In our in vitro study, we have revealed the potent beneficial activity of Berberine-LCNs against human lung cancer cell line (A549). The most notable activity observed with Berberine-LCNs were significant inhibition of proliferation, colony formation, invasion/migration and protein expression associated with these cancer progression events.
Researchers have widely explored various herbal plants extracts and their active chemical constituents for their potent anticancer activities, including lung cancer (Hardwick et al. 2021, Panth et al. 2017, Solanki et al. 2020). In this study, we have investigated a widely known and commercially available anti-inflammatory compound Berberine to investigate if it possesses anticancer potential against A549 cells. Berberine has very low dissolution rate and oral bioavailability owing to the fact that it is very slightly soluble in water, hence limiting its clinical application (Singh et al. 2020). Due to poor bioavailability, the oral dosage of Berberine prescribed for daily treatment of certain disease conditions ranges as high as 0.5–1.5 gram in multiple daily doses (Battu et al. 2010). Therefore, it is urgent necessary to find alternative formulation of Berberine utilizing advanced drug delivery system. Liquid crystalline nanoparticles (LCNs) offer versatility in designing and delivery of drug targeting chronic respiratory disease (Chan et al. 2021). We have previously shown that Rutin and Naringenin loaded LCNs offer better activity against oxidative stress and inflammation than using power form of these compound (Mehta et al. 2021, Paudel et al. 2020, Wadhwa et al. 2021). In line with the same hypothesis, we have designed Berberine-loaded LCNs and explored the activity against lung cancer proliferation and migration. To improve the physiochemical properties and enhanced efficacy, our Berberine LCNs formulation were tested for various characterization parameter (Table 1, Table 3, Fig. 5 and Fig. 6). These pharmaceutical tests showed a very favourable characterization data in terms of particle size, polydispersity index, entrapment efficiency and in vitro release.
The molecular modelling simulations revealed two important findings. Firstly, the drug molecule was incorporated within the geometrical space of the lipid molecule as a strained bimolecular complex. Secondly, both the bi- and tri-molecular complexes were stabilized by van der Waals forces against all other bonding interactions (bond, angle, and dihedral energies). This is interesting because vdW energy component represents intermolecular interactions, which are non-covalent and reversible in nature. This is evident from the preferable positioning of the drug within the lipid molecule and further encapsulation into the MOO-PF127 system under formulation conditions. It must be noted that this vdW attraction also brought into strained network structures, which may ultimately be responsible for the release of the drug once the environment changes such as in release medium or in vitro cell growth medium and conditions (Huo et al. 2021).
Next, we performed in vitro biological activity of our formulation against A549 cells. For cancer/tumor progression, two major event a) proliferation/growth and b) migration/invasion facilitate the entire tumorigenesis. Our colorimetric MTT assay that measure the cell viability and further cell count with trypan blue assay showed significant inhibition of A549 in a concentration dependent manner with an IC50 value of 10.10 µM (Fig. 7). An in vitro study by Qi Hw et al., 2014 observed the IC50 of Berberine powder to inhibit A549 cell proliferation using MTT assay was 56.15 ± 3.14 µM (Qi et al. 2014). This suggests that our Berberine-LCNs formulation was > 5 time more potent than Berberine powder. Inhibition of A549 cell growth was also observed in our colony formation assay where Berberine-LCNs treatment at a dose of 2.5 and 5 µM significantly inhibited the colony formation (Fig. 9). Likewise, in our anti-migratory assay performed by measuring the wound closure after 24 hours of Berberine-LCNs treatment and migration of A549 cells in trans-well chamber (Boyden’s chamber) assay revealed potent anti-migratory activity of Berberine-LCNs (Fig. 8A-D).
For mechanistic approach, we further elucidate the protein expression related with cancer cell migration and proliferation. Epithelial-mesenchymal transition (EMT) is a phenomenon where epithelial cells change to mesenchymal stem cells phenotype due to loss of their cell polarity and cell adhesion property that makes them more invasive and metastasis (Mittal 2016). Various proteins are involved in EMT in NSCLC that facilitate cancer cell invasion/metastasis (TSOUKALAS et al. 2017a). Among these proteins, the role of SNAIL (Yang et al. 2017b), p27 (Zhao et al. 2015) and Vimentin (Tsoukalas et al. 2017b) to promote EMT has been widely studied. Silencing of SNAIL was correlated with suppression of tumor cell invasion by reversing EMT in NSCLC (Yang et al. 2017b) while there was negative correlation between vimentin expression and overall survival in NSCLC (Tsoukalas et al. 2017b).
In our study, treatment of Berberine-LCNs at a dose of 5µM significantly downregulated the protein expression of SNAIL, p27 and vimentin (Fig. 10a, 10b, 10c). Likewise, certain proteins play key role in angiogenesis, proliferation, survival of cancer cell (Fig. 12). Platelet-derived growth factor-AA (PDGF-AA) is crucial autocrine regulator of vascular endothelial growth factor expression in NSCLC, and it facilitates the process of angiogenesis (Shikada et al. 2005). Galectin-3 is another protein that promotes angiogenesis (Newlaczyl &Yu 2011) and metastasis (Reticker-Flynn et al. 2012). Similarly, Axl (an oncogenic protein) expression is observed in 60% of NSCLC cell line and it promote in cell proliferation as well as adhesion (Kim et al. 2015, Wimmel et al. 2001). Progranulin expression is correlated with epithelial cell growth including A549 and promotes tumor growth in vivo (He &Bateman 1999). ERBB3 belongs to the member of epidermal growth factor receptor (EGFR) family, and it plays vital role in mediating NSCLC proliferation and differentiation (Guo et al. 2016). Protein such as Bcl-xL and Survivin are involved in cancer cell survival by inhibiting apoptosis of cancer cell (Hirano et al. 2015, Schott et al. 1995). Capthesin S (CTSS) protein is also strongly associated with NSCLC pathology owing to the fact that CTSS can degrade proteoglycan of interstitial matrix such as decorin (Kehlet et al. 2017) and nidogen-1 (Willumsen et al. 2017) to promote the NSCLC migration. Some protein such as CEA-related cell adhesion molecule 5 (CECAM5) stimulates both proliferation and migration of NSCLC (Zhang et al. 2020). In our protein array (Fig. 11), Berberine LCNs treatment at 5µM significantly inhibited the protein expression of PDGF-AA, Galectin 3, Axl, Progranulin, ERBB3, BCLx, Cathepsin S, Survivin, and CEACAM5. The details anti-cancer mechanism of action of Berberine LCNs is shown in Fig. 12. Taken together, the anti-cancer activity of Berberine LCNs against A549 cell was due to its potency to inhibited key protein involved in EMT, angiogenesis, metastasis, and proliferation.
There are few limitations as well as possibility of future direction from our study. Firstly, we can further prove the anti-cancer activity of Berberine-LCNs through gene approach. In current study, we have performed only protein related work and therefore to investigate if Berberine-LCNs inhibits key genes involved in A549 cell proliferation and migration would be interesting. Secondly, our study is entirely in vitro, and this opens a new platform for further in-depth studies in in vivo pre-clinical animal models of lung cancer. Third, we can also test the anti-cancer activity of Berberine-LCNs in other cell line of lung cancer such as Calu3, H460. Nevertheless, Berberine can a potential therapeutic alterative for lung cancer if we can formulate it appropriately using advance drug delivery system and manipulate the physiochemical parameters to increase efficacy, stability, cellular uptake.