Volume-7 , Issue-2 , Apr 2020, ISSN 2347-7520 (Online) Go Back
-
Open Access Article
Rediscovering the Diversity of Guatemalan Bumble Bees (Hymenoptera: Apoidea: Bombus Latreille, 1802)
Dardón MJ, Yurrita CL, Landaverde-González P, Vásquez M, Ayala R.
Research Paper | Journal-Paper (IJSRBS)
Vol.7 , Issue.2 , pp.1-14, Apr-2020
Abstract
Bumble bees (Bombus Latreille) play an essential role as pollinators of both wild and cultivated plants. Traditionally, these bees have been identified based on their color patterns, which in some cases vary among individuals of the same species, or different species, can share similar patterns and colorations allowing for inaccurate identifications. Despite these problems, Guatemalan reports on Bombus species have relied basically on color patterns. Here, we combine mitochondrial DNA barcode sequences and morphometric measurements, in an attempt to provide further information on the species limits of nine Bombus species occurring in Guatemala and also two species occurring in Mexico. When molecular analyses failed to distinguish between “morphological species” we applied traditional morphometrics using body measurements. Analyses of mitochondrial DNA sequences allowed for the discrimination of seven of the eleven “morphological species” evaluated. The remaining four species clustered into two pairs of species that have problematic taxonomy (B. sonorous–B. pensylvanicus, and B. ephippiatus–B. wilmattae). Morphometric analysis performed on the B. ephippiatus–B. wilmattae pair indicated non-discrimination between these forms, supporting the hypothesis that these are a single species. Therefore, based on our results we confirm the presence of the following species in Guatemala: B. ephippiatus (including B. wilmattae as synonym), Bombus (Cullumanobombus) macgregori Labougle & Ayala, 1985, Bombus (Thoracobombus) medius Cresson, 1863, Bombus (Thoracobombus) mexicanus Cresson, 1878, Bombus (Thoracobombus) pullatus Franklin, 1913, Bombus (Psithyrus) variabilis (Cresson, 1872), Bombus (Thoracobombus) weisi Friese, 1903, and Bombus (Thoracobombus) xelajuensis Asperen de Boer, 1992.Key-Words / Index Term
DNA barcoding, Bombini, Central America, Morphometry, TaxonomyReferences
[1] M.G. Park, R.A. Raguso, J.E. Losey, B.N. Danforth, “Per-visit pollinator performance and regional importance of wild Bombus and Andrena (Melandrena) compared to the managed honey bee in New York apple orchards”, Apidologie, Vol. 47, No. 2, pp. 145–160, 2015.
[2] C. Michener, “The Bees of the World”, John Hopkins University Press, Baltimore, USA, pp. 953, 2007
[3] P. Williams, “An annotated checklist of bumble bees with an analysis of patterns of description (Hymenoptera: Apidae, Bombini)”, Bulletin of the Natural History Museum (Entomology), Vol. 67, pp. 79–152, 1998.
[4] P. Williams, “A preliminary cladistic investigation of relationships among the bumble bees (Hymenoptera, Apidae)”, Systematic Entomology, Vol. 10, pp. 239–255, 1985.
[5] P. Rasmont, J. Devalez, A. Pauly, D. Michez, V.G. Radchenko, “Addition to the checklist of IUCN European wild bees (Hymenoptera: Apoidea)”, Annales de la Société entomologique de France, Vol. 53, No.1, pp. 17-32, doi:10.1080/00379271.2017.1307696, 2017.
[6] H.E: Milliron, “A Monograph of the Western Hemisphere Bumble bees (Hymenoptera: Bomninae), II. The Genus Megabombus Subgenus Megabombus”, Memoirs of the Entomological Society of Canada, Vol. 89, pp.81–237, 1972.
[7] J.M. Labougle, M. Ito, T. Okazawa, “The species of the genus Bombus (Hymenoptera: Apidae) of Chiapas, Mexico and Guatemala; with a morphometric and altitudinal analysis”, Folia Entomológica Mexicana, Vol. 64, pp. 55–72, 1985.
[8] J.M. Labougle, “Bombus of México and Central America (Hymenoptera, Apidae)”, University of Kansas University Science Bulletin, Vol. 54, No. 3, pp. 35–73, 1990.
[9] A.H. Abrahamovich, B. Díaz, “Bumble bees of the Neotropical region (Hymenoptera; Apidae)”, Biota colombiana, Vol. 3, pp. 199–214, 2002.
[10] E. Enríquez, C.L. Yurrita, R. Ayala, A. Marroquin, T. Griswold, “Diversidad de abejas silvestres (Hymenoptera: Apoidea) de Guatemala”. In: Cano E & Schuster JC (eds), Biodiversidad de Guatemala, Vol. 2, pp. 281–299. Universidad del Valle de Guatemala, Guatemala, 2012.
[11] M.J. Dardón, C.L. Yurrita, M. Vásquez, “Catalogue of the bees of the genus Bombus Latreille, 1802 (Hymenoptera: Apoidea: Apidae) in Guatemala”, Revista Científica, Vol. 26, No. 1, pp. 9–19, 2016.
[12] P. Williams, “The distribution of bumble bee colour patterns worldwide: possible significance for thermoregulation, crypsis, and warning mimicry”, Biological Journal of the Linnean Society, Vol. 92, pp. 97–118, 2007.
[13] H. Franklin, “The Bombidae of the new world (Continued)”, Transaction of the American Entomological Society, Vol. 39, pp. 73–200, 1913.
[14] J.R.J. Van Asperen de Boer, “Bombus krusemani - a new bumble bee species from Guatemala (Hymenoptera: Apidae)”, Entomologische Berichten, Vol. 50, pp. 1–3, 1990.
[15] J.S. Moure, D. Urban, G.A.R. Melo, “Catalogue of Bees (Hymenoptera, Apoidea) in the Neotropical Region”, Curitiba Sociedade Brasileira de Entomologia, xiv, 1058p. 2007.
[16] M. Duennes, J. Lozier, H. Hines, S. Cameron, “Geographical patterns of genetic divergence in the widespread Mesoamerican bumble bee Bombus ephippiatus (Hymenoptera: Apidae)”, Molecular Phylogenetics and Evolution, Vol. 64, No. 1, pp. 219–231, 2012.
[17] M. Duennes, C. Petranek, E. Pineda, J. Merida-Rivas, O. Martinez-Lopez, P. Sagot, R. Vandame, S.A. Cameron, “Population genetics and geometric morphometrics of the Bombus ephippiatus species complex with implications for its use as a commercial pollinator”, Conservation Genetics, Vol. 18, No. 3, pp. 553–572, 2017.
[18] B.C. Schlick-Steiner, F.M. Steiner, B. Seifert, C. Stauffer, E. Christian, R.H. Crozier, “Integrative taxonomy: a multisource approach to exploring biodiversity”, Annual Review of Entomology, Vol. 55, pp. 421–438, 2010.
[19] T. Lecocq, S. Dellicour, D. Michez, M. Dehon, A. Dewulf, T. De Meulemeester, N. Brasero, I. Valterová, J. Rasplus, P. Rasmont, “Methods for species delimitation in bumble bees (Hymenoptera:Apidae:Bombus):towards an integrative approach”, Zoologica Scripta, Vol. 44, pp. 281-97, 2015.
[20] N. Brasero, R. Vandame, P. Sagot, B. Martinet, I. Valterova, P. Rasmont, “Thoracobombus from Mexico: a description of the male species-specific cephalic labial gland secretions”, Apidologie, Vol. 50, pp.183-194, 2019.
[21] R. Ricklefs, J. Travis, “A morphological approach to the study of avian community organization”, The Auk, Vol. 97, pp. 321–338, 1980.
[22] T. Murray, Ú. Fitzpatrick, M.J.F. Brown, R. Paxton, “Cryptic species diversity in a widespread bumble bee complex revealed using mitochondrial DNA RFLPs”, Conservation Genetics, Vol. 9, No. 3, pp. 653–666, 2008.
[23] J. Gibbs, “Integrative taxonomy identifies new (and old) species in the Lasioglossum (Dialictus) tegulare (Robertson) species group (Hymenoptera, Halictidae)”, Zootaxa, Vol. 2032, pp. 1–38, 2009.
[24] P.H. Williams, M.J.F. Brown, J.C. Carolan, J. An, D. Goulson, A.M. Aytekin, L.R. Best, A.M. Byvaltsev, B. Cederberg, R. Dawson, J. Huang, M. Ito, A. Monfare, R.H. Raina, P. Schmid-Hempel, C.S. Sheffield, P. Šima, Z. Xie, “Unveiling cryptic species of the bumble bee subgenus Bombus s. str. worldwide with COI barcodes (Hymenoptera: Apidae)”, Systematics and Biodiversity, Vol. 10, pp. 21–56, 2012.
[25] P.H. Williams, A.M. Byvaltsev, B. Cederberg, M.V. Berezin, F. Ødegaard, C. Rasmussen, L.L. Richardson, J.X. Huang, C.S. Sheffield, S.T. Williams, “Genes suggest ancestral colour polymorphisms are shared across morphologically cryptic species in arctic bumble bees”, PLoS ONE, Vol. 10, pp. 1–26, 2015.
[26] P. Landaverde-González, H. Moo-Valle, T.E. Murray, R. Paxton, J.J. Quezada-Euán, M. Husemman, “Sympatric lineage divergence in cryptic Neotropical sweat bees (Hymenoptera: Halictidae: Lasioglossum)”, Organism Diversity and Evolution, Vol. 17, No. 1, pp. 251–265, 2017.
[27] R.C. Plowright, W.P. Stephen, “A numerical taxonomic analysis of the evolutionary relationships of Bombus and Psithyrus (Apidae: Hymenoptera)”, Canadian Entomologist, Vol. 105, pp. 733-743, 1973.
[28] A.M. Aytekin, N. Cagatay, “Systematical Studies on Megabombus (Apidae: Hymenoptera) Species in Central Anatolia”, Turkish Journal of Zoology, Vol. 27, pp. 195–204, 2003.
[29] A.M. Aytekin, M. Terzo, P. Rasmont, N. Cagatay, “Landmark based geometric morphometric analysis of wing shape in Sibiricobombus Vogt (Hymenoptera: Apidae: Bombus Latreille)”, Annales de la Societe entomologique de France, Vol. 43, No. 1, pp. 95-102, 2007.
[30] V.S. Ferreira, C.M.L. Aguiar, M.A. Costa, J.G. Silva, “Morphometric Analysis of Populations of Centris aenea Lepeletier (Hymenoptera: Apidae) from Northeastern Brazil”, Neotropical Entomology, Vol. 40, No. 1, pp. 97–102, 2011.
[31] W.J. May-Itzá, J.J.G. Quezada-Eúan, L. Iuit, C.M. Echazarreta, “Do morphometrics and allozymes reliable distinguish Africanized and European Apis mellifera drones in subtropical Mexico?”, Journal of Apicultural Research, Vol. 40, pp. 17–23, 2001.
[32] W.J. May-Itzá, J.J.G. Quezada-Eúan , L.A. Medina. E. Enríquez, P. De la Rúa, “Morphometric and genetic differentiation in isolated populations of the endangered Mesoamerican stingless bee Melipona yucatanica (Hymenoptera, Apoidea) suggest the existence of a two species complex”, Conservation Genetics, Vol. 11, No. 5, pp. 2079–2084, 2010.
[33] J.J.G. Quezada-Euán, R.J. Paxton, K.A. Palmer, W.J.M. May-Itzá, W.T. Tay, B.P. Oldroyd, “Morphological and molecular characters reveal differentiation in a Neotropical social bee, Melipona beecheii (Apidae: Meliponini)”, Apidologie, Vol. 38, pp. 247–258, 2007.
[34] J.J.G. Quezada-Euán, W.D.J. May-Itzá, M. Rincón, P. De la Rúa, R.J. Paxton,” Genetic and phenotypic differentiation in endemic Scaptotrigona hellwegeri (Apidae: Meliponini): implications for the conservation of stingless bee populations in contrasting environments”, Insect Conservation and Diversity, Vol. 5, pp. 433–443, 2012.
[35] L.A. Nunes, M.F.F. Costa-Pinto, P.L.S, Carneiro, D.G. Pereira, A.M. Waldschmidt, “Divergencia genética em Melipona scutellaris Latreille (Hymenoptera: Apidae) com base em caracteres morfológicos”, Bioscience Journal, Vol. 23, pp.1–9, 2007.
[36] S. Ratnasingham, P.D. Hebert, “BOLD: The Barcode of Life Data System (http://www.barcodinglife.org)”, Molecular Ecology Notes, Vol. 7, No. 3, pp. 355–364, 2007.
[37] P.D.N. Hebert, A. Cywinska, S.L. Ball, J.R. DeWaard, “Biological identifications through DNA barcodes”, Proceedings the Royal Society of London B, Biological Sciences, Vol. 270, pp. 313–21, 2003.
[38] O. Folmer, M. Black, W. Hoeh, R. Lutz, R. Vrijenhoek, “DNA primers for amplification of mitochondrial cytochrome c oxidase subunit I from diverse metazoan invertebrates”, Molecular Marine Biology and Biotechnology, Vol. 3, pp. 294–299, 1994.
[39] P.D.N. Hebert, E.H. Penton, J.M. Burns, D.H. Janzen, W. Hallwachs, “Ten species in one: DNA barcoding reveals cryptic species in the neotropical skipper butterfly Astraptes fulgerator”, Proceedings of the National Academy of Sciences of the United States of America, Vol. 101, pp. 14812–14817, 2004.
[40] T.A. Hall, “BioEdit: a user-friendly biological sequence alignment editor and analysis program for Windows 95/98/NT”, Nucleic Acids Symposium Series, Vol. 41, pp. 95–98, 1999.
[41] J. Rozas, C. Sánchez-DelBarrio, X. Messeguer, R. Rozas, “DnaSP, DNA polymorphism analyses by the coalescent and other methods”, Bioinformatics, Vol. 19, pp. 2496–2497, 2003.
[42] S. Tavaré, “Some probabilistic and statistical problems in the analysis of DNA sequences”, Lectures on mathematics in the life sciences, Vol. 17, No. 2, pp. 57–86, 1986.
[43] D. Darriba, G. Taboada, R. Doallo, D. Posada, “jModelTest 2: more models, new heuristics and parallel computing”, Nature Methods, Vol. 9, pp. 772, 2012.
[44] K. Tamura, G. Stecher, D. Peterson, A. Filipski, S. Kumar, “MEGA6: Molecular Evolutionary Genetics Analysis version 6.0”, Molecular Biology and Evolution, Vol. 30, pp. 2725–2729, 2013.
[45] P.D.N. Hebert, S. Ratnasingham, J.R. deWaard, “Barcoding animal life: cytochrome c oxidase subunit 1 divergences among closely related species”, Proceedings of the Royal Society London B, Vol. 270, pp. S96–S99, 2003.
[46] D.H. Janzen, M. Hajibabaei, J.M. Burns, W. Hallwachs, E. Remigio, P.D.N. Hebert, “Wedding biodiversity inventory of a large and complex Lepidoptera fauna with DNA barcoding”, Philosophical Transactions of the Royal Society B: Biological Sciences, Vol. 360, No. 1462, pp. 1835–1846, 2005.
[47] R. DeSalle, “Species discovery versus species identification in DNA barcoding efforts: response to Rubinoff”, Conservation Biology, Vol. 20, No. 5, pp.1545–1547, 2006.
[48] J. Gjershaug, A. Staverløkk, O. Kleven, F. Ødegaard, “Species status of Bombus monti-cola Smith (Hymenoptera:Apidae) supported by DNA barcoding”, Zootaxa, Vol. 3716, pp. 431–440, 2013.
[49] M.K. Fujita, A.D. Leaché, F.T. Burbrink, J.A. McGuire, C. Moritz, “Coalescent-based species delimitation in an integrative taxonomy”, Trends Ecology Evolution, Vol. 27, No. 9, pp. 480–488, 2012.
[50] S. Dellicour, J.F., Flot, “Delimiting species-poor data sets using single molecular markers: A study of Barcode Gaps, Haplowebs and GMYC”, Systematic Biology, Vol. 64, No. 6, pp. 900–908, 2015.
[51] N.M. Reid, B.C. Carstens, “Phylogenetic estimation error can decrease the accuracy of species delimitation: a Bayesian implementation of the general mixed Yule-coalescent model”, BMC Evolutionary Biology, Vol. 12, No. 196, pp.11, 2012.
[52] A. Drummond, M, Suchard, D. Xie, A. Rambaut, “Bayesian phylogenetics with BEAUti and the BEAST 1.7”, Molecular Biology and Evolution, Vol. 29, No. 8, pp. 1969–1973, 2012.
[53] P. Landaverde-González, L. Baltz, N. Escobedo, J. Mérida, R. Paxton, M. Husemann, “Recent low levels of differentiation in the native Bombus ephippiatus (Hymenoptera: Apidae) along two Neotropical mountain-ranges in Guatemala”, Biodiversity and conservation, Vol. 27, No. 13, pp. 3513–3531, 2018.
[54] N. Puillandre, A. Lambert, S. Brouillet, G. Achaz, “ABGD, Automatic Barcode Gap Discovery for primary species delimitation”, Molecular Ecology, Vol. 21, No. 8, pp. 1864–1877, 2011.
[55] M. Wiemers, K. Fiedler K, “Does the DNA barcoding gap exist? -a case study in blue butterflies (Lepidoptera: Lycaenidae)”, Frontiers in Zoology, Vol. 4, No. 1, pp. 8, 2007.
[56] T.L. Whitworth, R.D. Dawson, H. Magalon, E. Baudry, “DNA barcoding cannot reliably identify species of the blowfly genus Protocalliphora (Diptera: Calliphoridae)”, Proceedings of the Royal Society B, Vol. 274, No. 1619, pp. 1731-1739, 2007.
[57] C.P. Meyer, G. Paulay, “DNA Barcoding: Error rated based on comprehensive sampling”, Plos Biology, Vol. 3, No. 12, pp.:e422, 2005.
[58] L.F. Marcus, “Traditional morphometrics”. In Rohlf FJ & Bookstein FL (eds), Proceedings of the Michigan Morphometrics Workshop, pp. 77–122, University of Michigan Special Publication No. 2, Ann Arbor, 1990.
[59] F.J. Rohlf, L.F. Marcus, “A revolution in morphometrics”, Trends in Ecology and Evolution, Vol. 8, No. 4, pp. 129–132, 1993.
[60] M.D. Schwarzfeld, F.A.H. Sperling, “Species delimitation using morphology, morphometrics, and molecules: definition of the Ophion scutellaris Thomson species group, with descriptions of six new species (Hymenoptera, Ichneumonidae)”, ZooKeys, Vol. 462, pp. 59–114, 2014.
[61] C.A. Sanchez, “Bioestadística”, McGraw-Hill, México, pp.323, 2015.
[62] D. Wayne, “Bioestadística: base para el análisis de las ciencias de la salud”, Trad. León, F. Limusa, México, pp.755, 2007.
[63] E.T. Cresson, “List of the North American species of Bombus and Apathus”, Proceedings of the Entomological Society of Philadelphia, Vol. 2, pp. 83–116, 1863.
[64] E.T. Cresson, “Hymenoptera Texana”, Transaction of the American Entomological Society, Vol. 4, pp. 153–292, 1872.
[65] E.T. Cresson, “Description of Ichneumonidae, chiefly from the Pacific slope of the United States and British North America”, Proceedings of the Entomological Society of Philadelphia, Vol. 30, pp. 187, 1878
[66] H. Friese, „Neue bombus-arten aus der neotropischen region. (Hym)“, Zeitschrift für Systematische Hymenopterologie und Dipterologie, Vol. 3, pp. 253–255, 1903.
[67] J.R.J. Van Asperen de Boer, “Bombus xelajuensis -a new bumble bee species from Guatemala (Hymenoptera: Apidae)”, Entomologische Berichten, Vol. 52, pp. 162–164, 1992.
[68] T.D.A. Cockerell, “Descriptions and records of bees XLV”, Annals and Magazine of Natural History, Vol. 10, No. 8, pp. 21–31, 1912.
[69] P. Williams, S.A. Cameron, H.M. Hines, B. Cederberg, P. Rasmont, “A simplified subgeneric classification of the bumble bees (genus Bombus)”, Apidologie, Vol. 39, No. 1, pp. 46–74, 2008.
[70] P. Williams, “Bumble bees of the World”, Checklist. Natural History Museum. Available from http://nhm.ac.uk/research-curation/research/projects/B./ [accessed 14 Jun. 2018], 2018.
[71] J. Labougle, R. Ayala, “A new subgenus and species of Bombus (Hymenoptera, Apidae) from Guerrero, México”, Folia Entomológica Mexicana, Vol. 66, pp.47–55, 1985.
[72] R.W. Thorp, “Bumble bees and cuckoo bumble bees of California (Hymenoptera: Apidae), Bulletin of the California Insect Survey, Vol. 23, pp. 79, 1983.
[73] M. Riyaz, P. Mathew, G. Paulraj, S. Ignacimuthu, “Entomophily of Apple ecosystem in Kashmir valley, India: A review. International Journal of Scietific Research in Biological Sciences, Vol. 5, pp. 146-154, 2018.
Citation
Dardón MJ, Yurrita CL, Landaverde-González P, Vásquez M, Ayala R., "Rediscovering the Diversity of Guatemalan Bumble Bees (Hymenoptera: Apoidea: Bombus Latreille, 1802)," International Journal of Scientific Research in Biological Sciences, Vol.7, Issue.2, pp.1-14, 2020 -
Open Access Article
Reproductive Functions and Toxicology Following Scrotal Ultrasound Therapy in Rats
Abdul S. Ansari, Sonu, Rajeev Kumar Dhaked, Ayesha Badar, Barkha Khilwani, Nirmal Kumar Lohiya
Research Paper | Journal-Paper (IJSRBS)
Vol.7 , Issue.2 , pp.15-24, Apr-2020
Abstract
Therapeutic ultrasound involves direct application to the scrotum that might affect normal reproductive functions. We explore possibilities for development of a localized, non-invasive and reversible contraception method with ultrasound therapy in rat model. Animals were divided equally into five groups. Results observed with a gradual decrease in reproductive organs weight, testis volume, testosterone concentration and cauda epididymal sperm count, motility and viability following therapy, whereas sperm abnormalities were continuously enhanced throughout the study in groups II - V. Testis histopathology marked with duration and frequency dependent changes in groups II - V including conspicuous alterations in the seminiferous tubules, erupted germ cells, impaired spermatogenesis with pycnotic nuclei, vacuolization in the Sertoli cells, small Leydig cell nuclei, total necrosis and reduction in seminiferous tubules and wide interstitial space with fibroblast like cells. The fertility was totally lost at 180 days (groups II and III), 15 - 180 days (group IV) and 7 - 30 days of therapy (group V), respectively. A complete recovery was observed in all parameters in group V rats. Ultrasound application affects reproductive organs and fertility in rats. Hence, it may serve as a viable male contraceptive with further confirmation of dose and duration dependent efficacy, side effects and reversibility.Key-Words / Index Term
Fertility; Scrotum; Ultrasound; Toxicology; Wistar albino ratReferences
[1] K. Bowler, “The Effect of Repeated Applications of Heat on Spermatogenesis in the Rat: A Histological Study,” Journal of Reproduction and Fertility, Vol. 28, Issue.3, pp.325-333, 1972.
[2] M.S. Fahim, Z. Fahim , R. Der, D.G. Hall, J. Herman, “Heat in Male Contraception (Hot Water 60° C, Infrared, Microwave, and Ultrasound),” Contraception, Vol. 11, Issue.5, pp.549-562, 1975.
[3] M.S. Fahim, Z. Fahim, J. Harman, I. Thompson, J. Montie, D. G. Hall, “Ultrasound as a New Method of Male Contraception,” Fertility and Sterility, Vol. 28, Issue.8, pp.823-831, 1977.
[4] M.S. Fahim, Z. Fahim, F. Azzazi, “Effect of Ultrasound on Testicular Electrolytes (Sodium and Potassium),” Archives of Andrology, Vol. 1, Issue.2, pp.179-184, 1978.
[5] A. Dumontier, A. Burdick, B. Ewigman, M.S. Fahim, “Effects of Sonication on Mature Rat Testes,” Fertility and Sterility, Vol. 28, Issue.2, pp.195-204, 1977.
[6] R. Abadir, J. Harman, M.S. Fahim, “Enhancement of Ionizing Radiation Effect on the Testes of Rats by Microwave or Ultrasound-Induced Hyperthermia,” Journal of Medicine, Vol. 10, Issue.1-2, pp.1-12, 1979.
[7] J.K. Tsuruta, P.A. Dayton, C.M. Gallippi, G.O. Michael, M.A. Streicker, R.C. Gessner, T.S. Gregory, E.J. Silva, K.G. Hamil, G.J. Moser, D.C. Skal, “Therapeutic Ultrasound as a Potential Male Contraceptive: Power, Frequency and Temperature Required to Deplete Rat Testes of Meiotic Cells and Epididymides of Sperm Determined Using a Commercially Available System,” Reproductive Biology and Endocrinology, Vol. 10, pp.7, 2012.
[8] R. Leoci, G. Aiudi, S.A. De Sandro, F. Silvestre, F. Binetti, G.M. Lacalandra, “Ultrasound as a Mechanical Method for Male Dog Contraception,” Reproduction in Domestic Animals, Vol. 44, Suppl.2, pp.326-328, 2009.
[9] R. Leoci, G. Aiudi, F. Silvestre, E.A. Lissner, F. Marino, G.M. Lacalandra, “Therapeutic Ultrasound as a Potential Male Dog Contraceptive: Determination of the Most Effective Application Protocol,” Reproduction in Domestic Animals Vol. 50, Issue.5, pp.712-718, 2015.
[10] C. A. VandeVoort, T. L. Tollner, “The efficacy of ultrasound treatment as a Reversible Male Contraceptive in the Rhesus Monkey,” Reproductive Biology and Endocrinology Vol. 10, pp. 81, 2012.
[11] CPCSEA, “Guidelines on the Regulation of Scientific Experiments on Animals. Standard Operating Procedures (SOP) for Institutional Animals Ethics Committee (IAEC)”, Animal Welfare Division, Ministry of Environment, Forests and Climate Change. Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA), New Delhi,” India, 2010.
[12] WHO, “WHO Laboratory Manual for Examination and Processing of Human Semen,” 5th ed., World Health Organization (WHO), WHO Press, Department of Reproductive Health and Research, Geneva, 2010.
[13] P. Taylor, “Practical Teratology,” Ed. 1, Academic Press, London, 1986.
[14] M.S. Christian, “Test Methods for Assessing Female Reproductive and Developmental Toxicology,” Hayes AW (ed), Principles and Methods of Toxicology. Taylor and Francis Publishing, Philadelphia, pp.1301-1381, 2001.
[15] M.J. Derelanko, C.S. Auletta, “Handbook of Toxicology,” Ed. 3, CRC Press, Taylor & Francis Group, Boca Raton, USA, pp.355-359, 2014.
[16] V. Wasson, B. Singh, G. Wasson, “A Parallel Optimized Approach for Prostate Boundary Segmentation from Ultrasound Images,” International Journal of Scientific Research in Computer Science and Engineering, Vol.1, Issue.1, pp.14-19, 2013.
[17] J.K. Tsuruta, P. Dayton, R. Gessner, T.S. Gregory, M. Streicker, G. Moser, E.J. Silva, D. Sokal, “Can Therapeutic Ultrasound be Used as a Reversible Male Contraceptive?,” In the proceedings of the International Ultrasonics Symposium (IUS), San Deigo, CA, USA, Oct 11-14, 2010.
Citation
Abdul S. Ansari, Sonu, Rajeev Kumar Dhaked, Ayesha Badar, Barkha Khilwani, Nirmal Kumar Lohiya, "Reproductive Functions and Toxicology Following Scrotal Ultrasound Therapy in Rats," International Journal of Scientific Research in Biological Sciences, Vol.7, Issue.2, pp.15-24, 2020 -
Open Access Article
Physicochemical and analytical characterization of biosurfactant produced by Serratia rubidaea KAP
Anuradha S. Pendse, K. Aruna
Research Paper | Journal-Paper (IJSRBS)
Vol.7 , Issue.2 , pp.25-40, Apr-2020
Abstract
In an attempt to replace the chemical compounds that may potentially disrupt the environment, biological sources has been exploited extensively for characteristic recovery of their metabolic products. The present study was carried out with a similar attempt to characterize the biosurfactant produced by Serratia rubidaea KAP isolated from oil polluted soil sample. The biosurfactant sample was analyzed qualitatively by the Thin-layer chromatographic (TLC) technique which indicated the presence of proteins, carbohydrates (with rhamnose as major sugar) and lipids. This was further confirmed by High Performance-TLC technique. The concentration of proteins, carbohydrates and lipids were also quantified using suitable colorimetric and gravimetric techniques. They were estimated to be 11%, 37% and 51.09% respectively in the biosurfactant sample. It was further characterized to be ionic in nature, on the basis of agar double diffusion method. The analytical tests including Fourier Transformed Infra-Red spectrum and Gas Chromatography-Mass Spectroscopy confirmed the presence of rhamnolipids. The biosurfactant further showed a critical micelle concentration of 62%. Moreover, the stability studies indicated considerable biosurfactant activity at a wide range of temperature (from 4°C -100°C), alkaline pH and in an environment of salinity up to 12%. Thus, these results strongly suggest that the biosurfactant produced by S. rubidaea KAP can be useful in industries which operate at extreme conditions of pH, temperature and salinity e.g., pharmaceutical, cosmetics, food industries, and for bioremediation in the marine environment.Key-Words / Index Term
Biosurfactant, TLC, Rhamnolipid, Serratia rubidaea, StabilityReferences
[1] I.M. Banat, “Biosurfactants production and use in microbial enhanced oil recovery and pollution remediation: a review,” Bioresource Technology, Vol. 51, Issue.1, pp.1-12, 1995.
[2] J.D. Desai, I.M. Banat, “Microbial production of surfactants and their commercial potential,” Microbiology and Molecular Biology Reviews, Vol. 61, Issue.1, pp.47-64, 1997.
[3] N.G.K. Karanth, P.G. Deo, N.K. Veenanadig, “Microbial production of biosurfactants and their importance,” Current Science, Vol. 77, Issue.1, pp.116-126, 1999.
[4] E. Rosenberg, E.Z. Ron, “High- and low-molecular-mass microbial surfactants,” Applied Microbiology and Biotechnology, Vol. 52, Issue.2, pp.154-162, 1999.
[5] J.M. Campos, T.L. Stamford, L.A. Sarubbo, J.M. de Luna, R.D. Rufino, I.M. Banat, “Microbial biosurfactants as additives for food industries,” Biotechnology Progress, Vol. 29, Issue.5, pp.1097-1108, 2013.
[6] S. Sudhanshu, A. Sundaramanickam, T. Balasubramanian, “Biosurfactant producing microbes and their potential applications: A review,” Critical Reviews in Environmental Science and Technology, Vol. 45, Issue.14, pp.1522-1554, 2015.
[7] S. Vijayakumar, S. Varathrahjan, “Biosurfactants-types, sources and applications,” Research Journal of Microbiology, Vol. 10, Issue.5, pp.181-192, 2015.
[8] R.S. Makkar, S.S. Cameotra, “Effects of various nutritional supplements on biosurfactant production by a strain of Bacillus subtilis at 45°C,” Journal of Surfactants and Detergents, Vol. 5, Issue.1, pp.11-17, 2002.
[9] D.G. De Almeida, R.C. Soares Da Silva, J.M. Luna, R.D. Rufino, V.A. Santos, I.M. Banat, L.A. Sarubbo, “Biosurfactants: promising molecules for petroleum biotechnology advances,” Frontiers in Microbiology, Vol. 7, pp.1718, 2016.
[10] H.W.C. Araujo, R.F.S. Andrade, D. Montero-Rodríguez, D. Rubio-Ribeaux, C.A. Alves da Silva, G.M. Campos-Takaki, “Sustainable biosurfactant produced by Serratia marcescens UCP 1549 and its suitability for agricultural and marine bioremediation applications,” Microbial Cell Factories, Vol. 18, Issue.1, pp.2-13, 2019.
[11] A. Pendse, K. Aruna, “Optimisation of biosurfactant production by Serratia rubidaea KAP isolated from oil contaminated soil sample,” International Journal of Pharmaceutical Research and Bioscience, Vol. 8, Issue.4, pp.1-61, 2019.
[12] M. Dubois, K.A. Gilles, J.K. Hamilton, P.A. Rebers, F. Smith, “Colorimetric method for determination of sugars and related substances,” Analytical Chemistry, Vol. 28, pp.350-356, 1956.
[13] R.C.F.S. Silva, D.G. Almeida, R.D. Rufino, J.M. Luna, V.A. Santos, L.A. Sarubbo, “Applications of biosurfactants in the petroleum industry and the remediation of oil spills,” International Journal of Molecular Science, Vol. 15, Issue.7, pp.12523-12542, 2014.
[14] G. Soberon-Chavez, R.M. Maier, “Biosurfactants: a general overview,” Biosurfactants, Springer Berlin Heidelberg, pp.1-11, 2011.
[15] H. Jork, W. Funk, W. Fischer, H. Wimmer, “Thin layer chromatography: reagents and detection methods,” Physical and Chemical Detection Methods: Fundamentals, Reagents I. Vol. 1a, VCH Weinheira (Federal Republic of Germany), 1990.
[16] O.H. Lowry, N.J. Rosebrough, A.L. Farr, R.J. Randall, “Protein measurement with the Folin phenol reagent,” Journal of Biology and Chemistry, Vol. 193, Issue.1, pp.265-275, 1951.
[17] M. Folch, J.M. Lees, H.S. Stanly, “A simple method for the isolation and quantification of total lipids from animal tissues,” Journal of Biological Chemistry, Vol. 226, Issue.1, pp.497-501, 1956.
[18] E.J. Silva, N.M. Rocha, P. Silva, R.D. Rufino, J.M. Luna, R.O. Silva, L.A. Sarubbo, “Characterization of a biosurfactant produced by Pseudomonas cepacia CCT6659 in the presence of industrial wastes and its application in the biodegradation of hydrophobic compounds in soil,” Colloids and Surfaces; B, Biointerfaces, Vol. 117, pp.36-41, 2014.
[19] R.C.F.S. Silva, D.G. Almeida, H.M. Meira, E.J. Silva, C.B.B. Farias, R.D. Rufino, J.M. Luna, L. A. Sarubbo, “Production and characterization of a new biosurfactant from Pseudomonas cepacia grown in low-cost fermentative medium and its application in the oil industry,” Biocatalysis and Agricultural Biotechnology Vol. 12, pp.206-215, 2017.
[20] D.F. Gerson, J.E. Zajic, “Microbial biosurfactants,” Process Biochemistry, Vol. 14, pp.20-29, 1979.
[21] J.M. Luna, R.D. Rufino, C.D. Albuquerque, L.A. Sarubbo, G.M. Campos-Takaki, “Economic optimized medium for tensio-active agent production by Candida sphaerica UCP0995 and application in the removal of hydrophobic contaminant from sand,” International Journal of Molecular Science, Vol. 12, Issue.4, pp.2463-2478, 2011.
[22] Y. Zhang, R.M. Miller, “Enhanced octadecane dispersion and biodegradation by a Pseudomonas rhamnolipid surfactant (biosurfactant),” Applied and Environmental Microbiology, Vol. 58, Issue.10, pp.3276-3282, 1992.
[23] M. Abouseoud, A. Yatagheneb, A. Amranec, R. Maachib, “Effect of pH and salinity on the emulsifying capacity and naphthalene solubility of a biosurfactant produced by Pseudomonas fluorescens,” Journal of Hazardous Materials, Vol. 180, Issue.1-3, pp.131-136, 2010.
[24] T. Matsuyama, K. Kaneda, Y. Nakagawa, K. Isa, H. Hara-Hotta, I. Yano, “A novel extracellular cyclic lipopeptide which promotes flagellum-dependent and -independent spreading growth of Serratia marcescens,” Journal of Bacteriology, Vol. 174, Issue.6, pp.1769-1776, 1992.
[25] C.U. Anyanwu, S.K.C. Obi, B.N. Okolo, “Lipopetide biosurfactant production by Serratia marcescens SNK-1 strain isolated from petroleum contaminated soil,” Journal of Applied Science and Research, Vol. 7, Issue.1, pp.79-87, 2011.
[26] J. Ganley, G. Carr, T. Ioerger, J. Sacchettini, J. Clardy, E. Derbyshire, “Discovery of antimicrobial lipodepsipeptides produced by a Serratia sp. within mosquito microbiomes,” Chembiochem- A European Journal of Chemical Biology, Vol. 19, Issue.15, pp.1590-1594, 2018.
[27] T. Clements, T. Ndlovu, S. Khan, W. Khan, “Biosurfactants produced by Serratia species: classification, biosynthesis, production and application,” Applied Microbiology and Biotechnology, Vol. 103, Issue.2, pp.589-602, 2019.
[28] C.U. Anyanwu, S.K.C. Obi, B.N. Okolo, “Production of surface active glycolipid by Serratia marcescens NSK-1 isolated from petroleum contaminated soil,” Our Nature, Vol. 8, Issue.1, pp.1-11, 2010.
[29] D.H. Dusane, V.S. Pawar, Y.V. Nancharaiah, V.P. Venugopalan, A.R. Kumar, S.S. Zinjarde, “Anti-biofilm potential of a glycolipid surfactant produced by a tropical marine strain of Serratia marcescens,” Biofouling, Vol. 27, Issue.6, pp.645-654, 2011.
[30] N.S. Rosas-Galvan, F. Martınez-Morales, S. Marquina-Bahena, R. Tinoco-Valencia, L. Serrano-Carreon, B. Bertrand, R. Leon-Rodrıguez, R. Guzman-Aparicio, L. Alvarez-Berber, M.R. Trejo-Hernandez, “Improved production, purification, and characterization of biosurfactants produced by Serratia marcescens SM3 and its isogenic SMRG-5 strain,” Biotechnology and Applied Biochemistry, Vol. 65, Issue.5, pp.690-700, 2018.
[31] O. Munachimso, U.J. Josiah, C. Miracle, “Isolation, characterization and MEOR ability of the biosurfactant produced from Serratia marcescens UEO15,” Global Journal of Advanced Research, Vol. 2, Issue.6, pp.962-974, 2015.
[32] X. Hu, T. Cheng, J. Liu, “A novel Serratia sp. ZS6 isolate derived from petroleum sludge secretes biosurfactant and lipase in medium with olive oil as sole carbon source,” AMB Express, Vol. 8, Issue.1, pp.165, 2018.
[33] M. Abouseoud, R. Maachi, A. Amrane, S. Boudergua, A. Nabi, “Evaluation of different carbon and nitrogen sources in production of biosurfactant by Pseudomonas fluorescens,” Desalination, Vol. 223, Issue.1-3, pp.143-151, 2008.
[34] A.P. Kumar, A. Janardhan, B. Viswanath, K. Monika, J.Y. Jung, G. Narasimha, “Evaluation of orange peel for biosurfactant production by Bacillus licheniformis and their ability to degrade naphthalene and crude oil,” 3 Biotechnology, Vol. 6, Issue.1, pp.43, 2016.
[35] A. Hanano, M. Shaban, I. Almousally, “Biochemical, molecular, and transcriptional highlights of the biosynthesis of an effective biosurfactant produced by Bacillus safensis PHA3, a petroleum-dwelling bacteria,” Frontiers in Microbiology, Vol. 8, pp.77, 2017.
[36] R. Sharma, J. Singh, N. Verma, “Optimization of rhamnolipid production from Pseudomonas aeruginosa PBS towards application for microbial enhanced oil recovery,” 3 Biotechnology, Vol. 8, Issue.1, pp.20, 2017.
[37] G. Lan, Q. Fan, Y. Liu, C. Chen, G. Li, Y. Liu, X. Yu, “Rhamnolipid production from waste cooking oil using Pseudomonas SWP-4,” Biochemical Engineering Journal, Vol. 101, pp.44-54, 2015.
[38] Y. Al-Wahaibi, S. Joshi, S. Al-Bahry, A. Elshafie, A. Al-Bemani, B. Shibulal, “Biosurfactant production by Bacillus subtilis B30 and its application in enhancing oil recovery,” Colloids and Surfaces; B, Biointerfaces, Vol. 114, pp.324-333, 2014.
[39] S.J. Joshi, Y.M. Al-Wahaibi, S.N. Al-Bahry, A.E. Elshafie, A.S. Al-Bemani, A. Al-Bahri, M.S. Al-Mandhari, “Production, characterization, and application of Bacillus licheniformis W16 biosurfactant in enhancing oil recovery,” Frontiers in Microbiology, Vol. 7, pp.1853, 2016.
[40] A.E. Elshafie, S.J. Joshi, Y.M. Al-Wahaibi, A.S. Al-Bemani, S.N. Al-Bahry, D. Al-Maqbali, I.M. Banat, “Sophorolipids production by Candida bombicola ATCC 22214 and its potential application in microbial enhanced oil recovery,” Frontiers in Microbiology, Vol. 6, pp.1324, 2015.
[41] M. Geissler, C. Oellig, K. Moss, W. Schwack, M. Henkel, R. Hausmann, “High-performance thin-layer chromatography (HPTLC) for the simultaneous quantification of the cyclic lipopeptides surfactin, iturin A and fengycin in culture samples of Bacillus species,” Journal of Chromatography B: Analytical Technologies in the Biomedical and Life Sciences, Vol. 1044-1045 pp.214-224, 2017.
[42] R.M. Jain, K. Mody, A. Mishra, B. Jha, “Physicochemical characterization of biosurfactant and its potential to remove oil from soil and cotton cloth,” Carbohydrate Polymers, Vol. 89, Issue.4, pp.1110-1111, 2012.
[43] E. Rosenberg, A. Zuckerberg, C. Rubinovitz, D.L. Gutinck, “Emulsifier Arthrobacter RAG-1: Isolation and emulsifying properties,” Applied and Environmental Microbiology, Vol. 37, Issue.3, pp.402-408, 1979.
[44] C. Rubinovitz, D.L. Gutnick, E. Rosenberg, “Emulsan production by Acinetobacter calcoaceticus in the presence of chloramphenicol,” Journal of Bacteriology, Vol. 152, Issue.1, pp.126-132, 1982.
[45] R. Thavasi, V.R.M. Subramanyam, S. Jayalakshmi, T. Balasubramanian, I.M. Banat, “Biosurfactant production by Azotobacter chroococcum isolated from the marine environment,” Marine Biotechnology, Vol. 11, pp.551-556, 2009.
[46] N. Alwadani, P. Fatehi, “Synthetic and lignin-based surfactants: challenges and opportunities,” Carbon Resources Conversion, Vol. 1, Issue.2, pp.126-138, 2018.
[47] X. Sun, L. Wu, Y. Luo, “Application of organic agents in remediation of heavy metals- contaminated soil,” Ying Yong Sheng Tai Xue Bao, Vol. 17, Issue.6, pp.1123-1128, 2006.
[48] A.M. Davila, R. Marchel, J.P. Vandecasteele, “Sophorose lipid fermentation with differentiated substrate supply for growth and production phases,” Applied Microbiology and Biotechnology, Vol. 47, Issue.5, pp.496-501, 1997.
[49] S.A. Adebusoye, O.O. Amund, M.O. Ilori, D.O. Domeih, J. Okpuzor, “Growth and biosurfactant synthesis by Nigerian hydrocarbon-degrading estuarine bacteria,” Revista de Biologia Tropical, Vol. 56, Issue.4, pp.1603-1611, 2008.
[50] E. Ristau, F. Wanger, “Formation of novel trehalose lipids from Rhodococcus erythropolis under growth limiting conditions,” Biotechnology Letters, Vol. 5, pp.95-100, 1983.
[51] J.O. Kilburn, K. Takayama, “Effects of ethambutol on accumulation and secretion of trehalose mycolates and free mycolic acid in Mycobacterium smegmatis,” Antimicrobial Agents and Chemotherapy Vol. 20, Issue.3, pp.401-404, 1981.
[52] A. Kretschmer, H. Bock, F. Wagner, “Chemical and physical characterization of interfacial-active lipids from Rhodococcus erythropolis grown on n-alkanes,” Applied and Environmental Microbiology, Vol. 44, Issue.4, pp.864-870, 1982.
[53] M. Morikawa, H. Daido, T. Takao, S. Murata, Y. Shimonishi, T. Imanaka, “A new lipopeptide biosurfactant produced by Arthrobacter sp. strain MIS38,” Journal of Bacteriology, Vol. 175, Issue.20, pp.6459-6466, 1993.
[54] H.B.S. Sobrinho, R.D. Rufino, J.M. Luna, “Utilization of two agro industrial by-products for the production of a surfactant by Candida sphaerica UCP0995,” Process Biochemistry, Vol. 43, Issue.9, pp.912-917, 2008.
[55] J.M. Luna, A.S. Santos Filho, R.D. Rufino, L.A. Sarubbo, “Production of biosurfactant from Candida bombicola URM 3718 for environmental applications,” Chemical Engineering Transactions, Vol. 49, pp.583-588, 2016.
[56] L. Rodriguez-Lopez, at. Al.“Ionic behavior assessment of surface-active compounds from corn steep liquor by exchange resins,” Journal of Surfactant and Detergent, Vol. 20, Issue.1, pp.207-217, 2017.
[57] W.A. Eraqi, A.S. Yassin, A.E. Ali, M.A. Amin, “Utilization of crude glycerol s a substrate for the production of rhamnolipid by Pseudomonas aeruginosa,” Biotechnology Research International Article ID 3464509, 9 pages, 2016.
[58] S. Ferhat, R. Alouaoui, A. Badis, N. Moulaiostefa, “Production and characterization of biosurfactant by free and immobilized cells from Ochrobactrum intermedium isolated from the soil of southern Algeria with a view to environmental application,” Biotechnology & Biotechnological Equipment, Vol. 31, Issue.4, pp.733-742, 2017.
[59] K. Patowary, R. Patowary, M.C. Kalita, S. Deka, “Characterization of biosurfactant produced during degradation of hydrocarbons using crude oil as sole source of carbon,” Frontiers in Microbiology, Vol. 8, pp.279, 2017.
[60] A.M. Abdel-Mawgoud, M.M. Aboulwafa, N.A. Hassouna, “Characterization of surfactin produced by Bacillus subtilis isolates BS5,” Applied Biochemistry and Biotechnology, Vol. 150, Issue.3, pp.289-303, 2008.
[61] R.M. Jain, K. Mody, A. Mishra, B. Jha, “Isolation and structural characterization of biosurfactant produced by an alkaliphilic bacterium Cronobacter sakazakii isolated from oil contaminated wastewater,” Carbohydrate Polymers, Vol. 87, Issue.3, pp.2320-2326, 2012.
[62] P. Parthipan, E. Preetham, L.L. Machuca, P.K. Rahman, K. Murugan, A. Rajasekar, “Biosurfactant and degradative enzymes mediated crude oil degradation by bacterium Bacillus subtilis A1,” Frontiers in Microbiology, Vol. 8, pp.193, 2017.
[63] P. Parthipan, P. Elumalai, K. Sathishkumar, D. Sabarinathan, K. Murugan, G. Benelli, A. Rajasekar, “Biosurfactant and enzyme mediated crude oil degradation by Pseudomonas stutzeri NA3 and Acinetobacter baumannii MN3,” 3 Biotech, Vol. 7, Issue.5, pp.278, 2017.
[64] L.R. Rodrigues at. Al., “Physicochemical and functional characterization of a biosurfactant produced by Lactococcus lactis 53,” Colloids and Surfaces B: Biointerfaces Vol. 49, Issue.1, pp.79-86, 2006.
[65] L.M. Santa Anna, G.V. Sebastian, E.P. Menezes, T.L.M. Alves, A.S. Santos, N. Pereira, Jr. D.M.G, “Production of biosurfactants from Pseudomonas aeruginosa PA1 isolated in oil environments,” Brazilian Journal of Chemical Engineering, Vol. 19, Issue.2, pp.159-166, 2002.
[66] A. Singh, J.D. Van Hamme, O.P. Ward, “Surfactants in microbiology and biotechnology: Part 2. Application aspects,” Biotechnology Advances, Vol. 25, Issue.1, pp.99-121, 2007.
[67] M.L. Ibrahim, U.J.J. Ijah, S.B. Manga, L.S. Bilbis, S. Umar, “Production and partial characterization of biosurfactant produced by crude oil degrading bacteria,” International Journal of Biodeterioration and Biodegradation, Vol. 81, pp.28-34, 2013.
[68] F. Peng, Y. Wang, F. Sun, Z. Liu, Q. Lai, Z. Shao, “A novel lipopeptide produced by a Pacific Ocean deep-sea bacterium, Rhodococcus sp. TW53,” Journal of Applied Microbiology, Vol. 105, Issue.3, pp.698-705, 2008.
[69] A.M. Davila, R. Marchal, J.P. Vandecasteele, “Kinetics and balance of a fermentation free from product inhibition: sophorolipid production by Candida bombicola,” Applied Microbiology and Biotechnology, Vol. 38, Issue.1, pp.6-11, 1992.
[70] N.A. Bhawsar, M. Singh, “GC-MS profile of biosurfactant producing and hydrocarbon degrading P. aeruginosa NGB4 in liquid culture system,” Journal of Pharmacy Research, Vol. 10, Issue.11, pp.726-729, 2016.
[71] J. Liu, C. Vipulanandan, “Effects of Au/Fe and Fe nanoparticles on Serratia bacterial growth and production of biosurfactant,” Materials Science and Engineering C, Vol. 33, Issue.7, pp.3909-3915, 2013.
[72] D.K.F. Santos, R.D. Rufino, J.M. Luna, V.A. Santos, L.A. Sarubbo, “Biosurfactants: multifunctional biomolecules of the 21st century,” International Journal of Molecular Sciences, Vol. 17, Issue.3, pp.401, 2016.
[73] T.H. Mouafo, A. Mbawala, R. Ndjouenkeu, “Effect of different carbon sources on biosurfactants` production by three strains of Lactobacillus spp.,” BioMed Research International, Article ID 5034783, 15 pages, 2018.
[74] M. Pacwa-Płociniczak, G.A. Płaza, Z. Piotrowska-Seget, S.S. Cameotra, “Environmental applications of biosurfactants: Recent advances,” International Journal of Molecular Sciences, Vol. 12, Issue.1, pp.633-654, 2011.
[75] S.K.K. Randhawa, P.K. Rahman, “Rhamnolipid biosurfactants—past, present, and future scenario of global market,” Frontiers in Microbiology Vol. 5, pp.454, 2014.
[76] S.J. Varjani, V.N. Upasani, “Critical review on biosurfactant analysis, purification and characterization using rhamnolipid as a model biosurfactant,” Bioresource Technology, Vol. 232, pp.389-397, 2017.
[77] V. Pruthi, S.S. Cameotra, “Novel sucrose lipid produced by Serratia marcescens and its application in enhanced oil recovery,” Journal of Surfactants and Detergent, Vol. 3, Issue.4, pp.533-537, 2000.
[78] G.V. Moro, R.T.R. Almeida, A.P. Napp, C. Porto, E.J. Pilau, D.S. Ludtke, A.V. Moro, M.H. Vainstein, “Identification and ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry characterization of biosurfactants, including a new surfactin, isolated from oil-contaminated environments,” Microbial Biotechnology, Vol. 11, Issue.4, pp.759-769, 2018.
[79] D.G. Cooper, C.R. Macdonald, S.J. Duff, N. Kosaric, “Enhanced production of surfactin from Bacillus subtilis by continuous product removal and metal cation additions,” Applied and Environmental Microbiology, Vol. 42, Issue.3, pp.408-412, 1981.
[80] C. Syldatk, at. Al., “Chemical and physical characterization of four interfacial-active rhamnolipids from Pseudomonas spec. DSM 2874 grown on n-alkanes,” Z Naturforsch C, Vol. 40, Issue.1-2, pp.51-60, 1985.
[81] C.A.B. Gusmao, R.D. Rufino, L.A. Sarubbo, “Laboratory production and characterization of a new biosurfactant from Candida glabrata UCP1002 cultivated in vegetable fat waste applied to the removal of hydrophobic contaminant,” World Journal of Microbiology and Biotechnology, Vol. 26, Issue.9, pp.1683-1692, 2010.
[82] P. Datta, P. Tiwari, L.M. Pandey, “Isolation and characterization of biosurfactant producing and oil degrading Bacillus subtilis MG495086 from formation water of Assam oil reservoir and its suitability for enhanced oil recovery,” Bioresource Technology, Vol. 270, pp.439-448, 2018.
[83] A. Saimmai, at. Al.“Isolation and functional characterization of a biosurfactant produced by a new and promising strain of Oleomonas sagaranensis AT18,” World Journal of Microbiology and Biotechnology, Vol. 28, Issue.10, pp.2973-2986, 2012.
[84] S.K. Satpute, N.S. Mone, P. Das, I.M. Banat, A.G. Banpurkar, “Inhibition of pathogenic bacterial biofilms on PDMS based implants by L. acidophilus derived biosurfactant,” BMC Microbiology, Vol. 19, Issue.1, pp.39, 2019.
[85] R. Khademolhosseini, A. Jafari, S.M. Mousavi, H. Hajfarajollah, K.A. Noghabic, M. Manteghiana, “Physicochemical characterization and optimization of glycolipid biosurfactant production by a native strain of Pseudomonas aeruginosa HAK01 and its performance evaluation for the MEOR process,” RSC Advances, Vol. 9, Issue.14, pp.7932-7947, 2019.
[86] P. Renard, I. Canet, M. Sancelme, M. Matulova, I. Uhliarikova, B. Eyheraguibel, L. Nauton, J. Devemy, M. Traïkia, P. Malfreyt, A.M. Delort, “Cloud microorganisms, an interesting source of biosurfactants,” Surfactant and Detergents, IntechOpen Online, 2019.
[87] E. Haba, A. Pinazo, O. Jauregui, M.J. Espuny, M.R. Infante, A. Manresa, “Physicochemical characterization and antimicrobial properties of rhamnolipids produced by Pseudomonas aeruginosa 47T2 NCBIM 40044,” Biotechnology and Bioengineering, Vol. 81, Issue.3, pp.316-322, 2003.
[88] A. Khopade, R. Biao, X. Liu, K. Mahadik, L. Zhang, C. Kokare, “Production and stability studies of the biosurfactant isolated from marine Nocardiopsis sp. B4,” Desalination, Vol. 285, pp.198-204, 2012.
[89] M.C. Cirigliano, G.M. Carman, “Isolation of a bioemulsifier from Candida lipolytica,” Applied and Environmental Microbiology, Vol. 48, Issue.4, pp.747-750, 1984.
[90] A.M. Elazzazy, T.S. Abdelmoneim, O.A. Almaghrabi, “Isolation and characterization of biosurfactant production under extreme environmental conditions by alkali-halo-thermophilic bacteria from Saudi Arabia,” Saudi Journal of Biological Sciences, Vol. 22, Issue.4, pp.466-475, 2015.
[91] D. Hentati, A. Chebbi, F. Hadrich, I. Frikha, F. Rabanal, S. Sayadi, A. Manresac, M. Chamkha, Production, characterization and biotechnological potential of lipopeptides biosurfactants from a novel marine Bacillus stratosphericus strain FLU5,” Ecotoxicology and Environmental Safety, Vol. 167, pp.441-449, 2019.
[92] R.C.F. Soares da Silva, D.G. Almeida, H.M. Meira, E.J. Silva, C.B. Farias, R.D. Rufino, J.M. Luna, L.A. Sarubbo, “Production and characterization of a new biosurfactant from Pseudomonas cepacia grown in low-cost fermentative medium and its application in the oil industry,” Biocatalysis Agricultural Biotechnology, Vol. 12, pp.206-215, 2017.
[93] D.K.F. Santos, H.M. Meirab, R.D. Rufino, J.M. Luna, L.A. Sarubbo, “Biosurfactant production from Candida lipolytica in bioreactor and evaluation of its toxicity for application as a bioremediation agent,” Process Biochemistry, Vol. 54, pp.20-27, 2017.
[94] S.S. Jha, S.J. Joshi, S.J. Geetha, “Lipopeptide production by Bacillus subtilis R1 and its possible applications,” Brazilian Journal of Microbiology, Vol. 47, Issue.4, pp.955-964, 2016.
[95] A.J. Das, R. Kumar, “Utilization of agro-industrial waste for biosurfactant production under submerged fermentation and its application in oil recovery from and matrix,” Bioresource Technology, Vol. 260, pp.233-240, 2018.
[96] A. Aparna, G. Srinikethan, H. Smitha, “Production and characterization of biosurfactant produced by a novel Pseudomonas sp. 2B,” Colloids and Surfaces B: Biointerfaces, Vol. 95, pp.23-29, 2012.
[97] M. Govindammal, “Effect of carbon and nitrogen sources on the production of biosurfactant by Pseudomonas fluorescens isolated from mangrove ecosystem,” International Journal of Pharmaceutical & Biological Archives, Vol. 5, Issue.8, pp.108-115, 2014.
[98] A.M. Abdel-Mawgoud, M.M. Aboulwafa, N.A. Hassouna, “Characterization of rhamnolipid produced by Pseudomonas aeruginosa isolate Bs20,” Applied Biochemistry and Biotechnology, Vol. 157, pp.329-345, 2009.
[99] E.J. Gudina, E.C. Fernandes, A.I. Rodrigues, J.A. Teixeira, L.R. Rodrigues, “Biosurfactant production by Bacillus subtilis using corn steep liquor as culture medium,” Frontiers in Microbiology, Vol. 6, pp.59, 2015.
[100] R.M. Jain, K. Mody, N. Joshi, A. Mishra, B. Jha, “Production and structural characterization of biosurfactant produced by an alkaliphilic bacterium, Klebsiella sp.: evaluation of different carbon sources,” Colloids and Surfaces B: Biointerfaces, Vol. 108, pp.199-204, 2013.
[101] H. Abbasi, M.M. Hamedi, T.B. Lotfabad, H.S. Zahiri, H. Sharafi, F. Masoomi, A.A Moosavi-Movahedi, A. Ortiz, M Amanlou, K.A. Noghabi, “Biosurfactant-producing bacterium, Pseudomonas aeruginosa MA01 isolated from spoiled apples: physicochemical and structural characteristics of isolated biosurfactant,” Journal of Bioscience and Bioengineering, Vol. 113, Issue.2, pp.211-219, 2012.
[102] H.T. Ronning, E.H. Madslien, T.N. Asp, P.E. Granum, “Identification and quantification of lichenysin-a possible source of food poisoning,” Food Additives and Contamination, Vol. 32, Issue.12, pp.2120-2130, 2015.
[103] A.P. Karlapudi, T.C. Venkateswarulu, J. Tammineedi, L. Kanumuri, B.K. Ravuru, V.R. Dirisala, V.P. Kodali, “Role of biosurfactants in bioremediation of oil pollution-a review,” Petroleum, Vol. 4, Issue.3, pp.241-249, 2018.
[104] P. Singh, S.S. Cameotra, “Potential applications of microbial surfactants in biomedical sciences,” Trends in Biotechnology, Vol. 22, Issue.3, pp.142-146, 2004.
[105] S. Techaoei, S. Lumyong, W. Prathumpai, D. Santiarwarm, P. Leelapornoisid, “Screening, characterization and stability of biosurfactant produced by Pseudomonas aeruginosa SCMU 106 isolated from soil in Northern Thailand,” Asian Journal of Biological Sciences, Vol. 4, Issue.4, pp.340-351, 2011.
Citation
Anuradha S. Pendse, K. Aruna, "Physicochemical and analytical characterization of biosurfactant produced by Serratia rubidaea KAP," International Journal of Scientific Research in Biological Sciences, Vol.7, Issue.2, pp.25-40, 2020 -
Open Access Article
Effect of Unani Medicine in Iltehab-e-Jild Huzaazi (Seborrheic Dermatitis) of Head: A case study
Misbahuddin Azhar, Zamir Ahmad, Mustehasan
Research Paper | Journal-Paper (IJSRBS)
Vol.7 , Issue.2 , pp.41-43, Apr-2020
Abstract
Iltihab-e-Jild Huzazi (IJH) or Seborrheic dermatitis (SD) is a mild inflammatory disorder of skin that mainly affects scalp. Central face nasolabial folds, eyebrows and central chest are other commonly affected areas in adults. It is due to Pityrosporum ovale infection of the skin. According to Unani system of medicine IJH is due to predominance of certain humors at the site of balgham (phlegm) and sauda (black bile).Here in we report a case of Iltihab-e-Jild Huzazi and reduction in sign and symptoms following treatment with Pharmacopoeial Unani medicine Habb Musaffi Khoon along with a proprietary medicine Eczenil ointment. There is almost complete clearance of scaly patches on the head after the treatment for 15 days with this combination. It may be concluded that the above combination may be used as a cost effective and safe treatment option for IJH (SD).Key-Words / Index Term
Iltihab-e-Jild Huzazi, Seborrheic dermatitis, Unani medicine, Musaffi-e-Khoon, Eczenil ointmentReferences
[1] J.Q. Del Rosso, “Adult Seborrheic Dermatitis: A Status Report on Practical Topical Management”, J Clin. Aesthet. Dermatol. Vol. 4, issue., 5, pp.32-38, 2011.
[2] L.J. Borda, T.C. Wikramanayake, “Seborrheic Dermatitis and Dandruff: A Comprehensive Review”, J Clin Investig Dermatol., vol. 3, issue., 3, pp 1-22, 2015.
[3] C. Haslett, E.R. Chilvers, N.A. Boon, N.R. Colledge, “Davidson’s Principles and Practice of Medicine”, 19th edition, Churchill Livingstone New York, pp. 1073,1075, 2002.
[4] A. Majoosi, “Kamil us Sana”, Vol-II, (Urdu translation), Munshi Nawal Kishore Lucknow, pp. 252, 1889.
[5] M. Kabiruddin, “Sharah Asbab wa Alamat”, vol.-IV, Hikmat Book Depot Hyderabad, pp. 47-48, 1916.
[6] Z. Ahmad, “Moalajat-e-Jild”, A & U Tibbiya College, Karol Bagh Delhi, pp. 87-89, 2004.
[7] T. Burns, S. Breathnach, N. Cox, C. Griffiths ,“Rook’s Textbook of Dermatology”, ed 8. Blackwell, Oxford, pp. 23.29-23.34, 2010.
[8] M. Picardo, N. Cameli, “Seborrheic dermatitis. In: Williams H,Bigby M, Diepgen T, Herxheimer A, Naldi L, Rzany B, eds. Evidence-Based Dermatology”, Second Edition. Blackwell Publishing Massachusetts, pp. 164-170, 2008.
[9] Anonymous, “Unani System of Medicine (The Science of Health and Healing)”, Department of AYUSH, Ministry of Health & Family Welfare, Government of India, New Delhi, pp. 29-32, 39, 2013.
[10] Anonymous, “National Formulary of Unani Medicine”, Vol-I, part-III, The Controller of Publications, Civil Lines, Delhi, pp. 32, 2001.
Citation
Misbahuddin Azhar, Zamir Ahmad, Mustehasan, "Effect of Unani Medicine in Iltehab-e-Jild Huzaazi (Seborrheic Dermatitis) of Head: A case study," International Journal of Scientific Research in Biological Sciences, Vol.7, Issue.2, pp.41-43, 2020 -
Open Access Article
Manal M. E. Ahmed, Jakeen Eljakee, Tarek Mahran
Research Paper | Journal-Paper (IJSRBS)
Vol.7 , Issue.2 , pp.44-50, Apr-2020
Abstract
Emergence of multidrug-resistance (MDR) P. aeruginosa for respiratory tract infections represents a major critical challenge to the health care community worldwide. It is associated with progressive chronic respiratory infection in patients with cystic fibrosis (CF). While, the antibiotic resistance has been a global growing problem and vaccines development is impeded by several limitations, an urgent need to develop novel approaches is growing. In this study, we developed anti- P. aeruginosa IgYs as an alternative to meet this need. Preclinical evaluation revealed that immunization of Balb/c mice with the developed IgYs resulted in 95%, 90%, 70% protective effect among group (A) that was immunized intranasal one hour before challenge, group (B) that was immunized intranasal six hours after challenge and group (C) that was immunized orally, respectively. Furthermore, the pulmonary bacterial load in treated mice was lower than in the controls by more than 2 log after 24 hours of infection. Thus, the prepared IgYs had anti-P. aeruginosa immuno-reactivity that increasing its potential for use as a prophylactic therapy.Key-Words / Index Term
P. Aeruginosa, Immunoglobulins Y, Cystic fibrosis, protective efficacy, challenge test, prophylactic therapyReferences
[1] P.B. Davis, M. Drumm, M.W. Konstan, "Cystic fibrosis", Am. J. Respir. Crit. Care Med, Vol. 154, Issue. 5, pp. 1229–1256, 1996.
[2] P.H. Gilligan, "Microbiology of airway disease in patients with cystic fibrosis", J Clin Microbiol, Vol. 4, pp. 35–51, 1991.
[3] U.B. Schaad, A.B. Lang, J. Wedgwood, A. Ruedeberg, J.U. Que, E. Furer, S.J.J. Cryz, "Safety and immunogenicity of Pseudomonas aeruginosa conjugate A vaccine in cystic fibrosis", Lancet , Vol. 338, pp. 1236–1237, 1991.
[4] J.J. Smith, S.M. Travis, E.P. Greenberg, M.J. Welsh, "Cystic fibrosis airway epithelia fail to kill bacteria because of abnormal airway surface fluid", Cell, Vol. 85, pp. 229–236, 1996.
[5] J.L. McCaskill, S. Ressel, A. Alber, J. Redford, U.F. Power, J. Schwarze, B.M. Dutia, A.H. Buck, "Broad-spectrum inhibition of respiratory virus infection by MicroRNA mimics targeting p38 MAPK signaling", Mol Ther Nucl Acids, Vol. 7, pp. 256–266, 2017.
[6] D. Corti, N. Passini, A. Lanzavecchia, M. Zambon, "Rapid generation of a human monoclonal antibody to combat Middle East respiratory syndrome", J Infect Public Health , Vol. 9, pp. 231–235, 2016.
[7] A.W. Cripps, M.L. Dunkley, R.L. Clancy, J. Kyd, "Pulmonary immunity to Pseudomonas aeruginosa", Immunol Cell Biol, Vol. 73, pp. 418–424, 1995.
[8] M.A. Keller, E.R. Stiehm, "Passive immunity in prevention and treatment of infectious diseases", Clin Microbiol Rev, Vol. 13, pp. 602-614, 2000.
[9] B.S. Gaham, D.M. Ambrosino, "History of passive antibody administration for prevention and treatment of infectious diseases", Curr Opin HIV AIDS. Vol. 10, pp. 129–134, 2015.
[10] B. Kelley, "Industrialization of mAb production technology: the bioprocessing industry at a crossroads", mAbs, Vol. 1, pp. 443–452, 2009.
[11] A. Casadevall, E. Dadachova, L.A. Pirofski, "Passive antibody therapy for infectious diseases", Nat Rev Microbiol, Vol. 2, pp. 695–703, 2004.
[12] G.W. Warr, K.E. Magor, D.A. Higgins, "IgY: clues to the origins of modern antibodies", Immunol Today, Vol. 16, pp. 392–398, 1995.
[13] D. Carlander, J. Stalberg, A. Larsso, "Chicken antibodies: a clinical chemistry perspective", Ups J Med Sci, Vol. 104, pp. 179–189, 1999.
[14] K. Thomsen, L. Christophersen, T. Bjarnsholt, P.O. Jensen, C. Moser, N. Hoiby, "Anti-Pseudomonas aeruginosa IgY antibodies augment bacterial clearance in a murine pneumonia model", J Cyst Fibr, Vol. 15, pp. 171-178, 2016.
[15] H.H. Nguyen, T.M. Tumpey, H. J. Park, Y.H. Byun, L.D. Tran, V.D. Nguyen, P.E. Kilgore, C. Czerkinsky, J.M. Katz, B.L. Seong, J.M. Song, Y.B. Kim, H.T. Do, T. Nguyen, C.V. Nguyen, "Prophylactic and therapeutic efficacy of avian antibodies against Influenza virus H5N1 and H1N1 in mice", PLoS One, Vol. 5, pp. e10152, 2010.
[16] E.M. Akita, S. Nakai, "Comparison of four purification methods for the production of immunoglobulins from eggs laid by hens immunized with an enterotoxigenic E. coli strain.", J Immunol Methods, Vol. 160, pp. 207-214, 1993.
[17] R. Chalghoumi, A. Thewis, Y. Beckers, C. Marcq, D. Portetelle, Y.J. Schneider, "Adhesion and growth inhibitory effect of chicken egg yolk antibody (IgY) on Salmonella enterica serovars Enteritidis and Typhimurium in vitro", Food borne Pathog Dis, Vol. 6, pp. 593-604, 2009.
[18] M. Kuroki, Y. Ikemori, H. Yokoyama, R.C. Peralta, F.C. Icatlo, Y. Kodama, "Passive protection against bovine rotavirus-induced diarrhea in murine model by specific immunoglobulins from chicken egg yolk.", Vet Microbiol, Vol. 37, pp. 135-146, 1993.
[19] P.H. Gilligan, "Infections in patients with cystic fibrosis: diagnostic microbiology update. Clinics in Laboratory Medicine", Vol. 34, Issue. 2, pp. 197–217, 2014.
[20] N. Safdar, C. Dezfulian, H.R. Collard, S. Saint, "Clinical and economic consequences of ventilator-associated pneumonia: a systematic review", Critical Care Medicine, Vol. 33, Issue. 10, pp. 2184–2193, 2005.
[21] B.K Chan., M. Sistrom, J.E. Wertz, K.E. Kortright, D. Narayan, P.E. Turner, "Phage selection restores antibiotic sensitivity in MDR Pseudomonas aeruginosa", Scientific Reports , Vol. 6, pp. 26717, 2016.
[22] M.W. Douglas, K. Mulholland, V. Denyer, T. Gottlieb, "Multi-drug resistant Pseudomonas aeruginosa outbreak in a burns unit—an infection control study", Burns: Journal of the International Society for Burn. Injuries, Vol. 27, Issue. 2, pp. 131–135, 2001.
[23] M. Mudau , R. Jacobson, N. Minenza, L. Kuonza, V. Morris, H. Engelbrecht, M.P. Nicol, C. Bamford, "Outbreak of multidrug resistant Pseudomonas aeruginosa bloodstream infection in the haematology unit of a South African academic hospital", PLoS One, Vol. 8, Issue. 3, pp. e55985, 2013.
[24] G.P. Priebe, G.J. Meluleni, F.T. Coleman, J.B. Goldberg, G.B. Pier, "Protection against fatal Pseudomonas aeruginosa pneumonia in mice after nasal immunization with a live, attenuated aroA deletion mutant", Infect. Immun, Vol. 71, pp. 1453–1461, 2003.
[25] J. Kovacs-Nolan, Y. Mine. "Egg yolk antibodies for passive immunity", Annu Rev Food Sci Technol, Vol. 3, pp. 163–182, 2012.
[26] R. Weltzin, T.P. Monath, "Intranasal antibody prophylaxis for protection against viral disease", Clin Microbiol Rev, Vol. 12, pp. 383–393, 1999.
[27] R. Schade, A. Hlinak, "Egg yolk antibodies, state of the art and future prospects", Altex, Vol. 13, pp. 5–9, 1996.
[28] K. Selvan, R. Sentila, . A. Michael, "eneration and characterization of chicken egg yolk antibodies against propionibacterium acnes for the prevention of acne vulgaris", Indian J Dermatol, Vol. 57, pp. 15–19, 2012.
[29] El S.M. Ibrahim, A.K. Rahman, R. Isoda, K. Umeda, N. Van Sa, Y. Kodama", In vitro and in vivo effectiveness of egg yolk antibody against Candida albicans (anti-CA IgY)", Vaccine, Vol. 26, pp. 2073–2080, 2008.
[30] S.V. Nguyen, F.C. Jr. Icatlo, T. Nakano, E. Isogai, K. Hirose, H. Mizugai, M. Kobayashi-Sakamoto, H. Isogai, I. Chiba, "Anti-cell-associated glucosyl transferase immunoglobulin Y suppression of salivary mutans streptococci in healthy young adults", J Am Dent Assoc, Vol. 142, pp. 943–949, 2011.
[31] K. Yokoyama, N. Sugano, T. Shimada, R.A.K.M. Shofiqur, El. S.M. Ibrahim, R. Isoda, K. Umeda, N.V. Sa, Y. Kodama, K. Ito, "Effects of egg yolk antibody against Porphyromonas gingivalis gingipains in periodontitis patients", J Oral Sci, Vol. 49, pp. 201–206, 2007.
[32] H. Suzuki, S. Nomura , T. Masaoka, H. Goshima, N. Kamata, Y. Kodama, H. Ishii, M. Kitajima, K. Nomoto, T. Hibi, "Effect of dietary anti-Helicobacter pylori-urease immunoglobulin Y on Helicobacter pylori infection", Aliment Pharmacol Ther, Vol. 20, Issue. Suppl 1, pp. 185–192, 2004.
[33] K.S. Hong, M.R. Ki, H.M.A. Ullah, E.J. Lee, Y.D. Kim, M.J. Chung, A.K. Elfadl, J.K. Park, K.S. Jeong , "Preventive effect of anti-VacA egg yolk immunoglobulin (IgY) on Helicobacter pylori-infected mice", Vaccine, Vol. 36, pp. 371–380, 2018.
[34] N. Gujral, R. Lobenberg, M. Suresh, H. Sunwoo, "In-vitro and in-vivo binding activity of chicken egg yolk immunoglobulin Y (IgY) against gliadin in food matrix", J Agric Food Chem, Vol. 60, pp. 3166–3172, 2012.
[35] K. Hirai, H. Arimitsu, K. Umeda, K. Yokota, L. Shen, K. Ayada, Y. Kodama, T. Tsuji, Y. Hirai, K. Oguma, "Passive oral immunization by egg yolk immunoglobulin (IgY) to Vibrio cholerae effectively prevents cholera", Acta Med Okayama , Vol. 64, pp. 163–170, 2010.
[36] S. Rahman, K. Higo-Moriguchi, K.W. Htun, K. Taniguchi, F.C. Icatlo, T. Tsuji, Y. Kodama, S. Van Nguyen, K. Umeda, H. N. Oo, et al.,"Randomized placebo-controlled clinical trial of immunoglobulin Y as adjunct to standard supportive therapy for rotavirus-associated diarrhea among pediatric patients`, Vaccine, Vol. 30, pp. 4661–4669, 2012.
[37] M. Hirose, T. Ando, R. Shofiqur, K. Umeda, Y. Kodama, S. Nguyen, T. Goto, M. Shimada, S. Nagaoka, "Anti-obesity activity of hen egg anti lipase immunoglobulin yolk, a novel pancreatic lipase inhibitor", Nutr Metab (Lond), Vol. 10, Issue. 1, pp. 70, 2013.
[38] A.T. Abbas, S.A. El-Kafrawy, S.S. Sohrab, E.I.A. Azhar, "EIA. IgY antibodies for the immunoprophylaxis and therapy of respiratory infections`, Human Vaccines & Immunotherapeutics, Vol. 15, Issue. 1, pp. 264-275, 2019.
[39] J. Emerson, M. Rosenfeld, S. McNamara, B. Ramsey, R.L. Gibson, "Pseudomonas aeruginosa and other predictors of mortality and morbidity in young children with cystic fibrosis", Pediatr Pulmonol, Vol. 34, pp. 91–100, 2002.
[40] G.M. Nixon, D.S. Armstrong, R. Carzino, J. B. Carlin, A. Olinsky , C.F. Robertson, K. Grimwood, "Clinical outcome after early Pseudomonas aeruginosa infection in cystic fibrosis", J Pediatr, Vol. 138, pp. 699–704, 2001.
[41] E. Nilsson, A. Larsson, H.V. Olesen, P.E. Wejaker, H. Kollberg, "Good effect of IgY against Pseudomonas aeruginosa infections in cystic fibrosis patients", Pediatr Pulmonol, Vol. 43, pp. 892–899, 2008.
[42] M.M.E. Ahmed, W. Nazmy, J. Eljakee, "Preparation and Evaluation of Novel Anti-Obesity Immunoglobulins for Immunoprophylaxis and Therapy", International Journal of Scientific Research in Biological Sciences, Vol.6, Issue.6, pp.81-88, (2019).
[43] A.M. Abdou, M.M.E. Ahmed, Y. Yamashita, M. Kim, "Immunoglobulin: A Natural Way to Suppress Helicobacter pylori in Humans", Health, Vol.6, pp.781-791, 2014.
Citation
Manal M. E. Ahmed, Jakeen Eljakee, Tarek Mahran, "Developement of anti- P. aeruginosa Immunoglobulin Y antibodies as prophylacic therapy for cystic fibrosis patients," International Journal of Scientific Research in Biological Sciences, Vol.7, Issue.2, pp.44-50, 2020 -
Open Access Article
Bhaskor Kolita, Amlan Jyoti Bora, Pinaki Hazarika
Research Paper | Journal-Paper (IJSRBS)
Vol.7 , Issue.2 , pp.51-61, Apr-2020
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease that causes respiratory illness in human and has now become a major challenge for all over the world. As no drug is approved yet for COVID-19, it is strongly demanding to search lead compounds. Therefore, the current investigation is an attempt to screen lead molecule for inhibition of Covid-19 main protease. Herein, we considered 98 plant derived alkaloids having antimalarial activity, quinine and docked with COVID-19 main protease taking Chloroquine, hydroxychloroquine as reference. Finally, compounds Bidebiline E, Bisnordihydrotoxiferine and Thalifaberine were screened as lead molecule on the basis of Moldock score, H-bond interaction and ADMET study, and recommended for in vitro investigation.Key-Words / Index Term
COVID-19; Chloroquine; Hydroxychloroquine; Alkaloids; ADMETReferences
[1] X. Xu, P. Chen, J. Wang, et al. “Evolution of the novel coronavirus from the ongoing Wuhan outbreak and modeling of its spike protein for risk of human transmission” Sci China Life Sci. 63, 457–460,2020.
[2] J.E.L. Wong, Y.S. Leo, C.C. Tan.”COVID-19 in Singapore-Current Experience: Critical Global Issues That Require Attention and Action” JAMA.323(13),1243-1244, 2020.
[3] Li, X. Guan, P. Wu, et al. “Early Transmission Dynamics in Wuhan, China, of Novel Coronavirus-Infected Pneumonia”. N Engl J Med. 382,1199-1207, 2020.
[4] A.K. Singh, A. Singh, A. Shaikh, R. Singh, A. Misra.”Chloroquine and hydroxychloroquine in the treatment of COVID-19 with or without diabetes: A systematic search and a narrative review with a special reference to India and other developing countries” Diabetes Metab Syndr. 26;14(3).241-246. 2020 .
[5] M. Wang, R. Cao, L. Zhang, X. Yang, J. Liu, M. Xu, et al.”Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro”. Cell Resear, volume 30, pp 269–271, 2020.
[6] F. Touret, X. de Lamballerie “Of chloroquine and COVID-19”. Antiviral Res.177:104762, 2020.
[7] V.M.Quintana, B.Selisko, J.E.Brunetti,C.Eydoux,n J.C.Guillemot, B.Canard, E.B.Damonte, J.G.Julander, V.Castilla. “Antiviral activity of the natural alkaloid anisomycin against dengue and Zika viruses” Antiviral Research, Volume 176:104749 2020.
[8] K.P.Mishra, N. Sharma, D. Diwaker , L. Ganju and S.B. Singh.” Plant Derived Antivirals: A Potential Source of Drug Development” J Virol Antivir Res, 2:2, 2013.
[9] Pelletier and Caventoz. “Suite: Des recherches chimiques sur les quinquinas, Annales de Chimie et de Physique”, 15: 337-365, 1820.
[10] Z. Xu, C. Peng, Y. Shi, Z. Zhu, K. Mu, and X. Wang, “Nelfinavir was predicted to be a potential inhibitor of 2019-nCov main protease by an integrative approach combining homology modelling , moleculardocking and binding free energy calculation” bioRxiv.1201,0–2,2020.
[11] S. Khaerunnisa, H. Kurniawan, R. Awaluddin , S. Suhartati , S. Soetjipto. “Potential Inhibitor of COVID-19 Main Protease (Mpro) from Several Medicinal Plant Compounds by Molecular Docking Study” Preprints 2020030226, 2020. (doi: 10.20944/preprints202003.0226.v1).
[12] I. M. Said, A. Latiff, S. J. Partridge and J. D. Phillipson,., “Alkaloids from Dehaasia incrassata.” Planta Med., 57, 389, 1991.
[13] S. Kanokmedhakul et al. “Bioactive constituents of the roots of Polyalthia cerasoides.” J Nat Prod.70: 1536–1538, 2007.
[14] G.M. Rukunga et al. “The antiplasmodial activity of spermine alkaloids isolated from Albizia gummifera” Fitoterapia 78: 455–459, 2007.
[15] C. W. Wright, D. Allen, Ya Cai, J. D. Phillipson, I. M. Said, G. C. Kirby, D. C. Warhurst. “In vitro anti of amoebic and antiplasmodial activities of alkaloids isolated from Alstonia angustifolia roots” Phytother Res., 6, 121–124. 1992.
[16] K. Likhitwitayawuid, C. K. Angerhofer, G. A. Cordell, and J. M. Pezzuto,.. “Cytotoxic and antimalarial bisbenzylisoquinoline alkaloids from Stephania erects.” J. Nat. Prod., 56, 30–38, 1993.
[17] K. Likhitwitayawuid, C. K. Angerhofer, H.Chai, J. M. Pezzuto and G. A. Cordell “Cytotoxic and antimalarial alkaloids from the bulbs of Crium amabile” J. Nat. Prod. 56, 1331–1338, 1993.
[18] E. Osorio et al. “Actividad antiplasmodial de alcaloidesaporfinicos de Rollinia pittieri y Pseudomalmea boyacana(Annonaceae)” Vitae. Rev Fac Quimica Farm. 13: 49–54. 2006.
[19] K. Likhitwitayawuid, C. K. Angerhofer, H.Chai, J. M. Pezzuto and G. A. Cordell “Cytotoxic and antimalarial alkaloids from the tubers of Stephania pierrei” J. Nat. Prod. 56,1468–1478. 1993 .
[20] L. Z. Lin,. et al. “Cytotoxic and antimalarial bisbenzylisoquinoline alkaloids from Cyclea barbata.” J. Nat. Prod., 56, 22–29. 1993.
[21] N,T. Nguyen et al. “Antiplasmodial alkaloids from Desmos rostrata” J Nat Prod. 71: 2057–2059. 2008.
[22] A. Phrutivorapongkul et al. “Anti-plasmodial activity of bisbenzylisoquinoline alkaloids from Michelia figo leaves” Thai J Health Res. 20: 121–128. 2006.
[23] L.Z. Lin et al. “ Thalifaberidine, a cytotoxic aporphine-benzylisoquinoline alkaloid from Thalictrum faberi” J. Nat. Prod., 57, 1430–1436, 1994.
[24] G. C. Kirby, A. Paine, D. C. Warhurst, B. K Noamese, and J. D. Phillipson “In vitro and in vivo antimalarial activity of cryptolepine, a plant-derived indoloquinoline” Phytother. Res, 9, 359–363,1995.
[25] M. Sauvain, et al., “Antimalarial activity of alkaloids from Pogonopus tubulosus”. Phytother. Res. 10, 198–201. 1996.
[26] I. Kitagawa, T. Mahmud, P Simanjuntak, K. Hori, T. Uji and H. Shibuya “Indonesian medicinal plants. VIII. Chemical structures of three new triterpenoids, bruceajavanin A, dihydrobruceajavanin A, and bruceajavanin B, and a new alkaloidal glycoside, bruceacanthinoside, from the stems of Brucea javanica (Simaroubaceae)”Chem. Pharm. Bull., 42, 1416–1421, 1994 .
[27] P. Grellier et al., “Antimalarial activity of cryptolepine and isocryptolepine,alkaloids isolated from Cryptolepis sanguinolenta” Phytother. Res. 10, 317–321, 1996,
[28] Y. F Hallock et al., “Michellamines D-F, new HIV-inhibitorydimiric naphthylisoquinoline alkaloids and korupensamine E, a new antimalarial monomer, from Anchistrocladus korupensis” J.Nat. Prod., 60(7), 677–-83, 199 .
[29] Y. F. Hallock, J. H. Cordellina, M. Schaffer, Bringmann, G. Francois, and M. R. Boyd, A. Korundamine, “ a novel HIVinhibitory and antimalarial ‘hybrid’ naphthylisoquinoline alkaloid heterodime from Ancistrocladus korupensis” Bioorg. Med.Chem. Lett., 8, 1729–1734, 1998.
[30] A. Chea et al. “Antimalarial activity of alkaloids isolated from Stephania rotunda” J Ethnopharmacol 112: 132–137, 2007.
[31] T. Takaya “New type of Febrifugine analogues, bearing a quinolizidine moiety, show antimalarial activity against Plasmodium malaria parasite” J. Med. Chem.42, 3163–3166, 1999.
[32] I. Muhammad, D.C. Dunbar, S. Takamatsu, L. A. Walker, and , A. M. Clark “Antimalarial, cytotoxic, and antifungal alkaloids from Duguetia hadrantha” J. Nat. Prod.,64,559–562. 2001.
[33] H. Morita et al. “Cassiarins A and B, novel antiplasmodial alkaloids from Cassia siamea” Org Lett; 9: 3691–3693, 2007.
[34] G. Bringmann, C. Gunther, W. Saeb, J. Mies, A. Wickramasinghe, V. Mudogo and R. Brun, “Ancistrolikokines A–C: New 5,8¢-coupled naphthylisoquinoline alkaloids from Ancistrocladus likoko” J. Nat. Prod. 63, 1333–1337, 2000.
[35] V. Munoz,. et al., “Antimalarial activity and cytotoxicity of (–)- roemrefidine isolated from the stem bark of Sparattanthelium amazonum” Planta Med., 65, 448–449, 1999 .
[36] V. Jullian et al.“Validation of use of a traditional antimalarial remedy from French Guiana, Zanthoxylum rhoifolium Lam” J Ethnopharmacol, 106: 348–352, 2006.
[37] Osorio E et al. “Actividad antiplasmodial de alcaloides aporfinicos de Rollinia pittieri y Pseudomalmea boyacana (Annonaceae)” Vitae. Rev Fac Quimica Farm 13: 49–54, 2006.
[38] Toriizuka Y et al. “New lycorine-type alkaloid from Lycoris traubii and evaluation of antitrypanosomal and antimalarial activities of lycorine derivatives” Bioorg Med Chem. 16(24):10182-9.. 2008.
[39] E.Osorio et al. “Actividad antiplasmodial de alcaloides aporfinicos de Rollinia pittieri y Pseudomalmea boyacana (Annonaceae)” Vitae. Rev Fac Quimica Farm, 13: 49–54, 2006.
[40] N. Keawpradub, G.C. Kirby, J. C. P. Steele and P. J. Houghton “Antiplasmodial activity of extracts and alkaloids of three Alstonia species from Thailand” Planta Med., 65, 690–694, 1999.
[41] D. Staerk, E Lemmich, J. Christensen, A. Kharazmi, C. E Olsen and J. W. Jaroszewski “Leishmanicidal, antiplasmodial and cytotoxic activity of indole alkaloids from Corynanthe pachyceras” Planta Med. 66, 531–536, 2000,
[42] L. Mambu, M. T.Martin, D. Razafi-Mahefa, D. Ramanitrahasimbola, P. Rasoanaino and F. Frappier “Spectral characterization and antiplasmodial activity of bisbenzylisoquinolines from Solona ghesquiereina” Planta Med. 66, 537–540. 2000.
[43] M. Frederich, et al., “New antimalarial and cytotoxic sungucine derivatives from Strychnos icaja roots” Planta Med., 66,262–269, 2000.
[44] G. Bringmann et al., “Ancistrobertsonines B, C, and D as well as 1,2-didehydroancistrobertsonine D from Ancistrocladus robertsoniorum” Phytochemistry,52,321–332, 1999.
[45] Z. Li, H. Wan, Y. Shi, P. Ouyang “Personal experience with four kinds of chemical structure drawing software: review on ChemDraw, ChemWindow, ISIS/Draw, and ChemSketch” J Chem Inf Comput Sci, 44:1886-90, 2004.
[46] X. Liu, B. Zhang, Z. Jin, H Yang, Z. Rao “The crystal structure of COVID-19 main protease in complex with an inhibitor N3”. doi: 10.2210/pdb6LU7/pdb
[47] R. Thomsen, M.H..Christensen “MolDock: a new technique for high-accuracy molecular docking” J Med Chem. 49:3315-3321., 2006.
[48] H.A.DRajendran, B.Malgija, N.S.Ebenezer, U.Maheswari, Victor Roch G, J.Priyakumari, S.Ignacimuthu, “Homology modeling and molecular docking studies of Purple acid Phosphatase from Setaria italica (Foxtail millet),” International Journal of Scientific Research in Biological Sciences, Vol.5, Issue.4, pp.119-124, 2018.
[49] D.K. Gehlhaar, D. Bouzida, P. A. Rejto “Proceedings of the Seventh International Conference on Evolutionary Programming” ISBN:3-540-64891-7, 1998.
[50] J.M. Yang, C.C. Chen “GEMDOCK: A Generic Evolutionary Method for Molecular Docking” Proteins Structure Function and Bioinformatics. 55(2):288-304, 2004.
[51] F. Cheng, W. Li, Y. Zhou, J Shen, Z. Wu, G. Liu, P.W. Lee, Y. Tang “admetSAR: a comprehensive source and free tool for assessment of chemical ADMET properties.” J Chem Inf Model. 52:3099-105, 2012.
[52] F. Cheng, J Shen, Y. Yu, W Li, G. Liu, P.W. Lee, Y. Tang “In silico prediction of Tetrahymena pyriformis toxicity for diverse industrial chemicals with substructure pattern recognition and machine learning methods” Chemosphere. 82:1636−1643, 2011.
[53] F. Cheng, Y. Yu, J. Shen, L. Yang, W. Li, G. Liu , P.W. Lee, Y. Tang “Classification of Cytochrome P450 Inhibitors and non Inhibitors using Combined Classifiers” J Chem Inf Model, 51:996−1011. 2011.
[54] F. Cheng, Y. Yu, Y. Zhou, J. Shen, W. Xiao, L. Yang, G. Liu, W. Li , W.P. Lee and Y. Tang.”Insights into molecular basis of cytochrome p450 inhibitory promiscuity of compounds” J Chem Inf Model. 51:2482−2495, 2011.
[55] F. Cheng, J. Shen, W. Li, P.W. Lee, Y. Tang “In Silico prediction of terrestrial and aquatic toxicities for organic chemicals” Chin J Pestic Sci. 12:477−488. 2011.
[56] F. Cheng, Y. Ikenaga, Y. Zhou, Y. Yu, W. Li, J. Shen, Z. Du, L. Chen, C. Xu, G. Liu, P.W. Lee, Y.Tang “In Silico assessment of chemical biodegradability” J Chem Inf Model.52:655−669, 2012.
[57] F. Broccatelli, E. Carosati, A. Neri, M. Frosini, L. Goracci, T.I. Oprea , G. Cruciani. “A novel approach for predicting Pglycoprotein (ABCB1) inhibition using molecular interaction fields” J Med Chem.; 54:1740−1751, 2011.
[58] C.A. Lipinski, F. Lombardo, B.W. Dominy, P.J..Feeney “Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings.” Adv Drug Deliv Rev. 1;46(1-3):3-26. 1997.
[59] G.A. Jeffrey “An introduction to hydrogen bonding”Oxford University Press, USA. 1992; ISBN 0-19-509549.
[60] J.A. Williams, R. Hyland, B.C. Jones, D.A. Smith, S. Hurst, T.C. Goosen, V. Peterkin, J.R. Koup, S.E. Ball “Drug-drug interactions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios” Drug Metab Dispos. 32:1201–1208. 2004.
[61] M. Kacevska, G.R. Robertson, S.J. Clarke and C. Liddle “Inflammation and CYP3A4-mediated drug metabolism in advanced cancer: impact and implications for chemotherapeutic drug dosing” Expert Opin Drug Metab Toxicol.4: 137-149, 200.
[62] K.E. Lasser, P.D. Allen, S.J. Woolhandler, D.U. Himmelstein, S.M. Wolfe, D.H. Bor “Timing of new black box warnings and withdrawals for prescription medications” JAMA, 287:2215–2220. 2009.
[63] L.C. Wienkers, T.G. Heath “Predicting in vivo drug interactions from in vitro drug discovery data” Nat Rev Drug Discovery. 4(10):825-33, 2005.
Citation
Bhaskor Kolita, Amlan Jyoti Bora, Pinaki Hazarika, "A Search of COVID-19 main Protease Inhibitor from Plant Derived Alkaloids using Chloroquine and Hydroxychloroquine as Reference: An In-silico Approach," International Journal of Scientific Research in Biological Sciences, Vol.7, Issue.2, pp.51-61, 2020 -
Open Access Article
Mariam ADRA, Badr ALALI and Ahmad KARA ALI
Research Paper | Journal-Paper (IJSRBS)
Vol.7 , Issue.2 , pp.62-68, Apr-2020
Abstract
Antimicrobial substances are so widespread that they are likely to play an important therapeutic role. Many researches showed that microorganisms produce important biochemical compounds, which were used in different domains (medical, pharmaceutical, industrial). The present study aimed to isolate a marine bacterium and study the bioactivity of its methanolic crude extracts. Marine water samples were collected for the isolation of bacteria based on their growth density and then identified depended on morphological, microscopic characterization and biochemical tests. The antibiotics sensitivity was estimated by DL-96II Auto Microbial ID/AST System, While the effectiveness of the methanolic extract was determined by agar- well diffusion method. The compounds in the organic extract of the isolated bacteria were identified by Gas chromatography – mass spectrometry (GC-MS), and their bioactivity was studied. The results showed that these bacteria is Enterobacter cloacae and showed resistance to six from twenty antibiotics. The bioactivity of extract was higher against Pseudomonas aeruginosa and Escherichia coli than Staphylococcus aureus. The chemical analysis of the organic extract showed that eighteen chemical compounds have medical and pharmaceutical importance.Key-Words / Index Term
Marine Enterobacter cloacae, Antibiotics, Bioactivity, GC-MS analysisReferences
[1] Schupp, C. Eder, P. Proksch, V. V. Warry, B. Schneider, M. Herderich, et al., " Staurosporine derivatives from the ascidi- can Eudistomatoealensis and its predatory flatworm Pseudceros sp.," J. Nat. Prod, vol. 62, issue. pp. 795-798, 1999.
[2] Proksch, R. A. Edrada, and R. Ebel, " Drugs from the seas-current status and microbiological implications," Appl. Microbiol. Biotechnol, vol. 59, issue. pp. 125-134, 2002.
[3] J. G. Burgess, E. M. Jordan, M. Bregu, Mearns-Spragg, and K. G. Boyd, "Microbial antagonism: a neglected avenue of natural products research," J. Biotechnol, vol. 70, issue. pp. 27-32, 1999.
[4] D. Jayammal and T. Sivakumar, "Antibacterial activity of protein extract of marine Pseudomonas aeruginosa against bacterial pathogens," Int.J.Curr.Microbiol.App.Sci, vol. 2, issue. 7, pp. 207-216, 2013.
[5] P. Jeganathan, K. M. Rajasekaran, N. K. A. D. and, and S. Karuppusamy, "Antimicrobial activity and Characterization of Marine bacteria," Indian Journal of Pharmaceutical and Biological Research, vol. 1, issue. 4, pp. 38-44, 2013.
[6] A. M. Gad, E. A. Beltagy, U. M. Abdul-Raouf, M. A. El-Shenawy, and S. S. Abouelkheir, "Screening of Marine Fatty Acids Producing Bacteria with Antimicrobial Capabilities," Chemistry of Advanced Materials (CAM), vol. 1, issue. 2, pp. 41-54, 2016.
[7] P. R. Burkholder, R. M. Pfister, and F. P. Leitz, "Production pyrrole antibiotic by a marine bacterium," Appl. Microbioloy, vol. 14, issue. pp. 649 - 653, 1966.
[8] U. Holler, A. D. Wright, G. F. Matthee, G. M. Konig, S. Draeger, H. J. Aust, et al., "Fungi from marine sponges: diversity, biological activity and secondary metabolites," Mycol. Res, vol. 104, issue. pp. 1354-1365, 2000.
[9] C. Holmstrom and S. Kjelleberg, "Marine Pseudoalteromonas species are associated with higher organisms and produce bio- logically active extracellular agents," FEMS Microbiol.EcoL, vol. 30, issue. pp. 285-293, 1999.
[10] A. Isnansetyo and Y. Kamei, "MC21-A, a bactericidal antibiotic produced by a new marine bacterium, Pseudoaltermononasphenolica sp. nov. O-BC30T, against Methicillin-Resistant Staphylococcus aureus," Antimicrob. Agents Ch, vol. 47, issue. pp. 480-488, 2003.
[11] T. Kohler, J. C. Pechere, and P. Plesiat, "Bacterial antibiotic efflux systems of medical importance," Cell. Mol. Life Sci, vol. 56, issue. pp. 771-778, 1999.
[12] S. Octavia and R. Lan, "The Family Enterobacteriaceae," in The Prokaryotes: Gammaproteobacteria, E. Rosenberg, E. F. DeLong, S. Lory, E. Stackebrandt, and F. Thompson, Eds., ed Berlin, Heidelberg: Springer Berlin Heidelberg, pp. 225-286, 2014.
[13] A. Davin-Regli, J.-P. Lavigne, and J.-M. Pagès, "Enterobacter spp.: Update on taxonomy, clinical aspects, and emerging antimicrobial resistance," Clinical Microbiology Reviews, vol. 32, issue. 4, pp. e00002-19, 2019.
[14] S. Krishnamoorthy and N. Ekambaram, "Isolation and molecular characterization of Enterobacter species ENKS2 and its phosphate solubilisation from rhizospheric soil," Int. J. Sci. Res. in Biological Sciences Vol, vol. 5, issue. 6, pp. 20-25, 2018.
[15] A. Mearns-Spragg, M. Bregu, K. G. Boyd, and J. G. Burgess, "Cross-species induction and enhancement of antimicrobial activity produced by epibiotic bacteria from marine algae and invertebrates, after exposure to terrestrial bacteria," Letters in Applied Microbiology, vol. 27, issue. 3, pp. 142-146, 1998.
[16] G. Schwartsmann, A. B. da Rocha, R. G. Berlinck, and J. Jimeno, "Marine organisms as a source of new anticancer agents," The Lancet Oncology, vol. 2, issue. 4, pp. 221-225, 2001.
[17] V. K. Verma, N. Sehgal, and O. Prakash, "Characterization and screening of bioactive compounds in the extract prepared from aerial roots of Ficus benghalensis," International Journal of Pharmaceutical Sciences and Research, vol. 6, issue. 12, p. 5056, 2015.
[18] A. Shettima, Y. Karumi, O. Sodipo, H. Usman, and M. Tijjani, "Gas Chromatography–Mass Spectrometry (GC-MS) Analysis of Bioactive Components of Ethyl acetate Root Extract of Guiera senegalensis J.F. Gmel," Journal of Applied Pharmaceutical Science, vol. 3, issue. 3, pp. 146-150, 2013.
[19] J. Devakumar, V. Keerthana, and S. S. Sudha, "Identification of bioactive compounds by gas chromatography-mass spectrometry analysis of Syzygium jambos (L.) collected from Western Ghats region Coimbatore, Tamil Nadu," Asian Journal of Pharmaceutical and Clinical Research vol. 10, issue. 1, pp. 364-369 2017
[20] V. Aparna, K. V. Dileep, P. K. Mandal, P. Karthe, C. Sadasivan, and M. Haridas, "Anti-inflammatory property of n-hexadecanoic acid: Structural evidence and kinetic assessment," Chem. Biol. Drug Des, vol. 80, issue. pp. 434–439, 2012.
[21] P. P. Kumar, at. al "Screening of antioxidant activity, total phenolics and GC-MS study ofVitexnegundo," Afr. J. Biochem. Res., vol. 4, issue. pp. 191-195, 2010.
[22] A. A. Rahuman, G. Gopalakrishnan, B. S. Ghouse, S. Arumugam, and B. Himalayan, " Effect of Feronialimonia onmosquito larvae," Fitoterapia vol. 71, issue. pp. 553-555, 2000.
[23] S. M. Colegate and R. J. Molyneux, Bioactive Natural Products: Detection, Isolation, and Structural Determination, Second Edition: CRC Press, 2007.
[24] M. Kanjana, G. Kanimozhi, R. Udayakumar, A. Panneerselvam, and J. Kanjana, "GC-MS Analysis of Bioactive Compounds of Endophytic Fungi Chaetomium globosum, Cladosporium tenuissimum and Penicillium janthinellum," Biomedical and Pharmaceutical Sciences vol. 2, issue. 1, 2019.
[25] E. P. Awa, S. Ibrahim, and D. A. Ameh, "GC/MS analysis and antimicrobial activity of diethyl ether fraction ofmethanolic extract from the stem bark of Annonasenegalensis Pers," Int. J. Pharm. Sci. Res., vol. 3, issue. pp. 4213-4218, 2012.
[26] E. Vadivel and S. Gopalakrishnan, "GC-MS analysis of some bioactive constituents of Mussaenda frondosa Linn," Inter J Pharm Bio Sci, vol. 2, issue. pp. 313-320, 2011.
[27] K. Rakstys, S. Paek, G. Grancini, P. Gao, V. Jankauskas, A. M. Asiri, et al., "Low‐cost perovskite solar cells employing dimethoxydiphenylamine‐substituted bistricyclic aromatic enes as hole transport materials," ChemSusChem, vol. 10, issue. 19, pp. 3825-3832, 2017.
[28] J. D. Drazba, K. Chen, and J. P. Rolland, "Blocked silicone dual cure resins for additive manufacturing," ed: Google Patents, 2019.
[29] A. Abd Sharad, G. Usup, F. K. Sahrani, and A. Ahmad, "Antimicrobial activity and determination of bioactive components from marine Alcaligenes faecalis extract against a sulfate-reducing bacteria," AIP Conference Proceedings, vol. 1784, issue. 1, pp. 1-5, 2018.
[30] T. Tyagi and M. Agarwal, "Phytochemical screening and GC-MS analysis of bioactive constituents in the ethanolic extract of Pistia stratiotes L. and Eichhornia crassipes (Mart.) solms," Journal of Pharmacognosy and phytochemistry, vol. 6, issue. 1, pp. 195-206, 2017.
[31] A. Maruthupandian and V. Mohan, "GC-MS analysis of some bioactive constituents of Pterocarpus marsupium Roxb," Int J Chem Tech Res, vol. 3, issue. 3, pp. 1652-1657, 2011.
[32] P. Amudha, M. Jayalakshmi, N. Pushpabharathi, and V. Varadharaj, "Identification of bioactive components in enhalus acoroides seagrass extract by gas chromatography–mass spectrometry," Asian Journal of Pharmaceutical and Clinical Research, vol. 11, issue. 10, pp. 313-317, 2018.
[33] F. B. H. Kether, M. A. Mahjoub, S. A. Mahjoub, K. B. Salah, A. N. Helal, and Z. Mighri, "Antioxidant activities of essential oil from Cotulacoronopifolia L. growing in Tunisia.," African journal of microbiology research, vol. 6, issue. 20, pp. 4388-4395, 2012.
[34] R. P. Shobha and R. Agrawal, "Volatile Compounds of Therapeutic Importance Produced by Leuconostoc paramesenteroides, a Native Laboratory Isolate," Turk J Biol, vol. 31, issue. pp. 35-40 2007.
[35] A. Dhiman, "Gas chromatography-mass spectroscopy analysis of bioactive compounds in the whole plant parts of ethanolic extract of Asclepias Curassavica L," International Journal of Green Pharmacy (IJGP), vol. 12, issue. 02, 2018.
[36] T. Sudha, S. Chidambarampillai, and V. Mohan, "GC-MS analysis of bioactive components of aerial parts of kirganelia reticulata poir (Euphorbiaceae)," J. Curr. Chem. Pharm. Sc, vol. 3, issue. 2, pp. 113-122, 2013.
[37] G. M. Abou-Elela, H. Abd-Elnaby, H. A. H. Ibrahim, and M. A. Okbah, "Marine Natural Products and Their Potential Applications as Anti-Infective Agents," World Applied Sciences Journal, vol. 7, issue. 7, pp. 872-880, 2009.
[38] B. Parthipan, M. Suky, and V. R. Mohan, "GC-MS Analysis of Phytocomponents in Pleiospermium alatum (Wall. ex Wight & Arn.) Swingle, (Rutaceae)," Journal of Pharmacognosy and Phytochemistry vol. 4, issue. 1, pp. 216-222, 2015.
[39] S. Terés, G. Barceló-Coblijn, M. Benet, R. Álvarez, R. Bressani, J. Halver, et al., "Oleic acid content is responsible for the reduction in blood pressure induced by olive oil," Proc Natl Acad Sci U S A, vol. 105, issue. 37, pp. 13811-13816, 2008.
AUTHORS PROFILE
Mrs. Mariam Adra has obtained B. of Microbiology from Tishreen University in 2016, and is studying MSc of Marine Biology science at the Higher Institute for Marine Research. She works as a science teacher at the secondary level and teaches bacteriology and antibiotics at the university. Her main research work focuses on bacteria. she has 5 years of teaching experience and 3 years of research experience.
Citation
Mariam ADRA, Badr ALALI and Ahmad KARA ALI, "GC-MS Analysis of Bioactive Compounds in Methanolic Extract of Marine Enterobacter Cloacae Mariam ADRA1*, Badr ALALI2 and Ahmad KARA ALI3," International Journal of Scientific Research in Biological Sciences, Vol.7, Issue.2, pp.62-68, 2020 -
Open Access Article
Bacteriocin Induced Milk as a Vehicle against Dental Cavities
Parul Thapar, R.K. Malik, M.K. Salooja
Research Paper | Journal-Paper (IJSRBS)
Vol.7 , Issue.2 , pp.69-76, Apr-2020
Abstract
The prevalence of dental caries is consistently increasing, in every age across the globe, and today the concern for the “Oral health” is almost at the same level as for “General health”. The oral health care demands are beyond the capacities of the health care systems in most low and middle income countries. Therefore, it was thought to explore the natural ways by which the problems of dental caries can be prevented in a cost-effective manner. It was found that a few studies have set the path through use of natural bacteriocins against inhibition of caries causing organisms. The inhibition of caries causing organisms can be done by naturally occurring substances like bacteriocins found in lactic acid bacteria. Bacteriocins are potent protein toxins produced by virtually every bacterial and archeal species. In the present study, prevention of dental caries by utilization of bacteriocins from lactic acid bacteria has been done by incorporating bacteriocin in milk. The bacteriocin producing five strains of Lactobacillus brevis isolated from curd were used for the production of bacteriocin. The minimum inhibitory concentration (MIC) of the bacteriocins was carried out to find the best bacteriocin producing strain. Out of five bacteriocin producers, the strain LB11(1) showed maximum percentage of MIC. The characterization of the bacteriocin was done using various optimization conditions. The partial purification was done using ammonium sulphate precipitation and centrifugal filteration. The purified bacteriocin was then incorporated in the milk to check the effect of inhibition on caries causing organisms (Streptococcus mutans and Streptococcus sobrinus isolated from dental cavities). It was confirmed that the bacteriocin induced milk can be applied to prevent dental caries directly by the consumer and the milk itself can become a vehicle against dental cavities.Key-Words / Index Term
Bacteriocins, lactic acid bacteria, Streptococcus mutans, Streptococcus sobrinus, Lactobacillus brevisReferences
[1] S. Mathews and G.V. Mathew, “War inside the mouth,” Journal of Dental and Medical Sciences, Vol. 13, Issue. 1, pp. 23-25, 2014.
[2] C. Janakiram, B. Antony, J. Joseph and V. Ramanarayanan, “Prevalence of dental caries in India among the WHO index age groups: a meta- analysis,” Journal of Clinical Diagnostic and Research, Vol. 12, Issue. 1, pp. 8-13, 2018.
[3] A. R. Hosseinpoor, L. Itani and P. E. Petersen, “Socio-economic inequality in oral healthcare coverage: results from the World Health Survey,” Journal of Dental Research, Vol. 91, Issue. 3, pp. 275-281, 2012.
[4] L. R. Dorit, M. S. Roy and A. M. Riley, “The bacteriocins current knowledge and future prospects book,” Caister Academic Press, USA, pp. 103-126, 2016.
[5] H. P. Rodney, Z. Takeshi and S. Kenji, “Novel bacteriocins from lactic acid bacteria (LAB): various structures and applications,” Microbial Cell Factories, Vol. 13, Issue 5, pp. 1-3, 2014.
[6] L. L. Burlanek, and A. E. Yousef, “Solvent extraction of bacteriocins from liquid cultures,” Journal of Applied Microbiology, Vol. 31, Issue. 1, pp. 193-197, 2000.
[7] J. M. Andrews, “Determination of minimum inhibitory concentrations,” Journal of Antimicrobial Chemotherapy, Vol. 48, Issue. 3, pp. 5-16, 2002.
[8] P. Jean, W. Melvin, E. George, J. Stephen, L. James, L. Brandi, M. Amy, M. Tony, P. Robin, R. Sandra, S. Michael, S. Jana, T. Maria and Z. Barbara, “Performance standards for antimicrobial susceptibility testing,” Clinical and Laboratory Standards, Vol. 37, No. 1, pp. 144-249, 2017.
[9] B. G. Hall, H., Acar, A. Nandipati and M. Barlow, “Growth rates made easy,” Journal of Molecular Biology and Evolution, Vol. 31, Issue. 5, pp. 232-238, 2013.
[10] E. E. Hartree, “Determination of protein: a modification of the lowry method that gives a linear photometric response,” Analytical Biochemistry, Vol. 48, Issue. 1, pp. 422, 1972.
[11] O. H. Lowry, N. J. Rosebrough, A. L. Farr and R. J. Randall, “Protein measurement with the Folin phenol reagent,” Journal of Biological Chemistry, Vol. 19, Issue. 3, pp. 265, 1951.
[12] K. Wilson, and J. Walker, “Practical Biochemistry: Principles and Techniques,” Cambridge University Press, USA, pp. 50, 2000.
[13] R. Yang, M.C. Johnson and B. Ray, “Novel method to extract large amounts of bacteriocins from lactic acid bacteria,” Applied Environmental Microbiology, Vol. 58, Issue 5, pp. 3355-3359, 1992.
[14] N. Gautam and N. Sharma, “Purification and characterization of bacteriocin produced by strain of Lactobacillus brevis MTCC 7539,” Indian Journal of Biochemistry and Biophysics, Vol. 46, Issue 1, pp. 337-341, 2009.
[15] S. F. Barefoot, and T. R. Klaenhammer, “Purification and characterization of Lactobacillus acidophilus bacteriocin lactacin B,” Antimicrobial Agents and Chemotherapy, vol. 26, Issue. 7, pp. 328-334, 1984.
[16] M. M. Ahmed, A. M. Baher, I. M. Zainab, N. F. Tawfiek and Z. M. R. Hassan et al., “Antibacterial activity of some lactic acid bacteria isolated from egyptian dairy products,” International Journal of Chemical Technology and Research, Vol. 6, Issue. 1, pp. 1139-1150, 2014.
[17] N. Agrawal and A. Prakash, “Isolation of lactic acid bacteria from fermented milk products and their antimicrobial activity against Staphylococcus aureus,” Journal of Food Safety, Vol. 15, Issue. 4, pp. 39-42, 2013.
[18] C. R. Toro, Use of probiotic lactic bacteria in feeding Litopenaeus vannamei shrimp as inhibitors of pathogenic microorganisms and immune system stimulants. PhD Thesis, Federal University of Paraná, Curitiba, Brazil, 2005.
[19] H. Wang, H. Gong, X. Meng and L. Man, “A Bacteriocin Produced by Lactobacillus brevis KLDS1.0373 isolated from “Jiaoke”, a traditional fermented cream from China”. Advanced Materials and Research, Vol. 18, Issue 3, pp. 1132-1136, 2011.
[20] S. K. R. Namasivayam, J. C. R. Angel, R. S. A. Bharani and M. Y. Karthik, “Effect of Media on bacteriocin production by Lactobacillus brevis and evaluation of anti-bacterial activity,” Research Journal of Pharmaceutical, Biological Chemical Sciences, Vol. 5, Issue. 10 pp. 1129-1136, 2014.
[21] M. Mohankumar and M. Murugalatha, “Characterization and antibacterial activity of bacteriocin producing Lactobacillus isolated from raw cattle milk sample,” International Journal of Biology, Vol. 3, Issue. 1, pp. 128-143, 2011.
[22] A. A. Rushdy at. al. “Antimicrobial compounds produced by probiotic Lactobacillus brevis isolated from dairy products,” Annal of Microbiology, Vol. 63, Issue 6, pp. 81-90, 2013.
Citation
Parul Thapar, R.K. Malik, M.K. Salooja, "Bacteriocin Induced Milk as a Vehicle against Dental Cavities," International Journal of Scientific Research in Biological Sciences, Vol.7, Issue.2, pp.69-76, 2020 -
Open Access Article
Role of H+ and Ca++ concentrations on Vorticella stalk Contraction Determination
Amit Kumar Verma, Ashok Kumar Singh
Research Paper | Journal-Paper (IJSRBS)
Vol.7 , Issue.2 , pp.77-81, Apr-2020
Abstract
Vorticella stalks contraction mechanism is based upon the behaviour of novel types of motor proteins involved in the process and thus determined positive sensitive effects when in [H+] from 14.80 × 10-10 to 7.15 × 10-10 mole whereas [Ca++] from 0.5 to 4.5 mM reflected negative effects on contraction dynamics in terms of birefringence. The protoosmotic applications on the stalk contraction provided information about biochemical nature of motor proteins involved in contraction dynamics of the stalk in terms of frequency and duration profiles in reversible reactions have potential future applications in controlling patterns of structural modifications of motor proteins involved throughout the system.Key-Words / Index Term
H+ & Ca++ concentrations, Vorticella stalk, contraction determination, frequencies and durations profiles.References
[1]. Amos, W. B. Routledge, L. M. & Yew, F. F., “Calcium-binding proteins in a vorticellid contraction organelle”, J. Cell Sci. Vol. 19, Issue, 1, pp. 203-213, 1975.
[2]. Mahadevan, L. & Matsudaira, P., “Motility Powered by Supramolecular Springs and Ratchets”, Science, Vol. 288, No. 5463, pp. 95-99, 2000.
[3]. Singh, A. K. & Amin, M., “Oscillation in Nitella Cell membrane potential and its Effect on Cytoplasmic Streaming”, Current Science Vol. 58, No. 12, pp 669-672, 1989.
[4]. Shantosh, M. & Sarojnalini, Ch., “Nutritional Quality of Three Cobitid Fishes of Manipur, India: With Special Reference to Essential Mineral Elements”, International Journal of Scientific Research in Biological Sciences, Vol. 5, Issue. 2, pp. 24 – 33, 2018.
[5]. Misra, G. Dickinson, R.B. & Ladd, A.J., “Mechanics of Vorticella contraction”, Biophysical Journal, 98(12), 2923-2932, 2010.
[6]. Verma, A. K. & Singh, A. K., “Effect of extracellular pH and Ca++ - concentration on the contractility and force generation as verified on Vorticella stalk concentration pattern”, International Journal of Global Science Research, Vol. 4, No. 1, pp. 503 – 511, 2017.
[7]. Page F., “The culture and use of free-living protozoa in teaching” Institute of terrestrial ecology natural environment research council, culture centre of algae and protozoa, 36, Storey’s Way Cambridge CB3 0DT England 1981.
[8]. Naitoh, Y. & Kaneko, H., “Reactivated Triton-extracted models of Paramecium: modification of ciliary movement by calcium ions”, Science, Vol. 176, pp. 523-524, 1972.
[9]. Ochiai, A. Asai, H. & Fukui, K., “Hysteresis of Contraction-Extension Cycle of Glycerinated Vorticella”, Protozool, pp. 420-425, 1979.
[10]. Singh, A. K. & Amin, M., “Effect of extracellular pH on membrane potential and cytoplasmic streaming in nitella and chara cells” Indian Journal of Experimental Biology, Vol. 29, No. 3, 201-204, 1991.
[11]. Sokolovsky, M. Riordan, J. F. and Vallee, B. L., “Tetranitromethane. A reagent for the nitration of tyrosyl residues in proteins”, Biochemistry, Vol. 5, pp. 3582-3589, 1996.
[12]. Riordan, J. F. Sokolovsky, M. and Vallee, B. L., “Tetranitromethane, A reagent for nitration of tyrosyl residues in proteins”, J. Am. Chem. Soc., Vol. 88, pp. 4104-4105, 1996.
[13]. Gogendeau, D. Beisson, J. Garreau de Loubresse, N. Caer, J-PL. Ruiz, F. Cohen, J. Sperling, L. Koll, F. and Koltz, C.. “An Sfi-Like Centrin-Binding Protein Mediated Centrin-Based Ca2+-dependent Contractility in Paramecium tetraaurelia”, Eukaryotic., Vol. 6, No. 11, p. 1992-2000, 2007.
[14]. Ashley, C. C. & Moisescu, D. G., “The influence of Mg++ concentration and pH upon the relationship between steady-state isometric tension and Ca++ concentration in isolated bundles of barnacle myofibrils”, J. Physiol., Vol. 239, No. 2, pp. 112P-114P, 1974.
[15]. Chase, P. B. & Kushmerick, M. J., “Effects of pH on Contraction of Rabbit Fast and Slow Skeletal Muscle Fibers”, Biophys. J., Vol. 53, No. 6, pp. 935-946, 1988.
Citation
Amit Kumar Verma, Ashok Kumar Singh, "Role of H+ and Ca++ concentrations on Vorticella stalk Contraction Determination," International Journal of Scientific Research in Biological Sciences, Vol.7, Issue.2, pp.77-81, 2020 -
Open Access Article
H. Deepa and A. Subhashini
Research Paper | Journal-Paper (IJSRBS)
Vol.7 , Issue.2 , pp.82-91, Apr-2020
Abstract
Capsaicin is a nutritional factor, the active component of chilli, which is responsible for the pungent component of chilli. Chilli has diverse uses as a spice, condiment, culinary supplement, Medicine, vegetable and ornamental plant. Aim of the study: Stimulation and enhancement of capsaicin content by polyploidy induction, quantification of enhanced capsaicin, its cytotoxicity and Bioactivity studies. Materials and methods: Healthy 3 varieties (CO-1, K2, Jwala) of Capsicum annum L. (chilli) was selected for enhancement of capsaicin content by polyploidy induction. The variety showing high capsaicin content in the fifth generation was chosen for further study. The enhanced capsaicin was isolated by silica gel column chromatography and purified using HPLC. Cytotoxicity was studied in onion root tips by root tip squash method and Vero cell lines. Anti-inflammatory activity by inhibition of Albumin denaturation method and Anti-cancer activity by MTT assay was studied using the enhanced capsaicin. Results: Of the 3 chilli varieties selected, CO-1 showed high and enhanced capsaicin content in the fifth generation. By HPLC, with a retention time of 1.368 min, 72.93% purity was obtained against control having a retention time of 1.337min of 41.4% purity recorded. Cytotoxicity of onion root tips showed no cell abnormalities and hence was found to be non-toxic to the cells. In the Vero cell line assay, the IC50 value and the viability of the cell line was 60.39% in treated and 58.79% in control of 100 µg/ml concentration. Anti-inflammatory activity of enhanced Capsaicin also showed maximum inhibition rate of 81.23% against 76.26% in control using 100 µg/ml. In Anti-cancer activity, IC50 dose was 6.25μg/ml in treated whereas untreated capsaicin showed 25μg/ml. Thus, enhanced Capsaicin shows a significant anti-inflammatory and anti-cancer activity.Key-Words / Index Term
chilli, capsaicin, HPLC, inhibition, anti-inflammatory, anti-cancerReferences
[1]. Perucka, M. Materska, “Phenylalanine ammonia-lyase and antioxidant activities of a lipophilic fraction of fresh pepper fruits Capsicum annuum L”, Innovat. Food Sci. Emerg. Tech 2, pp.189-192, 2001.
[2]. Deal, C.L. Schnitzer, T.J. Lipstein, E. Seibold, J.R. Stevens, R.M.; Levy, M.D. Albert, D.s Renold, F, “Treatment of arthritis with topical capsaicin: A double-blind trial”. Clin. Ther. 13, pp.383-395, 1999.
[3]. Y. Zewdie, P.W. Bosland, “Evaluation of genotype, environment and genotype-by-environment interaction for capsaicinoids in Capsicum annuum L”. Euphytica, 111 pp.185-190, 2000.
[4]. D.J. Moore, D.M. Moore, “Synergistic Capsicum-tea mixtures with anticancer activity”, J. Pharm. Pharmacol. 55, pp.987-994, 2003.
[5]. J. Szolcsanyi, “Forty years in capsaicin research for sensory pharmacology and physiology”. Neuropeptides, 38, pp.377-384, 2004.
[6]. Bali, P.N. and Tandon, S.L, “Colchicine induced tetraploidy in Linaria vulgaris”. Curr. Sci., 26, pp.256-258, 1956.
[7]. Raghuvanshi, S. and Joshi, S, “Cytomorphological studies on the colchiploids of Capsicum frutescens L”. Cytologia, 225, pp.61-78, 1964.
[8]. Swanson, C.P, wilian Co., N.Y. and Kihlman, B.A. “The chromosomal aberration by ionizing radiations and chemical mutagens”. pp. 239-252, 1957.
[9]. Bhattacharya A, Chattopadhyay A, Mazumdar D, Chakravarty A and Pal S. “Antioxidant constituents and enzyme activities in chilli peppers. International Journal of Vegetable Science” 16, pp.201–211, 2010.
[10]. Luo XJ, Peng J and Li YJ. “Recent advances in the study on capsaicinoids and capsinoids”. European Journal of Pharmacology; 650, pp.1–7, 2011.
[11]. Adams, D. O., & Hamilton, T. A. “The cell biology of macrophage activation”. Annual Review of Immunology, 2, pp.283–318, 1984.
[12]. Tsai, T. H., Tsai, P. J., & Ho, S. C, “Antioxidant and anti- inflammatory activities of several commonly used spices”. Journal of Food Science, 70, pp.93–97, 2005.
[13]. Park, E., Lee, S. M., Lee, J., & Kim, J. H. “Anti-inflammatory activity of mulberry leaf extract through inhibition of NF-kB”. Journal of Functional Foods, 5, pp.178–186, 2013.
[14]. Skidgel, R. A., Gao, X. P., Brovkovych, V., Rahman, A., Jho, D., Predescu, S., Standiford, T. J., & Malik, A. B, “Nitric oxide stimulates macrophage inflammatory protein-expression in sepsis”. Journal of Immunology, 169, pp.2093–2101, 2002.
[15]. Morre, D.J., Chueh, P.J. and Morre, D.M, “Capsaicin inhibits preferentially the NADH oxidase and growth of transformed cells in culture”. Proceedings of the National Academy of Sciences. 92: pp.1831-1835, 1995.
[16]. . Morre, D.J., Sun, E., Geilen, C., Wu, L.Y., de-Cabo, R., Krasagakis, K., Orfanos, C.E. and Morre, D.M. “Capsaicin inhibits plasma membrane NADH oxidase and growth of human and mouse melanoma lines”. European Journal of Cancer. 32A, pp.1995-2003, 1996.
[17]. Macho, A., Blazquez, M.V., Navas, P. and Munoz, E, “Induction of apoptosis by vanilloid compounds does not require de novo gene transcription and activator protein 1 activity”. Cell Growth and Differentiation. 9, pp.277-286, 1998
[18]. . Macho, A., Calzado, M.A., Munoz-Blanco, J., Gomez-Diaz, C., Gajate, C., Mollinedo, F., Navas, P. and Munoz, E, “Selective induction of apoptosis by capsaicin in transformed cells: the role of reactive oxygen species and calcium”. Cell Growth and Differentiation. 6, pp.155-165, 1999.
[19]. Macho, A., Lucena, C., Calzado, M.A., Blanco, M., Donnay, I., Appendino, G. and Munoz, E, “Phorboid 20-homovanillates induce apoptosis through a VR1-independent mechanism”. Chemistry and Biology. 7, pp.483-492, 2000.
[20]. Wolvetang, E.J., Larm, J.A., Moutsoulas, P. and Lawen, A, “Apoptosis induced by inhibitors of the plasma membrane NADH-oxidase involves Bcl-2 and calcineurin”. Cell Growth and Differentiation. 7, pp.1315-1325, 1996.
[21]. Matsufuji, H., Nakamuro, H., Chino, M. and Mitsuharo, T, “Antioxidant activity of capsanthin and the fatty acid esters in paprika (Capsicum annuum)”. Journal of Agricultural and Food Chemistry. 46: pp.3462-3472, 1998.
[22]. J. Zheng, Y.Zhou, Y, Li, D.-P. Xu, S. Li, H.-B.Li, “Spices for prevention and treatment of cancers”, Nutrients 8 (8), pp.495, 2016.
[23]. M.D. Collins, L. Mayer-Wasmund, P.W. Bosland, “Improved method for quantifying capsaicinoids in Capsicum using high-performance liquid chromatography”, Hort. Sci., 30, pp.137-139, 1995.
[24]. H.W. Lewis, C.J. Moody, “Experimental Organic Chemistry: Principles and Practice (Illustrated ed.)”, pp.159–173, 1989.
[25]. T. Mosmann, “Rapid colourimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays”. J. Immun. Method., 65, pp.55–63, 1983.
[26]. Y. Mizushima, M. Kobayashi M. “Interaction of anti‐inflammatory drugs with serum proteins, especially with some biologically active proteins”. J. Pharm. Pharmacol. 20, 169‐ 173, 1968.
[27]. S. Sakat, A.R. Juvekar, M.N. Gambhire, “In vitro antioxidant and anti-inflammatory activity of methanol extract of Oxalis corniculata Linn”. Int. J. Pharm. Pharmacol. Sci. 2, pp.146-155, 2010.
[28]. Yacine Badjah Hadj Ahmed, Zeid Abdullah Al Othman, Mohamed Abdelaty Habila and Ayman Abdel Ghafar. “Determination of Capsaicin and Dihydrocapsaicin in Capsicum Fruit Samples using High-Performance Liquid Chromatography”. Molecules, 16, pp.8919-8929, 2011.
[29]. Bindu Thapa, Natasa Skalko-Basnet, Akihito Takano, Kazuo Masuda, and Purusotam Basnet, “High-Performance Liquid Chromatography Analysis of Capsaicin Content in 16 Capsicum Fruits from Nepal”. J Med Food 12 (4), pp.908–913, 2009.
[30]. Saksit chanthai, Jureerat Juangsamoot, Chalerm Ruangviriyachai and Suchila Techawongstien 2012. “Determination of Capsaicin and Dihydrocapsaicin in Some Chilli Varieties using Accelerated Solvent Extraction Associated with Solid-Phase Extraction Methods and RP-HPLC-Fluorescence”. E-Journal of Chemistry, 9(3), pp.1550-1561, 2012.
[31]. Mônica Rosa Bertão, Milena Cristina Moraes, Darío Abel Palmieri, Luciana Pereira Silva and Regildo Márcio Gonçalves da Silva, “Cytotoxicity, genotoxicity and antioxidant activity of extracts from Capsicum spp. Res. J. Med. Plants, 10, pp.265-275, 2016.
[32]. Liljana Koleva Gudeva, Sasa Mitrev, Viktorija Maksimova, Dusan Spasov, “Content of capsaicin extracted from hot pepper (Capsicum annuum ssp. microcarpum L.) and its use as an ecopesticide”. Hem. Ind. 67 (4) pp.671–675, 2013.
[33]. Adnan Jassim Mohammed, Al–Fartosy Sameerah, Ahmed Zearah, “Antioxidant, Antibacterial and Cytotoxicity Activities of Flavonoid Extract from Capsicum annum L”. Seeds Iraqi national journal of chemistry, volume 49, pp.100-112, 2013.
[34]. Reshma, K.P. Arun P. Brindha, “In Vitro Anti-Inflammatory, Antioxidant and Nephroprotective Studies on Leaves of Aegle marmelos and Ocimum sanctum”s, Asian J Pharm Clin Res, 7, 2014.
[35]. Manjunatha H. thriveni Vasanth Kumar, Rajesh KP, “Anti-inflammatory activity of curcumin and capsaicin augmented in combination”. Int J Pharm Pharm Sci, Vol 9, Issue 6, pp.145-149, 2017.
[36]. Jolayemi AT, Ojewole JAO, “Comparative anti-inflammatory properties of Capsaicin and ethyl- acetate extract of Capsicum frutescens Linn [Solanaceae] in rats”. African Health Sciences; 13(2): pp.357 – 361, 2013.
[37]. Muraleedharan G. Nair and Yunbao Liu, “Capsaicinoids in the Hottest Pepper Bhut Jolokia and its Antioxidant and Anti-inflammatory Activities”. Natural Product Communications Vol. 5 (1) pp.91-95, 2010.
[38]. N.J. Amruthraj, J.P. Preetam Raj, S. Saravanan, L. Antoine Lebel, “In Vitro Studies on Anticancer Activity of Capsaicinoids from Capsicum Chinense Against Human Hepatocellular Carcinoma Cells”, Int. J. Pharmacy Pharm. Sci. 6, pp.254-558, 2014.
[39]. Raj, John, “In Vitro Studies on Anticancer Activity Of Capsaicinoids From Capsicum Chinense Against Human Hepatocellular Carcinoma Cells. Int J Pharm Pharm Sci. 6. pp. 254-558, 2014.
[40]. Rosa Raybaudi-Massiliaa, Al´ırica I. Su´arezb, Francisco Arveloc,d, Alexandra Zambrano, Felipe Sojoc,d, Marıa I. Calderon-Gabaldon, and Jonathan Mosqueda-Melgara, “Cytotoxic, Antioxidant and Antimicrobial Properties of Red Sweet Pepper (Capsicum Annuum L. var. Llanero´n) Extracts: In Vitro Study”. International Journal of Food Studies, Volume 6, pp.222–231, 2017.
[41]. Mehmet Berkoza, Metin Yildirim, Gulhan Arvasc, Omer Turkmen, Oruc Allahverdiyeve, “Effect of capsaicin on transcription factors in 3T3-L1 cell line”. Eastern Journal of Medicine 20, pp.34-45, 2015.
Citation
H. Deepa and A. Subhashini, "Extraction and Isolation of capsaicin from Capsicum annum L (CO-1) variety by High-performance liquid chromatography and its Bioactivities," International Journal of Scientific Research in Biological Sciences, Vol.7, Issue.2, pp.82-91, 2020
Next Last |
You do not have rights to view the full text article.
Please contact administration for subscription to Journal or individual article.
Mail us at support@isroset.org or view contact page for more details.