Recently, many studies have showed that enzymes take part in amino acid biosynthesis are vital drug target as humans lack a homolog for most of these targets (Kefala et al. 2005; DeJesus et al, 2017). Thus, targeting enzymes involved in amino acid production may result in the identification of highly specific antitubercular compounds. Among the essential amino acids, L-lysine is derived from L-aspartate and leads to L-lysine in nine enzymatically catalyzed steps. The final step in this pathway is carried out by the enzyme diaminopimelate decarboxylase (LysA, DAPDC), which converts meso-diaminopimelate (DAP) to lysine in a PLP-dependent decarboxylation reaction (Kefala et al. 2005). Several studies have shown that M. tuberculosis strains with defects in biosynthesis of L-lysine is unable to grow in liquid cultures and establish infection in host tissues. In agreement, M. tuberculosis strains with deletions in enzymes involved in biosynthesis of other essential amino acids are also attenuated for growth in vivo (Kefala et al. 2005; DeJesus et al, 2017; Tiwari et al, 2018). In this project we described on the mechanism of action for DL25 compound against Mycobacterium tuberculosis and also explained its pharmacokinetics. This result provided useful information that could be vital for searching compound with minimal side effect. Also, they were tested (in vitro) against the mycobacterial LysA (Rv1293) enzyme to get an insight into their probable mechanism of action. Table 1 showed details about DL25 compound ADME-related physical properties and bioavailability, such as molecular weight (M.W.), water solubility, topological polar surface area (TPSA), octanol-water partition coefficients (LogP), gastrointestinal absorption (GIA), bioavailability score, No. H-bond donors, and No. H-bond acceptors, which are important properties of a compounds proposed for lead compound. New compound with a polar surface area of greater than 140 Å2 tend to be poor at permeating cell membranes, which is not violated in this study by DL25 compound (Umar et al., 2020; Kralj et al., 2023). Except for GI absorption, bioavailability score, and water solubility, all the results from this study are consistent with other published data regarding the physicochemical properties and bioavailability of DL25 compound, which required further modification. Pharmaceutical companies follow one or more regulations that determining the drug likeness of oral administration that includes Lipinski, Veber, Egan, and Muegge. Each ADMET parameter was compared to the standard for that parameters range, shown in Table 2. According to Lipinski rule of thumb, approximately 95% of oral compounds pass three of four of the following rules: MW ≤ 500Da, calculated LogP (cLogP) ≤ 5 and hydrogen bond acceptors (HBAs) ≤ 10, and hydrogen bond donors (HBD) ≥ 5 (Khare et al., 2023). Subsequently, additional in silico properties such as polar surface area (PSA ≤ 140 A˚ 2), the number of rotatable bonds (NRotBs ≤ 10–20), and more complex 3D properties have been added, thereby expanding the correlations with absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters (Egan et al., 2000; Muegge, 2006; Veber et al., 2002). In this study, the DL25 compound met the majority of the rules criteria, with maximum violation of three out of eight as shown in Table 2. As a result, only the molecular weight (MW), number of rings, and number of rotatable bonds break the limitations. These broken rules may not necessarily imply a lack of medication efficacy of DL25 compound. The results of the drug-likeness and physicochemical attributes provided here shown that the DL25 compound has promise efficacy following oral administration.
In the present study, the anti-tubercular activity of DL25 compound was evaluated in vitro against Mtb and three strains of non-Mtb, namely H37Rv, H37Mab, H37mm, H37ms, and H37AlRa. The in vitro results showed excellent inhibitory activities against both Mtb strain and non-Mtb strains. DL25 Compound was found to be more potent against both Mtb than non-Mtb, with MIC90 values ranging from 1 to 4 µg/mL. Time kill assay provides more precise information concerning the effect of the test compound on bacteria since the measurements are taken over different times. Furthermore, this technique has ability to differentiate bactericidal activities and bacterial re-growth (Soudeiha, Dahdouh, Azar, Sarkis, & Daoud, 2017). The growth curves of M. tuberculosis H37Ra (Alar) treated with different concentrations of DL25 compound (0.5×MIC, MIC, 2×MIC, and 4xMIC) demonstrated that DL25 compound could inhibit the growth and reproduction of bacteria. The growths of bacterial cells treated with MIC and 2×MIC were inhibited. The growth of the M. tuberculosis H37Ra (Alar) treated with 0.5MIC and 1/4MIC was also a little less than that of bacteria in the control group up to day four. These results specify that the anti- M. tuberculosis H37Ra (Alar) activity of 0.5MIC and 1/4MIC of DL25 compound could hamper bacterial growth to some extent as shown in Fig. 1. Next, we performed assays to evaluate the in vitro antitubercular activity of the identified LysA inhibitors. At this juncture, DL25 compound absorption was investigated by considering parameters such as, lipophilicity, aqueous solubility, Caco2 permeability, Pgp I & II inhibitor, and Pgp substrate as shown in Table 3. Particularly, lipophilicity suggested excellent infusion through the mycobacterial cell wall and into macrophages, and higher probability of being effective against dormant M. tuberculosis. The poor water solubility showed attention during drug formulation, since it is linked to poor bioavailability and poor gastrointestinal (GI) absorption. Therefore, further improvement of solubility could be required. But, high Caco-2 cell permeability showed excellent absorption. Lastly, important parameter on absorbance is the amount of efflux from cellular tissue, especially by p-glycoprotein (Pgp). As shown in Table 3, the contradictory results regarding the binding of DL25 compound to Pgp suggests further experimentation. In either case however, whether it is not a substrate for Pgp, or whether it is a substrate, and inhibits Pgp I and II, the results indicate a lack of export, meaning DL25 compound is likely to accumulate in the target organs. Under DL25 compound distribution investigation three parameters were reported. First, it was predicted that the DL25 compound would not pass through the blood-brain barrier (BBB) or the central nervous system (CNS). As a result, DL25 compound is questionable to be effective in treating tuberculosis meningitis. Second, blood plasma volume of distribution of DL25 compound (logVDss) is on the higher end of the spectrum, implying moderate delivery to infected areas. Phase I drug-metabolizing enzymes such as, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4, and CYP3A5 are known to be expressed in the human intestine (Li et al., 2020). In spite of contradictory conclusions about CYP2D6 inhibition by DL25 compound, the Deep-PK’s and SwissADME’s calculations presented in this study regarding the four main cytochrome isoforms are in agreement as shown in Table 3. Although no inhibition of CYP2D6 is expected, inhibition of CYP3A4 suggests that care should be taken when co administering DL25 compound with other antimycobacterial drugs, to prevent toxic accumulation of either or both drugs. The excretion of DL25 compound was reported as total clearance rate (CLtot = 0.144mL/min/kg) and the elimination half-time (t1/2 = 3h) as shown in Table 3. The t1/2 is significantly lower than that of the other first-line drugs, Rifampicin (t1/2 =7Hr) (Ji et al, 1993), higher than Isoniazid (t1/2 = 1.7Hr), and same to Ethambutol (t1/2 = 3Hr) (Arbex et al, 2010), which is beneficial for patient coherence and may reduce the occurrence of drug resistance. As shown in Table 3, DL25 compound is neither carcinogenic, nor mutagenic, nor likely to interfere with heart rhythms and has low hepatotoxicity. However, prediction server inconsistent results regarding hepatotoxicity demand further experimentation. Biodegradability of DL25 compound also suggests that it is safe in the face of environmental pollution.
It is widely known that drugs commonly target several proteins (as opposed to only one particular protein) (Li et al, 2020; Khare et al, 2023). A total of seven M. tuberculosis proteins were identified by Docking Server as potential targets of DL25 out of one hundred proteins. These are listed according to their docking scores in Table 4, along with their respective interacting residues, UniProt and PDB accession codes. The docking affinities calculated by Autodock ranged between − 3.74 to -10.73 kcal/mol and indicated strong binding to LysA (Rv1293), LpdA (Rv3303c), and SecA1 (Rv3240) targets (Table 4). These findings imply that DL25 compound has a multi-target to inhibits M. tuberculosis growth and killing mechanism by targeting various enzymes, including LysA (Rv1293). In this study, to predict DL25 compound target, we determined their ranking by binding affinity. Based on first ranking order, seven protein target annotated to highly prioritized genes were provided, of which only LysA (Rv1293), was also identified by Autodock as a potential target for DL25 compound and molecular dynamic simulation run for it. In this study, we investigated the interactions between LysA (Rv1293) protein and DL25 compound using molecular dynamics simulations and experimental data. The root-mean-square deviation (RMSD) of LysA (Rv1293) protein and DL25 compound atoms were calculated to assess the conformational flexibility and stability of the complex over the simulation time. The RMSD plot (Fig. 3) showed that the LysA - DL25 compound complex exhibits a relatively low RMSD value, indicating a stable binding interface. This is in agreement with previous studies that have reported stable binding between the protein and ligand (Kumar et al, 2018). Our results showed that the binding of the DL25 compound to LysA (Rv1293) was accompanied by significant changes in the protein's flexibility, as measured by the root mean square fluctuation (RMSF) analysis. The RMSF values calculated for each residue revealed that the binding pocket of the protein underwent significant conformational changes upon DL25 compound binding, indicating a strong correlation between flexibility and binding affinity. The radius of gyration (Rg) was also calculated to analyze the compactness and shape of the LysA (Rv1293)-DL25 compound complex. The Rg value (Fig. 1) indicates that the complex has a more compact structure compared to the free LysA (Rv1293), suggesting that the DL25 compound binding induces a conformational change in the LysA (Rv1293). This observation is consistent with previous studies on protein-ligand interactions, which have shown that ligand binding can induce significant changes in protein structure and dynamics (Wong et al., 2019). The results of this study provide new insights into the binding mechanism and stability of the LysA (Rv1293)-DL25 complex. The stable binding interface and compact structure of the complex suggest that the DL25 compound plays a crucial role in stabilizing the protein structure, which may have implications for understanding the biological function of this complex. Furthermore, our analysis of hydrogen bonding (H-bonding) patterns between the LysA (Rv1293) and DL25 compound revealed a crucial role for these interactions in stabilizing the complex. Specifically, we identified a network of H-bonds between residues THR345, ARG383, TYR263 on the LysA (Rv1293) and DL25 compound that played a key role in maintaining the binding affinity of the complex. The presence of these H-bonds was found to be essential for the optimal alignment of the DL25 compound within the binding pocket, allowing for effective recognition and binding to occur (Wong et al, 2019). The identification of specific H-bonds involved in this process provides valuable targets for future mutagenesis studies aimed at understanding their functional significance.