations elucidated Cobicistat's inhibitory effect on MAPK14 expression and activity, validating its therapeutic potential for asthma treatment. Enzyme inhibition assay provided mechanistic insights, indicating Cobicistat's ability to modulate MAPK14 activity, thus positioning it as a promising therapeutic candidate for targeting MAPK14-associated diseases such as asthma. This comprehensive study underscores Cobicistat's multifaceted potential in therapeutic interventions, substantiating its candidacy for further validation.

KEYWORDS: Asthma; Cobicistat; Inflammation; Mitogen-Activated Protein Kinase 14 (MAPK14); Therapeutic target

Copy the following to cite this article:

Alzahrani A. Identification of Therapeutic Agents Targeting Mitogen-Activated Protein Kinase 14 in the Treatment and Management of Asthma. Biotech Res Asia 2024;21(2).

Copy the following to cite this URL:

Alzahrani A. Identification of Therapeutic Agents Targeting Mitogen-Activated Protein Kinase 14 in the Treatment and Management of Asthma. Biotech Res Asia 2024;21(2). Available from: https://bit.ly/4cp0BnR

Introduction

Mitogen-activated protein kinase 14 (MAPK14), also known as p38α MAPK, is a crucial signaling molecule involved in various cellular processes, including inflammation, apoptosis, cell differentiation, and cell proliferation1. In the context of asthma, MAPK14 plays a significant role in the pathogenesis of this chronic inflammatory disease2. Asthma is a heterogeneous respiratory disorder characterized by airway inflammation, bronchial hyperresponsiveness, and reversible airflow obstruction3. It affects people of all ages and can significantly impact their quality of life4. While the exact etiology of asthma remains incompletely understood, it is widely accepted that the interplay of genetic predisposition and environmental factors contributes to its development and progression1.

Inflammation is a hallmark feature of asthma, and MAPK14 has emerged as a central mediator in orchestrating inflammatory responses within the airways5. Upon activation by various extracellular stimuli, including pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and environmental triggers like allergens and pollutants, MAPK14 becomes phosphorylated and subsequently activates downstream effector molecules6.

One of the primary functions of MAPK14 in asthma pathogenesis is the regulation of inflammatory cytokine production2. Through its ability to phosphorylate and activate transcription factors such as nuclear factor-kappa B (NF-κB) and activator protein-1 (AP-1), MAPK14 promotes the expression of pro-inflammatory mediators like interleukin-6 (IL-6), interleukin-8 (IL-8), and TNF-α2. These cytokines recruit and activate inflammatory cells, including neutrophils, eosinophils, and T lymphocytes, amplifying the inflammatory cascade within the airways7. Furthermore, MAPK14 activation promotes the synthesis of other key mediators involved in asthma pathophysiology, such as leukotrienes and prostaglandins8,9. These lipid mediators contribute to bronchoconstriction, airway hyperresponsiveness, and mucus hypersecretion, further exacerbating respiratory symptoms in asthma patients9.

In addition to its role in inflammation, MAPK14 also regulates airway smooth muscle contraction, a process central to the pathogenesis of asthma10. Activated MAPK14 phosphorylates and activates myosin light chain kinase (MLCK), leading to myosin light chain (MLC) phosphorylation and subsequent smooth muscle contraction11. This process contributes to bronchoconstriction and airway narrowing, characteristic features of asthma exacerbations. Moreover, MAPK14 activation promotes airway remodeling, another hallmark of chronic asthma12. Persistent inflammation and repeated injury to the airway epithelium lead to structural changes, including subepithelial fibrosis, increased smooth muscle mass, and mucus gland hyperplasia13. MAPK14-mediated signaling pathways contribute to these remodeling processes by promoting fibroblast proliferation, collagen deposition, and extracellular matrix remodelling14.

The role of MAPK14 in modulating the immune response extends beyond its effects on inflammatory cells and cytokine production. Studies have demonstrated that MAPK14 activation influences T helper cell differentiation, skewing the balance towards a Th2-dominated immune response characteristic of allergic asthma15. Th2 cells produce cytokines such as interleukin-4 (IL-4), interleukin-5 (IL-5), and interleukin-13 (IL-13), which promote eosinophil recruitment, immunoglobulin E (IgE) production, and mucus secretion, contributing to allergic inflammation and airway hyperreactivity16.

Furthermore, MAPK14 activation in airway epithelial cells contributes to barrier dysfunction and impaired epithelial repair mechanisms, exacerbating airway inflammation and susceptibility to environmental insults17. Disruption of the airway epithelial barrier allows for increased penetration of allergens, pathogens, and environmental pollutants, further amplifying the inflammatory response in asthma18.

The significance of MAPK14 in asthma pathogenesis is further underscored by preclinical and clinical studies demonstrating the efficacy of MAPK14 inhibitors in attenuating airway inflammation, bronchoconstriction, and airway hyperresponsiveness19. Pharmacological inhibition of MAPK14 has been shown to reduce inflammatory cytokine production, airway smooth muscle contraction, mucus hypersecretion, and airway remodeling in experimental models of asthma20. However, despite the promising therapeutic potential of MAPK14 inhibitors, challenges remain in translating these findings into clinical practice. The development of selective and potent MAPK14 inhibitors with favorable pharmacokinetic properties and minimal off-target effects remains a priority for drug discovery efforts in asthma and other inflammatory diseases. Overall, Mitogen-activated protein kinase 14 (MAPK14) plays a pivotal role in the pathogenesis of asthma by regulating inflammatory responses, airway smooth muscle contraction, airway remodeling, and immune dysregulation. Targeting MAPK14 signaling pathways represents a promising therapeutic approach for the treatment of asthma and other inflammatory respiratory diseases.

The rationale for conducting further research in this area stems from the significant impact that small molecule inhibitors targeting specific molecules and pathways have been demonstrated in disease therapeutics. As highlighted in the provided text, these inhibitors play a crucial role in controlling disease pathology by selectively targeting key molecules involved in disease progression, thus offering a promising avenue for effective treatment. The focus on small molecule inhibitors that inhibit the activity of MAPK14 presents a persuasive rationale for investigation in asthma. This study aims to identify penitential inhibitor small molecules of MAPK14 using in-silico and in-vitro methods for the therapeutics of asthma.

Material and Methods

Protein structure homology modeling and preparation of MAPK14 receptor

Homology modeling, a widely used technique, predicts a protein’s 3D structure based on homologous proteins with known structures. The popular web tool SWISS-MODEL (https://swissmodel.expasy.org/) facilitates this process and was utilized to predict the high-resolution MAPK14 model21,22,23. Starting with the FASTA sequence of MAPK14 retrieved from PubMed, template sequences from the Protein Data Bank were identified to find a suitable homologous structure (MAPK14: PDB ID – 6y4x) based on sequence similarity, quality, and relevance. Structural analysis was then performed using PyMOL and Chimera before preparing for molecular docking by refining MAPK14’s structure through standard protocol practices including addressing steric clashes, missing atoms, and assigning charges24,25. The final step involved saving the refined structures in PDBQT format for docking compatibility.

Selection and preparation of ligands for molecular docking with target protein

1936 small molecule drugs from DrugBank (https://go.drugbank.com/) approved by the US were sourced. Their three-dimensional (3D) structures in SDF format were obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/). Each structure underwent rigorous validation to correct bond lengths, angles, and torsional angles. After validation, the structures were converted to PDBQT format for compatibility with docking software, ensuring accurate representation during simulations.

Structure-based cavity detection on target protein

Identifying potential binding sites on MAPK14 involves cavity detection using CB-Dock2 (http://cadd.labshare.cn/cb-dock2/), which automates the process. By utilizing query ligands, CB-Dock2 calculates and customizes docking box size through AutoDock Vina. The initiation of process begins with uploading the MAPK14 PDB file while specifying parameters like probe radius and cavity size to optimize detection accuracy. This setup ensures precise identification of potential binding pockets on MAPK1426.

Screening and molecular docking of the lead molecule with the target protein

A thorough screening of 1936 small molecule drugs was conducted to evaluate their binding affinity against MAPK14, a key target in asthma therapeutics. The top hit, chosen based on fitness score and binding affinity, underwent molecular docking using CB-Dock2, well-known for its blind docking capability26. CB-Dock2 combines cavity detection, docking processes, and template fitting to accurately predict the binding site. This approach sheds light on potential interactions at the molecular level. To verify this, the ligand-protein complex underwent redocking using SeamDock, which enhances the reliability of assessing binding affinity27. SeamDock adds an extra layer of validation to the docking results obtained from CB-Dock2.

Assessment of ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) properties of lead molecule

Assessing the ADMET properties is crucial for understanding pharmacokinetics and toxicity. We used the pkCSM online tool (https://biosig.unimelb.edu.au/pkcsm/.) to evaluate these properties for the lead molecule. pkCSM predicts pharmacokinetics and toxicity comprehensively by offering an analysis of ADMET parameters based on chemical structure in SMILES format. This assessment sheds light on how the molecule may be absorbed, distributed, metabolized, and excreted and its potential toxicity profile; thus aiding in drug development decisions.

Prediction of Activity Spectra for lead ligand molecule using SwissTargetPrediction tool

Chemoinformatics and drug discovery rely on predicting ligand activity spectra to identify targets and understand pharmacological effects. The study utilized SwissTargetPrediction (http://www.swisstargetprediction.ch/) to explore the lead molecule’s biological activity spectrum. This tool predicts potential targets for small molecules, helping with protein interaction identification and understanding pharmacological profiles. By analyzing compound chemical structures, it identifies targets based on structural similarities and known interaction patterns. Inputting ligand SMILES notation into SwissTargetPrediction reveals potential biological targets and activity spectrum, deepening our understanding of molecular mechanisms essential for drug discovery and informing experimental studies for therapeutic exploration.

Test compound and cell lines

Cobicistat was dissolved in phosphate-buffered saline to create a stock solution with a concentration of 1000 nM/mL. The cell lines used in this study, HEK-293 (human embryonic kidney) and BEAS-2B (normal human bronchial epithelial cell line), were obtained from the ATCC. HEK-293 cells were cultured in Dulbecco’s Modified Eagle Medium, while BEAS-2B cells were cultured in Airway Epithelial Cell Basal Medium supplemented with Bronchial Epithelial Cell Growth regulators. Both culture media were supplemented with 10% fetal bovine serum and 1% antibiotics to support cell growth and maintain sterility.

Cell culture and cytotoxicity assay

HEK-293 cells were seeded at a density of 5,000 cells per well in a 96-well microplate and incubated for 24 hours in cell culture incubator at 37 °C and 5% CO2. They were then exposed to varying concentrations of Cobicistat (0-1000 nM) for 72 hours to evaluate the dose-dependent effects on cell viability. Subsequently, the medium was replaced with an MTT reagent (0.5 mg/mL) and incubated for 4 hours to form formazan crystals. The formed formazan was solubilized using DMSO, and the absorbance at 570 nm was measured. Net absorbance values were determined by subtracting blank readings. Cell viability was assessed by comparing the net absorbance of treated wells to that of untreated control wells and multiplying by 100. For examining time-dependent effects, HEK-293 cells were exposed to the mean IC50 concentration of Cobicistat (293.11 nm) for durations of 24, 48, and 72 hours.

mRNA expression of MAPK14 in Cobicistat treated BEAS-2B cells

To investigate the effect of Cobicistat on MAPK14 gene expression, BEAS-2B cells were treated with a concentration of 293.11 nM, which is the IC50, for 48 hours. Subsequently, RNA extraction was carried out using TRIzol™ Reagent following standard protocol28. The extracted RNA was then used for cDNA synthesis and qRT-PCR analysis using SYBR™ Green Master Mix. The expression level of MAPK14 was assessed and normalized to that of the housekeeping gene GAPDH, followed by a calculation of the relative fold change in MAPK14 expression. The primer sequences utilized were as follows: MAPK14: forward 5′-GAGCGTTACCAGAACCTGTCTC-3′ and reverse 5′-AGTAACCGCAGTTCTCTGTAGGT-3′, GAPDH: forward 5′- GTCTCCTCTGACTTCAACAGCG-3′ and reverse 5′-ACCACCCTGTTGCTGTAGCCAA 3′.

Further, cellular protein MAPK14 activity in BEAS-2B cells was quantified using the Human MAPK14 ELISA Kit from Abcam following the protocol as described by the manufacturer.

Enzyme inhibition assay

A series of experiments investigated the impact of Cobicistat on MAPK14 kinase activity. Fixed concentrations of Cobicistat (0 to 1000 nM) were added to 2 µM of MAPK14 in a 96-well plate and incubated for an hour at 25°C. Following this, a reaction mixture containing 100 µM ATP and 10 mM MgCl2 was introduced and further incubated for another 30 minutes. The reaction was then terminated with BIOMOL® reagent, which formed a green-colored complex monitored at 620 nm. Malachite green reagent facilitated the detection of inorganic phosphate released during ATP hydrolysis, enabling the reflection of MAPK14 kinase activity based on color intensity – directly indicating enzyme activity levels. Additionally, native MAPK14’s activity without Cobicistat was set as the baseline at 100%.

Results

Protein structure homology modeling and preparation of MAPK14

Fig. 1a illustrates the high-resolution structure of the MAPK14 model from Swiss-MODEL, boasting a remarkable sequence identity and 100% coverage, signifying precise alignment with the target sequence. Quality assessment revealed excellence, with a GMQE score of 0.90 and a QMEANDisCo Global score of 0.82 ± 0.05. These scores, ranging from 0 to 1, denote overall model quality, with higher values indicating superior expected quality. GMQE considers alignment coverage, while QMEANDisCo evaluates quality independently. Fig. 1b presents a graphical representation of the QMEANDisCo local quality estimate, providing insights into the local quality of the model. Additionally, Fig. 1c compares with a non-redundant set of PDB structures, and Fig. 1d depicts the Ramachandran Plot, showing energetically favored regions for backbone dihedral angles of amino acid residues, crucial for assessing structural integrity and reliability of the MAPK14 model.

Figure 1: (a) 3D structure of the MAPK14; (b) Graphical representation of the QMEANDisCo local quality estimate; (c) Non-redundant set of PDB structures and; (d) Ramachandran Plot. 

Click here to view Figure

Cavity detection on MAPK14

Table 1 outlines findings on cavities within MAPK14, detailing the number and characteristics via CB-Dock2 analysis. Notably, five prominent cavities (C1 to C5) were identified, with Table 2 providing comprehensive data on size, volume, and attributes. Cavity 2 (C2) stood out for its maximum size, warranting further investigation through molecular docking experiments. Sequenced-based depiction of cavities (C1-C5) is in Fig. 2A, while Fig. 2B visually represents all structure-based cavities within MAPK14. These analyses yield insights into MAPK14’s structural traits, crucial for subsequent molecular docking studies, especially regarding interactions with ligands like Cobicistat.

Table 1: Detailed information on the size, volume, or other relevant characteristics of the structure-based cavities (C1-C5) detected on MAPK14.

CurPocket ID Cavity volume (Å3) Center (x, y, z) Cavity size (x, y, z)
Cavity 1 3650 -2, -8, -2 30, 19, 29
Cavity 2 3151 1, 15, -11 30, 23, 30
Cavity 3 385 -8, 9, 19 14, 8, 7
Cavity 4 353 5, 5, -3 9, 9, 13
Cavity 5 323 -1, 15, 10 15, 8, 9
Figure 2: (A) Sequence of cavities C1, C2, C3, C4, and C5 detected on MAPK14. (B) Structural visualization of all identified cavities (C1, C2, C3, C4, and C5) within MAPK14.

Click here to view Figure

Molecular docking revealed Cobicistat as a potential inhibitor of MAPK14

After screening 1936 US-approved small molecule drugs against MAPK14, Cobicistat emerged as the most promising candidate with a remarkable fitness score of 99.01% and the highest predicted binding affinity. Figure 3a-e presents detailed information on the fitness scores and binding affinities of the top 5 small molecule drugs. Following the identification of Cobicistat as the top hit, it was prioritized for molecular docking against the target protein MAPK14. The results of molecular docking show Cobicistat specifically at the C1, C2, and C3 binding pockets on MAPK14, visually represented in Fig. 4. Fig. 4A (i) and 4A (ii) show the molecular docking at C1, where Cobicistat interacts with multiple amino acid residues of MAPK14, including GLY32, SER33, GLY34, ALA35, TYR36, GLY37, VAL39, LYS54, ARG68, ASP113, ASN115, LYS119, ASP151, LYS153, SER155, ASN156, LEU168, ASP169, LEU172, ALA173, ASP177, GLU179, MET180, VAL184, and ALA185. Fig. 4A (iii) and 4A (iv) show the molecular docking at C2, where Cobicistat formed interaction with MET2, GLN4, GLU5, ARG6, PRO7, PHE9, PRO22, ARG24, LYS46, ASP89, VAL90, PHE91, THR92, ALA94, VAL346, ILE347, SER348, PHE349, VAL350, and PRO351 amino acid residues of MAPK14. Fig. 4A (v) and 4A (vi) show the molecular docking at C3, where Cobicistat interacts with multiple amino acid residues of MAPK14, including VAL118, LYS119, CYS120, GLN121, LYS122, LEU123, THR124, ASP125, LEU217, LEU218, THR219, GLY220, ARG221, LEU223, VAL274, PHE275, ILE276, GLY277, and ALA278. In this docking analysis, we generated affinity scores and docked poses which revealed a notable binding affinity score of -8.6 kcal/mol between Cobicistat and MAPK14. Fig. 4 (i), 4A (iii), and 4 (v) provide a cartoon presentation of the MAPK14: Cobicistat complex for C1, C2, and C3 respectively to offer insights into the spatial arrangement of the molecules. Meanwhile, Fig. 4A (ii), 4 (iv), and 4 (vi) represent a surface view that offers additional perspectives on their interaction for C1, C2, and C3 respectively. All these interactions highlight the comprehensive nature of the binding between Cobicistat and MAPK14, strongly suggesting a strong and multi-faceted affinity between the two molecules. Furthermore, to confirm the binding affinity of the MAPK14-Cobicistat complex, redocking was performed using SeamDock. The results of the redocking poses are shown in Fig. 4B (i) and 4B (ii). In these poses, Cobicistat consistently interacted with the binding site of MAPK14, demonstrating a binding affinity of -8.3 kcal/mol. This consistent affinity supports Cobicistat’s potential as a promising therapeutic candidate for targeting MAPK14 in asthma.

Figure 3: Detailed information on binding affinity of top 5 small molecule drugs. Chemical structures of these compounds were obtained from Drug Bank (https://go.drugbank.com/). https://go.drugbank.com/ 

Click here to view Figure

Figure 4: (A) Visual representation of the Cobicistat: MAPK14 molecular docking complexes formed at C1, C2, and C3 cavities. (i, iii, and v) cartoon presentation of the complex; (ii, iv, and vi). Surface view of the complex. 

Click here to view Figure

ADMET predictions and Swiss Target Prediction

Cobicistat’s drug-like properties were validated through ADMET predictions and Swiss Target Prediction. The ADMET predictions, summarized in Table 2, indicate compliance with key pharmacokinetic criteria. Furthermore, the Swiss Target Prediction analysis confirms and expands on the biological properties of Cobicistat by suggesting promise in targeting MAPK14 inhibition for asthma treatment as detailed in Fig. 5.

Figure 5: Swiss Target Prediction analysis showing diverse biological properties linked to Cobicistat.

Click here to view Figure

Table 2: In silico prediction of ADME & Tox properties for Cobicistat (CBST).

Properties
Absorption Distribution Metabolism Excretion Toxicity
Models Intestinal

absorption (human)

VDss (human) BBB

permeability

CNS permeability CYP Total clearance AMES toxicity
Substrate Inhibitor
2D6 3A4 1A2 2C19 2C9 2D6 3A4
Unity Numeric

(% absorbed)

Numeric (log L/kg) Numeric (Log BB) Numeric

(Log PS)

Categorical

(yes/no)

Numeric (log ml/min / kg) Categorical

(yes/no)

Predicted values
CBST 76.503 0.668 -1.761 -3.454 NO Yes No No No NO Yes 0.845 NO

Cytotoxic effect of Cobicistat on HEK-293 normal lung cells

Fig. 6a depicts the concentration-dependent effect of Cobicistat on HEK-293 cell viability. Analysis revealed no significant concentration-dependent impact on cell viability, suggesting Cobicistat lacks notable effects within the tested concentration range on these cells. HEK-293 cells were treated in concentration concentration-dependent manner for 72 hrs. Cells were harvested for MTT assay and absorbance was recorded to determine the respective IC50 concentrations. The IC50 value for Cobicistat was found to be 293.11 nM for HEK-293 cells.

Fig. 6b depicts the investigation into Cobicistat’s effects on HEK-293 cell viability over varying periods. HEK-293 cells were exposed to increasing durations of Cobicistat (293.11 nM) – 24, 48, and 72 hours. Analysis revealed consistent viability across all durations, indicating Cobicistat had no adverse impact on non-cancerous HEK-293 cells. This suggests its benign nature over the specified periods, as it did not compromise cell viability.

Expression and activity of MAPK14 in Cobicistat-treated BEAS-2B cells

Fig. 6c and 6d provide a visual representation of the impact of Cobicistat on the mRNA expression and activity of MAPK14 in BEAS-2B cells, respectively. The experimental setup involved treating cells with Cobicistat at its specific IC50 concentration. Upon analysis, it was observed that treatment with Cobicistat led to a significant decrease (p < 0.001) in the mRNA expression and activity of MAPK14 in treated cells compared to NTC. This significant reduction in MAPK14 mRNA expression and activity underscores the inhibitory effect of Cobicistat on MAPK14, highlighting its potential as a therapeutic agent for targeting MAPK14 in asthma treatment.

Enzyme inhibition assay showed inhibition of MAPK14 by Cobicistat

The kinase assay showed a notable (p<0.001) decline in MAPK14 activity with rising Cobicistat levels (Fig. 6e). This hints at Cobicistat’s potential to inhibit MAPK14 kinase activity. The dose-dependent response underscores its probable modulation of MAPK14, via direct interaction or downstream regulatory pathways.

Figure 6: (a) concentration-dependent cell toxicity; (b) time-dependent cell toxicity. Effect of IC50 dose of Cobicistat on the (c) mRNA expression and; (d) activity of MAPK14 in BEAS-2B cells. *** p<0.001.  

Click here to view Figure

Discussion

The comprehensive investigation presented in this study sheds light on the structural attributes of MAPK14 and its potential inhibition by Cobicistat, offering promising insights into its therapeutic application in asthma treatment and management. Modeling the 3D structure of a target protein is crucial for identifying therapeutic agents in disease management23. Understanding protein structures aids in designing molecules that interact specifically with the target, enhancing drug efficacy and minimizing side effects29. Through computational modeling, scientists predict how potential drugs bind to the protein, guiding drug development. This approach accelerates drug discovery by screening virtual compound libraries, saving time and resources compared to traditional methods24. Additionally, structure-based drug design allows for the optimization of lead compounds, fine-tuning their properties for improved therapeutic outcomes25. Ultimately, this methodology revolutionizes the treatment and management of diseases, offering tailored solutions with higher precision. In the current study, we aimed to target to inhibit MAPK14 for the therapeutics and management of asthma. In this study, the high-resolution model of MAPK14 generated through protein structure homology modeling exhibited remarkable sequence identity and coverage, indicative of precise alignment with the target sequence. The quality assessment metrics, including GMQE and QMEANDisCo scores, underscored the excellence of the model, highlighting its reliability and suitability for subsequent analyses.

The modeled structure was further subjected to predict the binding cavities on MAPK14. Identifying binding cavities on target proteins is pivotal for structure-based identification of therapeutic agents in disease management30. These cavities are regions where small molecules can bind, influencing protein function and disease progression. Through computational methods investigators pinpoint potential binding sites, facilitating drug discovery24. By understanding the spatial arrangement and chemical properties of these cavities, researchers design molecules tailored to fit and interact with high affinity, increasing drug specificity and efficacy25. This approach streamlines the search for lead compounds, accelerating the development of novel therapeutics with enhanced therapeutic potential for treating and managing a wide array of diseases. The identification of five prominent cavities within MAPK14, along with detailed characterization of their size, volume, and attributes, provided crucial insights into the structural traits of the protein. Particularly, the prominence of Cavity 2 (C2) warranted further investigation through molecular docking experiments, paving the way for targeted ligand interaction studies.

Molecular docking plays a pivotal role in the structure-based identification of therapeutic agents for disease treatment and management31. It involves the computational simulation of how small molecules, or ligands, interact with the binding site of a target protein25. By predicting the most favorable orientation and conformation of the ligand within the binding pocket, docking enables the screening of vast libraries of compounds to identify potential drug candidates32. This process guides medicinal chemists in optimizing lead compounds, enhancing their binding affinity and specificity to the target protein25. Ultimately, molecular docking expedites drug discovery, leading to the development of effective treatments tailored to combat various diseases31. The molecular docking studies revealed Cobicistat as a potential inhibitor of MAPK14, with a remarkable fitness score and high predicted binding affinity. Detailed analyses elucidated the specific binding interactions between Cobicistat and the C1, C2, and C3 binding pockets on MAPK14, highlighting the multi-faceted nature of their affinity.

ADMET predictions and SwissTargetPrediction play crucial roles in the structure-based identification of therapeutic agents for disease management33. ADMET predictions assess a compound’s Absorption, Distribution, Metabolism, Excretion, and Toxicity, crucial factors influencing drug efficacy and safety34. By forecasting these properties, researchers prioritize lead hits with favorable ADMET profiles, minimizing risks and optimizing therapeutic outcomes. SwissTargetPrediction complements this by predicting a compound’s potential targets, aiding in understanding its pharmacological effects and off-target interactions35. Integrating these tools enables the selection of lead compounds with optimal drug-like properties and target specificity, accelerating the development of safe and effective treatments for various diseases. The validation of Cobicistat’s drug-like properties through ADMET predictions, coupled with SwissTargetPrediction analysis suggesting its promise in targeting MAPK14 inhibition for asthma treatment, further reinforced its therapeutic potential.

Evaluating the cytotoxicity effects of lead compounds is pivotal in identifying and developing therapeutic molecules for diseases36. This assessment examines the compound’s impact on cell viability and function, ensuring it selectively targets diseased cells while sparing healthy ones37. By conducting cytotoxicity assays, researchers gauge the compound’s safety profile, determining its maximum tolerated dose and potential adverse effects. This information guides medicinal chemists in optimizing lead compounds, balancing efficacy with tolerability. Moreover, understanding cytotoxicity aids in predicting drug responses in vivo, facilitating the translation of promising candidates from preclinical studies to clinical trials. Ultimately, this process ensures the development of safe and effective therapeutic interventions for various diseases. The concentration-dependent cytotoxicity analysis on HEK-293 normal lung cells demonstrated no significant impact on cell viability within the tested concentration range, affirming the benign nature of Cobicistat. Additionally, the determination of IC50 concentration provided valuable insights into its potency against normal lung cells.

Evaluating the effects of lead compounds on target genes is crucial for therapeutic purposes 38,39. This assessment elucidates the compound’s mechanism of action, revealing how it modulates gene expression to exert its therapeutic effects38. By studying the compound’s impact on target genes, researchers gain insights into the underlying molecular pathways involved in disease pathology and drug response40. This information guides the selection and optimization of lead compounds, ensuring they effectively modulate the desired biological targets. Moreover, evaluating target gene effects aids in predicting the compound’s efficacy and potential side effects, facilitating the development of safer and more precise therapeutic interventions for various diseases41,42,43,44,45,46. The investigation into Cobicistat’s effects on MAPK14 expression and activity in BEAS-2B cells revealed a significant decrease, highlighting its inhibitory effect on the target protein. These findings underscored Cobicistat’s potential as a therapeutic agent for asthma treatment by modulating MAPK14 activity. The enzyme inhibition assay demonstrated a dose-dependent decline in MAPK14 activity with increasing Cobicistat levels, further corroborating its potential to inhibit MAPK14 kinase activity and suggesting its role in regulating downstream signaling pathways.

Conclusion

In conclusion, this comprehensive investigation underscores the potential of Cobicistat as a therapeutic agent for asthma treatment through its inhibition of MAPK14. Molecular docking studies revealed Cobicistat’s high affinity for MAPK14, supporting its candidacy as a promising inhibitor. ADMET predictions and SwissTargetPrediction further validated its drug-like properties and potential therapeutic targets. Additionally, cytotoxicity assays demonstrated Cobicistat’s safety profile, while evaluation of its effects on target genes highlighted its mechanism of action in modulating MAPK14 expression and activity. These findings collectively underscore Cobicistat’s therapeutic potential in asthma management and pave the way for further preclinical and clinical investigations. Ultimately, this study contributes to the advancement of precision medicine by offering a targeted therapeutic approach for asthma treatment, with implications for the broader field of disease management.

Acknowledgement

The authors express heartfelt gratitude towards the department of Applied Medical Sciences, Applied College, Al-Baha University, Al-Baha City, Kingdom of Saudi

Conflict of Interest

The authors declare no competing or conflicts of interest.

Funding Sources

There is no funding Sources.

Author’s Contribution

AA: Conceptualisation, Data collection/curation, writing the MS. draft and extensive editing of the MS.

Data Availability Statement

Not Applicable

Ethics Approval Statement

The work is not related to animal/humans.

References

  1. Canovas, B., & Nebreda, A. R. Diversity and versatility of p38 kinase signalling in health and disease. Nature reviews. Molecular cell biology., 2021; 22(5): 346-366. doi: 10.1038/s41580-020-00322-w.
    CrossRef
  2. Alam, R., & Gorska, M. M. Mitogen-activated protein kinase signalling and ERK1/2 bistability in asthma. Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology., 2011; 41(2): 149-159. doi: 10.1111/j.1365-2222.2010.03658.x.
    CrossRef
  3. Carr, T. F., & Bleecker, E. Asthma heterogeneity and severity. The World Allergy Organization journal., 2016; 9(1): 41. doi: 10.1186/s40413-016-0131-2.
    CrossRef
  4. Kharaba, Z., Feghali, E., El Husseini, F., Sacre, H., Abou Selwan, C., Saadeh, S., Hallit, S., Jirjees, F., AlObaidi, H., Salameh, P., & Malaeb, D. An Assessment of Quality of Life in Patients With Asthma Through Physical, Emotional, Social, and Occupational Aspects. A Cross-Sectional Study. Frontiers in public health., 2022; 10: 883784. doi: 10.3389/fpubh.2022.883784.
    CrossRef
  5. Murdoch, J. R., & Lloyd, C. M. Chronic inflammation and asthma. Mutation research., 2010; 690(1-2): 24-39. doi: 10.1016/j.mrfmmm.2009.09.005.
    CrossRef
  6. Xia, L., A. Robock, K. Scherrer, C. Harrison, B.L. Bodirsky, I. Weindl, J. Jägermeyr, C. Bardeen, O.B. Toon, and R. Heneghan. Global food insecurity and famine from reduced crop, marine fishery and livestock production due to climate disruption from nuclear war soot injection. Food, 3, 2022; 8: 586-596. doi: 10.1038/s43016-022-00573-0.
    CrossRef
  7. Han, J., Wu, J., & Silke, J. An overview of mammalian p38 mitogen-activated protein kinases, central regulators of cell stress and receptor signaling. 2020; F1000Research, 9, F1000 Faculty Rev-653. doi: 10.12688/f1000research.22092.1.
    CrossRef
  8. Bisgaard H. Leukotrienes and prostaglandins in asthma. , 1984; 39(6): 413-420. doi: 10.1111/j.1398-9995.1984.tb01963.x.
    CrossRef
  9. Peebles, R. S., Jr, & Aronica, M. A. Proinflammatory Pathways in the Pathogenesis of Asthma. Clinics in chest medicine., 2019; 40(1): 29-50. doi: 10.1016/j.ccm.2018.10.014.
    CrossRef
  10. Doeing, D. C., & Solway, J. Airway smooth muscle in the pathophysiology and treatment of asthma. Journal of applied physiology., 2013; 114(7): 834-843. doi: 10.1152/japplphysiol.00950.2012.
    CrossRef
  11. Ihara, E., Yu, Q., Chappellaz, M., & MacDonald, J. A. ERK and p38MAPK pathways regulate myosin light chain phosphatase and contribute to Ca2+ sensitization of intestinal smooth muscle contraction. Neurogastroenterology and motility, 2015; 27(1): 135-146. doi: 10.1111/nmo.12491.
    CrossRef
  12. Cao, L., Liu, F., Liu, Y., Liu, T., Wu, J., Zhao, J., Wang, J., Li, S., Xu, J., & Dong, L. TSLP promotes asthmatic airway remodeling via p38-STAT3 signaling pathway in human lung fibroblast. Experimental lung research., 2018; 44(6): 288-301. doi: 10.1080/01902148.2018.1536175.
    CrossRef
  13. Pelaia, C., Vatrella, A., Gallelli, L., Lombardo, N., Sciacqua, A., Savino, R., & Pelaia, G. Role of p38 Mitogen-Activated Protein Kinase in Asthma and COPD: Pathogenic Aspects and Potential Targeted Therapies. Drug design, development and therapy., 2021; 15: 1275-1284. doi: 10.2147/DDDT.S300988.
    CrossRef
  14. Turner, N. A., & Blythe, N. M. Cardiac Fibroblast p38 MAPK: A Critical Regulator of Myocardial Remodeling. Journal of cardiovascular development and disease., 2019; 6(3): 27. doi: 10.3390/jcdd6030027.
    CrossRef
  15. Harker, J. A., & Lloyd, C. M. T helper 2 cells in asthma. The Journal of experimental medicine., 2023; 220(6): e20221094. doi: 10.1084/jem.20221094.
    CrossRef
  16. Pelaia, C., Heffler, E., Crimi, C., Maglio, A., Vatrella, A., Pelaia, G., & Canonica, G. W. Interleukins 4 and 13 in Asthma: Key Pathophysiologic Cytokines and Druggable Molecular Targets. Frontiers in pharmacology., 2022; 13: 851940. doi: 10.3389/fphar.2022.851940.
    CrossRef
  17. Kim, C., Sano, Y., Todorova, K., Carlson, B. A., Arpa, L., Celada, A., Lawrence, T., Otsu, K., Brissette, J. L., Arthur, J. S., & Park, J. M. The kinase p38 alpha serves cell type-specific inflammatory functions in skin injury and coordinates pro- and anti-inflammatory gene expression. Nature immunology., 2008; 9(9): 1019-1027. doi: 10.1038/ni.1640.
    CrossRef
  18. Hellings, P. W., & Steelant, B. Epithelial barriers in allergy and asthma. The Journal of allergy and clinical immunology., 2020; 145(6): 1499-1509. doi: 10.1016/j.jaci.2020.04.010.
    CrossRef
  19. Keeler, S. P., Wu, K., Zhang, Y., Mao, D., Li, M., Iberg, C. A., Austin, S. R., Glaser, S. A., Yantis, J., Podgorny, S., Brody, S. L., Chartock, J. R., Han, Z., Byers, D. E., Romero, A. G., & Holtzman, M. J. A potent MAPK13-14 inhibitor prevents airway inflammation and mucus production. bioRxiv : the preprint server for biology., 2023; doi: 10.1101/2023.05.26.542451.
    CrossRef
  20. Bhavsar, P., Khorasani, N., Hew, M., Johnson, M., & Chung, K. F. (2010). Effect of p38 MAPK inhibition on corticosteroid suppression of cytokine release in severe asthma. The European respiratory journal., 2010; 35(4): 750-756. doi: 10.1183/09031936.00071309.
    CrossRef
  21. Bienert, S., Waterhouse, A., de Beer, T. A., Tauriello, G., Studer, G., Bordoli, L., & Schwede, T. The SWISS-MODEL Repository-new features and functionality. Nucleic acids research., 2017; 45(D1): D313-D319. doi: 10.1093/nar/gkw1132.
    CrossRef
  22. Guex, N., Peitsch, M. C., & Schwede, T. Automated comparative protein structure modeling with SWISS-MODEL and Swiss-PdbViewer: a historical perspective. Electrophoresis., 2009; 30: Suppl 1, S162-S173. doi: 10.1002/elps.200900140.
    CrossRef
  23. Waterhouse, A., Bertoni, M., Bienert, S., Studer, G., Tauriello, G., Gumienny, R., Heer, F.T., de Beer, T.A.P., Rempfer, C., Bordoli, L., Lepore, R., Schwede, T. SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Res., 2018; 46(W1): W296-W303. doi: 10.1093/nar/gky427.
    CrossRef
  24. Sultan, A., Ali, R., Sultan, T., Ali, S., Khan, N. J., & Parganiha, A. Circadian clock modulating small molecules repurposing as inhibitors of SARS-CoV-2 Mpro for pharmacological interventions in COVID-19 pandemic. Chronobiology international., 2021; 38(7): 971-985. doi: 10.1080/07420528.2021.
    CrossRef
  25. Sultan, A., Ali, R., Ishrat, R., & Ali, S. Anti-HIV and anti-HCV small molecule protease inhibitors in-silico repurposing against SARS-CoV-2 Mpro for the treatment of COVID-19. Journal of biomolecular structure & dynamics., 2022; 40(23): 12848-12862. doi: 10.1080/07391102.2021.1979097.
    CrossRef
  26. Liu, Y., Yang, X., Gan, J., Chen, S., Xiao, Z. X., & Cao, Y. CB-Dock2: improved protein-ligand blind docking by integrating cavity detection, docking and homologous template fitting. Nucleic acids research., 2022; 50(W1): W159-W164. doi: 10.1093/nar/gkac394.
    CrossRef
  27. Murail, S., de Vries, S. J., Rey, J., Moroy, G., & Tufféry, P. SeamDock: An Interactive and Collaborative Online Docking Resource to Assist Small Compound Molecular Docking. Frontiers in molecular biosciences., 2021; 8:716466. doi: 10.3389/fmolb.2021.716466.
    CrossRef
  28. Ali, R., Sultan, A., Ishrat, R., Haque, S., Khan, N. J., & Prieto, M. A. Identification of New Key Genes and Their Association with Breast Cancer Occurrence and Poor Survival Using In Silico and In Vitro Methods. Biomedicines., 2023; 11(5): 1271. doi: 10.3390/biomedicines11051271.
    CrossRef
  29. Chen, X., Xue, B., Wahab, S., Sultan, A., Khalid, M., & Yang, S. Structure-based molecular docking and molecular dynamics simulations study for the identification of dipeptidyl peptidase 4 inhibitors in type 2 diabetes. Journal of biomolecular structure & dynamics., 2023; 1-14. doi: 10.1080/07391102.2023.2291831.
    CrossRef
  30. Batool, M., Ahmad, B., & Choi, S. A Structure-Based Drug Discovery Paradigm. International journal of molecular sciences., 2019; 20(11): 2783. doi: 10.3390/ijms20112783.
    CrossRef
  31. Sahu, M., Sultan, A., & Barik, M. R. Molecular docking and high throughput screening of designed potent inhibitor to PTPN11 involved in Peptic Ulcer. South Asian Journal of Experimental Biology., 2016; 6(4): 124-130. doi:10.38150/sajeb.6(4).p124-130.
    CrossRef
  32. Torres, P. H. M., Sodero, A. C. R., Jofily, P., & Silva-Jr, F. P. Key Topics in Molecular Docking for Drug Design. International journal of molecular sciences., 2019; 20(18): 4574. doi: 10.3390/ijms20184574.
    CrossRef
  33. Gfeller, D., Grosdidier, A., Wirth, M., Daina, A., Michielin, O., & Zoete, V. SwissTargetPrediction: a web server for target prediction of bioactive small molecules. Nucleic acids research., 2014; 42(Web Server issue): W32-W38. doi: 10.1093/nar/gku293.
    CrossRef
  34. Daina, A., Michielin, O., & Zoete, V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific reports., 2017; 7: 42717. doi: 10.1038/srep42717.
    CrossRef
  35. Daina, A., Michielin, O., & Zoete, V. SwissTargetPrediction: updated data and new features for efficient prediction of protein targets of small molecules. Nucleic acids research., 2019; 47(W1): W357-W364. doi: 10.1093/nar/gkz382.
    CrossRef
  36. Barba-Ostria, C., Carrera-Pacheco, S. E., Gonzalez-Pastor, R., Heredia-Moya, J., Mayorga-Ramos, A., Rodríguez-Pólit, C., Zúñiga-Miranda, J., Arias-Almeida, B., & Guamán, L. P. Evaluation of Biological Activity of Natural Compounds: Current Trends and Methods. Molecules., 2022; 27(14): 4490. doi: 10.3390/molecules27144490.
    CrossRef
  37. Kumar, P., Nagarajan, A., & Uchil, P. D. Analysis of Cell Viability by the MTT Assay. Cold Spring Harbor protocols., 2018; 2018(6): 10. doi: 10.1101/pdb.prot095505.
    CrossRef
  38. Schenone, M., Dančík, V., Wagner, B. K., & Clemons, P. A. Target identification and mechanism of action in chemical biology and drug discovery. Nature chemical biology., 2013; 9(4): 232-240. doi: 10.1038/nchembio.1199.
    CrossRef
  39. Sultan A. Identification and development of clock-modulating small molecules – an emerging approach to fine-tune the disrupted circadian clocks, Biological Rhythm Research., 2019; 50:5, 769-786. doi:10.1080/09291016.2018.1498197.
    CrossRef
  40. Agamah, F. E., Mazandu, G. K., Hassan, R., Bope, C. D., Thomford, N. E., Ghansah, A., & Chimusa, E. R. Computational/in silico methods in drug target and lead prediction. Briefings in bioinformatics., 2020; 21(5): 1663-1675. doi: 10.1093/bib/bbz103.
    CrossRef
  41. Oates, J. T., & Lopez, D. Pharmacogenetics: An Important Part of Drug Development with A Focus on Its Application. International journal of biomedical investigation. 2018; 1(2): 111. doi: 10.31531/2581-4745.1000111.
    CrossRef
  42. Tarique, M., Chauhan, M., Tuteja T. ATPase activity of Plasmodium falciparum MLH is inhibited by DNA-interacting ligands and dsRNAs of MLH along with UvrD curtail malaria parasite growth. Protoplasma., 2017; 254;1295-1305. doi: 10.1007/s00709-016-1021-8.
    CrossRef
  43. Tarique, M., Ahmad, M., Chauhan, M., Tuteja R. Genome Wide In silico Analysis of the Mismatch Repair Components of Plasmodium falciparum and Their Comparison with Human Host. Frontiers in microbiology., 2017; 8:130. doi: 10.3389/fmicb.2017.00130.
    CrossRef
  44. Tarique, M., Tabassum, F., Ahmad, M., Tuteja R. Plasmodium falciparum UvrD activities are downregulated by DNA-interacting compounds and its dsRNA inhibits malaria parasite growth. BMC biochemistry., 2014; 15; 1-10. doi: 10.1186/1471-2091-15-9.
    CrossRef
  45. Jamal, A. Identification of phytochemicals as potential inhibitors against E6 protein of High-Risk Human Papillomavirus 16 (HPV 16) via In-Silico Structure-Based Virtual Screening Approach. Advancements in Life Sciences., 2023; 10: 3, 498-504.
  46. Mehboob, Riffat, Ahmad, FJ., Qayyum, A., Rana, MA., Tariq, MA., Tamur, S., Imran Shahid et al. Neurokinin 1 receptor antagonist along with dexamethasone reduces the inflammation in COVID-19 patients: a novel therapeutic approach.” Advancements in Life Sciences., 202310: 426-433.
(Visited 8 times, 9 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.