Virtual Screening Studies on Bone Healing Phytochemicals Isolated from Cissus quadrangularis (round variant) and Cissus rotundifolia
Sabeerali Ansarali1, Ganapathy Murugan Alagu Lakshmanan2*, Selvarasuvasuki Manikandan3, Shagufta Rashid4
1Assistant Professor, PG Department of Botany, Sri Vidya Mandir Arts and Science College,
Uthangarai 636 902, Krishnagiri, Tamil Nadu, India.
2Department of Botany, Thiru Kolanjiappar Government Arts College,
Vridhachalam - 606001, Tamil Nadu, India.
3Guest Lecturer in Botany, Government Arts and Science College, Thittakudi - 606106, Tamil Nadu, India.
4Department of Botany, Annamalai University, Annamalai Nagar - 608002, Chidambaram, Tamil Nadu, India.
*Corresponding Author E-mail: gmalakshmanan@gmail.com
ABSTRACT:
Bone fractures are a prevalent orthopaedic issue that many people experience. While the body can naturally repair a broken bone, the duration of the healing process can range from weeks to years, depending on the injury's severity and potential complications. Different techniques are used in the process of bone healing, including both experimental and computational modeling methods. Computational modeling and simulation have been used to address the limitations of experimental methods. This study focuses on screening and investigating bone healing compounds found in the leaves of Cissus rotundifolia and the stem of Cissus quadrangularis (round-stemmed variant) using molecular docking studies to identify potent compounds for bone healing treatment. GC-MS analysis revealed two active compounds: alpha-methylglucofuranoside and tetradecanoic acid. These compounds were tested against the bone healing protein ABMP2 (ID: 4MID) sourced from the Protein Data Bank (PDB) and showed good docking scores and reasonable stability. In vivo and in-vitro approaches are recommended to further elucidate the molecular mechanisms of these compounds and develop them into potent drugs for treating bone fractures. The present study clearly observed with the screened compounds could possibly develop into potent drugs for treating bone fractures.
KEYWORDS: GC-MS, Bone healing, ABMP2 Chimera, Molecular docking, PDB.
INTRODUCTION:
Bone fractures are a common injury often referred to as "the silent disease" because they occur without early symptoms but result from gradual bone loss due to conditions like osteoporosis. The bone healing process is physiologically intricate, including tissue differentiation, tissue synthesis, encompassing cell migration, and the release of cytokines and growth factors, all governed by the mechanical environment, Although the body can naturally repair a broken bone, the healing process can span weeks, months, or even years, based on the injury's severity and potential complications, depending on the injury's severity and complications.
Elderly patients, smokers, and those with metabolic diseases such as osteoporosis or diabetes experience even longer healing times and a higher risk of non-union (non-healing of the bone)1,2.
Studying bone healing through experimental methods requires advanced equipment, precise accuracy, and controlled conditions, and is often costly. These methods are further complicated by factors like unknown subject backgrounds, comorbidities, and genetic variations3,4. To address these issues, computational models and simulations of the bone healing process have been developed. However, these approaches also have limitations in clinical applications and need further refinement and validation to produce clinically relevant results5,6. Computational models deepen our comprehension of the mechanisms of bone healing by demonstrating how the mechanical environment influences the healing process7. These models also predict how mechanical environments and drug treatment strategies affect the biological and cellular processes essential for the healing of bones8,9. Furthermore, these models offer critical insights for the optimization, design, and outcome predictions of future treatment strategies. To reach clinically relevant results, continued development and refinement are necessary10,11.
Cissus rotundifolia and Cissus quadrangularis, both belonging to the grape family (Vitaceae), are plants used in traditional African and Ayurveda medicine. C. rotundifolia leaves and the fleshy stem of C. quadrangularis are primarily used to treat bone fractures, gout, rheumatoid arthritis, osteoporosis, cancer, upset stomachs, and haemorrhoids. This study aims to delve deeper into the bone healing constituents present in C. rotundifolia leaves and C. quadrangularis stem (round variant) using molecular docking studies to identify potent compounds that can be developed into drugs for bone healing treatment.
METHODS:
Plant Source Materials:
The plants were meticulously gathered from their natural habitats, and herbarium specimens were created and stored at the Department of Botany, Annamalai University. The accession numbers assigned were AUBOT360 for C. quadrangularis and AUBOT362 for C. rotundifolia. For experimental use plants were thoroughly washed with running tap water to eliminate soil particles, after which the plant parts were separated and dried in the shade. The shade-dried and powdered stem of C. quadrangularis (round-stemmed variant) and leaves of C. rotundifolia were extracted with methanol, then filtered and concentrated by vacuum distillation. GC-MS analysis results were compared and used for molecular docking studies to achieve the study's objective.
Molecular Docking:
Target Protein Identification and Ligand Structure Preparation:
For docking studies, the X-ray crystal and NMR structures of the bone-inducing protein ABMP2 (PDB ID: 4MID) were obtained from the Protein Data Bank (PDB). These structures served as the foundation for detailed analysis and interactions with potential ligands. Missing loops in the protein molecule were filled by adding hydrogen atoms and removing water molecules from the structure. The RMSD value was minimized to 0.30 Å. Using the bioactive compound structures retrieved from PubChem and ChemSketch, ligand molecules were generated and saved in SDF format12.
High-throughput virtual screening:
Major plant-derived compounds found in C. quadrangularis (round stemmed variant) and C. rotundifolia were selected as ligands for target-based virtual screening using PyRx. Prior to screening, the compounds were assessed for drug-likeness according to Lipinski's Rule of Five13. PyRx, incorporating the Open Babel program for protein and ligand visualization, was employed in this process. Both the ligands and the protein target underwent energy minimization and charge assignment. Subsequently, they were converted to pdbqt format to enable docking with the Auto Dock Vina program14.
In-silico Pharmacokinetics:
Undesirable properties of pharmacokinetic can lead to the failure of most drugs during development. Addressing these issues early in the drug discovery process is advantageous. The pharmacokinetic properties of the identified compounds, such as absorption, distribution, metabolism, and excretion (ADME) toxicity, were determined using the ADMET tool in BIOVIA Accelrys Discovery Studio v4.5 (2016) under the "Calculate Molecular Properties" section and visualized accordingly15.
RESULTS:
High-throughput virtual screening:
The PyRx virtual screening software was utilized for high throughput virtual screening. Ten major compounds identified through GC-MS analysis were carefully chosen for computational screening analyses. The 2D and 3D structures of these compounds were obtained by using online tool PubChem. The high throughput virtual screening of C. quadrangularis (round variant) revealed pharmacological activities in three compounds: stigmast-5-en-3-ol, alpha-methylglucofuranoside, and stigmasterol. In C. rotundifolia, tetradecanoic acid and 7-tetradecenal were found. Overall, five biological compounds showed drug-likeness. The binding affinities observed from the virtual screening were -5.2, -6.0, -8.2, -6.2, and -7.2 for the respective compounds (Table 1).
Table 1. Virtual screening results of C. quadrangularis (round variant) and C. rotundifolia derived compounds
Name of the Species |
Name of the Compound |
Hepato toxicity Binding Affinity |
Cissus quadrangularis (round stem variant) |
Stigmast-5-en-3-ol, (3.beta.) |
-5.2 |
Alpha-Methylglucofuranoside |
-6.0 |
|
Stigmasterol |
-8.2 |
|
|
Ergost-5-en-3-ol |
-5.3 |
-9.1 |
||
Cissus rotundifolia |
Tetradecanoic acid |
-6.2 |
7-Tetradecenal |
-7.2 |
|
Beta.-Amyrin |
-8.4 |
|
Olean-18-En-3-ol |
-5.4 |
|
Squalene |
-8.1 |
Screening of Compounds through Pharmacokinetic Properties:
Pharmacokinetics uses mathematical principles to study how drugs enter the body, spread through it, are broken down, and are removed. This analysis helps determine the drug's duration and impact. (ADMET). Understanding these parameters is crucial for designing an appropriate drug regimen. Based on the virtual screening results, five compounds were filtered from the initial ten and tested for their ADMET properties (Table 2).
The ADMET properties of all ten compounds were evaluated using the pkCSM software, which can be accessed at http://biosig.unimelb.edu.au/pkcsm. Among these five compounds, two exhibited drug-likeness. Alpha-Methylglucofuranoside showed a Log P value of -3.2214, and tetradecanoic acid showed a Log P value of -2.7247, both with very loss toxicity levels and adherence to Lipinski's Rule of Five (Figures 1 and 2). Alpha-methylglucofuranoside from C. quadrangularis (round variant) and tetradecanoic acid identified in C. rotundifolia were selected for molecular docking analysis using the bone healing target protein (Figure 3). The compounds exhibited different interactions, including conventional hydrogen bonds and alkyl-shaped interactions with the target protein.
Table 2. ADMET Properties of GC-MS derived compounds from the Cissus quadrangularis (round stem variant) and Cissus rotundifolia
S. No |
Compound Name |
Absorption |
||||||
Water Solubility |
Permeability of CaCO2 |
Human Intestinal absorption Percentage |
Permeability of Skin |
P.glyco protein substrate |
P.glyco protein I inhibitor |
P.glyco protein II inhibitor |
||
1 |
Stigmast-5-en-3-ol |
-6.773 |
1.201 |
94.464 |
-2.783 |
No |
Yes |
Yes |
2 |
Alpha-Methylglucofuranoside |
-0.581 |
-0.153 |
45.236 |
-3.276 |
No |
No |
No |
3 |
Stigmasterol |
-6.682 |
1.213 |
94.97 |
-2.783 |
No |
Yes |
Yes |
4 |
Tetradecanoic acid |
-4.952 |
1.56 |
92.691 |
-2.705 |
No |
No |
No |
5 |
7-Tetradecenal, (Z)- |
-1.119 |
-0.258 |
32.274 |
-2.737 |
No |
No |
No |
|
Distribution |
Metabolism |
Excretion |
|||||||||||
Human VDSS |
Unbound Fraction |
Permeability of BBB |
Permeability of CNS |
CYP3A4 substrate |
CYP1A2 inhibitor |
Clearance Total |
||||||||
1 |
Stigmast-5-en-3-ol |
0.193 |
0 |
0.781 |
-1.705 |
Yes |
No |
-0.628 |
||||||
2 |
Alpha-Methylglucofuranoside |
-0.07 |
0.915 |
-0.756 |
-4.001 |
No |
No |
0.696 |
||||||
3 |
Stigmasterol |
0.178 |
0 |
0.771 |
-1.652 |
Yes |
No |
0.618 |
||||||
4 |
Tetradecanoic acid |
-0.578 |
0.171 |
-0.027 |
-1.925 |
No |
No |
1.693 |
||||||
5 |
7-Tetradecenal, (Z)- |
-0.217 |
0.821 |
-0.894 |
-3.667 |
No |
No |
0.639 |
||||||
|
Toxicity |
|||||||||||||
Toxicity of AMES |
Max. tolerated dose in human |
Inhibitor of hERG II |
Oral rat acute toxicity (LD50) |
Oral rat chronic toxicity (LDAEL) |
Toxicity of T.pyriformis |
Minnow toxicity |
Log P value |
|||||||
1 |
Stigmast-5-en-3-ol |
No |
-0.621 |
Yes |
2.552 |
0.855 |
0.43 |
-1.802 |
8.0248 |
|||||
2 |
Alpha-Methylglucofuranoside |
No |
1.951 |
No |
1.288 |
3.113 |
0.285 |
4.272 |
-3.2214 |
|||||
3 |
Stigmasterol |
No |
-0.664 |
Yes |
2.54 |
0.872 |
0.433 |
-1.675 |
7.8008 |
|||||
4 |
Tetradecanoic acid |
No |
-0.559 |
No |
1.477 |
3.034 |
0.978 |
-0.601 |
-2.7247 |
|||||
5 |
7-Tetradecenal, (Z)- |
No |
1.626 |
No |
1.128 |
3.529 |
0.285 |
4.869 |
-2.3214 |
|||||
Figure 1. 3D (three-dimensional) Structure of Alpha-Methylglucofuranoside molecule
Figure 2. 3D (three-dimensional) Structure of Tetradecanoic acid molecule
Figure 3. 3D Structure of bone healing protein (ID:4MID)
In the in-silico docking results, alpha-methyl gluco furanoside showed interactions with phenylalanine, tryptophan, methionine, and isoleucine amino acids, with a docking energy of -8.04418 kcal/mol (Figure 4).
Tetradecanoic acid interacted with cysteine amino acids in the bone healing protein, with a docking energy of -11.5209 kcal/mol (Figure 5). These two compounds showed maximum docking energy with the bone healing protein based on the molecular docking results (Tables 3 and 4).
Figure 4. 3D interaction of Alpha-Methylglucofuranoside with target protein (4MID)
Figure 5. 3D interaction of Tetradecanoic acid with target protein (4MID)
Table 3. Molecular docking of Alpha-Methylglucofuranoside interact with a target protein
Diseases/ Protein |
PDB – ID |
Docking energy (kcal/mol) |
Interactions |
Amino acid |
Binding site of amino acid |
Bone healing |
4MID |
-8.04418 |
Conventional Hydrogen Bond |
Cysteine |
111 |
Unfavorable Acceptor- Acceptor |
- |
- |
Table 4. Molecular docking of Tetradecanoic acid interact with a target protein
Diseases/ Protein |
PDB - ID |
Docking energy (kcal/mol) |
Interactions |
Amino acid |
Binding site of amino acid |
Bone healing |
4MID |
-11.5209 |
Unfavorable Bump |
Phenylalanine |
23 |
Conventional Hydrogen Bond |
Tryptophan |
28 |
|||
Alkyl |
Methionine |
89, 106 |
|||
Isoleucine |
103 |
DISCUSSION:
Drug development and discovery is a lengthy, complex, costly, in addition risky process. Computer-aided drug design (CADD) methodologies are extensively employed in the industry pharmaceutical to expedite this process. Applying computational tools in hit-to-lead optimization expands the exploration of chemical space while minimizing the number of compounds needing synthesis and in vitro testing16. The ADME profiles of the selected five compounds from C. quadrangularis (round variant) and C. rotundifolia showed good bioavailability. The two active components identified in this study, alpha-methylglucofuranoside and tetradecanoic acid, were subjected to further in-silico studies to analyse their effectiveness against the target protein. Commonly, rhBMP2 (recombinant bone morphogenetic protein) is used in clinical treatment for bone fracture patients but can cause side effects. The novel activin ABMP2 chimera promotes bone healing more effectively than rhBMP217. The structures of the target protein, ABMP2 chimera (ID: 4MID), were sourced from PDB (Protein Data Bank). These two compounds showed good docking scores and reasonable stability. The ADMET profile and Log P values support the bioavailability of these compounds compared to others. This investigation demonstrates significant bone healing activity in two Cissus species.
CONCLUSION:
In this study, phytocompounds from C. quadrangularis (round stem variant) and C. rotundifolia leaves were docked with the bone healing protein (ABMP2 chimera). Alpha-methylglucofuranoside and tetradecanoic acid were identified as key compounds. Based on molecular docking analysis, tetradecanoic acid showed promising target formation against bone healing. Both in-vivo and in-vitro approaches were recommended to elucidate the molecular mechanisms of this compound, aiming to develop it into a potent drug for treating bone fractures.
REFERENCE:
1. Abd Jalil MA, Shuid AN, Muhammad N. Role of medicinal plants and natural products on osteoporotic fracture healing. Evidence-Based Complementary and Alternative Medicine (eCAM). 2012; (2012): 714512. doi:10.1155/2012/714512.
2. Basim Mohammed Dhahir, Imad Hadi Hameed, Ahmed Radee Jaber. Prospective and Retrospective Study of Fractures According to Trauma Mechanism and Type of Bone Fracture. Research Journal of Pharmacy and Technology. 2017; 10(11): 3810-3818. doi: 10.5958/0974-360X.2017.00692.8
3. Moran CJ, Ramesh A, Brama PA, O'Byrne JM, O'Brien FJ, Levingstone TJ. The benefits and limitations of animal models for translational research in cartilage repair. Journal of Experimental Orthopaedics. 2016; 3(1): 1-12. doi:10.1186/s40634-015-0037-x.
4. Anil Malik, Neelam Malik, Priyanka Dhiman, Anurag Khatkar, Saloni Kakkar. Molecular docking, synthesis, α-amylase inhibition, urease inhibition and antioxidant evaluation of 4-hydroxy-3-methoxy benzoic acid. Research Journal of Pharmacy and Technology. 2019; 12(12): 5653-5663. doi: 10.5958/0974-360X.2019.00978.8
5. Padmini R, Sitrarasi R, Razia M. Molecular Docking Studies of Bioactive Compounds from Allium sativum Against EML4-ALK Receptor. Research Journal of Pharmacy and Technology. 2017; 10(11): 3741-3747. doi: 10.5958/0974-360X.2017.00679.5
6. Carlier A, Geris L, Lammens J, Van Oosterwyck H. Bringing computational models of bone regeneration to the clinic. Wiley Interdisciplinary Reviews: System Biology and Medicine. 2015; 7(4): 183-194. doi:10.1002/wsbm.1299.
7. Habeela Jainab N, Mohan Maruga Raja, MK. In Silico Molecular Docking Studies on the Chemical Constituents of Clerodendrum phlomidis for its Cytotoxic Potential against Breast Cancer Markers. Research Journal of Pharmacy and Technology, 2018; 11(4): 1612-1618. doi: 10.5958/0974-360X.2018.00300.1
8. Geris L, Reed AA, Vander Sloten J, Simpson AH, Van Oosterwyck H. Occurrence and treatment of bone atrophic non-unions investigated by an integrative approach. PLoS Computational Biology. 2010; 6(9): 1-11. doi:10.1371/journal.pcbi.1000915.
9. Carlier A, van Gastel N, Geris L, Carmeliet G, Van Oosterwyck H. Size does matter: an integrative in vivo-in silico approach for the treatment of critical size bone defects. PLoS Computational Biology. 2014; 10(11): 1-21. doi:10.1371/journal.pcbi.1003888.
10. Mohini Upadhye, Parikshit Gandhi, Mohini Phanse. Pharmacognostical, Phytochemical Studies and Comparative Extraction Techniques of Daucus carota. Research Journal of Pharmacognosy and Phytochemistry. 2014; 6(3): 115-117.
11. Firoj Alam, Badruddeen, Anil Kumar Kharya, Akhtar Juber, Mohammad Irfan Khan. Naringin: Sources, Chemistry, Toxicity, Pharmacokinetics, Pharmacological Evidences, Molecular Docking and Cell line Study. Research Journal of Pharmacy and Technology. 2020; 13(5): 2507-2515. doi: 10.5958/0974-360X.2020.00447.3
12. Kannadasan R, Arnold Emerson I, Saleem Basha MS. Docking of HIV-1 with Neem using Autodock in Bioinformatics. Research Journal of Pharmacy and Technology. 2017; 10(11): 3877-3880. doi: 10.5958/0974-360X.2017.00704.1
13. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Advanced Drug Delivery Reviews. 2001; 46(1-3): 3-26. doi:10.1016/s0169-409x(00)00129-0.
14. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal Computational Chemistry. 2010; 31(2): 455-461. doi:10.1002/jcc.21334.
15. Enyedy IJ, Egan WJ. Can we use docking and scoring for hit-to-lead optimization?. Journal of computer-aided molecular design. 2008; 22(3-4): 161-168. doi:10.1007/ s10822-007-9165-4.
16. Jeyabaskar Suganya, Viswanathan T, Mahendran Radha, Nishandhini Marimuthu. In silico Molecular Docking studies to investigate interactions of natural Camptothecin molecule with diabetic enzymes. Research Journal of Pharmacy and Technology. 2017; 10(9): 2917-2922. doi: 10.5958/0974-360X.2017.00515.7.
17. Yoon BH, Esquivies L, Ahn C, et al. An activin A/BMP2 chimera, AB204, displays bone-healing properties superior to those of BMP2. Journal of bone and Mineral Research. 2014; 29(9): 1950-1959. doi:10.1002/jbmr.2238.
Received on 11.06.2022 Revised on 13.11.2023 Accepted on 23.09.2024 Published on 24.12.2024 Available online from December 27, 2024 Research J. Pharmacy and Technology. 2024;17(12):5724-5728. DOI: 10.52711/0974-360X.2024.00871 © RJPT All right reserved
|