Molecular modeling studies of some phytoligands from Ficus sycomorus fraction as potential inhibitors of cytochrome CYP6P3 enzyme of Anopheles coluzzii

Authors

  • Abba Babandi Bayero University, Nigeria
  • Chioma A. Anosike University of Nigeria, Nigeria
  • Lawrence U.S. Ezeanyika University of Nigeria, Nigeria
  • Kemal Yelekçi Kadir Has University, Turkey
  • Abdullahi Ibrahim Uba Beijing Computational Science Research Center, China

DOI:

https://doi.org/10.35516/jjps.v15i2.324

Keywords:

Homology modeling, CYP6P3, Molecular docking, Molecular dynamics simulation ligand efficiency, CYP6P3 inhibitors

Abstract

The major obstacle in controlling malaria is the mosquito’s resistance to insecticides, including pyrethroids. The resistance is mainly due to the over-expression of detoxification enzymes such as cytochromes. Insecticides tolerance can be reduced by inhibitors of P450s involved in insecticide detoxification. Here, to design potential CYP6P3 inhibitors, a homology model of the enzyme was constructed using the crystal structure of retinoic acid-bound cyanobacterial CYP120A1 (PDB ID: 2VE3; Resolution: 2.1 Å). Molecular docking study and computational modeling were employed to determine the inhibitory potentials of some phytoligands isolated from Ficus sycomorus against Anopheles coluzzii modeled P450 isoforms, CYP6P3, implicated in resistance. Potential ligand optimization (LE) properties were analyzed using standard mathematical models. Compounds 5, 8,and 9 bound to the Heme iron of CYP6P3 within 3.14, 2.47 and 2.59 Å, respectively. Their respective binding energies were estimated to be -8.93, -10.44, and -12.56 Kcal/mol. To examine the stability of their binding mode, the resulting docking complexes of these compounds with CYP6P3 were subjected to 50 ns MD simulation. The compounds remained bound to the enzyme and Fe (Heme):O (Ligand) distance appeared to be maintained over time. The coordination of a strong ligand to the heme iron shifts the iron from the high- to the stable low-spin form and prevented oxygen from binding to the heme thereby inhibiting the catalytic activity. The LE index showed the high potential of these compounds (5 and 8) to provide a core fragment for optimization into potent P450 inhibitors.

Author Biographies

Abba Babandi, Bayero University, Nigeria

Department of Biochemistry

Department of Biochemistry, University of Nigeria, Nsukka, Nigeria

Chioma A. Anosike, University of Nigeria, Nigeria

Department of Biochemistry

Lawrence U.S. Ezeanyika, University of Nigeria, Nigeria

Department of Biochemistry

Kemal Yelekçi, Kadir Has University, Turkey

Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Science, Kadir Has University, Turkey

Abdullahi Ibrahim Uba, Beijing Computational Science Research Center, China

Complex Systems Division

References

(1) World Health Organization (WHO, 2014). Division of Malaria and Other Parasitic Diseases. From Malaria Control to Malaria Elimination: A Manual for Elimination Scenario Planning. World Health Organization; 2014.

(2) World Health Organization(WHO, 2013). Larval source management: a supplementary measure for malaria vector control: an operational manual. Geneva: World Health Organization; 2013. p. 116. Available from: www.who.int/malaria/publications/atoz/9789241505604/en/%0A%0A

(3)Rose, R.I. Pesticides and Public Health: Integrated Methods of Mosquito Management. Emerg Infect Dis J., 2001; 7:17.

(4) Katagi, T. Photodegradation of pesticides on plant and soil surfaces. Rev Environ Contam Toxicol., 2004; 182: 1-189.

(5) Feyereisen, R. Insect P450 enzymes. Ann. Rev. Entomol, 1999; 44: 507-533.

(6) Scott J. G. Cytochromes P450 and insecticides resistance. Insect Biochem Mol Biol. 1999; 29: 757-777.

(7) Holt, R. A., Subramanian, G. M., Halpern, A., Sutton, G. G., Charlab, R., Nusskern, D. R., Wincker, P., Clark, A. G., Ribeiro, J. C. and Wides, R. The genome sequence of the malaria mosquito Anopheles gambiae. Science, 2002; 298: 129-149.

(8) Nene, V., Wortman, J. R., Lawson, D., Haas, B., Kodira, C., Tu, Z. J., Loftus, B., Xi, Z., Megy, K. & Grabherr, M. Genome sequence of Aedes aegypti, a major arbovirus vector. Science, 2007; 316: 1718-1723.

(9) Arensburger, P., Megy, K., Waterhouse, R. M., Abrudan, J., Amedeo, P., Antelo, B., Bartholomay, L., Bidwell, S., Caler, E. & Camara, F. Sequencing of Culex quinquefasciatus establishes a platform for mosquito comparative genomics. Science, 2010; 330: 86-88.

(10) Nikou, D., Ranson, H. & Hemingway, J. An adult-specific CYP6 P450 gene is overexpressed in a pyrethroid-resistant strain of the malaria vector, Anopheles gambiae. Gene, 2003; 318: 91-102.

(11) David, J.P., Strode, C, Vontas, J., Nikou, D., Vaughan, A., Pignatelli, P.M., Louis, C., Hemingway, J., Ranson, H. The Anopheles gambiae detoxification chip: a highly specific microarray to study metabolic-based insecticide resistance in malaria vectors. Proc Natl Acad. Sci. 2005; 102 (11): 4080-4084. 10.1073/pnas.0409348102.

(12) Muller P, Donnelly MJ, Ranson H. Transcription profiling of a recently colonised pyrethroid resistant Anopheles gambiae strain from Ghana. BMC Genomics, 2007; 8: e36

(13) Mulamba, C., Irving, H., Riveron, J.M., Mukwaya, L.G., Birungi, J, Wondji, C.S. Contrasting Plasmodium infection rates and insecticide susceptibility profiles between the sympatric sibling species Anopheles parensis and Anopheles funestus s.s: a potential challenge for malaria vector control in Uganda. Parasit Vect., 2014; 7: 71.

(14) Mansuy, D. The great diversity of reactions catalyzed by cytochrome P450. Comp Biochem Physiol Part C, 1998; 121:5-14.

(15) Guengerich, F. P. Mechanisms of Cytochrome P450-Catalyzed Oxidations. ACS Catal., 2018; 8(12):10964–10976. doi:10.1021/acscatal.8b03401

(16) Correia, M. A., and Ortiz de Montello, P.R. Inhibition of cytochrome P450 enzymes, In P. R. Ortiz de Montello (ed.), Cytochrome P450: Structure, Mechanism, and Biochemistry, 3rd ed. Kluwer Academic/Plenym Publishers, New York. 2005; Pp 247-332.

(17) N’Guessan, R., A. Asidi, P. Boko, A. Odjo, M. Akogbeto, O. Pigeon, and Rowland, M. (2010). An experimental evaluation of PermaNet 3.0, a deltamethrin-piperonyl butoxide combination net, against pyrethroid-resistant Anopheles gambiae and Culex quinquefasciatus mosquitoes in southern Benin. Trans. R. Soc. Trop. Med. Hyg. 2010; 104: 758-765.

(18)Tungu, P., S. Magesa, C. Maxwell, R. Malima, D. Masue, W. Sudi, J. Myamba, O. Pigeon, and Rowland, M. Evaluation of PermaNet 3.0 a deltamethrin-PBO combination net against Anopheles gambiae and pyrethroid resistant Culex quinquefasciatus mosquitoes: an experimental hut trial in Tanzania. Malar. J., 2010; 9: 21.

(19) Darriet, F. and Chandre. F. Combining piperonyl butoxide and dinotefuran restores the efficacy of deltamethrin mosquito nets against resistant Anopheles gambiae (Diptera: Culicidae). J. Med. Entomol. 2011; 48: 952-955.

(20) Anosike, C.A., Babandi, A. and Ezeanyika, L.U.S. Potentiation Effects of Ficus sycomorus Active Fraction Against Permethrin-Resistant Field-Population of Anopheles coluzzii (Diptera: Culicidae). Neotrop. Entomol., 2021; 50: 484-496.

(21) Tanaka, T, Fujitani, T, Takahashi, O, Oishi, S. Developmental toxicity evaluation of piperonyl butoxide in CD-1 mice. Toxicol Lett. 1994; 71:123–129.

(22) Takahashi, O, Oishi, S, Fujitani, T, Tanaka, T, Yoneyama, M. Chronic toxicity studies of piperonyl butoxide in CD-1 mice: Induction of hepatocellular carcinoma 1. Toxicology, 1997; 124: 95–103.

(23) Djouaka, R. F., Bakare, A. A., Coulibaly, O. N., Akogbe T.O.M.C., Ranson, H., Hemingway, J. and Strode, C. Expression of the cytochrome P450s, CYP6P3 and CYP6M2 are significantly elevated in multiple pyrethroid resistant populations of Anopheles gambiae s.s. from Southern Benin and Nigeria. BMC Genomics, 2008; 9: 538.

(24) Muller, P, Warr, E, Stevenson, B.J, Pignatelli, P.M, Morgan, J.C, Steven, A. et al. Field-caught permethrin-resistant Anopheles gambiae over-express CYP6P3, a P450 that metabolises pyrethroids. PLoS Genet. 2008; 4: e1000286.

(25) Edi, C.V., Djogbenou, L., Jenkins, M.A., Regna, K., et al. CYP6 P450 enzymes and ACE-1 duplication produce extreme and multiple insecticide resistance in the malaria mosquito Anopheles gambiae. PLoS Genet. 2014; 10: 1-12.

(26) Ranson, H. and Lissenden, N. Insecticide resistance in African Anopheles mosquitoes: a worsening situation that needs urgent action to maintain malaria control. Trends in Parasitol., 2016; 32:187–96.

(27) Miresmailli, S. and Isman, M.B. Botanical insecticides inspired by plant-herbivore chemical interactions. Trends in Plant Sci., 2014; 19:29-35.

Biondi, A., Mommaerts, V., Smagghe, G., Viñuela, E., Zappalà, L., Desneux, N. The non-target impact of spinosyns on beneficial arthropods. Pest Mgt. Sci., 2012; 68:1523-1536.

(29) Rhome, A.A. Phytochemicals from Ficus sycomorus L. leaves act as insecticides and acaricides. African J Agric Res; 2013; 8(27):3571–9. doi: 10.5897/AJAR2013.7243.

(30) Govindarajan, M. Larvicidal and repellent properties of some essential oils against Culex tritaeniorhynchus Giles and Anopheles subpictus Grassi (Diptera: Culicidae). Asian Pac J. Trop Med., 2010; 4:106–111.

(31) Pavela, R. Acute toxicity and synergistic and antagonistic effects of the aromatic compounds of some essential oils against Culex quinquefasciatus Say larvae. Parasitol. Res., 2015; 114: 3835–3853.

(32) Pavela, R and Benelli, G. Essential oils as eco-friendly biopesticide? Challenges and constraints. Trend Plant Sci., 2016; 21:1000-1007.

(33) Sedjati, S., Ambariyanto, A., Trianto A., Supriyantini, E., Ridlo, A., Yudiati, E., Firmansyah, T. Anti-Vibrio from Ethyl Acetate Extract of Sponge-Associated Fungus Trichoderma longibrachiatum. Jordan Journal of Pharmaceutical Sciences, 2021; 4:435-443

(34) Ali, H., Alkowni, N., Jaradat, N., Masri, M. Evaluation of phytochemical and pharmacological activities of Taraxacum syriacum and Alchemilla arvensis. Jordan Journal of Pharmaceutical Sciences, 2021; 14(4):457-471.

(35) Perumalsamy, H., Chang, K.S., Park, C. and Ahn, Y. J. Larvicidal activity of compounds isolated from Asarum heterotropoides root constituents against insecticide-susceptible and -resistant Culex pipiens pallens, and Aedes aegypti and Ochlerotatus togoi. J. Agric. Food Chem. 2010; 58:10001-10006.

(36) Wang, Z., Kim, J. R., Wang, M., Shu, S. and Ahn, Y. J. Larvicidal activity of Cnidium monnieri fruit coumarins and structurally related compounds against insecticide susceptible and -resistant Culex pipiens pallens and Aedes aegypti. Pest Mgt Sci., 2012; 68:1041-1047.

(37) Pethuan, S., Duangkaew, P., Sarapusit, S., Srisook, E. and Rongnopart, P. Inhibition against Mosquito cytochrome P450 enzymes by Rhinacanthin-A, -B, and -C elicits synergism on cypermethrin cytotoxicity in Spodoptera frugiperda Cells. J. Med. Entomol., 2012; 49(5): 992-1000.

(38) Abdel-Tawab H.M., Samia, M.M.M. and Natarajan, C. Safety of Natural Insecticides: Toxic Effects on Experimental Animals. Biomed Res. Int. 2018:1-17. https://doi.org/10.1155/2018/4308054.

(39) Antonious, G.F. Residues and half lives of pyrethrins on field- grown pepper and tomato. J. Environ. Sci. Health B., 2004; 39:491–503.

(40) Jones, R. T., Bakker, S. E., Stone, D., Shuttleworth, S. N., Boundy, S., Mccart, C., Daborn, P. J. and Van Den Elsen, J. M. Homology modeling of Drosophila cytochrome P450 enzymes associated with insecticide resistance. Pest Mgt. Sci., 2010; 66:1106-1115.

(41) de Graaf, C. Cytochrome P450-Drug interactions computational binding mode and affinity predictions in CYP2D6. PhD thesis, Department of Chemistry and Pharmacochemistry, Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands.2006.

(42) Magrane, M. and Consortium, U. UniProt Knowledgebase: a hub of integrated protein data. Database (Oxford), 2011: P. 9.

(43) Kuhnel, K., Ke, N., Cryle, M.J., Sligar, S.G., Schuler, M.A., Schlichting, I. Crystal Structures of Substrate-Free and Retinoic Acid-Bound Cyanobacterial Cytochrome P450 Cyp120A1. Biochemistry, 2008; 47: 6552.

(44) Webb, B. and Sali, A. Comparative Protein Structure Modeling Using Modeller. Current Protocols in Bioinformatics 54, John Wiley & Sons, Inc., 5.6.1-5.6.37, 2016, P 54.

(45) Friesner, R. A.; Banks, J. L., Murphy, R. B., Halgren, T. A., Klicic, J. J., Mainz, D. T., Repasky, M. P., Knoll, E. H., Shelley, M.., Perry, J. K., Shaw, D. E., Francis, P., Shenkin, P. S. Glide: A new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J. Med. Chem., 2004; 47:1739-1749.

(46) Caporuscio, F., Rastelli, G.; Imbriano, C., del Rio, A. Structure-based design of potent aromatase inhibitors by high-throughput docking. J. Med. Chem. 2011; 54: 4006–4017.

(47) Abdul-Hay, S.O., Lane, A.L., Caulfield, T.R., Claussin, C., Bertrand, J., Masson, A., Choudhry, S., Fauq, A.H., Maharvi, G.M., Leissring, M.A. Optimization of peptide hydroxamate inhibitors of insulin-degrading enzyme reveals marked substrate-selectivity. J. Med. Chem., 2013; 56:2246–2255.

(48) Harder, E., Damm, W., Maple, J., Wu, C., Mark, R., Jin, Y. X., Wang, L., et al. (2016). OPLS3: A Force Field Providing Broad Coverage of Drug-like Small Molecules and Proteins. J. Chem. Theory Comput., 2016; 12(1):281-296.

(49) Onawole, A. T., Sulaiman, K. O., Adegoke, R. O., and Kolapo, T. U. Identification of potential inhibitors against the Zika virus using consensus scoring. J. Mol. Graphics Model., 2017; 73:54–61. https://doi.org/10.1016/j. jmgm.2017.01.018.

(50) Kuntz, I. D., Chen, K., Sharp, K. A., Kollman, P. A. The maximal affinity of ligands. Proc. Nat. Acad. Sci., U. S. A. 1999; 96: 9997-10002.

(51) Shultz, M.D. Setting expectations in molecular optimizations: strengths and limitations of commonly used composite parameters. Bioorg. Med. Chem. Lett. 2013; 23: 5980−5991.

(52) Hopkins, A. L., Groom, C.R., Alex, A. Ligand efficiency: a useful metric for lead selection. Drug Discov. Today, 2004; 9:430-1.

(53) Leeson, P.D. and Springthorpe B. The influence of drug-like concepts on decision-making in medicinal chemistry. Nat. Rev. Drug Discov. 2007; 6: 881–890.

(54) Reynolds, C. H., Bembenek, S. D., Tounge, B. A. The role of molecular size in ligand efficiency. Bioorg. Med. Chem. Lett. 2007; 17: 4258-4261.

(55) Nissink, J. W. M. Simple size-independent measure of ligand efficiency. J. Chem. Inf. Model. 2009; 49:1617-22.

(56) Lee, J., Cheng, X., Swails, J,M., Yeom, M.S., Eastman, P.K., Lemkul, J.A., et al. CHARMM-GUI Input Generator for NAMD, GROMACS, AMBER, OpenMM, and CHARMM/OpenMM Simulations Using the CHARMM36 Additive Force Field. J. Chem. Theory and Comput., 2016; 12(1):405-413.

(57) Kim, S., Lee, J., Jo, S., Brooks, C.L., Lee, H.S. and Im, W. CHARMM-GUI ligand reader and modeler for CHARMM force field generation of small molecules. J. Comput. Chem., 2017; 38(21):1879-1886.

(58)Phillips, J. C., Braun, R., Wang, W., Gumbart, J., Tajkorshid, E., Villa, E., et al. Scalable molecular dynamics with NAMD. J. Comput. Chem. 2005; 26(16):1781-1802.

(59) Pettersen, E. F., Thomas, D.G., Conrad, C.H., Gregory, S.C., Daniel, M.G., Elaine, C.M., et al. UCSF Chimera-A visualization system for exploratory research and analysis. J. Comput. Chem. 2004; 25(13):1605-1612.

(60) Uba, A. I., Weako J., Keskin, Ö., Gürsoy, A., Yelekçi, K. Examining the stability of binding modes of the co-crystallized inhibitors of human HDAC8 by molecular dynamics simulation. J. Biomol. Struct. Dyn. 2020; 38(6):1751-1760. doi: 10.1080/07391102.2019.1615989.

(61) Uba, A. I., Yelekçi, K. Crystallographic structure versus homology model: a case study of molecular dynamics simulation of human and zebrafish histone deacetylase 10. J. Biomol. Struct. Dyn. 2020; 38(15): 4397-4406. doi: 10.1080/07391102.2019.1691658.

(62) Chandor-Proust, A., Bibby, J., Regent-Kloeckner, M., Roux, J., Guittard-Crilat, E., Poupardin, R., Riaz, M.A., Paine, M., Dauphin-Villemant, C., Reynaud, S., David, J.P. The central role of mosquito cytochrome P450 CYP6Zs in insecticide detoxification revealed by functional expression and structural modelling. Biochem. J. 2013; 455:75e85.

(63) McLaughlin, L.A., Niazi, U., Bibby, J., David, J.P., Vontas, J., Hemingway, J., Ranson, H., Sutcliffe, M.J., Paine, M.J. Characterization of inhibitors and substrates of Anopheles gambiae CYP6Z2. Insect Mol. Biol. 2008; 17:125e135.

(64) Szklarz, G. D., He, Y. A. and Halpert, J. R. Site-directed mutagenesis as a tool for molecular modeling of cytochrome P450 2B1. Biochemistry, 1995; 34:14312-14322.

(65) Lewis, D.F., Lake, B.G. and Dickins, M. Quantitative structure-activity relationships (QSARs) in inhibitors of various cytochromes P450: The importance of compound lipophilicity. J. Enzym. Inhib. Med. Chem., 2007; 22:1–6.

(66) Sridhar, J., Ellis, J., Dupart, P., Liu, J., Stevens, C.L., Foroozesh, M. Development of flavone propargyl ethers as potent and selective inhibitors of cytochrome P450 enzymes 1A1 and 1A2. Drug Metab. Lett., 2012; 6:275–284.

(67) Farid, R., Day, T., Friesner, R. A., Pearlstein, R. A. New Insights about HERG Blockade Obtained from Protein Modeling, Potential Energy Mapping, and Docking Studies. Bioorg. Med. Chem., 2006; 14:3160-3173.

(68) Sherman, W., Day, T., Jacobson, M. P.; Friesner, R. A. and Farid, R. Novel Procedure for Modeling Ligand/Receptor Induced Fit Effects. J. Med. Chem., 2006; 49:534-553.

(69) Krovat, E. M., Steindl, T., Langer, T. Recent Advances in Docking and Scoring. Curr. Comput.-Aided Drug Des. 2005; 1: 93-102.

(70) Meunier, B., de Visser, S. P., Shaik, S. Mechanism of oxidation reactions catalyzed by cytochrome P450 enzymes. Chem Rev, 2004; 104:3947-3980.

(71) Hritz, J., deRuiter, A., and Oostenbrink, C. Impact of plasticity and flexibility on docking results for cytochrome P4502D6: a combined approach of molecular dynamics and ligand docking. J. Med. Chem. 2008; 51:7469–7477.doi: 10.1021/jm801005m.

(72) Vasanthanathan, P., Hritz, J., Taboureau, O., Olsen, L., Jørgensen, F.S., Vermeulen, N.P., et al. Virtual screening and prediction of site of metabolism for cytochrome P4501A2 ligands. J. Chem. Inf. Model. 2009; 49:43–52. doi: 10.1021/ci800371f.

(73) White, R.E.; Coon, M.J. Oxygen Activation by Cytochrome P450. Ann. Rev. Biochem., 1980; 49:315–356.

(74) Ortiz de Montellano, P.R.; Correia, M.A. Inhibition of Cytochrome P450 Enzymes. In Cytochrome P450: Structure, Mechanism, and Biochemistry; 2nd Ed.; Ortiz de Montellano, P.R., Ed.; Plenum Press: New York, 1995; Pp305–364.

(75) Schenkman, J.B., Sligar, S.G., Cinti, D.L. Substrate Interactions with Cytochrome P-450. Pharmacol. Ther., 1981; 12:43–71.

(76) Guengerich, F.P. Oxidation–Reduction Properties of Rat Liver Cytochromes P450 and NADPH-Cytochrome P-450 Reductase Related to Catalysis in Reconstituted Systems. Biochemistry, 1983; 22: 2811–2820.

(77) Hollenberg, P.E. Characteristics and common properties of inhibitors, inducers, and activators of cyp enzymes. Drug Metab Rev, 2002; 34(1&2):17–35.

(78) Pelkonen, O., Turpeinen, M., Hakkola, J., Honkakoski, P., Hukkanen, J., and Raunio, H. Inhibition and induction of human cytochrome P450 enzymes: current status. Arch. Toxicol., 2008; 82:667–715.doi:10.1007/s00204-008- 0332-8.

(79) Orr, S.T., Ripp, S.L., Ballard, T.E., Henderson, J.L., Scott, D.O., Obach, R.S., et al. Mechanism-based inactivation (MBI) of cytochrome P450 enzymes: structure-activity relationships and discovery strategies to mitigate drug-drug interaction risks. J. Med. Chem. 2012; 55:4896–4933.doi:10.1021/jm300065h.

(80) Nickerson, D.P., Hardford-Cross, C.F., Fulcher, S.R., Wong, L.L. The catalytic activity of cytochrome P450 (cam) toward styrene oxidation is increased by site specific mutagenesis. FEBS Lett., 1997; 405:153-156.

(81) Kitchen, D.B., Decornez, H., Furr, J.R., Bajorath, J. Docking and scoring in virtual screening for drug discovery: Methods and applications. Nature Rev Drug Discov., 2004; 3: 935–949.

(82) Reynolds, C. H., Tounge, B.A., Bembenek, S.D. Ligand binding efficiency: trends, physical basis, and implications. J. Med. Chem. 2008; 51: 2432-8.

(83) Loving, K., Alberts, I., Sherman, W. Computational approaches for fragment-based and de novo design. Curr. Top. Med. Chem. 2012; 10: 14-32.

(84) Ferenczy, G. G., Keseru, G. M. Enthalpic efficiency of ligand binding. J. Chem. Inf. Mod. 2010; 50:1536-1541.

(85) Reynolds, C. H., Holloway, M. K. Thermodynamics of Ligand Binding and Efficiency. ACS Med. Chem. Lett. 2011; 2: 433-7.

Downloads

Published

2022-06-01

How to Cite

Babandi, A. ., Anosike, C. A. ., Ezeanyika, L. U. ., Yelekçi, K. ., & Uba, A. I. . (2022). Molecular modeling studies of some phytoligands from Ficus sycomorus fraction as potential inhibitors of cytochrome CYP6P3 enzyme of Anopheles coluzzii. Jordan Journal of Pharmaceutical Sciences, 15(2), 258–275. https://doi.org/10.35516/jjps.v15i2.324

Issue

Section

Articles